Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-34799399

ABSTRACT

BACKGROUND: NIZ985 is a recombinant heterodimer of physiologically active interleukin (IL-)15 and IL-15 receptor alpha. In preclinical models, NIZ985 promotes cytotoxic lymphocyte proliferation, killing function, and organ/tumor infiltration, with resultant anticancer effects. In this first-in-human study, we assessed the safety, pharmacokinetics, and immune effects of NIZ985 in patients with metastatic or unresectable solid tumors. METHODS: Single agent NIZ985 dose escalation data are reported from a phase I dose escalation/expansion study of NIZ985 as monotherapy. Adult patients (N=14) received 0.25, 0.5, 1, 2 or 4 µg/kg subcutaneous NIZ985 three times weekly (TIW) for the first 2 weeks of each 28-day cycle, in an accelerated 3+3 dose escalation trial design. IL-15 and endogenous cytokines were monitored by ELISA and multiplexed electrochemiluminescent assays. Multiparameter flow cytometry assessed the frequency, phenotype and proliferation of peripheral blood mononuclear cells. Preliminary antitumor activity was assessed by overall response rate (Response Evaluation Criteria in Solid Tumors V.1.1). RESULTS: As of March 2, 2020, median treatment duration was 7.5 weeks (range 1.1-77.1). Thirteen patients had discontinued and one (uveal melanoma) remains on treatment with stable disease. Best clinical response was stable disease (3 of 14 patients; 21%). The most frequent adverse events (AEs) were circular erythematous injection site reactions (100%), chills (71%), fatigue (57%), and fever (50%). Treatment-related grade 3/4 AEs occurred in six participants (43%); treatment-related serious AEs (SAEs) in three (21%). The per-protocol maximum tolerated dose was not reached. Pharmacokinetic accumulation of serum IL-15 in the first week was followed by significantly lower levels in week 2, likely due to more rapid cytokine consumption by an expanding lymphocyte pool. NIZ985 treatment was associated with increases in several cytokines, including interferon (IFN)-γ, IL-18, C-X-C motif chemokine ligand 10, and tumor necrosis factor-ß, plus significant induction of cytotoxic lymphocyte proliferation (including natural killer and CD8+ T cells), increased CD16+ monocytes, and increased CD163+ macrophages at injection sites. CONCLUSIONS: Subcutaneous NIZ985 TIW was generally well tolerated in patients with advanced cancer and produced immune activation paralleling preclinical observations, with induction of IFN-γ and proliferation of cytotoxic lymphocytes. Due to delayed SAEs at the two highest dose levels, administration is being changed to once-weekly in a revised protocol, as monotherapy and combined with checkpoint inhibitor spartalizumab. These alterations are expected to maximize the potential of NIZ985 as a novel immunotherapy. TRIAL REGISTRATION NUMBER: NCT02452268.


Subject(s)
Interleukin-15/administration & dosage , Interleukin-15/agonists , Neoplasms/drug therapy , Receptors, Interleukin-15/administration & dosage , Adult , Aged , Female , Humans , Immunotherapy , Male , Middle Aged , Neoplasm Metastasis , Protein Multimerization , Recombinant Proteins/administration & dosage
2.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32073845

ABSTRACT

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.


Subject(s)
Benzoic Acid/chemistry , Complement Factor B/antagonists & inhibitors , Indoles/chemistry , Atypical Hemolytic Uremic Syndrome/metabolism , Atypical Hemolytic Uremic Syndrome/pathology , Benzoic Acid/metabolism , Benzoic Acid/pharmacokinetics , Binding Sites , Catalytic Domain , Complement Factor B/metabolism , Crystallography, X-Ray , Drug Evaluation, Preclinical , Half-Life , Humans , Indoles/metabolism , Indoles/pharmacokinetics , Inhibitory Concentration 50 , Macular Degeneration/metabolism , Macular Degeneration/pathology , Molecular Dynamics Simulation , Structure-Activity Relationship
3.
J Med Chem ; 62(9): 4656-4668, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30995036

ABSTRACT

Complement factor D (FD), a highly specific S1 serine protease, plays a central role in the amplification of the alternative complement pathway (AP) of the innate immune system. Dysregulation of AP activity predisposes individuals to diverse disorders such as age-related macular degeneration, atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis type II, and paroxysmal nocturnal hemoglobinuria. Previously, we have reported the screening efforts and identification of reversible benzylamine-based FD inhibitors (1 and 2) binding to the open active conformation of FD. In continuation of our drug discovery program, we designed compounds applying structure-based approaches to improve interactions with FD and gain selectivity against S1 serine proteases. We report herein the design, synthesis, and medicinal chemistry optimization of the benzylamine series culminating in the discovery of 12, an orally bioavailable and selective FD inhibitor. 12 demonstrated systemic suppression of AP activation in a lipopolysaccharide-induced AP activation model as well as local ocular suppression in intravitreal injection-induced AP activation model in mice expressing human FD.


Subject(s)
Benzylamines/pharmacology , Complement Pathway, Alternative/drug effects , Serine Proteinase Inhibitors/pharmacology , Animals , Benzylamines/chemical synthesis , Benzylamines/metabolism , Binding Sites , Complement Factor D/antagonists & inhibitors , Complement Factor D/chemistry , Complement Factor D/metabolism , Dogs , Drug Design , Humans , Mice, Inbred C57BL , Mice, Transgenic , Molecular Docking Simulation , Protein Conformation , Rats , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/metabolism
4.
J Med Chem ; 61(4): 1622-1635, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29400470

ABSTRACT

A noninvasive topical ocular therapy for the treatment of neovascular or "wet" age-related macular degeneration would provide a patient administered alternative to the current standard of care, which requires physician administered intravitreal injections. This manuscript describes a novel strategy for the use of in vivo models of choroidal neovascularization (CNV) as the primary means of developing SAR related to efficacy from topical administration. Ultimately, this effort led to the discovery of acrizanib (LHA510), a small-molecule VEGFR-2 inhibitor with potency and efficacy in rodent CNV models, limited systemic exposure after topical ocular administration, multiple formulation options, and an acceptable rabbit ocular PK profile.


Subject(s)
Administration, Topical , Indoles/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Wet Macular Degeneration/drug therapy , Animals , Choroidal Neovascularization , Drug Discovery , Indoles/pharmacokinetics , Indoles/therapeutic use , Ophthalmic Solutions , Protein Kinase Inhibitors , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Rabbits , Rodentia , Structure-Activity Relationship
5.
SLAS Discov ; 22(10): 1239-1245, 2017 12.
Article in English | MEDLINE | ID: mdl-28787579

ABSTRACT

Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) is becoming an important technology to determine the distribution of drugs and their metabolites in the tissue of preclinical species after dosing. Interest in IMS is growing in the ophthalmology field, but little work to this point has been done to investigate ocular drug transit using this technology. Information on where and how a drug is distributing through the eye is important in understanding efficacy and whether it is reaching the desired target tissue. For this study, ocular distribution of brimonidine was investigated in rabbits following topical administration. Brimonidine has been shown to lower intraocular pressure and is approved to treat glaucoma, the second leading cause of blindness in the world. We have developed IMS methods to assess transit of topically administered brimonidine from the anterior to the posterior segment of rabbit eyes. Using IMS, brimonidine was detected in the cornea, aqueous humor, iris, and posterior segments of the eye. The distribution of brimonidine suggests that the route of transit following topical administration is mainly through the uvea-scleral route. This study demonstrates that IMS can be applied to assess ocular transit and distribution of topically administered drugs.


Subject(s)
Brimonidine Tartrate/pharmacology , Eye/drug effects , Imaging, Three-Dimensional , Mass Spectrometry , Animals , Rabbits , Signal Processing, Computer-Assisted , Time Factors
6.
Nat Commun ; 8: 14837, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332616

ABSTRACT

Protein drugs that neutralize vascular endothelial growth factor (VEGF), such as aflibercept or ranibizumab, rescue vision in patients with retinal vascular diseases. Nonetheless, optimal visual outcomes require intraocular injections as frequently as every month. Here we report a method to extend the intravitreal half-life of protein drugs as an alternative to either encapsulation or chemical modifications with polymers. We combine a 97-amino-acid peptide of human origin that binds hyaluronan, a major macromolecular component of the eye's vitreous, with therapeutic antibodies and proteins. When administered to rabbit and monkey eyes, the half-life of the modified proteins is increased ∼3-4-fold relative to unmodified proteins. We further show that prototype long-acting anti-VEGF drugs (LAVAs) that include this peptide attenuate VEGF-induced retinal changes in animal models of neovascular retinal disease ∼3-4-fold longer than unmodified drugs. This approach has the potential to reduce the dosing frequency associated with retinal disease treatments.


Subject(s)
Bevacizumab/administration & dosage , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Retinal Diseases/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Bevacizumab/chemistry , Bevacizumab/pharmacokinetics , Disease Models, Animal , Female , Half-Life , Humans , Hyaluronic Acid/chemistry , Intravitreal Injections , Macaca fascicularis , Male , Rabbits , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Retinal Diseases/metabolism
7.
J Med Chem ; 58(23): 9273-86, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26568411

ABSTRACT

The benefit of intravitreal anti-VEGF therapy in treating wet age-related macular degeneration (AMD) is well established. Identification of VEGFR-2 inhibitors with optimal ADME properties for an ocular indication provides opportunities for dosing routes beyond intravitreal injection. We employed a high-throughput in vivo screening strategy with rodent models of choroidal neovascularization and iterative compound design to identify VEGFR-2 inhibitors with potential to benefit wet AMD patients. These compounds demonstrate preferential ocular tissue distribution and efficacy after oral administration while minimizing systemic exposure.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Wet Macular Degeneration/drug therapy , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Animals , Choroid/drug effects , Choroid/pathology , Choroidal Neovascularization/pathology , Female , Humans , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Rats , Wet Macular Degeneration/pathology
8.
J Med Chem ; 56(13): 5464-72, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23738526

ABSTRACT

Glaucoma is a leading cause of vision loss and blindness, with increased intraocular pressure (IOP) a prominent risk factor. IOP can be efficaciously reduced by administration of topical agents. However, the repertoire of approved IOP-lowering drug classes is limited, and effective new alternatives are needed. Agonism of the cannabinoid receptors CB1/2 significantly reduces IOP clinically and experimentally. However, development of CB1/2 agonists has been complicated by the need to avoid cardiovascular and psychotropic side effects. 1 is a potent CB1/2 agonist that is highly excluded from the brain. In a phase I study, compound 1 eyedrops were well tolerated and generated an IOP-lowering trend but were limited in dose and exposure due to poor solubility and ocular absorption. Here we present an innovative strategy to rapidly identify compound 1 prodrugs that are efficiently metabolized to the parent compound for improved solubility and ocular permeability while maintaining low systemic exposures.


Subject(s)
Ophthalmic Solutions/pharmacology , Prodrugs/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Area Under Curve , Eye/metabolism , Eye/physiopathology , Glaucoma/metabolism , Glaucoma/physiopathology , Glaucoma/prevention & control , Humans , Intraocular Pressure/drug effects , Male , Metabolic Clearance Rate , Models, Chemical , Molecular Structure , Ophthalmic Solutions/chemical synthesis , Ophthalmic Solutions/pharmacokinetics , Permeability , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
9.
J Biol Chem ; 286(52): 44965-75, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22072717

ABSTRACT

Pathological neovascularization occurs when a balance of pro- and anti-angiogenic factors is disrupted, accompanied by an amplifying inflammatory cascade. However, the interdependence of these responses and the mechanism triggering the initial angiogenic switch have remained unclear. We present data from an epithelial debridement model of corneal neovascularization describing an initial 3-day period when a substantial component of neovascular growth occurs. Administration of selective inhibitors shows that this initial growth requires signaling through VEGFR-2 (vascular endothelial growth factor receptor-2), independent of the accompanying inflammatory response. Instead, increased VEGF production is found prominently in repair epithelial cells and is increased prior to recruitment of neutrophil/granulocytes and macrophage/monocytes. Consequently, early granulocyte and monocyte depletion has little effect on corneal neovascularization outgrowth. These data indicate that it is possible to pharmacologically uncouple these mechanisms during early injury-driven neovascularization in the cornea and suggest that initial tissue responses are coordinated by repair epithelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Epithelium/metabolism , Animals , Cornea/metabolism , Cornea/pathology , Corneal Neovascularization/pathology , Epithelium/pathology , Female , Macrophages/metabolism , Macrophages/pathology , Mice , Monocytes/metabolism , Monocytes/pathology , Neutrophils/metabolism , Neutrophils/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...