scholarly journals Preclinical and clinical characterization of the RORγt inhibitor JNJ-61803534

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Xiaohua Xue ◽  
Aimee De Leon-Tabaldo ◽  
Rosa Luna-Roman ◽  
Glenda Castro ◽  
Michael Albers ◽  
...  

AbstractThe nuclear receptor retinoid-related orphan receptor gamma t (RORγt) plays a critical role in driving Th17 cell differentiation and expansion, as well as IL-17 production in innate and adaptive immune cells. The IL-23/IL-17 axis is implicated in several autoimmune and inflammatory diseases, and biologics targeting IL-23 and IL-17 have shown significant clinical efficacy in treating psoriasis and psoriatic arthritis. JNJ-61803534 is a potent RORγt inverse agonist, selectively inhibiting RORγt-driven transcription versus closely-related family members, RORα and RORβ. JNJ-61803534 inhibited IL-17A production in human CD4+ T cells under Th17 differentiation conditions, but did not inhibit IFNγ production under Th1 differentiation conditions, and had no impact on in vitro differentiation of regulatory T cells (Treg), nor on the suppressive activity of natural Tregs. In the mouse collagen-induced arthritis model, JNJ-61803534 dose-dependently attenuated inflammation, achieving ~ 90% maximum inhibition of clinical score. JNJ-61803534 significantly inhibited disease score in the imiquimod-induced mouse skin inflammation model, and dose-dependently inhibited the expression of RORγt-regulated genes, including IL-17A, IL-17F, IL-22 and IL-23R. Preclinical 1-month toxicity studies in rats and dogs identified doses that were well tolerated supporting progression into first-in-human studies. An oral formulation of JNJ-61803534 was studied in a phase 1 randomized double-blind study in healthy human volunteers to assess safety, pharmacokinetics, and pharmacodynamics. The compound was well tolerated in single ascending doses (SAD) up to 200 mg, and exhibited dose-dependent increases in exposure upon oral dosing, with a plasma half-life of 164 to 170 h. In addition, dose-dependent inhibition of ex vivo stimulated IL-17A production in whole blood was observed, demonstrating in vivo target engagement. In conclusion, JNJ-61803534 is a potent and selective RORγt inhibitor that exhibited acceptable preclinical safety and efficacy, as well as an acceptable safety profile in a healthy volunteer SAD study, with clear evidence of a pharmacodynamic effect in humans.

Author(s):  
Manuel J. Del Rey ◽  
Carolina Meroño ◽  
Cristina Municio ◽  
Alicia Usategui ◽  
María Mittelbrunn ◽  
...  

Mitochondrial dysfunction in different cell types is associated to several pathological processes and potentially contributes to chronic inflammatory and ageing-related diseases. Mitochondrial Transcription Factor A (TFAM) plays a critical role in maintaining mtDNA integrity and function. Taking advantage of the Tfamfl/fl UBC-Cre/ERT2+/+ mice, we sought to develop a cellular in vitro system to investigate the role of mitochondrial dysfunction in the stromal cell component. We describe an inducible model of mitochondrial dysfunction by stable depletion of TFAM in primary mouse skin fibroblast (SK-FB) after 4-hydroxytamoxifen (4-OHT) administration. Tfam gene deletion caused a sustained reduction of Tfam and mtDNA-encoded mRNA expression in Cre(+) cultured for low (LP) and high passages (HP). Ultimately, Tfam knockout translated into a loss of TFAM protein. TFAM depletion led to a substantial reduction of the mitochondrial respiratory chain (MRC) complexes that was exacerbated in HP SK-FB cultures. The assembly pattern showed that the respiratory complexes fail to reach the respirasome in 4-OHT Cre(+) SK-FB. Functionally, we determined the mitochondrial function and the glycolytic activity by mito-stress and glycolysis-stress test respectively. These analysis showed that mitochondrial dysfunction was developed after long-term 4-OHT treatment in HP Cre(+) SK-FB and was compensated by an increase in the glycolytic capacity. Finally, expression analysis revealed that 4-OHT-treated HP Cre(+) SK-FB showed a senescent and pro-inflammatory phenotype. In conclusion, we have generated and validated the first ex vivo model of fibroblast mitochondrial dysfunction that results in a pro-inflammatory phenotype applicable to explore this process in other cell types in a variety of pathological conditions.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4698-4698
Author(s):  
Noelia Purroy ◽  
Pau Abrisqueta ◽  
Júlia Carabia ◽  
Eva Calpe ◽  
Cecilia Carpio ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) is characterized by the accumulation and proliferation of monoclonal CD5+ mature B cells in peripheral blood, lymph nodes (LN), and bone marrow (BM). The microenvironment found in BM and LN induces proliferation of CLL cells and protects them from spontaneous and chemotherapy-induced apoptosis. Syk protein is a tyrosine kinase essential for the BCR signaling pathway that also participates in signaling from chemokine receptors and has been shown to be deregulated in CLL. Therefore Syk has been hypothesized to be a rational candidate for targeted therapy in CLL and its inhibition has been tested with the non-specific Syk inhibitor fostamatinib (R406). Against this background we tested the effectiveness of the highly specific Syk inhibitor TAK-659 in suppressing the induction of survival, proliferation and migration of CLL cells by the microenvironment. To mimic the microenvironment of the proliferative centers ex vivo, we co-cultured primary CLL cells with the BM stromal cells (BMSC), CD40L, CpG ODN and anti-IgM (BCR stimulation). This co-culture system protected CLL cells from apoptosis (mean % of viable cells relative to suspension: 137.52±26.17, P<0.05); proliferative responses were significantly observed after 72 hours (mean % of Ki-67-positive cells: 0.91±0.22 in suspension vs. 7.00±1.49 in co-culture, P<0.001); CLL cells activation according to CD69, CD38, and CD86 expression was markedly induced (mean MFI of CD69: 137.2±26.3 in suspension vs. 339.1±41.4 in co-culture, P<0.01; mean MFI of CD38: 14.1±1.67 in suspension vs. 29.8±6.26 in co-culture, P<0.01; mean MFI of CD86: 27.3±2.99 in suspension vs. 80.8±15.1 in co-culture, P<0.01). Moreover, in this system CLL cells became chemoresistant to fludarabine and bendamustine. TAK-659 inhibited SykTyr525, Btk, Akt and ERK1/2 phosphorylation after BCR cross-linking with anti-IgM in the B cell line Ramos and in primary CLL cells, as assessed by western blot. Syk inhibition by TAK-659 translated into an induction of apoptosis in primary CLL cells, obtaining a LD50 for CLL cells in suspension of 40.39μM (95%CI 21.7-75.2μM) vs. 16.99μΜ (95%CI 7.67-37.67μM) for CLL cells in co-culture. Interestingly, TAK-659 displayed stronger capacity to induce apoptosis than R406 especially in co-cultured CLL cells (LD50 TAK-659 16.99μΜ vs. LD50 R406 not achieved). In addition, combination of TAK-659 with fludarabine, ibrutinib or idelalisib showed a synergistic effect in inducing apoptosis especially in co-cultured CLL cells (Cooperative index for TAK-659 0.1μΜ combined with fludarabine 1μM: 0.62, with ibrutinib 0.1μM: 0.68, and with idelalisib 0.1μM: 0.18). Treatment with TAK-659 also resulted in almost complete abrogation of co-culture-induced proliferation in a dose-dependent manner (mean % of Ki-67-positive cells: 7.00±1.49 in untreated controls vs. 3.39±0.76 after 0.1μM TAK-659 vs. 1.72±0.20 after 1μM TAK-659 vs. 1.27±0.18 after 10μM TAK-659, P<0.01), and CLL cells activation (mean MFI of CD38: 29.7±6.26 in untreated controls vs. 23.9±4.39 after 0.1μM TAK-659, P<0.01; mean MFI of CD86: 80.8±15.1 in untreated controls vs. 58.7±8.99 after 0.1Μm TAK-659, P<0.05). Since BCR signaling also promotes CLL cell chemotaxis toward BMSCs and the chemokines CXCL12 and CXCL13, we next evaluated the effect of treatment with TAK-659 on the migratory capacity of primary CLL cells and we observed that TAK-659 markedly decreased chemotaxis of CLL cells toward CXCL12, CXCL13 and BMSCs. Given the significant homology between Syk and ZAP-70 protein and the critical role of the latter in T cell signaling we aimed to assess the effects of TAK-659 on Jurkat T cells. Surprisingly, we observed that although TAK-659 inhibited ZAP-70Tyr493 phosphorylation, this inhibition did not translate into inhibition of downstream signaling elements, such as Itk, Akt or ERK. We next analyzed the effects of TAK-659 in viability and activation of primary T cells and we observed that TAK-659 did not induce significant apoptosis neither inhibition of activation in terms of CD69 and CD38 expression. In conclusion, these findings demonstrate that, in this ex vivo system, the specific inhibition of Syk by TAK-659 effectively overcomes the microenvironment signals that promote proliferation, activation, survival and chemoresistance of primary CLL cells. Altogether, this study provides a rationale for the clinical development of TAK-659 in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2364-2364
Author(s):  
Anwar A. Sayed ◽  
Amna Malik ◽  
Grace Ayoola ◽  
Elisa Lucchini ◽  
Sasfia Candrianita ◽  
...  

Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by a skewed proinflammatory T cell profile. Thrombopoietin-receptor agonists (TPO-RA) have largely replaced immunosuppressants in the management of this disorder, with some patients achieving remission after a period of treatment with TPO-RA. The potential immune modulatory role of TPO-RA has not been fully investigated. The two current TPO-RA licensed for use in ITP; Eltrombopag (Elt) and Romiplostim (Romi) act on different parts of the TPO-R and have similar response rates. However, patients can respond to one agent but not the other. Elt has been described to have a strong iron chelating effect, and hence we propose that it may have an additive immunomodulatory effect on the T cells, absent in Romi. We determined the immunomodulatory effect of Elt by assessing the proliferation and functionality of T-cell lines and primary T-cells. T cell proliferation was assessed using both CFSE proliferation assay and MTT cell viability assay. T cell phenotype and functionality were assessed by multicolor surface and intracellular flow cytometric staining. Cells were co-cultured with Elt and Romi in vitro and ex vivo with both Jurkat and DG75 cells lines as well as primary cells, respectively. Deferoxamine (DFX) was used as a positive control for iron-chelation, and human TPO was used as a positive control for TPO-RA. All treatment doses were based on their calculated therapeutic serum levels. Mann Whitney U and Kruskal-Wallis H statistical tests were applied where applicable, and a P value of less than 0.05 were considered significant. Elt significantly decreased Jurkat T cells proliferation in a dose-dependent manner compared to no treatment and Romi. DFX, an iron chelator, also decreased Jurkat T cell proliferation to comparable levels of Elt. Interestingly, this anti-proliferative effect of Elt was only observed on Jurkat T cells, but not DG75 B cell line. Ex vivo CFSE proliferation assay was performed on primary CD4 and CD8 T cells assessing the antiproliferative effect of Elt. Elt significantly reduced proliferation compared to no treatment. DFX exhibited a similar antiproliferative effect on primary T cells, however, less potent compared to Elt. Neither Romi nor TPO affected the proliferation of Jurkat cells, DG75 cells or primary T cells. The functionality of CD4 and CD8 T cells was assessed based on the capacity of T cells to produce intracellular TNFα, IFNγ and Granzyme B. Elt significantly reduced the percentages of TNFα+/IFNγ+ CD4+ and CD8+ T cells in a dose-dependent manner. This reduction was also observed, albeit to a lesser extent, when T cells were treated with DFX. Furthermore, Granzyme B expression in CD8+ T cells was significantly reduced when cells were treated Elt, compared to no treatment. Romi did not affect the frequency of CD8+ TNFα+/IFNγ+ populations nor the expression of Granzyme B in CD8+ T cells. CD4+ and CD8+ T cells did not express TPO-R on their surface. To confirm the immunomodulatory role of Elt in vivo, the terminally-differentiated effector (CD45RA+CD62L-) CD8+ T cells were assessed in 13 Elt-treated patients and 11 Romi-treated patients. Patients on Elt had significantly reduced frequency of effector CD8 T cells compared to Romi-treated patients (44% vs 76.8%; p<0.01). Taken together, these novel findings suggest an off target immunomodulatory nature of Elt besides its thrombopoietic effect. This dose-dependent immunomodulatory effect is not TPO-R dependent and targets T cells primarily. This study is the first to display such property of Elt and could explain why there is a differential response to Elt and Romi. We hypothesise that Elt may be more effective in patients with T cell mediated disease, whilst patients with predominantly antibody mediated disease are more likely respond to Romi. These findings can also offer an explanation for Elt effectiveness in other T cell-mediated autoimmune conditions such as Aplastic Anemia. Disclosures Cooper: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Rigel: Consultancy, Membership on an entity's Board of Directors or advisory committees; Principia: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees.


2020 ◽  
Vol 4 (18) ◽  
pp. 4483-4493
Author(s):  
Iosifina P. Foskolou ◽  
Laura Barbieri ◽  
Aude Vernet ◽  
David Bargiela ◽  
Pedro P. Cunha ◽  
...  

Abstract Cancer immunotherapy is advancing rapidly and gene-modified T cells expressing chimeric antigen receptors (CARs) show particular promise. A challenge of CAR-T cell therapy is that the ex vivo–generated CAR-T cells become exhausted during expansion in culture, and do not persist when transferred back to patients. It has become clear that naive and memory CD8 T cells perform better than the total CD8 T-cell populations in CAR-T immunotherapy because of better expansion, antitumor activity, and persistence, which are necessary features for therapeutic success and prevention of disease relapse. However, memory CAR-T cells are rarely used in the clinic due to generation challenges. We previously reported that mouse CD8 T cells cultured with the S enantiomer of the immunometabolite 2-hydroxyglutarate (S-2HG) exhibit enhanced antitumor activity. Here, we show that clinical-grade human donor CAR-T cells can be generated from naive precursors after culture with S-2HG. S-2HG–treated CAR-T cells establish long-term memory cells in vivo and show superior antitumor responses when compared with CAR-T cells generated with standard clinical protocols. This study provides the basis for a phase 1 clinical trial evaluating the activity of S-2HG–treated CD19-CAR-T cells in patients with B-cell malignancies.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1933-1933
Author(s):  
Said Dermime ◽  
Cynthia Lehe ◽  
Hazem Ghebeh ◽  
Abdullah Al-Sulaiman ◽  
Ghofran Al Qudaihi ◽  
...  

Abstract Compelling evidences indicate a key role for regulatory T cells (Tregs) on the host response to cancer and recent studies indicated that the generation of effective WT1-specific cytotoxic T cells can be largely affected by the presence of Tregs. This is the first study to describe human Tregs generated specifically against the WT1 antigen which is overexpressed in several human leukemias and provide the mechanism by which these anti-WT1 Tregs inhibit the immune response in leukemia patients. We have generated T cell lines and clones that specifically recognized a WT1-84 peptide in an HLA DRB1*0402/TCR-Vb8-restricted manner. Importantly, they recognized HLADRB1* 04-matched fresh leukemic cells expressing the WT1 antigen. These clones exerted a Th2 cytokine profile, had a CD4+CD25+Foxp3+GITR+CD127− Tregs phenotype, and significantly inhibited the proliferative activity of allogeneic T cells independently of cell-contact. Priming of allo-reactive T cells in the presence of Tregs strongly inhibited the expansion of NK; NK-T and CD8+ T cells, had an inhibitory effect on NK/NK-T cytotoxic activity but not on CD8+ T cells. Furthermore, priming of T cells with the WT1- 126 HLA-A0201-restricted peptide in the presence of Tregs strongly inhibited the induction of anti-WT1-126 CD8+ CTL responses as evidenced by both very low cytotoxic activity and IFN-g production. Moreover, these Tregs clones specifically produced Granzyme-B and selectively induced apoptosis in WT1-84 pulsed-autologous APCs but not in apoptoticresistant DR4-matched leukemic cells. Importantly, we have also detected anti-WT1-84 IL-5+/Granzyme-B+/Foxp3+ CD4+ Tregs in 5 out of 8 HLA-DR4+ AML patients. These findings suggest a critical role for anti-WT1 Tregs in the inhibition of T cell responses against leukemia. This study may have important implications for the clinical manipulation of Tregs such as immuno-targeting of TCR-Vb-8 with mAbs to eliminate anti-WT1 Tregs in leukemia patients in order to enhance GVL before vaccination with the WT1 antigen. On the other hand, leukemia patients with GVHD should be clinically-tried for vaccination with the current WT1-84 peptide or adoptively-treated with ex-vivo anti-WT1 Treg cells to specifically enhance their frequency, which is known to be very low in these patients.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A316-A316
Author(s):  
Thomas Marron ◽  
Julia Kodysh ◽  
Alex Rubinsteyn ◽  
John Finnigan ◽  
Ana Blazquez ◽  
...  

BackgroundThe efficacy of T cell directed immunotherapies relies on adequate priming of T cells to tumor-specific neoantigens, which some studies have augmented with synthetic neoantigen vaccines. This is the first report of a personalized genomic vaccine (PGV-001) in multiple histologies in the adjuvant setting.MethodsTumor and germline RNA and DNA were sequenced, and neoantigen peptides were selected using our OpenVax custom computation pipeline that identifies and ranks mutant sequences by a combination of predicted MHC-I binding affinity and neoantigen abundance within tumor. Up to 10 peptides were synthesized per patient and were administered over the course of 27 weeks in combination with the poly-ICLC. Primary objectives were to determine 1) the safety and tolerability; 2) the feasibility of PGV-001 production and administration; and 3) the immunogenicity of PGV-001. Secondary objectives included immunophenotyping neoantigen-specific T cells in peripheral blood, and characterization of peripheral blood lymphoid, myeloid and humoral responses. We report here for the first time on the primary endpoints.ResultsVaccine was synthesized for 15 patients. A mean of 1619 somatic variants (range 521–5106) were detected. Our pipeline identified a mean of 67.1 neoantigens/patient (range 8–193) and 9.7 peptides/patient were synthesized (range 7–10). 13 patients received PGV-001 (11 patients received all 10 doses and 2 patients received at least 8 doses) while 2 had progressive disease before vaccine initiation. Transient grade 1 injection site reactions were seen in 31% of patients, and one patient experienced grade 1 fever. There were no other significant adverse events. Ex vivo ELISpot analysis of patient blood demonstrated significant induction of T cell responses following receipt of 10 vaccines that were not present after the 6th vaccine, supporting the need for a prolonged dosing schedule. Robust responses were seen in both CD4 and CD8 T cells by intracellular cytokine staining for TNF-a, IFN-a, and IL-2 following in vitro expansion in the presence of vaccine antigens. Additional studies are ongoing to define the most immunogenic antigens.ConclusionsA personalized neoantigen vaccine of synthetic mutant peptides and adjuvant poly-ICLC was successfully synthesized for 15 patients and administered successfully to 87% patients over the course of 27 weeks. The vaccine was well tolerated, and T cell expansion and reactivity to synthetic neoantigens confirms immunogenicity of neoantigens identified with OpenVax.Trial RegistrationNCT02721043Ethics ApprovalThis study was approved by the IRB of The Mount Sinai Hospital in accordance with Federal law. HSM #15-00841.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4373-4373
Author(s):  
Valentina Ferrari ◽  
Tiffany N Tanaka ◽  
Alison Tarke ◽  
Hanna Fields ◽  
Luca Ferrari ◽  
...  

Abstract Background: There are few therapeutic options for higher risk patients with myelodysplastic syndrome (MDS) who fail standard therapy, and their 2-year survival rate is approximately 15%. Here we report on a recently initiated collaborative (industry-academia) first-in-human phase 1 clinical trial to assess the safety and tolerability of a novel form of adoptive T cell immunotherapy for such patients that targets patient and disease-specific, mutation-derived neoantigens. This experimental therapy is based on the concept that a) cancer is caused by somatic mutations that may generate novel immunogenic proteins (ie, neopeptides and possible neoantigens), b) that the adaptive immune system can be trained ex vivo to recognize neopeptides as neoantigens and c) that infusion of culture-expanded, neoantigen-immunized autologous T cells may be safe and therapeutically effective. Methods: This is an open-label, phase 1, 3+3 dose escalation trial with 3 cell doses (0.3, 1, and 3.0 × 107nucleated cells/kg) in cohorts of 3 patients each (see www.clinicaltrials.gov, NCT-03258359). Eligible subjects are 18 years of age or older and will have Intermediate, High, or Very High risk MDS by the revised International Prognostic Scoring System, with at least one cytopenia, and will have failed or relapsed after 6 cycles of standard hypomethylating therapy or declined such therapy, an ECOG status 0-2, and adequate organ function. Each patient's MDS-related mutations are identified and autologous T cells are immunized ex vivo with peptides corresponding to the mutated protein(s), then expanded and suitability-tested for experimental infusion (referred to as PACTN). Importantly, the T cells must demonstrate neoantigen specificity and must kill autologous MDS stem-progenitor cells prior to qualification for infusion. Each eligible subject receives a single infusion of autologous PACTN followed by intensive monitoring for adverse events (AEs) for 4 weeks and periodic monitoring for 1 year. The primary study end-point (EP) is assessment of dose-limiting toxicity (DLT) and maximum tolerated PACTN dose (MTD). Secondary EPs include disease response 1 month after PACTN infusion, overall and progression-free survival at 6 and 12 months, and assessment of the peak abundance and persistence of the infused T cells in peripheral blood. Exploratory EPs include an assessment of the effect of PACTN infusion on the allele frequencies of the targeted and non-targeted MDS mutations in blood and marrow leukocytes. Results: At this time, two subjects have been infused with PACTN in the first dose cohort. Neither subject had an infusion reaction, severe AE, or DLT after follow-up for 2-3 months, nor has a disease response occurred in these subjects. Of interest, the infused PACTN product in the first subject showed 59% clonal dominance by a single T cell receptor (TCR) clone that was present at only 0.002% in patient's blood prior to T cell immunization with neoantigen related peptides. Multiple additional expanded TCR clones were also identified in the infused PACTN product. The presence of a dominant TCR clone in the PACTN product enabled the assessment of the in vivo abundance and persistence of the clone after PACTN infusion. The dominant clone expanded between day 1 and day+4 after PACTN infusion to a peak frequency of 0.13%, representing a 64-fold expansion of this TCR compared with the pre-infusion sample of blood leukocytes, then decreased to 0.09% by day +8. The clone was also demonstrated in bone marrow on day +15 at a frequency of 0.03%, representing a 20-fold expansion of this TCR clone compared with the pre-infusion marrow sample. Similar studies on the second subject are in progress, and will be continued in future subjects as the clinical trial continues. Finally, our studies show that it has been possible to effectively immunize autologous T cells to patient-specific neoantigens in all patients studied with MDS (n=4) and also all patients with AML (n=3) studied to date. Conclusion: The early results of this clinical trial support the feasibility and safety of this novel approach to adoptive T cell mediated immunotherapy for patients with higher-risk MDS and encourages continuation of the trial in the higher dose level cohorts. Disclosures Ferrari: Persimmune, Inc.: Employment. Tarke:Persimmune, Inc.: Employment. Fields:Persimmune, Inc.: Employment. Ferrari:Persimmune, Inc.: Employment. Ni:Persimmune, Inc.: Employment. Ferrari:Persimmune, Inc.: Employment. Warner:Persimmune, Inc.: Employment. Jochelson:PersImmune, Inc.: Consultancy. Bejar:Celgene: Consultancy, Honoraria; AbbVie/Genentech: Consultancy, Honoraria; Takeda: Research Funding; Genoptix: Consultancy; Modus Outcomes: Consultancy; Foundation Medicine: Consultancy; Astex/Otsuka: Consultancy, Honoraria. Vitiello:Persimmune, Inc.: Employment. Lane:PersImmune, Inc.: Employment.


2021 ◽  
Vol 15 ◽  
Author(s):  
Yifat Zaychik ◽  
Nina Fainstein ◽  
Olga Touloumi ◽  
Yehuda Goldberg ◽  
Liel Hamdi ◽  
...  

Background: Exercise training induces beneficial effects on neurodegenerative diseases, and specifically on multiple sclerosis (MS) and it’s model experimental autoimmune encephalomyelitis (EAE). However, it is unclear whether exercise training exerts direct protective effects on the central nervous system (CNS), nor are the mechanisms of neuroprotection fully understood. In this study, we investigated the direct neuroprotective effects of high-intensity continuous training (HICT) against the development of autoimmune neuroinflammation and the role of resident microglia.Methods: We used the transfer EAE model to examine the direct effects of training on the CNS. Healthy mice performed HICT by treadmill running, followed by injection of encephalitogenic proteolipid (PLP)-reactive T-cells to induce EAE. EAE severity was assessed clinically and pathologically. Brain microglia from sedentary (SED) and HICT healthy mice, as well as 5-days post EAE induction (before the onset of disease), were analyzed ex vivo for reactive oxygen species (ROS) and nitric oxide (NO) formation, mRNA expression of M1/M2 markers and neurotrophic factors, and secretion of cytokines and chemokines.Results: Transfer of encephalitogenic T-cells into HICT mice resulted in milder EAE, compared to sedentary mice, as indicated by reduced clinical severity, attenuated T-cell, and neurotoxic macrophage/microglial infiltration, and reduced loss of myelin and axons. In healthy mice, HICT reduced the number of resident microglia without affecting their profile. Isolated microglia from HICT mice after transfer of encephalitogenic T-cells exhibited reduced ROS formation and released less IL-6 and monocyte chemoattractant protein (MCP) in response to PLP-stimulation.Conclusions: These findings point to the critical role of training intensity in neuroprotection. HICT protects the CNS against autoimmune neuroinflammation by reducing microglial-derived ROS formation, neurotoxicity, and pro-inflammatory responses involved in the propagation of autoimmune neuroinflammation.


2019 ◽  
Author(s):  
Azlann Arnett ◽  
Keagan G Moo ◽  
Kaitlin J Flynn ◽  
Thomas B Sundberg ◽  
Liv Johannessen ◽  
...  

AbstractImmune health requires innate and adaptive immune cells to engage precisely balanced pro- and anti-inflammatory forces. A holistic understanding of how individual small molecules affect this balance is essential to anticipate immune-related side effects, select mitigating immunomodulatory therapies and highlight novel utility as immunomodulators. We previously showed that the high-specificity, low-toxicity cyclin dependent kinase 8 (CDK8) inhibitor DCA promotes tolerogenic effects in innate immune cells. Here, we demonstrate that DCA exerts a novel profile of tolerogenic activity on CD4+ T cells, promoting Treg and Th2 while inhibiting Th1 and Th17 differentiation. DCA enhances human Treg differentiation and our models demonstrate clear tolerogenic function of DCA-driven Tregs in the absence of confounding contribution from DCA-innate immune interactions. DCA engages unique mechanisms, including specifically enhancing early Foxp3 expression via regulating c-Jun phosphorylation, to promote Treg differentiation. CDK8 inhibitors are currently being developed to treat cancer; our findings suggest that the potential blunting of host-versus-tumor effects may warrant ancillary pro-inflammatory agents. Importantly, these results highlight novel utility of DCA as an immunomodulator, not only in vivo, but also in ex vivo cellular therapy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1940-1940 ◽  
Author(s):  
David M. Foureau ◽  
Manisha Bhutani ◽  
Myra Robinson ◽  
Fei Guo ◽  
Duy Pham ◽  
...  

Abstract INTRODUCTION: The human B-Cell maturation antigen (BCMA) is a surface marker that is highly expressed on plasma cells and has been recognized as a novel target in multiple myeloma (MM). TNB-383B is a fully human bispecific monoclonal IgG4 antibody. TNB-383B consists of 2 heavy and 1 light chain(s) paired through knob-in-hole technology. The first heavy chain and a kappa light chain form the paratope to recognize and bind human CD3. The second heavy chain is comprised of two identical VH domains in sequence and targets human BCMA with high affinity and avidity. Herein, we describe the ability of TNB-383B to mediate killing of patient-derived tumor cell lysis by endogenous T-cells was assessed ex vivo. METHODS: Bone marrow mononuclear cells (BMMCs) were isolated by density gradient centrifugation from 7 relapsed MM patients enrolled in an IRB-approved biospecimen collection protocol. Freshly isolated BMMC subsets were characterized by flow cytometry, specifically plasma cell (PC) / cytotoxic T cell (CTL) distribution, PC BCMA expression and PC viability were determined. BMMCs were then incubated for 24h (± 2h) with TNB-383B, or negative control, at concentrations ranging from 0.001-1μg. Following incubation, PC lysis, viability, BCMA expression, as well as CTL distribution and degranulation were assessed by flow cytometry. Tukey's sequential trend test was performed for each measured variable, utilizing ANOVA models and contrast statements, to detect linear dose response trends to TNB-383B or negative control treatments. Additionally, a parametric model (EMax) was used for each measured variable to estimate dose response curves, interpolating between tested doses. Two-way factorial ANOVA was utilized to compare the main effects of E:T ratio (or PC phenotype) and dose level and the interaction effect between E:T ratio and dose level on measured variables. RESULTS: Dose-dependent PC lysis was triggered by TNB-383B at concentrations as low as 0.001μg (p=0.0102) while no significant loss of PC was observed with negative control (Figure). This effect was coupled with significant CTL degranulation as expressed by increased CD107a mean fluorescence intensity (MFI) specific to TNB-383B treatment (p=0.0153 at 1μg). Although apoptotic rates (7-AAD+, Annexin V+) of the remaining PC tend to increase among TNB-383B treated compared with isotype control-treated cells, this trend was not significant. As opposed to CTL degranulation, CTL proliferation was not significantly triggered by TNB-383B but was significantly increased when BMMCs were exposed to negative control antibody (p=0.0057 at 0.001 μg). BMMC containing effector to target (E:T) ratio >10 contained lower viable (7-AAD-) PC and higher apoptotic PC counts compared with BMMC specimen with E:T ratio <10 (p<0.001). Using CD45 expression as a surrogate marker of PC maturation and BCMA expression, CD45+ PC displayed higher BCMA expression than CD45- PC (p=0.0189) and were more sensitive TNB-383B-induced killing (p<0.001). Noticeably, overall BCMA expression pre/post TNB-383B exposure remained unaltered. CONCLUSION: Taken together, our findings demonstrate TNB-383B triggers primary PC lysis and CTL degranulation in a dose-dependent fashion ex vivo. The ex vivo bispecific monoclonal antibody assay employed in this study allowed us to identify underlying biological drivers of PC killing efficacy by TNB-383B and may provide a valuable preclinical platform to screen bispecific antibodies and clinical platform to identify mechanism of primary or acquired resistance to the drug. Enrollment of patients with relapsed/refractory MM into a phase I clinical trial with TNB-383B is expected in early 2019. Figure. Figure. Disclosures Foureau: Teneobio Inc.: Research Funding. Pham:Teneobio Inc.: Employment. Force Aldred:Teneobio Inc.: Employment. Buelow:Teneobio Inc.: Employment. Voorhees:Oncopeptides: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: served on an IRC; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Consultancy, Other: served on an IRC; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: served on an IRC; Amgen Inc.: Speakers Bureau; TeneoBio: Consultancy, Membership on an entity's Board of Directors or advisory committees. Usmani:Abbvie, Amgen, Celgene, Genmab, Merck, MundiPharma, Janssen, Seattle Genetics: Consultancy; Amgen, BMS, Celgene, Janssen, Merck, Pharmacyclics,Sanofi, Seattle Genetics, Takeda: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document