Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Neuroimmunol ; 391: 578360, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38723578

ABSTRACT

OBJECTIVE: Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disorder of the peripheral nerves with an incompletely understood underlying pathophysiology. This investigation focused on defining B and T cell frequencies, T cell functional capacity and innate immune system analysis in patients with CIDP. METHODS: By using multi-parameter flow cytometry, we examined the phenotype and function of PBMCs in 25 CIDP patients who were relatively clinically stable on treatment who met EFNS/PNS criteria, 21 patients with genetically confirmed hereditary neuropathy and 25 healthy controls. We also evaluated the regulatory T cell (Treg) inhibitory capacity by co-culturing Treg and effector T cells. RESULTS: Proinflammatory CD4 T cells, especially type 1 helper T cell (Th1) and CD8 T cells in patients with CIDP were found to have an enhanced capacity to produce inflammatory cytokines. There was no difference in frequency of Th17 regulatory cells in CIDP patients versus healthy controls, however, Treg function was impaired in CIDP patients. There was no remarkable difference in innate immune system measures. Within B cell subsets, transitional cell frequency was decreased in CIDP patients. INTERPRETATION: Patients with CIDP clinically stable on treatment continued to show evidence of a proinflammatory state with impaired Treg function. This potentially implies an inadequate suppression of ongoing inflammation not addressed by standard of care therapies as well as persistent activity of disease while on treatment. Targeting T cells, especially inhibiting Th1 and polyfunctional CD8 T cells or improving Treg cell function could be potential targets for future therapeutic research.

2.
Breast Cancer Res Treat ; 201(2): 307-316, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37418031

ABSTRACT

PURPOSE: In a phase II trial in patients with metastatic triple-negative breast cancer (mTNBC; NCT02978716), administering trilaciclib prior to gemcitabine plus carboplatin (GCb) enhanced T-cell activation and improved overall survival versus GCb alone. The survival benefit was more pronounced in patients with higher immune-related gene expression. We assessed immune cell subsets and used molecular profiling to further elucidate effects on antitumor immunity. METHODS: Patients with mTNBC and ≤ 2 prior chemotherapy regimens for locally recurrent TNBC or mTNBC were randomized 1:1:1 to GCb on days 1 and 8, trilaciclib prior to GCb on days 1 and 8, or trilaciclib alone on days 1 and 8, and prior to GCb on days 2 and 9. Gene expression, immune cell populations, and Tumor Inflammation Signature (TIS) scores were assessed in baseline tumor samples, with flow cytometric analysis and intracellular and surface cytokine staining used to assess immune cell populations and function. RESULTS: After two cycles, the trilaciclib plus GCb group (n = 68) had fewer total T cells and significantly fewer CD8+ T cells and myeloid-derived suppressor cells compared with baseline, with enhanced T-cell effector function versus GCb alone. No significant differences were observed in patients who received GCb alone (n = 34). Of 58 patients in the trilaciclib plus GCb group with antitumor response data, 27 had an objective response. RNA sequencing revealed a trend toward higher baseline TIS scores among responders versus non­responders. CONCLUSION: The results suggest that administering trilaciclib prior to GCb may modulate the composition and response of immune cell subsets to TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Carboplatin , Triple Negative Breast Neoplasms/pathology , Pyrimidines , Pyrroles/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
J Gene Med ; 25(8): e3509, 2023 08.
Article in English | MEDLINE | ID: mdl-36994804

ABSTRACT

BACKGROUND: A major challenge to adeno-associated virus (AAV)-mediated gene therapy is the presence of anti-AAV capsid neutralizing antibodies (NAbs), which can block viral vector transduction even at very low titers. In the present study, we examined the ability of a combination immunosuppression (IS) treatment with bortezomib and a mouse-specific CD20 monoclonal antibody to suppress anti-AAV NAbs and enable readministration of AAV vectors of the same capsid in mice. METHODS: An AAV8 vector (AAV8-CB-hGAA) that ubiquitously expresses human α-glucosidase was used for initial gene therapy and a second AAV8 vector (AAV8-LSP-hSEAP) that contains a liver-specific promoter to express human secreted embryonic alkaline phosphatase (hSEAP) was used for AAV readministration. Plasma samples were used for determination of anti-AAV8 NAb titers. Cells isolated from whole blood, spleen, and bone marrow were analyzed for B-cell depletion by flow cytometry. The efficiency of AAV readministration was determined by the secretion of hSEAP in blood. RESULTS: In näive mice, an 8-week IS treatment along with AAV8-CB-hGAA injection effectively depleted CD19+ B220+ B cells from blood, spleen, and bone marrow and prevented the formation of anti-AAV8 NAbs. Following administration of AAV8-LSP-hSEAP, increasing levels of hSEAP were detected in blood for up to 6 weeks, indicating successful AAV readministration. In mice pre-immunized with AAV8-CB-hGAA, comparison of IS treatment for 8, 12, 16, and 20 weeks revealed that the 16-week IS treatment demonstrated the highest plasma hSEAP level following AAV8-LSP-hSEAP readministration. CONCLUSIONS: Our data suggest that this combination treatment is an effective IS approach that will allow retreatment of patients with AAV-mediated gene therapy. A combination IS treatment with bortezomib and a mouse-specific CD20 monoclonal antibody effectively suppressed anti-AAV NAbs in naïve mice and in mice with pre-existing antibodies, allowing successful readministration of the same AAV capsid vector.


Subject(s)
Antibodies, Neutralizing , Glycogen Storage Disease Type II , Humans , Mice , Animals , Bortezomib/pharmacology , Bortezomib/therapeutic use , Capsid , Antibodies, Viral , Genetic Vectors/genetics , Retreatment , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Dependovirus/genetics
4.
Front Immunol ; 14: 1301912, 2023.
Article in English | MEDLINE | ID: mdl-38250073

ABSTRACT

Introduction: The efficacy of enzyme replacement therapy (ERT) with alglucosidase alfa for infantile-onset Pompe disease (IOPD) is limited in some patients due to the development of high and sustained antibody titers (HSAT; ≥12,800). Methods: We carried out detailed immunophenotyping of IOPD patients (n=40), including analysis of circulating cell populations by flow cytometry and plasma cytokines by multiplex array, to determine whether patients with HSAT have unique immunological characteristics compared to those with low titers (LT; <12,800). Results: Compared to patients with LT, patients who develop HSAT were skewed toward a type 2 immune profile, with an increased frequency of Th2 cells that was positively correlated with levels of Th2 (IL-4, IL-5, IL-13) and pro-inflammatory (IL-6, TNF-α, MIP-1α, MIP-1ß) cytokines. B cells were increased in HSAT patients with a decreased fraction of unswitched memory B cells. Plasma GM-CSF concentrations were lower on average in HSAT patients, while CXCL11 was elevated. Finally, using principal components analysis, we derived an HSAT Signature Score that successfully stratified patients according to their antibody titers. Discussion: The immune profiles revealed in this study not only identify potential biomarkers of patients that developed HSAT but also provide insights into the pathophysiology of HSAT that will ultimately lead to improved immunotherapy strategies.


Subject(s)
Glycogen Storage Disease Type II , Humans , Glycogen Storage Disease Type II/drug therapy , Immunophenotyping , Enzyme Replacement Therapy , Cytokines , B-Lymphocytes
5.
Brain Behav Immun Health ; 26: 100558, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36439058

ABSTRACT

Mounting evidence suggests that chronic stress and subsequent distress can promote ovarian cancer progression. These altered psychological states have been linked to sustained release of stress hormones, activation of the ß-adrenergic receptors in ovarian cancer cells, and induction of pro-tumoral signaling pathways. In addition, data suggest that chronic stress promotes an inflammatory landscape highlighted by increased infiltration of tumor-associated macrophages into the ovarian tumor microenvironment (TME). In ovarian cancer, ascites is a unique TME comprised of tumor, and immune cells, which secrete pro-tumoral cytokines and chemokines that modulate tumor-associated immunity. However, our knowledge about how stress hormones impact the ascites TME remains limited. We hypothesized that the ascites harbors measurable levels of stress hormones, and accumulation of these in the ascites generates a pro-tumorigenic, inflammatory, and immunosuppressive TME. We evaluated ascites samples from 49 patients with high grade serous ovarian cancer (HGSOC) and quantified cortisol and stress hormones metabolites, metanephrine (MN), and normetanephrine (NMN) in all samples. We also measured 38 individual cytokines in the ascites, including several pro-inflammatory cytokines, such as IL-6, which were positively correlated to MN or NMN levels of those samples. Conversely, we found cortisol levels were negatively correlated to several pro-inflammatory cytokines. As T-cells are integral to the TME and our analyses identified cytokines in the ascites known to modulate T-cell function, we characterized ascites-derived T-cells and assessed the impact of stress hormones on the T-cell phenotype. Our data show an altered CD4+/CD8+ T-cell ratio and a heterogeneous expression of exhaustion markers in T-cells from the ascites, while ascites-derived CD8+ T-cells exposed to epinephrine had decreased co-expression CD38 and Granzyme B. To extend these findings to animal models, we subjected ovarian cancer-bearing mice to daily restraint stress, which resulted in increased tumor growth in two models. Congruent with our human analyses, we detected corticosterone, MN, and NMN in the ascites from tumor-bearing mice, and these stress hormones correlated with several inflammatory cytokines. Moreover, daily restraint stress leads to increased CD4+PD-1+/CD8+PD-1+ T-cell ratio in the ovarian tumor microenvironment. Overall, these data highlight a role of stress hormones in the ascites TME as a driver of tumor-associated inflammation, T-cell suppression, and disease progression.

6.
Future Oncol ; 18(33): 3701-3711, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36135712

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive malignancy for which cytotoxic chemotherapy remains the backbone of treatment. Trilaciclib is an intravenous cyclin-dependent kinase 4/6 inhibitor that induces transient cell cycle arrest of hematopoietic stem and progenitor cells and immune cells during chemotherapy exposure, protecting them from chemotherapy-induced damage and enhancing immune activity. Administration of trilaciclib prior to gemcitabine plus carboplatin (GCb) significantly improved overall survival (OS) compared with GCb alone in an open-label phase II trial in patients with metastatic TNBC, potentially through protection and direct activation of immune function. The randomized, double-blind, placebo-controlled, phase III PRESERVE 2 trial will evaluate the efficacy and safety of trilaciclib administered prior to GCb in patients with locally advanced unresectable or metastatic TNBC. Clinical Trial Registration: NCT04799249 (ClinicalTrials.gov).


Subject(s)
Gemcitabine , Triple Negative Breast Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/therapeutic use , Pyrimidines/therapeutic use , Randomized Controlled Trials as Topic , Triple Negative Breast Neoplasms/pathology
7.
Arthritis Res Ther ; 23(1): 280, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34736523

ABSTRACT

BACKGROUND: Anterior cruciate ligament (ACL) and meniscus tears are common knee injuries. Despite the high rate of post-traumatic osteoarthritis (PTOA) following these injuries, the contributing factors remain unclear. In this study, we characterized the immune cell profiles of normal and injured joints at the time of ACL and meniscal surgeries. METHODS: Twenty-nine patients (14 meniscus-injured and 15 ACL-injured) undergoing ACL and/or meniscus surgery but with a normal contralateral knee were recruited. During surgery, synovial fluid was aspirated from both normal and injured knees. Synovial fluid cells were pelleted, washed, and stained with an antibody cocktail consisting of fluorescent antibodies for cell surface proteins. Analysis of immune cells in the synovial fluid was performed by polychromatic flow cytometry. A broad spectrum immune cell panel was used in the first 10 subjects. Based on these results, a T cell-specific panel was used in the subsequent 19 subjects. RESULTS: Using the broad spectrum immune cell panel, we detected significantly more total viable cells and CD3 T cells in the injured compared to the paired normal knees. In addition, there were significantly more injured knees with T cells above a 500-cell threshold. Within the injured knees, CD4 and CD8 T cells were able to be differentiated into subsets. The frequency of total CD4 T cells was significantly different among injury types, but no statistical differences were detected among CD4 and CD8 T cell subsets by injury type. CONCLUSIONS: Our findings provide foundational data showing that ACL and meniscus injuries induce an immune cell-rich microenvironment that consists primarily of T cells with multiple T helper phenotypes. Future studies investigating the relationship between immune cells and joint degeneration may provide an enhanced understanding of the pathophysiology of PTOA following joint injury.


Subject(s)
Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament Reconstruction , Cartilage, Articular , Meniscus , Anterior Cruciate Ligament/surgery , Anterior Cruciate Ligament Injuries/surgery , Humans , Knee Joint , Magnetic Resonance Imaging , Synovial Fluid
8.
Nat Commun ; 12(1): 5456, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526511

ABSTRACT

Sensitized kidney transplant recipients experience high rates of antibody-mediated rejection due to the presence of donor-specific antibodies and immunologic memory. Here we show that transient peri-transplant treatment with the central complement component C3 inhibitor Cp40 significantly prolongs median allograft survival in a sensitized nonhuman primate model. Despite donor-specific antibody levels remaining high, fifty percent of Cp40-treated primates maintain normal kidney function beyond the last day of treatment. Interestingly, presence of antibodies of the IgM class associates with reduced median graft survival (8 vs. 40 days; p = 0.02). Cp40 does not alter lymphocyte depletion by rhesus-specific anti-thymocyte globulin, but inhibits lymphocyte activation and proliferation, resulting in reduced antibody-mediated injury and complement deposition. In summary, Cp40 prevents acute antibody-mediated rejection and prolongs graft survival in primates, and inhibits T and B cell activation and proliferation, suggesting an immunomodulatory effect beyond its direct impact on antibody-mediated injury.


Subject(s)
Antibodies/immunology , Complement C3/antagonists & inhibitors , Graft Rejection/prevention & control , Graft Survival/drug effects , Kidney Transplantation/methods , Macaca mulatta/immunology , Pyridones/pharmacology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Complement C3/immunology , Complement C3/metabolism , Cytokines/blood , Cytokines/immunology , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Survival/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
9.
Clin Immunol ; 231: 108830, 2021 10.
Article in English | MEDLINE | ID: mdl-34450290

ABSTRACT

Eculizumab (ECU), a C5 complement inhibitor, is approved to treat acetylcholine receptor autoantibody positive generalized myasthenia gravis (AChR MG). The clinical effect of ECU relies on inhibition of the terminal complement complex; however, the effect of ECU on lymphocytes is largely unknown. We evaluated innate and adaptive immunity among AChR MG patients (N = 3) before ECU and ≥3 months later while on stable therapy, and found reduced activation markers in memory CD4+ T cell subsets, increased regulatory T cell populations, and reduced frequencies of CXCR5+HLA-DR+CCR7+ Tfh subsets and CD11b+ migratory memory B cells. We observed increases within CD8+ T cell subsets that were terminally differentiated and senescent. Our data suggest complement inhibition with ECU modulates the adaptive immunity in patients with MG, consistent with preclinical data showing changes in complement-mediated signaling by T- and antigen-presenting cells. These findings extend our understanding of ECU's mechanism of action when treating patients with MG.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , B-Lymphocytes/drug effects , Complement Inactivating Agents/therapeutic use , Myasthenia Gravis/drug therapy , T-Lymphocytes/drug effects , Adaptive Immunity/drug effects , Aged , Female , Humans , Male , Middle Aged , Myasthenia Gravis/immunology
10.
Front Immunol ; 12: 636731, 2021.
Article in English | MEDLINE | ID: mdl-34220802

ABSTRACT

Infantile-onset Pompe disease (IOPD) is a glycogen storage disease caused by a deficiency of acid alpha-glucosidase (GAA). Treatment with recombinant human GAA (rhGAA, alglucosidase alfa) enzyme replacement therapy (ERT) significantly improves clinical outcomes; however, many IOPD children treated with rhGAA develop anti-drug antibodies (ADA) that render the therapy ineffective. Antibodies to rhGAA are driven by T cell responses to sequences in rhGAA that differ from the individuals' native GAA (nGAA). The goal of this study was to develop a tool for personalized immunogenicity risk assessment (PIMA) that quantifies T cell epitopes that differ between nGAA and rhGAA using information about an individual's native GAA gene and their HLA DR haplotype, and to use this information to predict the risk of developing ADA. Four versions of PIMA have been developed. They use EpiMatrix, a computational tool for T cell epitope identification, combined with an HLA-restricted epitope-specific scoring feature (iTEM), to assess ADA risk. One version of PIMA also integrates JanusMatrix, a Treg epitope prediction tool to identify putative immunomodulatory (regulatory) T cell epitopes in self-proteins. Using the JanusMatrix-adjusted version of PIMA in a logistic regression model with data from 48 cross-reactive immunological material (CRIM)-positive IOPD subjects, those with scores greater than 10 were 4-fold more likely to develop ADA (p<0.03) than those that had scores less than 10. We also confirmed the hypothesis that some GAA epitopes are immunomodulatory. Twenty-one epitopes were tested, of which four were determined to have an immunomodulatory effect on T effector response in vitro. The implementation of PIMA V3J on a secure-access website would allow clinicians to input the individual HLA DR haplotype of their IOPD patient and the GAA pathogenic variants associated with each GAA allele to calculate the patient's relative risk of developing ADA, enhancing clinical decision-making prior to initiating treatment with ERT. A better understanding of immunogenicity risk will allow the implementation of targeted immunomodulatory approaches in ERT-naïve settings, especially in CRIM-positive patients, which may in turn improve the overall clinical outcomes by minimizing the development of ADA. The PIMA approach may also be useful for other types of enzyme or factor replacement therapies.


Subject(s)
Computational Biology/methods , Glycogen Storage Disease Type II/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , alpha-Glucosidases/metabolism , Enzyme Replacement Therapy , Epitope Mapping , Female , HLA-DR Antigens/genetics , Humans , Immune Tolerance , Infant , Male , Precision Medicine , Prognosis , Regression Analysis , Risk , alpha-Glucosidases/genetics , alpha-Glucosidases/immunology
11.
Data Brief ; 35: 106857, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33665253

ABSTRACT

We determined normative data for plasma cytokines established from a cohort of 126 carefully screened healthy adults aged 18 to 64 years. Participants were enrolled to ensure an even age and sex distribution and to include at least 30% non-Caucasians. Plasma cytokines for 18 analytes were tested by multiplex immunoassay. The data are presented by age cohort (18-29 years, 30-39, 40-49, and 50-66), as well as by sex and racial background. This dataset complements published normative ranges of cellular subsets generated by comprehensive polychromatic flow cytometry analysis of the healthy human immune system [1]. These data are available to researchers and have value as a reference range for research involving peripheral cytokines.

12.
J Orthop Res ; 39(8): 1710-1721, 2021 08.
Article in English | MEDLINE | ID: mdl-33104263

ABSTRACT

The inflammatory response to joint injury, specifically intra-articular fracture, has been implicated in posttraumatic arthritis development. However, the role of T cells in regulating the development of posttraumatic arthritis is unclear. We hypothesized that the absence of T cells would lead to less severe posttraumatic arthritis following intra-articular fracture. T cell-deficient, athymic nude, and wild-type C57BL/6NJ mice were assessed at 8 weeks following closed articular fracture. Joints were assessed using histologic scores of arthritis, synovitis, and bone morphology via micro computed tomography. Cells were profiled in whole blood via flow cytometry, and plasma and synovial fluid derived cytokines were quantified by multiplex analysis. Compared to C57BL/6NJ mice, nude mice had significantly greater histologic evidence of arthritis and synovitis. Whole blood immune cell profiling revealed a lower percentage of dendritic cells but increased natural killer (NK) cells in nude mice. Concurrently, nude mice had significantly higher levels of NK cells in synovial tissue. Concentrations of plasma interleukin 1ß (IL-1ß) and tumor necrosis factor α, and synovial fluid IL-12, IL-17, and IL-6 in both knees were greater in nude mice. Outcomes of this study suggest that T cells may play a protective regulatory role against the development of posttraumatic arthritis. Clinical significance: Lack of functional T cells exacerbated the development of posttraumatic arthritis following intra-articular fracture suggesting that critical regulators of the immune responses, contained within the T cell population, are required for protection. Future research identifying the specific T cell subsets responsible for modulating disease immunopathogenesis will lead to new therapeutic targets to mitigate posttraumatic arthritis.


Subject(s)
Arthritis , Intra-Articular Fractures , Synovitis , Animals , Arthritis/etiology , Intra-Articular Fractures/complications , Mice , Mice, Inbred C57BL , Mice, Nude , Synovitis/etiology , X-Ray Microtomography
13.
Muscle Nerve ; 63(4): 577-585, 2021 04.
Article in English | MEDLINE | ID: mdl-33294984

ABSTRACT

BACKGROUND: The immunopathology of autoimmune seronegative myasthenia gravis (SN MG) is poorly understood. Our objective was to determine immune profiles associated with a diagnosis of SN MG. METHODS: We performed high-dimensional flow cytometry on blood samples from SN MG patients (N = 68), healthy controls (N = 46), and acetylcholine receptor antibody (AChR+) MG patients (N = 27). We compared 12 immune cell subsets in SN MG to controls using logistic modeling via a discovery-replication design. An exploratory analysis fit a multinomial model comparing AChR+ MG and controls to SN MG. RESULTS: An increase in CD19+ CD20- CD38hi plasmablast frequencies was associated with lower odds of being a SN MG case in both the discovery and replication analyses (discovery P-value = .0003, replication P-value = .0021). Interleukin (IL) -21 producing helper T cell frequencies were associated with a diagnosis of AChR+ MG (P = .004). CONCLUSIONS: Reduced plasmablast frequencies are strongly associated with a SN MG diagnosis and may be a useful diagnostic biomarker in the future.


Subject(s)
Autoantibodies/blood , Myasthenia Gravis/blood , Plasma Cells/cytology , Receptors, Cholinergic/blood , Adult , Aged , Biomarkers/blood , Female , Flow Cytometry/methods , Humans , Male , Middle Aged , Myasthenia Gravis/diagnosis , Receptors, Cholinergic/immunology , Young Adult
14.
Clin Cancer Res ; 27(5): 1287-1295, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33172894

ABSTRACT

PURPOSE: In this prospective trial, we sought to assess the feasibility of concurrent administration of ipilimumab and radiation as adjuvant, neoadjuvant, or definitive therapy in patients with regionally advanced melanoma. PATIENTS AND METHODS: Twenty-four patients in two cohorts were enrolled and received ipilimumab at 3 mg/kg every 3 weeks for four doses in conjunction with radiation; median dose was 4,000 cGy (interquartile range, 3,550-4,800 cGy). Patients in cohort 1 were treated adjuvantly; patients in cohort 2 were treated either neoadjuvantly or as definitive therapy. RESULTS: Adverse event profiles were consistent with those previously reported with checkpoint inhibition and radiation. For the neoadjuvant/definitive cohort, the objective response rate was 64% (80% confidence interval, 40%-83%), with 4 of 10 evaluable patients achieving a radiographic complete response. An additional 3 patients in this cohort had a partial response and went on to surgical resection. With 2 years of follow-up, the 6-, 12-, and 24-month relapse-free survival for the adjuvant cohort was 85%, 69%, and 62%, respectively. At 2 years, all patients in the neoadjuvant/definitive cohort and 10/13 patients in the adjuvant cohort were still alive. Correlative studies suggested that response in some patients were associated with specific CD4+ T-cell subsets. CONCLUSIONS: Overall, concurrent administration of ipilimumab and radiation was feasible, and resulted in a high response rate, converting some patients with unresectable disease into surgical candidates. Additional studies to investigate the combination of radiation and checkpoint inhibitor therapy are warranted.


Subject(s)
Chemoradiotherapy, Adjuvant/mortality , Ipilimumab/therapeutic use , Melanoma/therapy , Neoadjuvant Therapy/mortality , Neoplasm Recurrence, Local/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Recurrence, Local/pathology , Prognosis , Prospective Studies , Radiotherapy Dosage , Survival Rate , Young Adult
15.
J Neuroimmunol ; 345: 577279, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32497931

ABSTRACT

A detailed understanding of the role of Tfh cells in MuSK-antibody positive myasthenia gravis (MuSK-MG) is lacking. We characterized phenotype and function of Tfh cells in MuSK-MG patients and controls. We found similar overall Tfh and follicular regulatory (Tfr) T cell frequencies in MuSK-MG and healthy controls, but MuSK-MG patients exhibited higher frequencies of Tfh17 cells and a higher ratio of Tfh:Tfr cells. These results suggest imbalanced Tfh cell regulation, further supported by increased frequencies of CD4 T cells co-producing IL-21/IL-17 and IL-17/IFN-γ, and increased Tfh-supported IgG production. These results support a role for Tfh cell dysregulation in MuSK-MG immunopathology.


Subject(s)
Autoantibodies/immunology , Inflammation Mediators/immunology , Interleukin-17/immunology , Myasthenia Gravis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , Autoantibodies/blood , Coculture Techniques , Female , Humans , Inflammation Mediators/blood , Interleukin-17/blood , Male , Middle Aged , Myasthenia Gravis/blood , T-Lymphocytes, Helper-Inducer/metabolism , Young Adult
16.
Exp Neurol ; 325: 113146, 2020 03.
Article in English | MEDLINE | ID: mdl-31838097

ABSTRACT

IL-17 producing CD4 T cells (Th17) cells increase significantly with disease severity in myasthenia gravis (MG) patients. To suppress the generation of Th17 cells, we examined the effect of inhibiting retinoic acid receptor-related-orphan-receptor-C (RORγ), a Th17-specific transcription factor critical for differentiation. RORγ inhibition profoundly reduced Th17 cell frequencies, including IFN-γ and IL-17 co-producing pathogenic Th17 cells. Other T helper subsets were not affected. In parallel, CD8 T cell subsets producing IL-17 and IL-17/IFN-γ were increased in MG patients and inhibited by the RORγ inhibitor. These findings provide rationale for exploration of targeted Th17 therapies, including ROR-γ inhibitors, to treat MG patients.


Subject(s)
Immunologic Factors/pharmacology , Myasthenia Gravis/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Th17 Cells/drug effects , Th17 Cells/immunology , Adult , Aged , Aged, 80 and over , Autoantibodies/immunology , Autoantigens/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Female , Humans , Interleukin-17/immunology , Male , Middle Aged , Receptors, Cholinergic/immunology
17.
PLoS One ; 14(12): e0225512, 2019.
Article in English | MEDLINE | ID: mdl-31825961

ABSTRACT

Existing normative flow cytometry data have several limitations including small sample sizes, incompletely described study populations, variable flow cytometry methodology, and limited depth for defining lymphocyte subpopulations. To overcome these issues, we defined high-dimensional flow cytometry reference ranges for the healthy human immune system using Human Immunology Project Consortium methodologies after carefully screening 127 subjects deemed healthy through clinical and laboratory testing. We enrolled subjects in the following age cohorts: 18-29 years, 30-39, 40-49, and 50-66 and enrolled cohorts to ensure an even gender distribution and at least 30% non-Caucasians. From peripheral blood mononuclear cells, flow cytometry reference ranges were defined for >50 immune subsets including T-cell (activation, maturation, T follicular helper and regulatory T cell), B-cell, and innate cells. We also developed a web tool for visualization of the dataset and download of raw data. This dataset provides the immunology community with a resource to compare and extract data from rigorously characterized healthy subjects across age groups, gender and race.


Subject(s)
Cell Separation/standards , Flow Cytometry/standards , Immunity, Cellular/physiology , Leukocytes, Mononuclear/immunology , Lymphocyte Subsets/immunology , Adolescent , Adult , Age Factors , Aged , Cell Separation/methods , Female , Flow Cytometry/methods , Healthy Volunteers , Humans , Male , Middle Aged , Racial Groups , Reference Values , Young Adult
18.
J Am Soc Nephrol ; 30(12): 2399-2411, 2019 12.
Article in English | MEDLINE | ID: mdl-31658991

ABSTRACT

BACKGROUND: Patients with broad HLA sensitization have poor access to donor organs, high mortality while waiting for kidney transplant, and inferior graft survival. Although desensitization strategies permit transplantation via lowering of donor-specific antibodies, the B cell-response axis from germinal center activation to plasma cell differentiation remains intact. METHODS: To investigate targeting the germinal center response and plasma cells as a desensitization strategy, we sensitized maximally MHC-mismatched rhesus pairs with two sequential skin transplants. We administered a proteasome inhibitor (carfilzomib) and costimulation blockade agent (belatacept) to six animals weekly for 1 month; four controls received no treatment. We analyzed blood, lymph node, bone marrow cells, and serum before desensitization, after desensitization, and after kidney transplantation. RESULTS: The group receiving carfilzomib and belatacept exhibited significantly reduced levels of donor-specific antibodies (P=0.05) and bone marrow plasma cells (P=0.02) compared with controls, with a trend toward reduced lymph node T follicular helper cells (P=0.06). Compared with controls, carfilzomib- and belatacept-treated animals had significantly prolonged graft survival (P=0.02), and renal biopsy at 1 month showed significantly reduced antibody-mediated rejection scores (P=0.02). However, four of five animals with long-term graft survival showed gradual rebound of donor-specific antibodies and antibody-mediated rejection. CONCLUSIONS: Desensitization using proteasome inhibition and costimulation blockade reduces bone marrow plasma cells, disorganizes germinal center responses, reduces donor-specific antibody levels, and prolongs allograft survival in highly sensitized nonhuman primates. Most animals experienced antibody-mediated rejection with humoral-response rebound, suggesting desensitization must be maintained after transplantation using ongoing suppression of the B cell response.


Subject(s)
Abatacept/pharmacology , Graft Enhancement, Immunologic/methods , Graft Rejection/prevention & control , Kidney Transplantation , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Animals , B-Lymphocytes/immunology , Bone Marrow/immunology , Costimulatory and Inhibitory T-Cell Receptors/drug effects , Costimulatory and Inhibitory T-Cell Receptors/immunology , Drug Evaluation, Preclinical , Germinal Center/immunology , Graft Survival , Histocompatibility , Immunologic Memory/drug effects , Immunosuppressive Agents/therapeutic use , Isoantibodies/biosynthesis , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Macaca mulatta , Male , Plasma Cells/immunology , Preoperative Care , Skin Transplantation , T-Lymphocytes, Helper-Inducer/immunology
19.
J Am Soc Nephrol ; 30(7): 1206-1219, 2019 07.
Article in English | MEDLINE | ID: mdl-31227636

ABSTRACT

BACKGROUND: Donor-specific antibodies are associated with increased risk of antibody-mediated rejection and decreased allograft survival. Therefore, reducing the risk of these antibodies remains a clinical need in transplantation. Plasma cells are a logical target of therapy given their critical role in antibody production. METHODS: To target plasma cells, we treated sensitized rhesus macaques with daratumumab (anti-CD38 mAb). Before transplant, we sensitized eight macaques with two sequential skin grafts from MHC-mismatched donors; four of them were also desensitized with daratumumab and plerixafor (anti-CXCR4). We also treated two patients with daratumumab in the context of transplant. RESULTS: The animals treated with daratumumab had significantly reduced donor-specific antibody levels compared with untreated controls (57.9% versus 13% reduction; P<0.05) and prolonged renal graft survival (28.0 days versus 5.2 days; P<0.01). However, the reduction in donor-specific antibodies was not maintained because all recipients demonstrated rapid rebound of antibodies, with profound T cell-mediated rejection. In the two clinical patients, a combined heart and kidney transplant recipient with refractory antibody-mediated rejection and a highly sensitized heart transplant candidate, we also observed a significant decrease in class 1 and 2 donor-specific antibodies that led to clinical improvement of antibody-mediated rejection and to heart graft access. CONCLUSIONS: Targeting CD38 with daratumumab significantly reduced anti-HLA antibodies and anti-HLA donor-specific antibodies in a nonhuman primate model and in two transplant clinical cases before and after transplant. This supports investigation of daratumumab as a potential therapeutic strategy; however, further research is needed regarding its use for both antibody-mediated rejection and desensitization.


Subject(s)
Antibodies, Monoclonal/pharmacology , Kidney Transplantation , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/physiology , Adult , Animals , Antibody-Dependent Cell Cytotoxicity , Benzylamines , Cyclams , Graft Rejection , HLA Antigens/immunology , Heterocyclic Compounds/pharmacology , Humans , Isoantibodies/blood , Macaca mulatta , Male , T-Lymphocytes, Regulatory/drug effects
20.
Exp Neurol ; 312: 43-50, 2019 02.
Article in English | MEDLINE | ID: mdl-30472069

ABSTRACT

Muscle specific tyrosine kinase antibody positive myasthenia gravis (MuSK- MG) is characterized by autoantibodies against the MuSK protein of the neuromuscular junction resulting in weakness of bulbar and proximal muscles. We previously demonstrated that patients with MuSK-MG have increased pro-inflammatory Th1 and Th17 responses. Tacrolimus, an immunosuppressant used in AChR-MG and transplantation patients, inhibits T cell responses through interference with IL-2 transcription. The therapeutic efficacy and immunological effect of tacrolimus in MuSK-MG is unclear. In the current study we examined the proliferation, phenotype and cytokine production of CD4+ and CD8+ T cells in peripheral blood mononuclear cells of MuSK-MG following a 3-day in vitro culture with or without tacrolimus. We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-γ, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-γ and IL-2 producing CD8 T cells. Tacrolimus also inhibits pathogenic Th17 cells coproducing IL-17 and IFN-γ. In addition, tacrolimus suppressed follicular T helper cell (Tfh) and regulatory T helper cell (Treg) subsets. These findings provide preliminary support for tacrolimus as a potential alternative immunosuppressive therapy for MuSK-MG.


Subject(s)
Immunosuppressive Agents/therapeutic use , Myasthenia Gravis/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/metabolism , Tacrolimus/therapeutic use , Th1 Cells/metabolism , Th17 Cells/metabolism , Adult , Aged , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Tacrolimus/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...