15 results on '"Morton Lori C"'
Search Results
2. Defining the Activated Fibroblast Population in Lung Fibrosis Using Single-Cell Sequencing
- Author
-
Qin Ruan, Scott M. MacDonnell, Wen Zhang, Theodore J. Kaplan, Rebecca Peyser, Gabor Halasz, Yi Wei, Kishor Devalaraja-Narashimha, Morton Lori C, Yong Kim, Christina Adler, Min Ni, Yinglin Gao, Luis Cheng, and Justin C. Grindley
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Clinical Biochemistry ,Population ,Biology ,Extracellular matrix ,03 medical and health sciences ,chemistry.chemical_compound ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,Fibrosis ,Pulmonary fibrosis ,medicine ,education ,Fibroblast ,Molecular Biology ,education.field_of_study ,Cell Biology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,chemistry ,Cancer research ,Nintedanib ,Myofibroblast - Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disorder driven by unrelenting extracellular matrix deposition. Fibroblasts are recognized as the central mediators of extracellular matrix production in IPF; however, the characteristics of the underlying fibroblast cell populations in IPF remain poorly understood. Here, we use an unbiased single-cell RNA sequencing analysis of a bleomycin-induced pulmonary fibrosis model to characterize molecular responses to fibrotic injury. Lung cells were isolated on Day 11 to capture emerging fibrosis and gene expression was analyzed by three complementary techniques, which, together, generated a 49-gene signature that defined an activated subpopulation of fibroblasts. However, none of the identified genes were specific to the activated cells or to the disease setting, implying that the activated fibroblasts are not uniquely defined, but exhibit a similar, yet amplified, gene expression pattern to control cells. Our findings have important implications for fibrosis research, including: 1) defining myofibroblasts with any single marker will fail to capture much of the underlying biology; 2) fibroblast activation is poorly correlated with expression of transforming growth factor-β pathway genes; 3) single-cell analysis provides insight into the mechanism of action of effective therapies (nintedanib); 4) early events in lung fibrosis need not involve significant changes in fibroblast number; populations that do increase in number, such as macrophages, dendritic cells, and proliferating myeloid cells, may merit closer examination for their role in pathogenesis.
- Published
- 2019
3. A liver Hif-2α-Irs2 pathway sensitizes hepatic insulin signaling and is modulated by Vegf inhibition
- Author
-
Wei, Kevin, Piecewicz, Stephanie M., McGinnis, Lisa M., Taniguchi, Cullen M., Wiegand, Stanley J., Anderson, Keith, Chan, Carol W-m, Mulligan, Kimberly X., Kuo, David, Yuan, Jenny, Vallon, Mario, Morton, Lori C., Lefai, Etienne, Simon, M. Celeste, Maher, Jacquelyn J., Mithieux, Gilles, Rajas, Fabienne, Annes, Justin P., McGuinness, Owen P., Thurston, Gavin, Giaccia, Amato J., and Kuo, Calvin J.
- Subjects
Vascular endothelial growth factor -- Physiological aspects ,Insulin -- Physiological aspects ,Glucose metabolism -- Research ,Type 2 diabetes -- Research ,Biological sciences ,Health - Abstract
Insulin initiates diverse hepatic metabolic responses, including gluconeogenic suppression and induction of glycogen synthesis and lipogenesis (1), (2). The liver possesses a rich sinusoidal capillary network with a higher degree [...]
- Published
- 2013
- Full Text
- View/download PDF
4. Advances and unmet needs in genetic, basic and clinical science in Alport syndrome : report from the 2015 International Workshop on Alport Syndrome
- Author
-
Gross, Oliver, Kashtan, Clifford E., Rheault, Michelle N., Flinter, Frances, Savige, Judith, Miner, Jeffrey H., Torra Balcells, Roser, Ars, Elisabet, Deltas, Constantinos, Savva, Isavella, Perin, Laura, Renieri, Alessandra, Ariani, Francesca, Mari, Francesca, Baigent, Colin, Judge, Parminder, Knebelman, Bertrand, Heidet, Laurence, Lagas, Sharon, Blatt, Dave, Ding, Jie, Zhang, Yanqin, Gale, Daniel P., Prunotto, Marco, Xue, Yong, Schachter, Asher D., Morton, Lori C. G., Blem, Jacqui, Huang, Michael, Liu, Shiguang, Vallee, Sebastien, Renault, Daniel, Schifter, Julia, Skelding, Jules, Gear, Susie, Friede, Tim, Turner, A. Neil, Lennon, Rachel, and Universitat Autònoma de Barcelona
- Subjects
Chronic kidney disease ,Hereditary kidney disease ,Guidelines ,Nephroprotection ,Alport syndrome - Abstract
Alport syndrome (AS) is a genetic disease characterized by haematuric glomerulopathy variably associated with hearing loss and anterior lenticonus. It is caused by mutations in the COL4A3, COL4A4 or COL4A5 genes encoding the α3α4α5(IV) collagen heterotrimer. AS is rare, but it accounts for >1% of patients receiving renal replacement therapy. Angiotensin-converting enzyme inhibition slows, but does not stop, the progression to renal failure; therefore, there is an urgent requirement to expand and intensify research towards discovering new therapeutic targets and new therapies. The 2015 International Workshop on Alport Syndrome targeted unmet needs in basic science, genetics and diagnosis, clinical research and current clinical care. In three intensive days, more than 100 international experts including physicians, geneticists, researchers from academia and industry, and patient representatives from all over the world participated in panel discussions and breakout groups. This report summarizes the most important priority areas including (i) understanding the crucial role of podocyte protection and regeneration, (ii) targeting mutations by new molecular techniques for new animal models and potential gene therapy, (iii) creating optimal interaction between nephrologists and geneticists for early diagnosis, (iv) establishing standards for mutation screening and databases, (v) improving widespread accessibility to current standards of clinical care, (vi) improving collaboration with the pharmaceutical/biotech industry to investigate new therapies, (vii) research in hearing loss as a huge unmet need in Alport patients and (viii) the need to evaluate the risk and benefit of novel (including 'repurposing') therapies on an international basis.
- Published
- 2021
5. Development of a semi-automated segmentation tool for high frequency ultrasound image analysis of mouse echocardiograms
- Author
-
Kristi Powers, Michael E. Dunn, William Cupelo, Rhonda S. Silva, Morton Lori C, Justin Torello, Yannick Cadoret, and Raymond Chang
- Subjects
Male ,Test data generation ,Computer science ,Molecular biology ,Physiology ,Science ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,Cardiology ,Article ,Imaging ,Automation ,Mice ,Software ,Medical research ,Image Processing, Computer-Assisted ,Animals ,Computer vision ,Throughput (business) ,Ultrasonography ,Protocol (science) ,Multidisciplinary ,Modality (human–computer interaction) ,business.industry ,Drug discovery ,Ultrasound ,High-throughput screening ,Reproducibility of Results ,Scientific data ,High-Throughput Screening Assays ,Workflow ,Echocardiography ,Data quality ,Medicine ,Artificial intelligence ,business ,Structural biology ,Systems biology ,Algorithms - Abstract
Echocardiography is a widely used and clinically translatable imaging modality for the evaluation of cardiac structure and function in preclinical drug discovery and development. Echocardiograms are among the first in vivo diagnostic tools utilized to evaluate the heart due to its relatively low cost, high throughput acquisition, and non-invasive nature; however lengthy manual image analysis, intra- and inter-operator variability, and subjective image analysis presents a challenge for reproducible data generation in preclinical research. To combat the image-processing bottleneck and address both variability and reproducibly challenges, we developed a semi-automated analysis algorithm workflow to analyze long- and short-axis murine left ventricle (LV) ultrasound images. The long-axis B-mode algorithm executes a script protocol that is trained using a reference library of 322 manually segmented LV ultrasound images. The short-axis script was engineered to analyze M-mode ultrasound images in a semi-automated fashion using a pixel intensity evaluation approach, allowing analysts to place two seed-points to triangulate the local maxima of LV wall boundary annotations. Blinded operator evaluation of the semi-automated analysis tool was performed and compared to the current manual segmentation methodology for testing inter- and intra-operator reproducibility at baseline and after a pharmacologic challenge. Comparisons between manual and semi-automatic derivation of LV ejection fraction resulted in a relative difference of 1% for long-axis (B-mode) images and 2.7% for short-axis (M-mode) images. Our semi-automatic workflow approach reduces image analysis time and subjective bias, as well as decreases inter- and intra-operator variability, thereby enhancing throughput and improving data quality for pre-clinical in vivo studies that incorporate cardiac structure and function endpoints.
- Published
- 2020
6. Inhibition of complement pathway activation with Pozelimab, a fully human antibody to complement component C5
- Author
-
Irena Lovric, Hui Xiao, Jingtai Cao, Brian Zambrowicz, Carola Springer, Olav Olsen, Vishal Kamat, Marc W. Retter, Ming Yuan, Ashique Rafique, Ying Hu, George Ehrlich, Yashu Liu, William C. Olson, Carmelo Romano, Randi Foster, Pamela Krueger, Nina Liu, Henry Chen, William Poueymirou, Qian Zhang, Michael P. Rosconi, Morton Lori C, Marc Cao, Gang Chen, Robert Babb, Adrianna Latuszek, and Tammy T. Huang
- Subjects
0301 basic medicine ,Physiology ,Complement System ,Pharmacology ,Monkeys ,Biochemistry ,Antigen-Antibody Reactions ,Mice ,0302 clinical medicine ,Mathematical and Statistical Techniques ,Immune Physiology ,Medicine and Health Sciences ,Medicine ,Enzyme-Linked Immunoassays ,Complement Activation ,Complement component 5 ,Mammals ,Multidisciplinary ,Immune System Proteins ,biology ,Eukaryota ,Complement C5 ,Animal Models ,Eculizumab ,Hemolysis ,Curve Fitting ,Body Fluids ,Blood ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Monoclonal ,Vertebrates ,Antibody ,Anatomy ,medicine.drug ,Research Article ,Half-Life ,Primates ,Science ,Immunology ,Mouse Models ,Research and Analysis Methods ,Antibodies, Monoclonal, Humanized ,Antibodies ,03 medical and health sciences ,Inhibitory Concentration 50 ,Model Organisms ,Animals ,Humans ,Immunoassays ,Protein Structure, Quaternary ,Innate immune system ,Binding Sites ,business.industry ,Organisms ,Biology and Life Sciences ,Proteins ,Genetic Variation ,medicine.disease ,Complement system ,Macaca fascicularis ,030104 developmental biology ,Immune System ,Amniotes ,biology.protein ,Immunologic Techniques ,Animal Studies ,business ,Mathematical Functions ,Ex vivo - Abstract
Complement is a key component of the innate immune system. Inappropriate complement activation underlies the pathophysiology of a variety of diseases. Complement component 5 (C5) is a validated therapeutic target for complement-mediated diseases, but the development of new therapeutics has been limited by a paucity of preclinical models to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) properties of candidate therapies. The present report describes a novel humanized C5 mouse and its utility in evaluating a panel of fully human anti-C5 antibodies. Surprisingly, humanized C5 mice revealed marked differences in clearance rates amongst a panel of anti-C5 antibodies. One antibody, pozelimab (REGN3918), bound C5 and C5 variants with high affinity and potently blocked complement-mediated hemolysis in vitro. In studies conducted in both humanized C5 mice and cynomolgus monkeys, pozelimab demonstrated prolonged PK and durable suppression of hemolytic activity ex vivo. In humanized C5 mice, a switch in dosing from in-house eculizumab to pozelimab was associated with normalization of serum C5 concentrations, sustained suppression of hemolytic activity ex vivo, and no overt toxicity. Our findings demonstrate the value of humanized C5 mice in identifying new therapeutic candidates and treatment options for complement-mediated diseases.
- Published
- 2020
7. Kappa-on-Heavy (KoH) bodies are a distinct class of fully-human antibody-like therapeutic agents with antigen-binding properties
- Author
-
Pamela Krueger, James Shevchuk, Joannie Bautista, Macdonald Lynn, Natasha Levenkova, Saurabh Wadhwa, Cagan Gurer, Johanna Hansen, Douglas MacDonald, Tammy T. Huang, Ashique Rafique, Kenneth S. Graham, Lakeisha Esau, Matthew C. Franklin, George Ehrlich, Ashok Badithe, George D. Yancopoulos, Maggie Zhong, Andrew J. Murphy, Chunguang Guo, Vera Voronina, Robert Babb, Gang Chen, Naxin Tu, Morton Lori C, Craig Grant, William Poueymirou, Karoline A Meagher, Wojtek Auerbach, and John Mcwhirter
- Subjects
Models, Molecular ,Nicotine ,Protein Conformation ,Immunoglobulin Variable Region ,Endogeny ,Germline ,Antibodies ,chemistry.chemical_compound ,Immunoglobulin kappa-Chains ,Mice ,parasitic diseases ,Animals ,Humans ,Antigens ,Multidisciplinary ,Marinobufagenin ,biology ,Base Sequence ,Chemistry ,Antigen binding ,Small molecule ,Bufanolides ,Biochemistry ,PNAS Plus ,Monoclonal ,biology.protein ,Binding Sites, Antibody ,Antibody ,Genetic Engineering ,Immunoglobulin Heavy Chains ,Kappa - Abstract
We describe a Kappa-on-Heavy (KoH) mouse that produces a class of highly diverse, fully human, antibody-like agents. This mouse was made by replacing the germline variable sequences of both the Ig heavy-chain (IgH) and Ig kappa (IgK) loci with the human IgK germline variable sequences, producing antibody-like molecules with an antigen binding site made up of 2 kappa variable domains. These molecules, named KoH bodies, structurally mimic naturally existing Bence-Jones light-chain dimers in their variable domains and remain wild-type in their antibody constant domains. Unlike artificially diversified, nonimmunoglobulin alternative scaffolds (e.g., DARPins), KoH bodies consist of a configuration of normal Ig scaffolds that undergo natural diversification in B cells. Monoclonal KoH bodies have properties similar to those of conventional antibodies but exhibit an enhanced ability to bind small molecules such as the endogenous cardiotonic steroid marinobufagenin (MBG) and nicotine. A comparison of crystal structures of MBG bound to a KoH Fab versus a conventional Fab showed that the KoH body has a much deeper binding pocket, allowing MBG to be held 4 Å further down into the combining site between the 2 variable domains.
- Published
- 2019
8. Interim Analysis of an Open-Label, Ascending-Dose, Phase 1 Study of the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Single Doses of the Subcutaneously Administered Human Monoclonal Antibody Pozelimab in Combination with Single Doses of the Subcutaneously Administered siRNA Cemdisiran in Healthy Volunteers
- Author
-
Merideth L Rodgers, Lorah Perlee, Morton Lori C, David M. Weinreich, Oren Levy, Jonathan Weyne, Yiziying Chen, Olivier Harari, Tavé van Zyl, Karoline A Meagher, Jutta Miller, George D. Yancopoulos, and Umesh Chaudhari
- Subjects
business.industry ,medicine.drug_class ,Immunology ,Safety tolerability ,Cell Biology ,Hematology ,Pharmacology ,Monoclonal antibody ,Interim analysis ,Biochemistry ,Pharmacokinetics ,Healthy volunteers ,Medicine ,Open label ,business - Abstract
Introduction. Pozelimab (REGN3918) and cemdisiran (ALN-CC5) are C5 inhibitors under development for the treatment of paroxysmal nocturnal hemoglobinuria (PNH), myasthenia gravis (MG), and other diseases in which tissue damage is mediated by terminal complement pathway activity. Pozelimab is a fully human monoclonal immunoglobulin G4P (IgG4P) antibody directed against C5, and cemdisiran is a synthetic small interfering RNA (siRNA) targeting C5 mRNA. Both agents can be administered via subcutaneous (SC) injection. Pharmacokinetic (PK)/pharmacodynamic (PD) modeling based on observed data from both pozelimab and cemdisiran healthy volunteer studies (NCT03115996, NCT02352493) suggests that, by combining cemdisiran and pozelimab, the dose of both agents may be significantly reduced and the interval for SC dosing of pozelimab may be significantly increased. Objectives. To provide initial safety and tolerability data for a SC cemdisiran/pozelimab combination approach. Methods. This is an ongoing phase 1, open-label, parallel-dose cohort combination study (NCT04601844) assessing the safety and tolerability of ascending doses of SC pozelimab in combination with SC cemdisiran when administered on the same day or sequentially, on different days, in healthy subjects. Cohort Descriptions. Three parallel ascending-dose cohorts, consisting of six subjects each, were selected based on PK/PD modeling using observed data from both pozelimab and cemdisiran non-combination healthy volunteer studies: Cohort 1: Cemdisiran low dose SC followed by pozelimab low dose SC, administered on different days Cohort 2: Cemdisiran high dose SC followed by pozelimab low dose SC, administered on different days Cohort 3: Cemdisiran high dose SC and pozelimab high dose SC, both administered on the same day Dose regimens were designed to limit the duration of >50% complement inhibition for no longer than 8 weeks, to mitigate risk in the enrolled healthy subjects. An optional additional Cohort 4 was included in the study design and this interim analysis was planned to review safety data to guide the decision regarding whether to enroll this cohort and to inform its dose selection. Interim Analysis: An interim safety analysis was performed with a data cut-off corresponding to study day 127 for Cohort 1, study day 85 for Cohort 2, and study day 71 for Cohort 3. Eighteen healthy volunteer subjects, nine female and nine male, with a mean age of 36 years (standard deviation 9.5 years), were randomized to one of the three cohorts. Demographic and baseline characteristics were comparable for subjects in each cohort. A total of 47 treatment emergent adverse events (TEAEs) were reported, with comparable distribution across cohorts, all mild to moderate in severity with no severe TEAEs. "Nervous system disorders" was the TEAE System Organ Class with the most subjects for all three cohorts, occurring in 12 (66.7%) of the 18 subjects. "Headache" was the most frequent MedDRA Preferred Term for all three cohorts, occurring in 11 (61.1%) of the 18 subjects. All TEAEs were recovered or recovering at the time of data cut-off, and there were no study drug discontinuations or interruptions of treatment due to TEAEs. There were no serious TEAEs or deaths. There were no clinically meaningful findings or treatment-emergent changes reported on the vital signs or laboratory safety tests. Conclusions: The combined SC administration of pozelimab and cemdisiran represents a promising approach to achieve therapeutically significant complement inhibition for extended durations. An interim safety analysis demonstrated that the combined administration was generally safe and well tolerated. Final PK/PD analysis from this healthy volunteer study will allow for assessment of agreement between modeling predictions and observed data, and further inform combination dose regimens for studies in target patient populations. Disclosures van Zyl: Regeneron Pharmaceuticals, Inc.: Current Employment. Weyne: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Chaudhari: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Harari: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Levy: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Miller: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Chen: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Meagher: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Rodgers: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Perlee: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Morton: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Weinreich: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Yancopoulos: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company.
- Published
- 2021
9. Pozelimab, a Human Monoclonal Antibody Against Complement Factor C5, Provided Inhibition of Intravascular Hemolysis in Patients with Paroxysmal Nocturnal Hemoglobinuria
- Author
-
Jonathan Weyne, David M. Weinreich, Jun-Ho Jang, Bradley Dain, Morag Griffin, Umesh Chaudhari, Morton Lori C, Merideth L Rodgers, Olivier Harari, Karoline A Meagher, Jutta Miller, George D. Yancopoulos, and Lorah Perlee
- Subjects
business.industry ,medicine.drug_class ,Immunology ,Cell Biology ,Hematology ,Complement factor I ,medicine.disease ,Monoclonal antibody ,Biochemistry ,Intravascular hemolysis ,Paroxysmal nocturnal hemoglobinuria ,medicine ,In patient ,business - Abstract
INTRODUCTION Paroxysmal Nocturnal hemoglobuinuria (PNH) is a rare, acquired, life-threatening disorder characterised by intravascular hemolysis and increased risk of thrombosis. Current available treatments represent a significant burden to patients and include anti-complement factor 5 (C5) therapies, eculizumab and ravulizumab, both intravenous infusions, or more recently, twice weekly C3 inhibitor, Pegcetacoplan, a subcutaneous (SC) infusion treatment. Pozelimab (REGN3918), a fully human monoclonal immunoglobulin G4 antibody directed against C5, has been shown to bind with high affinity to wild-type and variant (R885H/C) human C5 and block its activity. In a healthy volunteer study (NCT03115996), SC administration of pozelimab was found to be generally well tolerated while providing complete inhibition of ex vivo-assessed hemolytic activity [Weyne J et al. Blood. 2018;S1:1039]. The data suggested that pozelimab may provide control of intravascular hemolysis in patients with active PNH, and thus could provide an important new alternative for patients supporting progression to a first-in-patient study of pozelimab in patients with active PNH. OBJECTIVE To demonstrate a clinically significant reduction in intravascular hemolysis by once-weekly SC administration of pozelimab over 26 weeks of treatment in patients with active PNH. METHODS This is a phase 2, open-label, single-arm, 26-week treatment study in 24 patients with active symptomatic PNH who are naïve to complement inhibitor therapy, or who have received prior treatment with a complement inhibitor but not within 6 months prior to the start of the study (NCT03946748). The treatment regimen consists of pozelimab as a single IV loading dose of 30 mg/kg followed thereafter by weekly SC 800 mg administrations. The effect of pozelimab on intravascular hemolysis (monitored via LDH levels) and transfusion avoidance, as well as safety, is to be assessed from baseline to week 26 (study day 183; only partial data were available for some patients at the time of abstract submission). Pozelimab pharmacodynamics was assessed utilizing a sheep red blood cell (RBC) complement activity assay (CH50) and rabbit RBC complement activity assay (AH50). RESULTS All 24 patients completed the 26-week treatment period with no study drug discontinuation and have been enrolled into an ongoing open-label extension study (NCT04162470). An interim analysis on the first 17 patients was previously reported. All 17 patients had at least 71 days of treatment, with 10 patients receiving treatment for up to 183 days. All enrolled patients had baseline LDH levels ≥2 x upper limit of normal (ULN). Participants were enrolled in two cohorts: Cohort A for dose confirmation and Cohort B for further evaluation of efficacy and safety. Interim Efficacy Analysis As previously reported, treatment with pozelimab led to a rapid and sustained reduction in LDH through study week 26. All 17 patients achieved LDH reduction to below the clinically significant threshold of LDH ≤1.5 x ULN. All but one patient achieved control of intravascular hemolysis (LDH ≤1.5 x ULN) at week 2, and all but one patient achieved normalization of LDH (LDH ≤1.0 x ULN) at week 4. Importantly, one patient who is a carrier of a C5 variant known to be resistant to blockade by eculizumab/ravulizumab demonstrated rapid and sustained normalization of LDH. Interim Safety As previously reported, no serious adverse events or adverse events leading to treatment discontinuation were reported. Common treatment-emergent adverse events included headache (4 patients; 23.5%) and nausea (2 patients; 11.8%). No breakthrough hemolysis events were observed. CONCLUSIONS Pozelimab administered SC once weekly led to the inhibition of intravascular hemolysis in patients with active PNH and was generally well-tolerated. An update to the previously reported interim analysis will be provided. Upon successful completion of the 26-week treatment period, patients are enrolled into an open-label extension study. Disclosures Weyne: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Chaudhari: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Harari: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Miller: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Dain: Regeneron Pharmaceuticals, Inc.: Other: Contractor. Meagher: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Rodgers: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Perlee: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Morton: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Weinreich: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Yancopoulos: Regeneron Pharmaceuticals, Inc.: Current Employment, Current holder of stock options in a privately-held company. Griffin: Alexion, AstraZeneca Rare Disease: Honoraria, Membership on an entity's Board of Directors or advisory committees; BioCryst Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sobi Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; Apellis: Other: Educational grant support.
- Published
- 2021
10. Pozelimab, a Human Antibody Against Complement Factor C5, Demonstrates Robust Inhibition of Alternative Complement Activity Both in Normal Human Serum and in Phase I Normal Healthy Volunteers
- Author
-
Ming-Dauh Wang, Jonathan Weyne, Yan G. Ni, Cong Huang, Andrew J Rankin, Olivier Harari, Kishor Devalaraja-Narashimha, Morton Lori C, Umesh Chaudhari, and Srinivasa Prasad
- Subjects
business.industry ,Immunology ,Cell Biology ,Hematology ,Complement factor I ,Eculizumab ,Pharmacology ,medicine.disease ,Biochemistry ,Loading dose ,Hemolysis ,Atypical hemolytic uremic syndrome ,Paroxysmal nocturnal hemoglobinuria ,medicine ,Alternative complement pathway ,business ,Ex vivo ,medicine.drug - Abstract
INTRODUCTION: Blockade of complement factor C5 has demonstrated benefit in paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, generalized myasthenia gravis and neuromyelitis optica. We have completed a Phase I study of pozelimab, a fully human anti-C5 IgG4, in healthy volunteers. Pozelimab was well tolerated and resulted in dose-dependent inhibition of hemolytic activity through the classical complement pathway in normal healthy volunteers. Complete inhibition of hemolytic activity was maintained over a 4-week dosing period by a weekly subcutaneous (SC) regimen following an intravenous (IV) loading dose (ASH2018 abstract). OBJECTIVE: To further characterize the impact of pozelimab on the alternative complement pathway activity, we investigated the effect of pozelimab on alternative pathway-mediated hemolysis using an AH50 assay in the completed first-in-human (FIH) study. In addition, we compared the effect of pozelimab in both alternative and classical pathway hemolysis assays with those of in-house eculizumab and in-house ravulizumab in pooled normal human serum (NHS) samples, ex vivo. METHODS: In total, 56 subjects were randomized to 4 sequential ascending IV single dose cohorts plus 2 sequential ascending SC single dose cohorts followed by 1 multiple dose cohort (consisting of an IV loading dose and weekly SC doses). Each cohort consisted of 8 subjects randomized to receive pozelimab or placebo (6 active: 2 placebo). Serum collected at multiple time-points was utilized to assess the effect of pozelimab on alternative pathway activity. For ex vivo spike experiments, pooled NHS was used to compare the hemolytic function of pozelimab, in-house produced eculizumab and in-house produced ravulizumab. Comparator antibodies were synthesized from published sequence. The alternative pathway (AP) and classical pathway (CP) hemolysis assays were performed based on lysis of rabbit red blood cells (RBCs) and sensitized sheep RBCs, respectively. Both assays measure the amount of hemoglobin released from red blood cells at 412 nm. RESULTS: In the FIH study, baseline AH50 was comparable across treatment groups with a mean of 110 U/mL (standard deviation = 19, n = 56). Pozelimab exposure led to dose-dependent inhibition of AH50. In all 4 IV dosing cohorts, peak suppression of hemolysis was observed at end of infusion (EOI). Maximal suppression of hemolysis was approximately −85% change from baseline. This was achieved with the 30 mg/kg IV group and the repeat dose 15 mg/kg IV + 400 mg SC QW group. In the 2 SC cohorts, peak suppression of hemolysis was observed 3-7 days post dosing, which was consistent with observed peak concentrations of pozelimab in serum. In an ex vivo spike study, pozelimab, in-house eculizumab and in-house ravulizumab were spiked into 10, 25 or 48% pooled NHS for AP, and 5, 10 or 25% for CP. The results from AP hemolysis assays showed that, for a given concentration of spiked antibody, the maximal suppression of hemolysis for all the antibodies decreased with increased percentage of serum (Figure). The maximal suppression of hemolysis was consistently higher (32-169%) for pozelimab compared with in-house eculizumab, and lower for in-house ravulizumab compared with pozelimab and in-house eculizumab at all serum percentages tested. The results from CP hemolysis assays showed that, although the maximal suppression of hemolysis was similar for all antibodies tested, in-house ravulizumab was required to be at least a log higher in concentration to achieve a similar effect as the other two anti-C5 antibodies. CONCLUSIONS: Ex vivo studies with pooled NHS demonstrate that pozelimab robustly blocks both CP and AP hemolysis. In-house ravulizumab appeared to be less potent compared with in-house eculizumab in both CP and AP hemolysis assays. The Phase I healthy volunteer study of pozelimab demonstrated dose-dependent and significant inhibition of alternative pathway hemolysis, with the maximal suppression of hemolysis approximately −85% change from baseline. Figure Disclosures Devalaraja-Narashimha: Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Ni:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Huang:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Wang:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Chaudhari:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Prasad:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Harari:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Rankin:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Morton:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Weyne:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. OffLabel Disclosure: The drug is pozelimab and it is a fully human anti-C5 IgG4 being tested a Phase 1 study in healthy volunteers.
- Published
- 2019
11. A Randomized, Double-Blind, Placebo-Controlled Phase 1 Study of the Pharmacokinetics and Pharmacodynamics of REGN3918, a Human Antibody Against Complement Factor C5, in Healthy Volunteers
- Author
-
Ming-Dauh Wang, Melissa Simek-Lemos, Srinivasa Prasad, Olivier Harari, Ronda Rippley, Richard DelGizzi, Yan G. Ni, Stephen Godin, Andrew J Rankin, Jonathan Weyne, and Morton Lori C
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Placebo ,Biochemistry ,Gastroenterology ,Loading dose ,03 medical and health sciences ,Regimen ,030104 developmental biology ,Pharmacokinetics ,Tolerability ,Internal medicine ,Pharmacodynamics ,Cohort ,medicine ,Dosing ,business - Abstract
INTRODUCTION: Blockade of complement factor C5 has demonstrated benefit in Paroxysmal Nocturnal Hemoglobinuria (PNH), atypical Hemolytic Uremic Syndrome and Generalized Myasthenia Gravis. We developed a human IgG4P antibody, REGN3918, that binds with high affinity to wild-type and variant (R885H/C) human C5. REGN3918 was well-tolerated in monkey toxicology studies with up to 26 weeks of dosing at up to 100 mg/kg/week. This finding was supportive of conducting this first-in-human study of REGN3918 in healthy volunteers. OBJECTIVE: The primary objective of this ongoing study is to evaluate the safety and tolerability of single ascending intravenous (IV) and subcutaneous (SC) doses and a multiple dose regimen consisting of an IV loading dose plus multiple weekly SC doses of REGN3918 administered in healthy volunteers. The secondary objectives of the study are to assess the pharmacokinetic (PK) and pharmacodynamic (PD) profile of REGN3918. METHODS: 56 subjects were randomized to 4 sequential ascending IV dose cohorts plus 2 sequential ascending SC cohorts followed by 1 multiple dose cohort (consisting of an IV loading dose and weekly SC doses). Each cohort consisted of 8 subjects randomized to receive REGN3918 or placebo (6 active: 2 placebo). An adaptive design was implemented to allow for dose level and dosing interval adjustment utilizing in-study PK and PD measures. The PD profile of REGN3918 was assessed utilizing a sheep red blood cell complement activity assay (CH50 assay) as well as serum concentrations of total C5. REGN3918 was administered as follows:Cohort 1: 1 mg/kg IV, single doseCohort 2a: 3 mg/kg IV, single doseCohort 2b: 300 mg SC, single doseCohort 3a: 10 mg/kg IV, single doseCohort 3b: 600 mg SC, single doseCohort 4: 30 mg/kg IV, single doseCohort 5: Loading dose of 15 mg/kg IV followed by 4 repeat SC doses of 400 mg administered once weekly for four weeks. RESULTS: REGN3918 was found to be well tolerated in single doses of up to 30 mg/kg IV and 600 mg SC. The multiple dose Cohort 5 has completed dosing in all subjects and is currently in safety follow-up. Thus far, there has been one SAE, salpingitis in a subject with an intra-uterine contraceptive device. The SAE occurred in a Cohort 5 subject after completion of dosing and has since resolved. REGN3918 exhibited dose-dependent increases in exposure in serum, with a trend toward prolonged serum concentrations at IV doses ≥10 mg/kg. Following SC administration, concentrations of REGN3918 in serum peaked at 4 to 8 days post dose and bioavailability was estimated as approximately 70%. REGN3918 exposure led to dose-dependent inhibition of CH50. In all 4 IV dosing cohorts, suppression of hemolysis was observed at 15 min post-injection. Complete suppression of hemolysis was achieved with ≥ 3mg/kg dosing. At 30 mg/kg, complete suppression of hemolysis was maintained for >6 weeks, consistent with observed prolonged REGN3918 concentrations following this dose. In the 2 SC cohorts, peak suppression of hemolysis was observed 3-7 days post dosing, again consistent with observed peak concentrations of REGN3918 in serum. In the multiple dose cohort 5, complete suppression of CH50 was observed over the four-week dosing period. CONCLUSIONS: REGN3918 was well tolerated and resulted in dose-dependent inhibition of hemolytic activity in normal healthy volunteers. A single IV infusion of R3918, at 30 mg/kg, blocked hemolytic activity completely for >6 weeks. Complete inhibition of hemolytic activity was maintained over a 4-week dosing period by a weekly SC regimen following an IV loading dose. Disclosures Weyne: Regeneron Pharmaceuticals: Employment, Equity Ownership. Ni:Regeneron Pharmaceuticals: Employment, Equity Ownership. DelGizzi:Regeneron Pharmaceuticals: Employment, Equity Ownership. Godin:Regeneron Pharmaceuticals: Employment. Morton:Regeneron Pharmaceuticals: Employment, Equity Ownership. Prasad:Regeneron Pharmaceuticals: Employment, Equity Ownership. Rankin:Regeneron Pharmaceuticals: Employment, Equity Ownership. Simek-Lemos:Regeneron Pharmaceuticals: Employment, Equity Ownership. Wang:Regeneron Pharmaceuticals: Employment, Equity Ownership. Rippley:Regeneron Pharmaceuticals: Employment, Equity Ownership. Harari:Regeneron Pharmaceuticals, Inc.: Employment.
- Published
- 2018
12. Novel approaches for modeling C3 glomerulopathy in mouse and rat
- Author
-
George D. Yancopoulos, Scott M. MacDonnell, Jingjing Wang, Brian Zambrowicz, Dara Lorn, Gabor Halasz, Joseph F. Hickey, William Poueymirou, John O’Brien, Theodore J. Kaplan, William C. Olson, Andrew J. Murphy, Karoline A Meagher, Morton Lori C, Wojtek Auerbach, John Mcwhirter, Michael Roos, Rebecca Peyser, Yifan Luo, Cong Huang, Macdonald Lynn, Kishor Devalaraja-Narashimha, Sarah Casanova, Jeffery Lee, and Eric Chiao
- Subjects
Pathology ,medicine.medical_specialty ,Glomerulopathy ,Immunology ,medicine ,Biology ,medicine.disease ,Molecular Biology - Published
- 2018
13. A liver Hif-2 alpha-Irs2 pathway sensitizes hepatic insulin signaling and is modulated by Vegf inhibition
- Author
-
Mario Vallon, Calvin J. Kuo, Morton Lori C, Fabienne Rajas, Etienne Lefai, Kevin Wei, Gilles Mithieux, Owen P. McGuinness, Keith D. Anderson, Jenny Yuan, Cullen M. Taniguchi, Lisa M. McGinnis, Jacquelyn J. Maher, Carol W.M. Chan, M. Celeste Simon, Kimberly X. Mulligan, Gavin Thurston, Amato J. Giaccia, Stanley J. Wiegand, Stephanie M. Piecewicz, David Kuo, Justin P. Annes, Stanford University School of Medicine [CA, USA], RegenerRegeneron Pharmaceut Inc, Partenaires INRAE, Regeneron Pharmaceut Inc, Vanderbilt University [Nashville], Mécanisme Moléculaire du Diabète (MMD), Institut National de la Recherche Agronomique (INRA)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Pennsylvania, University of California [San Francisco] (UCSF), University of California, Université de Lyon (COMUE), Stanford Medical Scientist Training Program, NIGMS US National Institutes of Health (NIH) [GM-07365], Stanford Cardiovascular Institute [1K12HL087746], Molecular and Cellular Immunobiology Program [5T32AI07290], NIH American Recovery and Reinvestment Act Supplement [1R01HL074267], Radiological Society of North America Research Resident [1018, 1111], NIH [DK084206, RO1DK043748, P60 DK020593, U24DK059637], Molecular Endocrinology Training Program [T32DK007563], Cell Biology Core Facility, University of California-San Francisco Liver Center [P30 DK026743], Sydney Frank Foundation, Stanford Developmental Cancer Research Award [NIH R01HL074267, R01NS064517, R01CA158528], ProdInra, Migration, University of California [San Francisco] (UC San Francisco), and University of California (UC)
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,HYPOXIA ,HIF-1-ALPHA ,Biology ,Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,ANGIOGENESIS ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,LIPID-METABOLISM ,Internal medicine ,Insulin receptor substrate ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Insulin ,Glycogen synthase ,IN-VIVO ,030304 developmental biology ,GENE-EXPRESSION ,0303 health sciences ,Glycogen ,RECEPTOR ,General Medicine ,IRS2 ,Insulin oscillation ,Mice, Inbred C57BL ,[SDV] Life Sciences [q-bio] ,Insulin receptor ,Endocrinology ,Diabetes Mellitus, Type 2 ,Liver ,chemistry ,030220 oncology & carcinogenesis ,Lipogenesis ,Insulin Receptor Substrate Proteins ,biology.protein ,GROWTH ,GLUCOSE-HOMEOSTASIS ,RESISTANCE ,Signal Transduction - Abstract
International audience; Insulin initiates diverse hepatic metabolic responses, including gluconeogenic suppression and induction of glycogen synthesis and lipogenesis1,2. The liver possesses a rich sinusoidal capillary network with a higher degree of hypoxia and lower gluconeogenesis in the perivenous zone as compared to the rest of the organ3. Here, we show that diverse vascular endothelial growth factor (VEGF) inhibitors improved glucose tolerance in nondiabetic C57BL/6 and diabetic db/db mice, potentiating hepatic insulin signaling with lower gluconeogenic gene expression, higher glycogen storage and suppressed hepatic glucose production. VEGF inhibition induced hepatic hypoxia through sinusoidal vascular regression and sensitized liver insulin signaling through hypoxia-inducible factor-2 alpha (Hif-2 alpha, encoded by Epas1) stabilization. Notably, liverspecific constitutive activation of HIF-2 alpha, but not HIF-1 alpha, was sufficient to augment hepatic insulin signaling through direct and indirect induction of insulin receptor substrate-2 (Irs2), an essential insulin receptor adaptor protein4-6. Further, liver Irs2 was both necessary and sufficient to mediate Hif-2 alpha and Vegf inhibition effects on glucose tolerance and hepatic insulin signaling. These results demonstrate an unsuspected intersection between Hif-2 alpha-mediated hypoxic signaling and hepatic insulin action through Irs2 induction, which can be co-opted by Vegf inhibitors to modulate glucose metabolism. These studies also indicate distinct roles in hepatic metabolism for Hif-1 alpha, which promotes glycolysis(7-9), and Hif-2 alpha, which suppresses gluconeogenesis, and suggest new treatment approaches for type 2 diabetes mellitus.
- Published
- 2013
14. Murine Transfer Factors: Dose-Response Relationships and Routes of Administration
- Author
-
Kirkpatrick, Charles H., primary, Hamad, Abdel Rahim, additional, and Morton, Lori C., additional
- Published
- 1995
- Full Text
- View/download PDF
15. Advances and unmet needs in genetic, basic and clinical science in Alport syndrome: report from the 2015 International Workshop on Alport Syndrome.
- Author
-
Gross O, Kashtan CE, Rheault MN, Flinter F, Savige J, Miner JH, Torra R, Ars E, Deltas C, Savva I, Perin L, Renieri A, Ariani F, Mari F, Baigent C, Judge P, Knebelman B, Heidet L, Lagas S, Blatt D, Ding J, Zhang Y, Gale DP, Prunotto M, Xue Y, Schachter AD, Morton LCG, Blem J, Huang M, Liu S, Vallee S, Renault D, Schifter J, Skelding J, Gear S, Friede T, Turner AN, and Lennon R
- Subjects
- Animals, Collagen Type IV genetics, Genetic Therapy, Humans, Mutation, Needs Assessment, Nephritis, Hereditary therapy, Podocytes, Quality Improvement, Nephritis, Hereditary genetics
- Abstract
Alport syndrome (AS) is a genetic disease characterized by haematuric glomerulopathy variably associated with hearing loss and anterior lenticonus. It is caused by mutations in the COL4A3, COL4A4 or COL4A5 genes encoding the α3α4α5(IV) collagen heterotrimer. AS is rare, but it accounts for >1% of patients receiving renal replacement therapy. Angiotensin-converting enzyme inhibition slows, but does not stop, the progression to renal failure; therefore, there is an urgent requirement to expand and intensify research towards discovering new therapeutic targets and new therapies. The 2015 International Workshop on Alport Syndrome targeted unmet needs in basic science, genetics and diagnosis, clinical research and current clinical care. In three intensive days, more than 100 international experts including physicians, geneticists, researchers from academia and industry, and patient representatives from all over the world participated in panel discussions and breakout groups. This report summarizes the most important priority areas including (i) understanding the crucial role of podocyte protection and regeneration, (ii) targeting mutations by new molecular techniques for new animal models and potential gene therapy, (iii) creating optimal interaction between nephrologists and geneticists for early diagnosis, (iv) establishing standards for mutation screening and databases, (v) improving widespread accessibility to current standards of clinical care, (vi) improving collaboration with the pharmaceutical/biotech industry to investigate new therapies, (vii) research in hearing loss as a huge unmet need in Alport patients and (viii) the need to evaluate the risk and benefit of novel (including 'repurposing') therapies on an international basis., (© The Author 2016. Published by Oxford University Press on behalf of ERAEDTA.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.