Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 373
Filter
1.
Arthritis Rheumatol ; 74(1): 124-133, 2022 01.
Article in English | MEDLINE | ID: mdl-34164952

ABSTRACT

OBJECTIVE: To assess the safety, tolerability, pharmacokinetics, and efficacy of rituximab (RTX) in pediatric patients with granulomatosis with polyangiitis (GPA) or microscopic polyangiitis (MPA). METHODS: The Pediatric Polyangiitis Rituximab Study was a phase IIa, international, open-label, single-arm study. During the initial 6-month remission-induction phase, patients received intravenous infusions of RTX (375 mg/m2 body surface area) and glucocorticoids once per week for 4 weeks. During the follow-up period, patients could receive further treatment, including RTX, for GPA or MPA. The safety, pharmacokinetics, pharmacodynamics, and exploratory efficacy outcomes with RTX were evaluated. RESULTS: Twenty-five pediatric patients with new-onset or relapsing disease were enrolled at 11 centers (19 with GPA [76%] and 6 with MPA [24%]). The median age was 14 years (range 6-17 years). All patients completed the remission-induction phase. During the overall study period (≤4.5 years), patients received between 4 and 28 infusions of RTX. All patients experienced ≥1 adverse event (AE), mostly grade 1 or grade 2 primarily infusion-related reactions. Seven patients experienced 10 serious AEs, and 17 patients experienced 31 infection-related AEs. No deaths were reported. RTX clearance correlated with body surface area. The body surface area-adjusted RTX dosing regimen resulted in similar exposure in both pediatric and adult patients with GPA or MPA. Remission, according to the Pediatric Vasculitis Activity Score, was achieved in 56%, 92%, and 100% of patients by months 6, 12, and 18, respectively. CONCLUSION: In pediatric patients with GPA or MPA, RTX is well tolerated and effective, with an overall safety profile comparable to that observed in adult patients with GPA or MPA who receive treatment with RTX. RTX is associated with a positive risk/benefit profile in pediatric patients with active GPA or MPA.


Subject(s)
Granulomatosis with Polyangiitis/drug therapy , Immunologic Factors/therapeutic use , Microscopic Polyangiitis/drug therapy , Rituximab/therapeutic use , Adolescent , Child , Female , Humans , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Male , Rituximab/adverse effects , Rituximab/pharmacokinetics , Treatment Outcome
2.
Article in English | MEDLINE | ID: mdl-34654708

ABSTRACT

BACKGROUND AND OBJECTIVES: Costimulation by CD40 and its ligand CD40L (CD154) is important for the functional differentiation of T cells. Preclinical studies have recognized the importance of this costimulatory interaction in the pathogenesis of experimental models of multiple sclerosis (MS). To determine safety, pharmacokinetics, and immune effect of a humanized monoclonal antibody (mAb) against CD40 ligand (toralizumab/IDEC-131) in patients with relapsing-remitting MS (RRMS). METHODS: This single-institution open-label dose-escalation study (phase I) enrolled 12 patients with RRMS to receive 4 doses of 1, 5, 10, or 15 mg/kg of humanized αCD40L (toralizumab) IV infusion every other week. Patients were followed up to 18 weeks, annually, and finally at 5 years. In addition to safety and pharmacokinetics, other secondary and exploratory measurements are immune effects, clinical, MRI, laboratory, and neuropsychological evaluations. RESULTS: Fifteen adverse events, all of mild to moderate severity, were considered to be of possible or of unknown relationship to treatment. No serious adverse events, including thromboembolic events, occurred during the 18-week defined study period. Annual and long-term follow-up at 5 years revealed no delayed toxicity. Pharmacokinetics were nonlinear between the 5 and 10 mg/kg dose groups. The serum half-life of toralizumab was consistent between the dose groups with a mean of 15.3 days (SD = 1.9). Flow cytometry revealed no depletion of lymphocyte subsets. An increase in the CD25+/CD3+ and CD25+/CD4+ ratio and a shift toward an anti-inflammatory cytokine response were seen after treatment. DISCUSSION: Our study suggests that blocking CD40L is safe and well tolerated in patients with RRMS while increasing CD25 + T cells and anti-inflammatory cytokine profile. These findings support further studies to assess the efficacy of blocking CD40L as a potential treatment of RRMS. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence on the safety, pharmacokinetics, and immune effects of an mAb to CD40L in patients with RRMS.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Immunologic Factors/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Adult , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , CD40 Ligand , Female , Follow-Up Studies , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Male , Middle Aged , Outcome Assessment, Health Care
3.
Int Immunopharmacol ; 100: 108044, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34601202

ABSTRACT

BACKGROUND: The pharmacokinetics of Ig20Gly, a 20% subcutaneous immunoglobulin (IG) therapy, is well characterized in IG-experienced patients with primary immunodeficiency diseases (PID). Data from IG-naïve patients are limited. OBJECTIVE: Simulate serum total immunoglobulin G (IgG) pharmacokinetic profiles in IG-naïve patients with PID for different Ig20Gly initiation and maintenance dosing regimens. METHODS: A population pharmacokinetic model developed with data from pivotal phase 2/3 trials of weekly Ig20Gly in PID (NCT01412385, NCT01218438) was used to simulate pharmacokinetic profiles of IgG in various scenarios with 400- or 800-mg/kg total loading doses (administered as split doses over 1-2 weeks) and corresponding 100- or 200-mg/kg weekly maintenance doses, respectively. Endogenous baseline IgG levels (1.5, 2.0, 4.0, 6.0 g/L) were evaluated for each scenario; time to putative therapeutic target IgG trough level (7 g/L) was determined. RESULTS: Serum IgG levels reached steady-state by approximately Week 12 for all scenarios and baseline endogenous IgG levels. Time to target trough level generally occurred sooner with 1-week versus 2-week loading schemes. Endogenous baseline IgG levels <4 g/L required a 1-week 800-mg/kg total loading dose to achieve target levels within 2 weeks. Both maintenance regimens sustained serum IgG above target level. CONCLUSIONS: Simulations indicated IG-naïve patients with PID can achieve protective serum IgG levels within 1-3 weeks using appropriate Ig20Gly loading regimens. Patients with low endogenous IgG may benefit most from an 800-mg/kg/month loading dose. 400- or 800-mg/kg/month Ig20Gly maintenance regimens appeared adequate to maintain stable IgG levels. Serum IgG monitoring and clinical status can guide dosing parameters.


Subject(s)
Immunoglobulin G/administration & dosage , Immunologic Factors/pharmacokinetics , Models, Biological , Primary Immunodeficiency Diseases/drug therapy , Adolescent , Adult , Biological Variation, Population , Child , Child, Preschool , Computer Simulation , Drug Administration Schedule , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Factors/administration & dosage , Immunologic Factors/blood , Injections, Subcutaneous , Male , Primary Immunodeficiency Diseases/immunology , Young Adult
4.
Ann Clin Transl Neurol ; 8(11): 2166-2173, 2021 11.
Article in English | MEDLINE | ID: mdl-34704393

ABSTRACT

OBJECTIVE: To assess the independent contributions of clinical measures (relapses, Expanded Disability Status Scale [EDSS] scores, and neuroperformance measures) and nonclinical measures (new brain magnetic resonance imaging [MRI] activity and serum neurofilament light chain [sNfL] levels) for distinguishing natalizumab-treated from placebo-treated patients. METHODS: We conducted post hoc analyses using data from the AFFIRM trial of natalizumab for multiple sclerosis. We used multivariable regression analyses with predictors (EDSS progression, no relapse, new or enlarging MRI activity, brain atrophy, sNfL levels, and neuroperformance worsening) to identify measures that independently discriminated between treatment groups. RESULTS: The multivariable model that best distinguished natalizumab from placebo was no new or enlarging T2 or gadolinium-enhancing activity on MRI (odds ratio; 95% confidence interval: 7.2; 4.7-10.9), year 2 sNfL levels <97.5th percentile (4.1; 2.6-6.2), and no relapses in years 0-2 (2.1; 1.5-3.0). The next best-fitting model was a two-component model that included no MRI activity and sNfL levels <97.5th percentile at year 2. There was little difference between the three- and two-component models. INTERPRETATION: Nonclinical measures (new MRI activity and sNfL levels) discriminate between treatment and placebo groups similarly to or better than clinical outcomes composites and have implications for patient monitoring.


Subject(s)
Immunologic Factors/pharmacokinetics , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/pharmacology , Outcome Assessment, Health Care , Adult , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neurofilament Proteins/blood , Precision Medicine
5.
Front Immunol ; 12: 715195, 2021.
Article in English | MEDLINE | ID: mdl-34512637

ABSTRACT

Objective: To determine the transfer of the monoclonal antibody natalizumab into breastmilk and to evaluate drug and serum neurofilament light chain ((s)NfL) levels in natalizumab exposed pregnancies and lactation periods. Methods: Eleven women with relapsing remitting multiple sclerosis treated with natalizumab during pregnancy and lactation were included in this study. Breastmilk samples were collected up to 302 days after delivery and analyzed for natalizumab concentration and NfL. Additionally, maternal drug levels and sNfL were determined preconceptually, in each trimester, at delivery and postpartum. Clinical and radiological disease activity was systemically assessed across pregnancy and postpartum period. Results: The mean average natalizumab concentration in breast milk was low at 0.06 µg/ml [standard deviation (SD) 0.05] in the eight patients who provided serial breastmilk samples with an estimated mean absolute infant dose of 0.007 mg/kg/d (SD 0.005). The relative infant dose (RID), a metric comparing the infant with maternal drug exposure was low as well with a mean of 0.04% (SD=0.03). Most patients had a maximum concentration in breast milk at one to eight days after infusion. Pregnancy was associated with a non-significant decline of the median natalizumab serum concentration. All patients exposed to natalizumab prior (n=10) and during pregnancy (n=11) kept free of disease activity during gestation. While pregnancy was associated with low sNfL levels in patients treated with natalizumab prior and during pregnancy, the postpartum period was linked to a transient sNfL increase in some patients without any evidence of clinical or radiological disease activity. NfL was detectable in the majority of breastmilk samples with a median concentration of 1.7 pg/ml (range 0.004-18.1). Conclusion: We determined transfer of natalizumab into breastmilk with an RID far below the threshold of concern of 10%. Studies including childhood development assessment are needed in order to gain safety data about natalizumab-exposed breastfeeding. SNfL assessment might be a useful adjunct to monitor silent disease activity and therapeutic response during pregnancy and postpartum period. However, further investigations regarding transient postpartum sNfL increases are required to determine its association to parturition per se or to a silent disease activity in people with multiple sclerosis.


Subject(s)
Immunologic Factors/pharmacokinetics , Milk, Human/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Natalizumab/pharmacokinetics , Neurofilament Proteins/metabolism , Pregnancy Complications , Adult , Biomarkers , Disease Management , Drug Monitoring , Female , Humans , Multiple Sclerosis/blood , Neurofilament Proteins/blood , Pregnancy
6.
Bioorg Med Chem Lett ; 50: 128320, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34400299

ABSTRACT

The atypical chemokine receptor C-X-C chemokine receptor type 7 (CXCR7) is an attractive therapeutic target for a variety of cardiac and immunological diseases. As a strategy to mitigate known risks associated with the development of higher molecular weight, basic compounds, a series of pyrrolidinyl-azolopyrazines were identified as promising small-molecule CXCR7 modulators. Using a highly enabled parallel medicinal chemistry strategy, structure-activity relationship studies geared towards a reduction in lipophilicity and incorporation of saturated heterocycles led to the identification of representative tool compound 20. Notably, compound 20 maintained good potency against CXCR7 with a suitable balance of physicochemical properties to support in vivo pharmacokinetic studies.


Subject(s)
Drug Discovery , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacology , Receptors, CXCR/antagonists & inhibitors , Animals , Drug Delivery Systems , Drug Design , Immunologic Factors/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Signal Transduction , Structure-Activity Relationship
7.
Res Vet Sci ; 138: 69-78, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34111716

ABSTRACT

Astaxanthin (AST) is a red pigment of carotenoid and is considered a high-quality keto-carotenoid pigment with food, livestock, cosmetic, therapeutic and nutraceutical proposes. Astaxanthin exists naturally in fish, crustacean, algae, and birds that naturally exists, principally as fatty acid esters. Many investigations have exhibited the beneficial impacts of astaxanthin when utilized as a pharmaceutical agent in animal nutrition. Astaxanthin has a variety of considerable biological actions, such as being antihypertensive, an antioxidant, anti-obesity properties, and anti-carcinogenic. Astaxanthin has recently acquired popularity as a powerful immunomodulator to maintain the health status and well-being of both animals and humans. The use of astaxanthin is broadly utilized in medical sciences and the nutrition pf aquatic species; however, it presently has limited applications in broader animal nutrition. Understanding astaxanthin's structure, source, and mode of action in the body provides a conceptual base for its clinical application and could enhance the screening of compounds associated with the treatment of many diseases. This review article aims to clarify the important aspects of astaxanthin such as its synthesis, bioavailability, and therapeutics actions, with special interest in practical applications. Awareness of this benefits and production is expected to aid the livestock industry to develop nutritional strategies that ensure the protection of animal health.


Subject(s)
Animal Husbandry , Immunologic Factors , Livestock , Animals , Biological Availability , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacokinetics , Immunologic Factors/therapeutic use , Xanthophylls/chemical synthesis , Xanthophylls/pharmacokinetics , Xanthophylls/therapeutic use
8.
Arch Dis Child ; 106(11): 1058-1065, 2021 11.
Article in English | MEDLINE | ID: mdl-34112638

ABSTRACT

Rituximab is a chimeric monoclonal antibody capable of depleting B cell populations by targeting the CD20 antigen expressed on the cell surface. Its use in oncology, initially in B cell lymphoma and post-transplant lymphoproliferative disorders, predates its current utility in various fields of medicine wherein it has become one of the safest and most effective antibody-based therapies. It was subsequently found to be effective for rheumatological conditions such as rheumatoid arthritis and antineutrophil cytoplasmic antibody-associated vasculitis. Over the past decade, rituximab has generated a lot of interest in nephrology and has become an emerging or accepted therapy for multiple renal conditions, including systemic lupus erythematosus, lupus nephritis, vasculitis, nephrotic syndrome and in different scenarios before and after kidney transplantation. This review outlines its current use in paediatric nephrology practice, focusing on the knowledge required for general paediatricians who may be caring for children prescribed this medication and reviewing them on a shared care basis.


Subject(s)
Antigens, CD20/drug effects , Immunologic Factors/pharmacokinetics , Nephrology/standards , Rituximab/pharmacokinetics , Administration, Intravenous , Antibodies, Monoclonal/therapeutic use , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Child , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Kidney Diseases/drug therapy , Kidney Diseases/immunology , Kidney Transplantation/adverse effects , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Nephrology/statistics & numerical data , Nephrotic Syndrome/drug therapy , Pediatricians/statistics & numerical data , Practice Guidelines as Topic , Rituximab/administration & dosage , Rituximab/pharmacology , Rituximab/therapeutic use , Vasculitis/drug therapy
9.
CPT Pharmacometrics Syst Pharmacol ; 10(8): 839-850, 2021 08.
Article in English | MEDLINE | ID: mdl-34085779

ABSTRACT

The two main objectives of this analysis were to (i) characterize the relationship between immunoglobulin (Ig) exposure and chronic inflammatory demyelinating polyneuropathy (CIDP) disease severity using data from 171 patients with CIDP who received either subcutaneous Ig (IgPro20; Hizentra® ) or placebo (PATH study), and to (ii) simulate and compare exposure coverage with various dosing approaches considering weekly dosing to be the reference dose. IgG pharmacokinetic (PK) parameters, including those from a previous population PK model, were used to predict individual IgG profile and exposure metrics. Treatment-related changes in Inflammatory Neuropathy Cause and Treatment (INCAT) scores were best described by a maximum effect (Emax ) model as a function of ΔIgG (total serum IgG at INCAT score assessment minus baseline IgG levels before intravenous Ig restabilization). Simulations indicate that flexible dosing from daily to biweekly (every other week) provide an exposure coverage equivalent to that of a weekly Ig dose.


Subject(s)
Immunoglobulin G/administration & dosage , Immunologic Factors/administration & dosage , Models, Biological , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy , Adult , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Humans , Immunoglobulin G/metabolism , Immunologic Factors/pharmacokinetics , Injections, Subcutaneous , Male , Middle Aged , Randomized Controlled Trials as Topic , Treatment Outcome , Young Adult
10.
J Nutr Biochem ; 97: 108787, 2021 11.
Article in English | MEDLINE | ID: mdl-34089819

ABSTRACT

The outbreak of mysterious pneumonia at the end of 2019 is associated with widespread research interest worldwide. The coronavirus disease-19 (COVID-19) targets multiple organs through inflammatory, immune, and redox mechanisms, and no effective drug for its prophylaxis or treatment has been identified until now. The use of dietary bioactive compounds, such as phenolic compounds (PC), has emerged as a putative nutritional or therapeutic adjunct approach for COVID-19. In the present study, scientific data on the mechanisms underlying the bioactivity of PC and their usefulness in COVID-19 mitigation are reviewed. In addition, antioxidant, antiviral, anti-inflammatory, and immunomodulatory effects of dietary PC are studied. Moreover, the implications of digestion on the putative benefits of dietary PC against COVID-19 are presented by addressing the bioavailability and biotransformation of PC by the gut microbiota. Lastly, safety issues and possible drug interactions of PC and their implications in COVID-19 therapeutics are discussed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , COVID-19/therapy , Dietary Supplements , Gastrointestinal Microbiome , Phenols/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Biological Availability , Curcumin/pharmacokinetics , Curcumin/pharmacology , Curcumin/therapeutic use , Dietary Supplements/analysis , Gastrointestinal Microbiome/drug effects , Humans , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Phenols/pharmacokinetics , Phenols/pharmacology , Quercetin/pharmacokinetics , Quercetin/pharmacology , Quercetin/therapeutic use , Resveratrol/pharmacokinetics , Resveratrol/pharmacology , Resveratrol/therapeutic use , SARS-CoV-2/drug effects
11.
PLoS One ; 16(6): e0248479, 2021.
Article in English | MEDLINE | ID: mdl-34115763

ABSTRACT

The Coronavirus disease (COVID-19) caused by the virus SARS-CoV-2 has become a global pandemic in a very short time span. Currently, there is no specific treatment or vaccine to counter this highly contagious disease. There is an urgent need to find a specific cure for the disease and global efforts are directed at developing SARS-CoV-2 specific antivirals and immunomodulators. Ayurvedic Rasayana therapy has been traditionally used in India for its immunomodulatory and adaptogenic effects, and more recently has been included as therapeutic adjuvant for several maladies. Amongst several others, Withania somnifera (Ashwagandha), Tinospora cordifolia (Guduchi) and Asparagus racemosus (Shatavari) play an important role in Rasayana therapy. The objective of this study was to explore the immunomodulatory and anti SARS-CoV2 potential of phytoconstituents from Ashwagandha, Guduchi and Shatavari using network pharmacology and docking. The plant extracts were prepared as per ayurvedic procedures and a total of 31 phytoconstituents were identified using UHPLC-PDA and mass spectrometry studies. To assess the immunomodulatory potential of these phytoconstituents an in-silico network pharmacology model was constructed. The model predicts that the phytoconstituents possess the potential to modulate several targets in immune pathways potentially providing a protective role. To explore if these phytoconstituents also possess antiviral activity, docking was performed with the Spike protein, Main Protease and RNA dependent RNA polymerase of the virus. Interestingly, several phytoconstituents are predicted to possess good affinity for the three targets, suggesting their application for the termination of viral life cycle. Further, predictive tools indicate that there would not be adverse herb-drug pharmacokinetic-pharmacodynamic interactions with concomitantly administered drug therapy. We thus make a compelling case to evaluate the potential of these Rasayana botanicals as therapeutic adjuvants in the management of COVID-19 following rigorous experimental validation.


Subject(s)
Antiviral Agents/metabolism , Asparagus Plant/chemistry , COVID-19/metabolism , Immunologic Factors/metabolism , Molecular Docking Simulation/methods , Plant Extracts/metabolism , SARS-CoV-2/enzymology , Tinospora/chemistry , Withania/chemistry , Antiviral Agents/pharmacokinetics , Binding Sites , COVID-19/virology , Coronavirus 3C Proteases/metabolism , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Herb-Drug Interactions , Humans , Immunologic Factors/pharmacokinetics , India , Medicine, Ayurvedic/methods , Phytotherapy/methods , Plant Extracts/pharmacokinetics , Protein Binding , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
12.
Front Immunol ; 12: 669005, 2021.
Article in English | MEDLINE | ID: mdl-33968075

ABSTRACT

Understanding individual responses to nutrition and medicine is of growing interest and importance. There is evidence that differences in bitter taste receptor (TAS2R) genes which give rise to two frequent haplotypes, TAS2R38-PAV (functional) and TAS2R38-AVI (non-functional), may impact inter-individual differences in health status. We here analyzed the relevance of the TAS2R38 receptor in the regulation of the human immune response using the TAS2R38 agonist allyl isothiocyanate (AITC) from Brassica plants. A differential response in calcium mobilization upon AITC treatment in leucocytes from healthy humans confirmed a relevance of TAS2R38 functionality, independent from cation channel TRPV1 or TRPA1 activation. We further identified a TAS2R38-dependence of MAPK and AKT signaling activity, bactericidal (toxicity against E. coli) and anti-inflammatory activity (TNF-alpha inhibition upon cell stimulation). These in vitro results were derived at relevant human plasma levels in the low micro molar range as shown here in a human intervention trial with AITC-containing food.


Subject(s)
Immunologic Factors/pharmacology , Isothiocyanates/pharmacology , Leukocytes/drug effects , Receptors, G-Protein-Coupled/agonists , Adaptive Immunity/drug effects , Adult , Calcium Signaling , Cells, Cultured , Diet , Escherichia coli K12/growth & development , Female , Humans , Immunity, Innate/drug effects , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Isothiocyanates/administration & dosage , Isothiocyanates/pharmacokinetics , Leukocytes/immunology , Leukocytes/metabolism , Male , Microbial Viability , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Polymorphism, Single Nucleotide , Precision Medicine , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
13.
Adv Sci (Weinh) ; 8(10): 2001879, 2021 05.
Article in English | MEDLINE | ID: mdl-34026426

ABSTRACT

Lymphoma is a heterogeneous disease with varying clinical manifestations and outcomes. Many subtypes of lymphoma, such as Burkitt's lymphoma and diffuse large B cell lymphoma, are highly aggressive with dismal prognosis even after conventional chemotherapy and radiotherapy. As such, exploring specific biomarkers for lymphoma is of high clinical significance. Herein, a potential marker, CD38, is investigated for differentiating lymphoma. A CD38-targeting monoclonal antibody (mAb, daratumumab) is then radiolabeled with Zr-89 and Lu-177 for theranostic applications. As the diagnostic component, the Zr-89-labeled mAb is highly specific in delineating CD38-positive lymphoma via positron emission tomography (PET) imaging, while the Lu-177-labeled mAb serves well as the therapeutic component to suppress tumor growth after a one-time administration. These results strongly suggest that CD38 is a lymphoma-specific marker and prove that 89Zr/177Lu-labeled daratumumab facilitates immunoPET imaging and radioimmunotherapy of lymphoma in preclinical models. Further clinical evaluation and translation of this CD38-targeted theranostics may be of significant help in lymphoma patient stratification and management.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal/pharmacology , Lutetium/pharmacokinetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Membrane Glycoproteins/immunology , Positron Emission Tomography Computed Tomography/methods , Precision Medicine/methods , Radioisotopes/pharmacokinetics , Zirconium/pharmacokinetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Humans , Immunologic Factors/pharmacokinetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, SCID , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Clin Transl Sci ; 14(5): 1756-1768, 2021 09.
Article in English | MEDLINE | ID: mdl-33834628

ABSTRACT

Safe and effective new oral therapies for autoimmune, allergic, and inflammatory conditions remain a significant therapeutic need. Here, we investigate the human pharmacokinetics, pharmacodynamics (PDs), and safety of the selective, covalent Bruton's tyrosine kinase (BTK) inhibitor, remibrutinib. Study objectives were explored in randomized single and multiple ascending dose (SAD and MAD, respectively) cohorts with daily doses up to 600 mg, and a crossover food effect (FE) cohort, in adult healthy subjects without (SAD [n =80]/FE [n =12]) or with asymptomatic atopic diathesis (MAD [n =64]). A single oral dose of remibrutinib (0.5-600 mg) was rapidly absorbed (time to maximum concentration = 0.5 h-1.25 h) with an apparent blood clearance of 280-560 L/h and apparent volume of distribution of 400-15,000 L. With multiple doses (q.d. and b.i.d.), no pronounced accumulation of remibrutinib was detected (mean residence time was <3 h). Food intake showed no clinically relevant effect on remibrutinib exposure suggesting no need for dose adaptation. With remibrutinib doses greater than or equal to 30 mg, blood BTK occupancy was greater than 95% for at least 24 h (SAD). With MAD, remibrutinib reached near complete blood BTK occupancy at day 12 predose with greater than or equal to 10 mg q.d. Near complete basophil or skin prick test (SPT) inhibition at day 12 predose was achieved at greater than or equal to 50 mg q.d. for CD63 and at greater than or equal to 100 mg q.d. for SPT. Remibrutinib was well-tolerated at all doses without any dose-limiting toxicity. Remibrutinib showed encouraging blood and skin PDs with a favorable safety profile, supporting further development for diseases driven by mast cells, basophils, and B-cells, such as chronic spontaneous urticaria, allergic asthma, or Sjögren's syndrome.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Food-Drug Interactions , Immunologic Factors , Protein Kinase Inhibitors , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Administration, Oral , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Cross-Over Studies , Dose-Response Relationship, Drug , Healthy Volunteers , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Skin Tests
15.
Clin Transl Sci ; 14(4): 1524-1534, 2021 07.
Article in English | MEDLINE | ID: mdl-33742764

ABSTRACT

RO6870868 is an oral prodrug of the toll-like receptor 7 (TLR7) specific agonist, RO6871765. TLR7 agonists augment host immune activity and are in development to treat hepatitis B infection. We evaluated the safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of RO6870868 in a first-in-human, phase I, randomized, single ascending oral dose study in 60 healthy volunteers at 6 dose levels (200-2000 mg). Single oral doses were generally well-tolerated with a predictable safety profile associated with dose-dependent increases in systemic interferon. No serious adverse events (AEs) were reported and no subject withdrew from the study due to an AE. No clinically significant changes were observed in vital signs, electrocardiograms, or laboratory parameters. Following oral RO6870868 doses, plasma RO6871765 concentrations increased rapidly, exhibiting mean terminal half-life ranging 2-6 h across all cohorts, with area under the plasma concentration versus time curve extrapolated to infinity (AUC0-∞ ) increasing proportionally with dose. A pattern of dose and time-dependent PD activity was demonstrated consistent with engagement of the TLR7 system. Single RO6870868 doses activated components of the TLR innate immune system in a dose-dependent manner with adequate safety and tolerability. Single-dose data in healthy volunteers are useful to evaluate safety, PK, and PD activity of TLR7 agonists and help to guide dose and regimen selection for further trials in patients with chronic hepatitis B.


Subject(s)
Immunologic Factors/adverse effects , Toll-Like Receptor 7/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Half-Life , Healthy Volunteers , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/immunology , Humans , Immunity, Innate/drug effects , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Interferons/blood , Interferons/metabolism , Male , Middle Aged , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Signal Transduction/drug effects , Signal Transduction/immunology , Young Adult
16.
Molecules ; 26(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546377

ABSTRACT

Dihydrochalcones, phlorizin (PZ) and its aglycone phloretin (PT), have evidenced immunomodulatory effects through several mechanisms. However, the differential metabolic signatures that lead to these properties are largely unknown. Since macrophages play an important role in the immune response, our study aimed to characterise human THP-1 macrophages under PZ and PT exposure. A multiplatform-based untargeted metabolomics approach was used to reveal metabolites associated with the anti-inflammatory mechanisms triggered by the dihydrochalcones in LPS-stimulated macrophages, for the first time. Results showed differential phenotypic response in macrophages for all treatments. Dihydrochalcone treatment in LPS-stimulated macrophages mimics the response under normal conditions, suggesting inhibition of LPS response. Antagonistic effects of dihydrochalcones against LPS was mainly observed in glycerophospholipid and sphingolipid metabolism besides promoting amino acid biosynthesis. Moreover, PT showed greater metabolic activity than PZ. Overall, the findings of this study yielded knowledge about the mechanisms of action PZ and PT at metabolic level in modulating inflammatory response in human cells.


Subject(s)
Immunologic Factors , Macrophages/immunology , Metabolomics , Phloretin , Phlorhizin , Humans , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Phloretin/pharmacokinetics , Phloretin/pharmacology , Phlorhizin/pharmacokinetics , Phlorhizin/pharmacology , THP-1 Cells
17.
Expert Rev Gastroenterol Hepatol ; 15(6): 633-641, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33440996

ABSTRACT

Introduction: Women with inflammatory bowel disease (IBD) endorse a tremendous amount of concern about medication exposure during pregnancy and their effects on the fetus. Medical providers caring for this patient population should be well informed and feel comfortable counseling their patients for the best pregnancy outcome possible.Areas covered: It is of particular importance to understand the implications of use of biologics in preconception, pregnancy, and postpartum timeframes. Herein, we aim to inform the clinician about the impact of uncontrolled inflammation during pregnancy, the mechanisms of biologic transport through the placenta, the effects of biologics in maternal and neonatal outcomes, and additional postpartum considerations such as breastfeeding and vaccination safety.Expert opinion: The groundwork already set by previous research in terms of safety of biologic therapy during pregnancy has been reassuring. With the advent of more mechanisms of action but similar protein structure, i.e. they are IgG1 antibodies; the authors anticipate the recommendation of continuation of therapy throughout pregnancy will be sustained.


Subject(s)
Biological Products/therapeutic use , Biological Therapy/methods , Immunologic Factors/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Maternal Health Services , Pregnancy Complications/drug therapy , Biological Products/pharmacokinetics , Breast Feeding , Drug Administration Schedule , Female , Humans , Immunologic Factors/pharmacokinetics , Infant, Newborn , Patient Safety , Pregnancy , Pregnancy Outcome , Treatment Outcome
18.
J Intern Med ; 289(5): 738-746, 2021 05.
Article in English | MEDLINE | ID: mdl-33511686

ABSTRACT

BACKGROUND: Published reports on tocilizumab in COVID-19 pneumonitis show conflicting results due to weak designs or heterogeneity in critical methodological issues. METHODS: This open-label trial, structured according to Simon's optimal design, aims to identify factors predicting which patients could benefit from anti-IL6 strategies and to enhance the design of unequivocal and reliable future randomized trials. A total of 46 patients with COVID-19 pneumonia needing of oxygen therapy to maintain SO2 > 93% and with recent worsening of lung function received a single infusion of tocilizumab. Clinical and biological markers were measured to test their predictive values. Primary end point was early and sustained clinical response. RESULTS: Twenty-one patients fulfilled pre-defined response criteria. Lower levels of IL-6 at 24 h after tocilizumab infusion (P = 0.049) and higher baseline values of PaO2/FiO2 (P = 0.008) predicted a favourable response. CONCLUSIONS: Objective clinical response rate overcame the pre-defined threshold of 30%. Efficacy of tocilizumab to improve respiratory function in patients selected according to our inclusion criteria warrants investigations in randomized trials.


Subject(s)
Antibodies, Monoclonal, Humanized , Biomarkers, Pharmacological/analysis , COVID-19 , Drug Monitoring/methods , Interleukin-6 , Pneumonia, Viral , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/physiopathology , COVID-19/therapy , Female , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Infusions, Intravenous , Interleukin-6/antagonists & inhibitors , Interleukin-6/blood , Italy/epidemiology , Male , Oximetry/methods , Oxygen Inhalation Therapy/methods , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Pneumonia, Viral/etiology , Predictive Value of Tests , Respiratory Function Tests/methods , SARS-CoV-2/isolation & purification , Treatment Outcome
19.
Mediators Inflamm ; 2021: 6699560, 2021.
Article in English | MEDLINE | ID: mdl-33505216

ABSTRACT

Licorice extract is a Chinese herbal medication most often used as a demulcent or elixir. The extract usually consists of many components but the key ingredients are glycyrrhizic (GL) and glycyrrhetinic acid (GA). GL and GA function as potent antioxidants, anti-inflammatory, antiviral, antitumor agents, and immuneregulators. GL and GA have potent activities against hepatitis A, B, and C viruses, human immunodeficiency virus type 1, vesicular stomatitis virus, herpes simplex virus, influenza A, severe acute respiratory syndrome-related coronavirus, respiratory syncytial virus, vaccinia virus, and arboviruses. Also, GA was observed to be of therapeutic valve in human enterovirus 71, which was recognized as the utmost regular virus responsible for hand, foot, and mouth disease. The anti-inflammatory mechanism of GL and GA is realized via cytokines like interferon-γ, tumor necrotizing factor-α, interleukin- (IL-) 1ß, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, and IL-17. They also modulate anti-inflammatory mechanisms like intercellular cell adhesion molecule 1 and P-selectin, enzymes like inducible nitric oxide synthase (iNOS), and transcription factors such as nuclear factor-kappa B, signal transducer and activator of transcription- (STAT-) 3, and STAT-6. Furthermore, DCs treated with GL were capable of influencing T-cell differentiation toward Th1 subset. Moreover, GA is capable of blocking prostaglandin-E2 synthesis via blockade of cyclooxygenase- (COX-) 2 resulting in concurrent augmentation nitric oxide production through the enhancement of iNOS2 mRNA secretion in Leishmania-infected macrophages. GA is capable of inhibiting toll-like receptors as well as high-mobility group box 1.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Glycyrrhetinic Acid/pharmacokinetics , Glycyrrhizic Acid/pharmacokinetics , Immunologic Factors/pharmacokinetics , Animals , Cytokines/metabolism , Drugs, Chinese Herbal/pharmacokinetics , Glycyrrhiza/chemistry , Humans , Inflammation , Interferons/metabolism , Interleukins/metabolism , Leishmania/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Macrophages/parasitology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , RNA, Messenger/metabolism , Th1 Cells/cytology , Toll-Like Receptors/metabolism
20.
Mol Pharm ; 18(2): 539-549, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32502346

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease that results in severe inflammatory microenvironments in the joint tissues. In clinics, disease-modifying antirheumatic drugs (DMARDs) are generally prescribed to patients with RA, but their long-term use often shows toxicity in some organs such as the gastrointestinal system, skin, and kidneys and immunosuppression-mediated infection. Nanomedicine has emerged as a new therapeutic strategy to efficiently localize the drugs in inflamed joints for the treatment of RA. In this Review, we introduce recent research in the area of nanomedicine for the treatment of RA and discuss how the nanomedicine can be used to deliver therapeutic agents to the inflamed joints and manage the progression of RA, particularly focusing on targeted delivery, controlled drug release, and immune modulation.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Drug Carriers/chemistry , Immunologic Factors/administration & dosage , Nanoparticles/chemistry , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antirheumatic Agents/pharmacokinetics , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Compounding/methods , Drug Liberation , Humans , Immunologic Factors/pharmacokinetics , Injections, Intra-Articular , Injections, Intravenous , Injections, Subcutaneous , Particle Size , Surface Properties , Synovial Membrane/immunology , Synovial Membrane/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...