Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
3.
Nat Commun ; 14(1): 5053, 2023 08 19.
Article En | MEDLINE | ID: mdl-37598178

Brain exposure of systemically administered biotherapeutics is highly restricted by the blood-brain barrier (BBB). Here, we report the engineering and characterization of a BBB transport vehicle targeting the CD98 heavy chain (CD98hc or SLC3A2) of heterodimeric amino acid transporters (TVCD98hc). The pharmacokinetic and biodistribution properties of a CD98hc antibody transport vehicle (ATVCD98hc) are assessed in humanized CD98hc knock-in mice and cynomolgus monkeys. Compared to most existing BBB platforms targeting the transferrin receptor, peripherally administered ATVCD98hc demonstrates differentiated brain delivery with markedly slower and more prolonged kinetic properties. Specific biodistribution profiles within the brain parenchyma can be modulated by introducing Fc mutations on ATVCD98hc that impact FcγR engagement, changing the valency of CD98hc binding, and by altering the extent of target engagement with Fabs. Our study establishes TVCD98hc as a modular brain delivery platform with favorable kinetic, biodistribution, and safety properties distinct from previously reported BBB platforms.


Blood-Brain Barrier , Brain , Animals , Mice , Tissue Distribution , Antibodies , Engineering , Macaca fascicularis
4.
J Exp Med ; 219(3)2022 03 07.
Article En | MEDLINE | ID: mdl-35226042

Delivery of biotherapeutics across the blood-brain barrier (BBB) is a challenge. Many approaches fuse biotherapeutics to platforms that bind the transferrin receptor (TfR), a brain endothelial cell target, to facilitate receptor-mediated transcytosis across the BBB. Here, we characterized the pharmacological behavior of two distinct TfR-targeted platforms fused to iduronate 2-sulfatase (IDS), a lysosomal enzyme deficient in mucopolysaccharidosis type II (MPS II), and compared the relative brain exposures and functional activities of both approaches in mouse models. IDS fused to a moderate-affinity, monovalent TfR-binding enzyme transport vehicle (ETV:IDS) resulted in widespread brain exposure, internalization by parenchymal cells, and significant substrate reduction in the CNS of an MPS II mouse model. In contrast, IDS fused to a standard high-affinity bivalent antibody (IgG:IDS) resulted in lower brain uptake, limited biodistribution beyond brain endothelial cells, and reduced brain substrate reduction. These results highlight important features likely to impact the clinical development of TfR-targeting platforms in MPS II and potentially other CNS diseases.


Iduronate Sulfatase , Mucopolysaccharidosis II , Receptors, Transferrin , Recombinant Fusion Proteins , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Iduronate Sulfatase/metabolism , Iduronate Sulfatase/pharmacology , Lysosomes/metabolism , Mice , Mucopolysaccharidosis II/metabolism , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tissue Distribution
5.
Cell ; 184(18): 4651-4668.e25, 2021 09 02.
Article En | MEDLINE | ID: mdl-34450028

GRN mutations cause frontotemporal dementia (GRN-FTD) due to deficiency in progranulin (PGRN), a lysosomal and secreted protein with unclear function. Here, we found that Grn-/- mice exhibit a global deficiency in bis(monoacylglycero)phosphate (BMP), an endolysosomal phospholipid we identified as a pH-dependent PGRN interactor as well as a redox-sensitive enhancer of lysosomal proteolysis and lipolysis. Grn-/- brains also showed an age-dependent, secondary storage of glucocerebrosidase substrate glucosylsphingosine. We investigated a protein replacement strategy by engineering protein transport vehicle (PTV):PGRN-a recombinant protein linking PGRN to a modified Fc domain that binds human transferrin receptor for enhanced CNS biodistribution. PTV:PGRN rescued various Grn-/- phenotypes in primary murine macrophages and human iPSC-derived microglia, including oxidative stress, lysosomal dysfunction, and endomembrane damage. Peripherally delivered PTV:PGRN corrected levels of BMP, glucosylsphingosine, and disease pathology in Grn-/- CNS, including microgliosis, lipofuscinosis, and neuronal damage. PTV:PGRN thus represents a potential biotherapeutic for GRN-FTD.


Biological Products/therapeutic use , Brain/metabolism , Lysosomal Storage Diseases/therapy , Progranulins/therapeutic use , Animals , Bone Morphogenetic Proteins/metabolism , Endosomes/metabolism , Female , Frontotemporal Dementia/blood , Frontotemporal Dementia/cerebrospinal fluid , Gliosis/complications , Gliosis/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/pathology , Lipid Metabolism , Lipofuscin/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Nerve Degeneration/pathology , Phenotype , Progranulins/deficiency , Progranulins/metabolism , Receptors, Immunologic/metabolism , Receptors, Transferrin/metabolism , Tissue Distribution
6.
Clin Cancer Res ; 27(15): 4435-4448, 2021 08 01.
Article En | MEDLINE | ID: mdl-34011561

PURPOSE: Ovarian cancer represents a major clinical hurdle for immune checkpoint blockade (ICB), with reported low patient response rates. We found that the immune checkpoint ligand PD-L2 is robustly expressed in patient samples of ovarian cancers and other malignancies exhibiting suboptimal response to ICB but not in cancers that are ICB sensitive. Therefore, we hypothesize that PD-L2 can facilitate immune escape from ICB through incomplete blockade of the PD-1 signaling pathway. EXPERIMENTAL DESIGN: We engineered a soluble form of the PD-1 receptor (sPD-1) capable of binding and neutralizing both PD-L2 and PD-L1 with ×200 and ×10,000 folds improvement in binding affinity over wild-type PD-1 leading to superior inhibition of ligand-mediated PD-1 activities. RESULTS: Both in vitro and in vivo analyses performed in this study demonstrated that the high-affinity sPD-1 molecule is superior at blocking both PD-L1- and PD-L2-mediated immune evasion and reducing tumor growth in immune-competent murine models of ovarian cancer. CONCLUSIONS: The data presented in this study provide justification for using a dual targeting, high-affinity sPD-1 receptor as an alternative to PD-1 or PD-L1 therapeutic antibodies for achieving superior therapeutic efficacy in cancers expressing both PD-L2 and PD-L1.


Immune Checkpoint Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Animals , Drug Resistance, Neoplasm , Female , Humans , Mice
7.
Sci Transl Med ; 12(545)2020 05 27.
Article En | MEDLINE | ID: mdl-32461331

Most lysosomal storage diseases (LSDs) involve progressive central nervous system (CNS) impairment, resulting from deficiency of a lysosomal enzyme. Treatment of neuronopathic LSDs remains a considerable challenge, as approved intravenously administered enzyme therapies are ineffective in modifying CNS disease because they do not effectively cross the blood-brain barrier (BBB). We describe a therapeutic platform for increasing the brain exposure of enzyme replacement therapies. The enzyme transport vehicle (ETV) is a lysosomal enzyme fused to an Fc domain that has been engineered to bind to the transferrin receptor, which facilitates receptor-mediated transcytosis across the BBB. We demonstrate that ETV fusions containing iduronate 2-sulfatase (ETV:IDS), the lysosomal enzyme deficient in mucopolysaccharidosis type II, exhibited high intrinsic activity and degraded accumulated substrates in both IDS-deficient cell and in vivo models. ETV substantially improved brain delivery of IDS in a preclinical model of disease, enabling enhanced cellular distribution to neurons, astrocytes, and microglia throughout the brain. Improved brain exposure for ETV:IDS translated to a reduction in accumulated substrates in these CNS cell types and peripheral tissues and resulted in a complete correction of downstream disease-relevant pathologies in the brain, including secondary accumulation of lysosomal lipids, perturbed gene expression, neuroinflammation, and neuroaxonal damage. These data highlight the therapeutic potential of the ETV platform for LSDs and provide preclinical proof of concept for TV-enabled therapeutics to treat CNS diseases more broadly.


Blood-Brain Barrier , Iduronate Sulfatase , Animals , Brain , Disease Models, Animal , Enzyme Replacement Therapy , Lysosomes , Mice
8.
Sci Transl Med ; 12(545)2020 05 27.
Article En | MEDLINE | ID: mdl-32461332

Effective delivery of protein therapeutics to the central nervous system (CNS) has been greatly restricted by the blood-brain barrier (BBB). We describe the development of a BBB transport vehicle (TV) comprising an engineered Fc fragment that exploits receptor-mediated transcytosis for CNS delivery of biotherapeutics by binding a highly expressed brain endothelial cell target. TVs were engineered using directed evolution to bind the apical domain of the human transferrin receptor (hTfR) without the use of amino acid insertions, deletions, or unnatural appendages. A crystal structure of the TV-TfR complex revealed the TV binding site to be away from transferrin and FcRn binding sites, which was further confirmed experimentally in vitro and in vivo. Recombinant expression of TVs fused to anti-ß-secretase (BACE1) Fabs yielded antibody transport vehicle (ATV) molecules with native immunoglobulin G (IgG) structure and stability. Peripheral administration of anti-BACE1 ATVs to hTfR-engineered mice and cynomolgus monkeys resulted in substantially improved CNS uptake and sustained pharmacodynamic responses. The TV platform readily accommodates numerous additional configurations, including bispecific antibodies and protein fusions, yielding a highly modular CNS delivery platform.


Amyloid Precursor Protein Secretases , Blood-Brain Barrier , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Haplorhini/metabolism , Immunoglobulin Fc Fragments , Mice , Receptors, Transferrin/metabolism
9.
J Clin Invest ; 127(1): 183-198, 2017 01 03.
Article En | MEDLINE | ID: mdl-27893463

The AXL receptor and its activating ligand, growth arrest-specific 6 (GAS6), are important drivers of metastasis and therapeutic resistance in human cancers. Given the critical roles that GAS6 and AXL play in refractory disease, this signaling axis represents an attractive target for therapeutic intervention. However, the strong picomolar binding affinity between GAS6 and AXL and the promiscuity of small molecule inhibitors represent important challenges faced by current anti-AXL therapeutics. Here, we have addressed these obstacles by engineering a second-generation, high-affinity AXL decoy receptor with an apparent affinity of 93 femtomolar to GAS6. Our decoy receptor, MYD1-72, profoundly inhibited disease progression in aggressive preclinical models of human cancers and induced cell killing in leukemia cells. When directly compared with the most advanced anti-AXL small molecules in the clinic, MYD1-72 achieved superior antitumor efficacy while displaying no toxicity. Moreover, we uncovered a relationship between AXL and the cellular response to DNA damage whereby abrogation of AXL signaling leads to accumulation of the DNA-damage markers γH2AX, 53BP1, and RAD51. MYD1-72 exploited this relationship, leading to improvements upon the therapeutic index of current standard-of-care chemotherapies in preclinical models of advanced pancreatic and ovarian cancer.


Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia/drug therapy , Neoplasms, Experimental/drug therapy , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Histones/genetics , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Leukemia/metabolism , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
10.
ACS Chem Biol ; 12(2): 336-341, 2017 02 17.
Article En | MEDLINE | ID: mdl-27997117

Affinity maturation of protein-protein interactions requires iterative rounds of protein library generation and high-throughput screening to identify variants that bind with increased affinity to a target of interest. We recently developed a multipurpose protein engineering platform, termed µSCALE (Microcapillary Single Cell Analysis and Laser Extraction). This technology enables high-throughput screening of libraries of millions of cell-expressing protein variants based on their binding properties or functional activity. Here, we demonstrate the first use of the µSCALE platform for affinity maturation of a protein-protein binding interaction. In this proof-of-concept study, we engineered an extracellular domain of the Axl receptor tyrosine kinase to bind tighter to its ligand Gas6. Within 2 weeks, two iterative rounds of library generation and screening resulted in engineered Axl variants with a 50-fold decrease in kinetic dissociation rate, highlighting the use of µSCALE as a new tool for directed evolution.


Protein Engineering , Proteins/metabolism , Protein Binding , Saccharomyces cerevisiae/metabolism , Single-Cell Analysis
11.
Nat Commun ; 7: 13898, 2016 12 23.
Article En | MEDLINE | ID: mdl-28008921

Increasing evidence suggests that ionizing radiation therapy (RT) in combination with checkpoint immunotherapy is highly effective in treating a subset of cancers. To better understand the limited responses to this combination we analysed the genetic, microenvironmental, and immune factors in tumours derived from a transgenic breast cancer model. We identified two tumours with similar growth characteristics but different RT responses primarily due to an antitumour immune response. The combination of RT and checkpoint immunotherapy resulted in cures in the responsive but not the unresponsive tumours. Profiling the tumours revealed that the Axl receptor tyrosine kinase is overexpressed in the unresponsive tumours, and Axl knockout resulted in slower growth and increased radiosensitivity. These changes were associated with a CD8+ T-cell response, which was improved in combination with checkpoint immunotherapy. These results suggest a novel role for Axl in suppressing antigen presentation through MHCI, and enhancing cytokine release, which promotes a suppressive myeloid microenvironment.


Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Animals , Cell Line, Tumor , Cell Proliferation , Combined Modality Therapy , Cytokines/metabolism , Histocompatibility Antigens Class I/metabolism , Immunity , Immunosuppression Therapy , Immunotherapy , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Radiation Tolerance/radiation effects , Axl Receptor Tyrosine Kinase
12.
Nat Chem Biol ; 10(11): 977-83, 2014 Nov.
Article En | MEDLINE | ID: mdl-25242553

Aberrant signaling through the Axl receptor tyrosine kinase has been associated with a myriad of human diseases, most notably metastatic cancer, identifying Axl and its ligand Gas6 as important therapeutic targets. Using rational and combinatorial approaches, we engineered an Axl 'decoy receptor' that binds Gas6 with high affinity and inhibits its function, offering an alternative approach from drug discovery efforts that directly target Axl. Four mutations within this high-affinity Axl variant caused structural alterations in side chains across the Gas6-Axl binding interface, stabilizing a conformational change on Gas6. When reformatted as an Fc fusion, the engineered decoy receptor bound Gas6 with femtomolar affinity, an 80-fold improvement compared to binding of the wild-type Axl receptor, allowing effective sequestration of Gas6 and specific abrogation of Axl signaling. Moreover, increased Gas6 binding affinity was critical and correlative with the ability of decoy receptors to potently inhibit metastasis and disease progression in vivo.


Genetic Engineering , Immunoglobulin Fc Fragments/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/pharmacology , Signal Transduction/drug effects , Animals , Binding Sites , Disease Progression , Dose-Response Relationship, Drug , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/isolation & purification , Immunoglobulin Fc Fragments/therapeutic use , Intercellular Signaling Peptides and Proteins/chemistry , Mice , Models, Molecular , Mutation/genetics , Neoplasm Metastasis/drug therapy , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Binding/drug effects , Protein Binding/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship , Axl Receptor Tyrosine Kinase
13.
Proc Natl Acad Sci U S A ; 111(37): 13373-8, 2014 Sep 16.
Article En | MEDLINE | ID: mdl-25187556

Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/secondary , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Cell Hypoxia , Cell Line, Tumor , Enzyme Activation , Hepatocyte Growth Factor/pharmacology , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Models, Biological , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Mas , Signal Transduction , Treatment Outcome , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Axl Receptor Tyrosine Kinase
14.
Curr Opin Biotechnol ; 24(6): 1072-7, 2013 Dec.
Article En | MEDLINE | ID: mdl-23587963

Protein-based biologics, which leverage the inherent affinity and specificity of protein-protein interactions, offer an effective strategy for targeting and modulating disease pathways. Despite the broad diversity of the proteome, monoclonal antibodies have been the major focus of such drug discovery efforts. While antibodies have shown great clinical value, the breadth and complexity of human disease highlight the need for alternatives that expand the therapeutic repertoire beyond this single class of proteins. The elucidation of molecular mechanisms underlying human disease has provided new opportunities for protein-based drugs to address challenging clinical problems. Natural ligands and receptors, which inherently modulate complex biological processes, have emerged as promising candidates for protein-based drug discovery efforts. Protein engineering strategies, guided by biological principles, are allowing ligands and receptors to be developed as next-generation therapeutics with improved safety and efficacy.


Drug Discovery/methods , Protein Engineering/methods , Proteins/therapeutic use , Antibodies, Monoclonal/therapeutic use , Drug Discovery/trends , Humans , Ligands , Protein Engineering/trends , Proteins/adverse effects , Proteins/metabolism , Receptor Cross-Talk/drug effects , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/therapeutic use
15.
J Mol Recognit ; 24(1): 127-35, 2011.
Article En | MEDLINE | ID: mdl-21194123

A truncated form of the Agouti-related protein (AgRP), a member of the cystine-knot family, has shown promise as a scaffold for engineering novel peptides with new molecular recognition properties. In this study, we replaced a constrained six amino acid loop in AgRP with a nine amino acid loop containing an Arg-Gly-Asp integrin recognition motif, and randomized the neighboring residues to create a library of approximately 20 million AgRP variants. We displayed the AgRP mutants as fusions on the surface of yeast and used high-throughput fluorescence-activated cell sorting (FACS) to isolate peptides that bound specifically to the platelet integrin α(IIb)ß(3), a clinically important target for the prevention and treatment of thrombosis. These AgRP peptides had equilibrium dissociation (K(D)) constants for α(IIb)ß(3) integrin ranging from 60 to 90 nM, and did not bind to α(v)ß(3), α(v)ß(5), or α(5)ß(1) integrins. Using an alternate library screening strategy, we identified AgRP peptides that bound to both α(IIb)ß(3) and α(v)ß(3) integrins with K(D) values ranging from 40 to 70 nM and 20 to 30 nM, respectively, and did not bind to α(v)ß(5) or α(5)ß(1) integrins. Unique consensus sequences were identified within both series of AgRP peptides suggesting alternative molecular recognition events that dictate different integrin binding specificities. In addition, the engineered AgRP peptides prevented platelet aggregation as well as or slightly better than the FDA-approved cyclic peptide eptifibatide. Collectively, these data demonstrate that cystine-knot peptides can be generated with high affinity and specificity to closely-related integrins, and provide insights into molecular interactions between small, structured peptide ligands and their receptors.


Agouti-Related Protein/genetics , Agouti-Related Protein/pharmacology , Blood Platelets/drug effects , Integrins/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Agouti-Related Protein/chemistry , Agouti-Related Protein/metabolism , Amino Acid Sequence , Humans , K562 Cells , Mutant Proteins , Peptide Library , Pichia/genetics , Pichia/metabolism , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
...