[go: up one dir, main page]

WO2025049603A1 - Procédés et compositions pour réduire la réponse à un corps étranger - Google Patents

Procédés et compositions pour réduire la réponse à un corps étranger Download PDF

Info

Publication number
WO2025049603A1
WO2025049603A1 PCT/US2024/044222 US2024044222W WO2025049603A1 WO 2025049603 A1 WO2025049603 A1 WO 2025049603A1 US 2024044222 W US2024044222 W US 2024044222W WO 2025049603 A1 WO2025049603 A1 WO 2025049603A1
Authority
WO
WIPO (PCT)
Prior art keywords
spp1
active agent
capsule
adm
foreign body
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/044222
Other languages
English (en)
Inventor
Michelle F. GRIFFIN
Norah LIANG
Jennifer B.L. PARKER
Ruth TEVLIN
Emily MEANY
Michael T. Longaker
Arash MOMENI
Derrick C. WAN
Eric A. APPEL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leland Stanford Junior University
Original Assignee
Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Leland Stanford Junior University filed Critical Leland Stanford Junior University
Publication of WO2025049603A1 publication Critical patent/WO2025049603A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels

Definitions

  • FBR foreign body response
  • ADM acellular dermal matrix
  • ECM extracellular matrix
  • the present disclosure provides methods for reducing foreign body response (FBR) in a subject. Aspects of the methods of reducing FBR in a subject include implanting a foreign body in the subject in combination with an effective amount of an SPP1 active agent to reduce FBR in the subject.
  • the SPP1 active agent comprises an SPP1 polypeptide.
  • the SPP1 active agent is a recombinant SPP1 polypeptide.
  • the SPP1 active agent is present in an SPP1 active agent composition.
  • the SPP1 active agent composition comprises a controlled release composition.
  • the SPP1 active agent composition comprises a gel. In some cases, the gel is a hydrogel.
  • the hydrogel comprises a polymer.
  • the polymer comprises a polysaccharide.
  • the polysaccharide comprises hydroxypropyl methylcellulose.
  • the SPP1 active agent composition comprises a nanoparticle.
  • the nanoparticle comprises a diblock copolymer.
  • the diblock copolymer comprises polyethylene glycol) and poly(lactic acid).
  • the foreign body is coated with a SPP1 active agent composition of the present disclosure. In some cases, the foreign body is an implantable device.
  • the methods of the present disclosure produce an implant with a reduced foreign body capsule thickness around the implant compared to a control.
  • the foreign body capsule has reduced collagen density compared to a control.
  • the foreign body capsule has increased elastin density compared to a control.
  • the foreign body capsule has increased elasticity compared to a control.
  • the subject is a mammal. In some cases, the subject is a human. In some cases, the human is an adult. In some cases, the subject is a mouse.
  • the present disclosure additionally provides implantable devices coated with a SPP1 active agent composition of the present disclosure. Also provided are kits including an implantable device and an SPP1 active agent composition of the present disclosure.
  • Figures 1A-1 F Acellular Dermal Matrix (ADM) alters extracellular matrix ultrastructure in human capsule specimens and leads to a reduction in fibrotic encapsulation.
  • A Schematic of paired capsule specimen retrieval obtained from human patients.
  • B Hematoxylin and eosin (H&E) staining of ADM (left) and Native (right) capsule. Black dotted regions indicate areas from which magnified images were captured.
  • C Trichrome staining of ADM and Native capsule. Quantification of collagen density in Native and ADM capsule (right).
  • ECM extracellular matrix
  • Elastin staining of ADM and Native capsule left. Quantification of percentage of elastin+ staining in ADM and Native capsule (right).
  • F Schematic of Luminex workflow (left).
  • Luminex secretion assay comparing median fluorescent intensity (MFI) of Stromal cell-derived factor 1 A and B (SDF1A+B), C-X-C motif chemokine ligand 10 (CXCL10), and lnterleukin-1 alpha (IL-1 a) secretion, between Native and ADM capsule (right).
  • Linked data points represent Native and ADM capsules retrieved from the same patient.
  • C, E Data shown as mean ⁇ standard deviation (S.D.).
  • FIGS 2A-2H Single-cell transcriptomic analyses reveal changes in myeloid cell dynamics during foreign body capsule formation in humans with Acellular Dermal Matrix (ADM) application.
  • ADM Acellular Dermal Matrix
  • scRNA-seq single cell RNA sequencing
  • UMAP Uniform-manifold approximation and projection
  • C UMAP of myeloid cells colored by Seurat cluster (0-3).
  • FIG. 3A-3G CODEX analysis of ADM and Native Human Capsules.
  • A Schematic of the CO-Detection by indEXing (CODEX) experiment.
  • B Example of Acellular Dermal Matrix (ADM) and Native capsules following CODEX conjugation.
  • C Example of staining of CODEX markers in ADM (top) and Native (bottom) capsules [Integrin ocX (CD11C); White, Collagen Type 1 (COL1); Green, CD45; Blue, Yes-associated protein (YAP); Yellow and Osteopontin-1 (SPP1); Red].
  • D Uniform-manifold approximation and projection (UMAP) of CODEX defined clusters.
  • FIG. 4A-4E Murine Acellular Dermal Matrix (ADM) FBR model recapitulates decreased fibrosis as observed in human ADM capsule specimens.
  • ADM Silicone implants either coated with ADM (mADM) or alone (mNative) were implanted subcutaneously in the dorsi of C57BL/6j (wild-type) mice.
  • Implant and peri-implant tissue were retrieved at postoperative day (POD) 28 for histological analyses.
  • B Hematoxylin and eosin (H&E) staining of mNative and mADM capsule (left and middle columns). Black dotted regions indicate areas from which magnified images were captured. Schematic representing skin layers and capsule (top right). Quantification of capsule thickness in mNative and mADM capsule (bottom right).
  • C Trichrome staining of mNative and mADM capsule (left and middle columns). Schematic representing skin layers and capsule (top right). Quantification of collagen density in mNative and mADM capsule (bottom right).
  • FIGS 5A-5H Single-cell transcriptomic analyses comparing murine Native and Acellular Dermal Matrix (ADM) capsule mimics myeloid cell dynamics in human FBR.
  • A Silicone implants either coated with ADM (mADM) or alone (mNative) were implanted subcutaneously in the dorsi of C57BL/6J (wild-type) mice. Implant and peri-implant tissue were retrieved at postoperative day (POD) 28 for single-cell RNA sequencing (scRNA-seq).
  • scRNA-seq single-cell RNA sequencing
  • UMAP Uniform-manifold approximation and projection
  • Black dotted region indicates cells transcriptomically classified as monocytes (i.e., in silica selection) that were used for downstream analysis.
  • C UMAP of monocytes colored by Seurat subcluster (0-6).
  • D Relative representation of monocytes belonging to Seurat subclusters 0-6 from mNative and mADM capsule.
  • E Dot plot of relative expression of the top 2 differentially expressed genes in each monocyte Seurat subcluster.
  • F Violin plots showing expression of Osteopontin (Spp1) in monocytes by Seurat subcluster (top) and experimental condition (right below).
  • G Inferred SPP1 signaling network in cells from mADM (above) and mNative (below) capsule.
  • FIGS 6A-6H Osteopontin (SPP1) is both necessary and sufficient to decrease fibrotic encapsulation due to foreign body response.
  • SPP1 Osteopontin
  • FIG. 6A Schematic of experimental flow of the murine FBR model used. Silicone implants either alone (KO mNative), coated with Acellular Dermal Matrix (KO mADM), or combined with an ipsilateral injection of recombinant SPP1 -loaded PNP hydrogel (KO +SPP1 ) were implanted subcutaneously in the dorsi of Spp1 Knock-Out (SPP1 KO) mice, Implant and peri-implant tissue retrieved at postoperative day (POD) 28 for histologic analyses.
  • POD postoperative day
  • Silicone implants either combined with an ipsilateral injection of empty PNP hydrogel (mNative -SPP1 ), coated with Acellular Dermal Matrix (mADM), or combined with an ipsilateral injection of recombinant SPP1 -loaded PNP hydrogel (mNative +SPP1 ) were implanted subcutaneously in the dorsi of C57BL/6J (Wildtype) mice. Implant and peri-implant tissue retrieved at postoperative day (POD) 28 for histologic analyses. (F) H&E staining of mNative -SPP1 (far left column), mADM (middle left column), and mNative +SPP1 (middle right column) capsule.
  • Black dotted regions indicate areas from which magnified images were captured. Schematic representing skin layers and capsule (far right column, top). Quantification of capsule thickness in mNative -SPP1 , mADM, and mNative +SPP1 capsule (bottom). (G) Trichrome staining of mNative-SPP1 (far left column), mADM (middle left column), and mNative +SPP1 (middle right column) capsule. Black dotted regions indicate areas from which magnified images were captured. Schematic representing skin layers and capsule (far right column, top). Quantification of collagen in mNative -SPP1 , mADM, and mNative+SPP1 capsule (bottom).
  • Figures 7A-7F (A) Intraoperative photograph following breast implant removal showing acellular dermal matrix (ADM right) and Native capsule (left) retrieval (black arrows). (B) Hematoxylin and eosin (H&E) staining of ADM alone. (C) Trichrome staining of ADM alone. (D) Elastin staining of ADM alone.
  • E Luminex secretion assay comparing median fluorescent intensity (MFI) of the following chemokine markers between implant alone (Native) and Acellular Dermal Matrix-coated (ADM) capsule: Eotaxin-2, Eotaxin-3, Interleukin 5 (IL-5), Chemokine ligand 2 (CCL2), Chemokine ligand 21 (CCL21 ), Chemokine ligand 22 (CCL22), and Chemokine ligand 27 (CCL27).
  • Linked data points represent Native and ADM capsules retrieved from the same patient.
  • F Tensile Young’s modulus of ADM and Native capsule.
  • E Data shown as absolute values.
  • F Data shown as mean modulus of elasticity (MPa) ⁇ standard deviation (S.D.). *P ⁇ 0.05.
  • Figures 8A-8F (A) Uniform-manifold approximation and projection (UMAP) of single-cell RNA-sequencing (scRNA-seq) data from all human capsule cells, colored by patient. (B) UMAP of all cells colored by experimental condition [implant alone (Native) or Acellular Dermal Matrix- coated (ADM) capsule]. (C) UMAP of fibroblast cells (i.e., in silico selection) colored by Seurat subcluster (0-6). (D) UMAP of fibroblasts colored by experimental condition (Native or ADM capsule). (E) UMAP of myeloid cells colored by experimental condition (Native or ADM capsule).
  • UMAP Uniform-manifold approximation and projection
  • FIGS 11A-11 E (A) Uniform-manifold approximation and projection (UMAPs) of macrophage cells colored by Seurat subcluster (0-4). (B) UMAP of macrophages colored by experimental condition (Native or ADM capsule). (C) Relative representation of macrophages belonging to Seurat subclusters 0-4 from ADM and Native capsule. (D) Violin plot displaying Osteopontin (SPP1) expression in macrophages by experimental condition (ADM or Native capsule). (E) Heatmap displaying top differentially expressed genes for each macrophage Seurat subcluster. The red square emphasizes that SPP1 is a gene highly expressed in macrophage Seurat subcluster 1 .
  • UMAPs Uniform-manifold approximation and projection
  • Figures 12A-12B (A) EnrichR analysis results for Pathways and Gene Ontology (GO) terms characteristic of cells in macrophage Seurat subclusters 0, 1 , and 4. (B) EnrichR analysis results for Pathways and Gene Ontology (GO) terms characteristic of cells in macrophage Seurat subclusters 2 and 3.
  • A, B Pathways: NCI-Nature 2016 (blue), KEGG 2021 Human (red), WikiPathway 2021 Human (yellow).
  • Figures 13A-13F Bar graphs of Osteopontin (SPP1 ), Integrin Alpha M (CD11 c), and Collagen Type 1 (COL1 ) protein expression in Acellular Dermal Matrix-coated (ADM) and implant alone (Native) capsules.
  • B Bar graph quantifying Myeloid 1 and Myeloid 3 communication in ADM and Native capsules.
  • C Differential interaction maps in ADM (top) and Native (bottom) capsules
  • D Bar graph quantifying Smooth muscle cell and Myeloid cell communication in ADM and Native capsules (left, top and bottom).
  • Bar graph quantifying cluster of differentiation 4 (CD4) cell and Myeloid cell communication in ADM and Native capsules (right, top and bottom).
  • E Ridge plot showing Yes-associated protein (YAP) expression in all cells.
  • Figures 14A-14B (A) Gross photograph of mNative and mADM capsule retrieval (left). Photos on the right show magnified images of mNative and mADM capsules. (B) Tensile Young’s modulus of PDMS and ADM.
  • Figures 15A-15F (A) Uniform-manifold approximation and projection (UMAPs) of fibroblasts colored by Seurat subcluster (0-4) (B) UMAP of fibroblasts colored by experimental condition [Acellular Dermal Matrix-coated (mADM) or implant alone (mNative) capsule]. (C) UMAP of monocytes colored by experimental condition (mNative or mADM capsule). (D) Heatmap displaying top differentially expressed genes for each monocyte Seurat subcluster. (E) Enrich R analysis results for Pathways and Gene Ontology (GO) terms characteristic of cells in monocyte Seurat subclusters 2, 3, and 4.
  • UMAPs Uniform-manifold approximation and projection
  • E Pathways: NCI-Nature 2016 (blue), KEGG 2021 Human (red), WikiPathway 2021 Human (yellow).
  • GO Terms GO Cellular Component 2021 (blue), GO Molecular Function 2021 (red), GO Biological Process (yellow).
  • (F) DAPI (4',6- diamidino-2-phenylindole), nuclear counterstain (blue signal), n 3 unless otherwise specified.
  • Pathways NCI-Nature 2016 (blue), KEGG 2021 Human (red), WikiPathway 2021 Human (yellow).
  • GO Terms GO Cellular Component 2021 (blue), GO Molecular Function 2021 (red), GO Biological Process (yellow).
  • FIGS 17A-17E (A) Schematic of Osteopontin-1 (SPP1) release from polymer- nanoparticle (PNP) hydrogel wherein 100 pL of PNP hydrogel with 15 pL of Spp1 -AF647 is injected into the bottom of a capillary tube and PBS buffer added (left). Buffer is sampled over time to quantify Spp1 release. Quantification of release assay (right).
  • ECM extracellular matrix
  • C Immunofluorescence (IF) co-staining of Osteopontin-1 (SPP1 ) (green signal) and Integrin alpha X (CD11c) (red signal) (top) and co-staining of SPP1 (green signal) and Collagen Type I (COL1A1) (red signal) bottom in KO mNative, KO mADM, and KO +SPP1 capsules.
  • IF Immunofluorescence
  • ECM extracellular matrix
  • Figures 18A-18B (A) Relative representation of fibroblasts belonging to Seurat clusters 0-6 from ADM and Native capsules. (B) Enrichr analysis results for Pathways and Gene Ontology (GO) terms characteristic to cells in fibroblast Seurat clusters 0 and 3. Pathways: NCI- Nature 2016 (blue), KEGG 2021 Human (red), WikiPathway 2021 Human (yellow). GO Terms: GO Cellular Component 2021 (blue), GO Molecular Function 2021 (red), GO Biological Process (yellow).
  • Figures 19A-19B (A) EnrichR analysis results for Pathways and Gene Ontology (GO) terms characteristic to cells in monocytes/macrophages cell Seurat clusters 1 and 2. (B) EnrichR analysis results for Pathways and Gene Ontology (GO) terms characteristic to cells in monocytes/macrophages Seurat clusters 0 and 3. (A, B) Pathways: NCI-Nature 2016 (blue), KEGG 2021 Human (red) WikiPathway 2021 Human (yellow). GO Terms: GO Cellular Component 2021 (blue), GO Molecular Function 2021 (red), GO Biological Process (yellow).
  • SPP1 Osteopontin
  • Figures 21A-21 B (A) All human cell single-cell RNA-sequencing Uniform-manifold approximation and projection (UMAP) colored by expression level for Integrin Alpha M (CD11b) (top) and Integrin Alpha X (CD11c) (bottom). (B) IF staining of SPP1 (green signal) and CDU b (red signal) in Native and ADM capsule. DAPI (4 Z ,6-diamidino-2-phenylindole), nuclear counterstain (blue signal). Scale bars 100 i m.
  • UMAP Uniform-manifold approximation and projection
  • Figures 22A-22B (A) Haematoxylin and eosin (H&E) staining of Native -ADM capsule.
  • B Immunofluorescence (IF) staining of Osteopontin (SPP1 ) (green signal) and Integrin alpha X (CD11c) (red signal, left). Quantification of SPP1 and CD11 c co-expression from IF staining (right).
  • Figures 23A-23D (A) Schematic of experimental flow of the murine FBR model applied using ADM on its own. Implant and peri-implant tissue retrieved at postoperative day (POD) 28 for histology analysis. (B) Hematoxylin and eosin (H&E) staining of ADM alone capsule. Quantification of capsule thickness in mADM and ADM alone capsule (right). (C) Trichrome staining of ADM alone capsule. Quantification of collagen density in mADM and ADM alone capsule (right). (D) Immunofluorescence (IF) staining of a-Smooth Muscle Actin (a-SMA) (red signal) in ADM alone (far left).
  • H&E Hematoxylin and eosin stain staining of ADM alone capsule. Quantification of capsule thickness in mADM and ADM alone capsule (right).
  • C Trichrome staining of ADM alone capsule. Quantification of collagen density in mADM and ADM alone capsule (right
  • Figures 24A-24B (A) Relative representation of fibroblasts belonging to fibroblast Seurat clusters 0-4 from mADM and mNative capsules. (B) EnrichR analysis results for Pathways and Gene Ontology (GO) terms characteristic to cells in fibroblast Seurat clusters 0 and 1 .
  • Pathways NCI-Nature 2016 (blue), KEGG 2021 Human (red), WikiPathway 2021 Human (yellow).
  • GO Terms GO Cellular Component 2021 (blue), GO Molecular Function 2021 (red), GO Biological Process (yellow).
  • FIGS 25A-25C (A) Violin plot displaying Osteopontin (SPP1) expression in macrophages by experimental condition [Acellular Dermal Matrix-coated (mADM) or implant alone (mNative) capsule]. (B) All mouse single cell RNA-sequencing (scRNA-seq) UMAP colored by expression level for Integrin alpha M (Cd11b) (top) and Integrin alpha X (Cd11c) (bottom). (C) IF staining of SPP1 (green signal) and CD11 b (red signal) in mNative and mADM capsule. Bright green tissue is autofluorescence of panniculus carnosus. (C) DAPI, nuclear counterstain (blue signal).
  • Figures 26A-26B (A) Summary of correlations between human monocyte/macrophage cluster 2 from Fig. 2 to mouse monocyte clusters 1 , 2, and 4 from Fig. 5. (B) Violin plot depicting the similarity of human monocyte/macrophage cluster 2 onto mouse monocyte derived Seurat clusters.
  • FIGS 27A-27F (A) Schematic of experimental flow of the murine FBR model used. Silicone implants coated with Acellular Dermal Matrix combined with an ipsilateral injection of empty PNP hydrogel (KO mADM -SPP1) and silicone implants coated with ADM combined with an ipsilateral injection of recombinant SPP1 -loaded PNP hydrogel (KO mADM +SPP1 ) were implanted subcutaneously in the dorsi of Spp1 Knock-Out (SPP1 KO) mice. Implant and periimplant tissue retrieved at postoperative day (POD) 28 for histologic analyses.
  • POD postoperative day
  • Figures 28A-28E (A) Hematoxylin and eosin (H&E) staining of mouse skin injected subcutaneously every other day with SPP1 resuspended in PBS. (B) Schematic of experimental flow of the murine FBR model used. Silicone implants combined with a subcutaneous injection of SPP1 in PBS (mNative + SPP1 PBS). Implant and peri-implant tissue retrieve at postoperative day (POD) 28 for histology analysis. (C) Hematoxylin and eosin (H&E) staining of mNative + SPP1 PBS capsule.
  • H&E Hematoxylin and eosin staining of mNative + SPP1 PBS capsule.
  • Quantification of capsule thickness in mNative, mNative + SPP1 , and mNative + SPP1 PBS capsule (right).
  • D Trichrome staining of mNative + SPP1 PBS. Quantification of collagen density in mNative, mNative + SPP1 , and mNative + SPP1 PBS capsule (right).
  • E Immunofluorescence (IF) staining of a-Smooth Muscle Actin (a-SMA) (red signal) in mNative+SPP1 PBS (left). Quantification of a-SMA expression from IF staining (right).
  • Figures 29A-29E (A) Violin plots displaying Integrin Subunit Alpha V (ITGAV) and Integrin Subunit Beta 3 (ITGB3) expression in human monocytes/macrophages by Seurat cluster (0-6). (B) Violin plot displaying Cluster of Differentiation 44 (CD44) expression in human monocytes/macrophages by Seurat cluster (0-6). (C) Immunofluorescence (IF) staining of CD44 (red signal) in ADM and Native capsule (left and right). (D) Violin plot displaying Matrix Metallopeptidase 9 (MMP9) in human monocytes/macrophages by experimental condition (ADM or Native capsule).
  • IGAV Integrin Subunit Alpha V
  • IGB3 Integrin Subunit Beta 3
  • a “foreign body” refers to any material, composition, or object that is foreign or exogenous to the tissue, and/or organism in which the foreign body is implanted.
  • a “foreign body” encompasses implantable devices.
  • aspects of the methods include implanting a foreign body in the subject in combination with an effective amount of a SPP1 active agent.
  • Secreted Phosphoprotein 1 also known as Osteopontin (OPN)
  • OPN Osteopontin
  • the SPP1 protein is secreted, can bind to hydroxyapatite with high affinity, and can function as a cytokine.
  • the SPP1 active agent is an SPP1 protein or a fragment thereof.
  • the SPP1 protein is encoded by the Secreted Phosphoprotein-1 gene.
  • Example gene, transcript, and protein sequences of SPP1 may be accessed at Genbank at NG_030362.1 , NM 000582.3, and NP 000573.1 , respectively.
  • a SPP1 polypeptide encompasses any natural variants, alternative sequences, isoforms, or mutant proteins that can be found in any mammal (e.g., that can be found in a human).
  • Examples of SPP1 protein isoforms found in humans include, without limitation, the protein sequences which may be accessed at Genbank at NP 001035147.1 , NP 001035149.1 , NP 001238758.1 , and NP 001238759.1 .
  • SPP1 protein variants found in the human population include, without limitation, L10I, T73A, D107N, F136S, and T152R.
  • Variants of SPP1 include SPP1 variants which are not glycosylated or substantially less glycosylated than endogenously secreted SPP1 proteins.
  • Additional variants of SPP1 include SPP1 proteins that are not phosphorylated or substantially less phosphorylated than endogenously secreted SPP1 proteins.
  • the SPP1 protein used in the methods and compositions described herein may be endogenously derived (e.g., from demineralized bone) or may be a recombinant SPP1 protein.
  • the SPP1 polypeptide is a fusion protein comprising a heterologous polypeptide.
  • the heterologous polypeptide is an affinity tag polypeptide that may aid in purification and isolation of a recombinant SPP1 polypeptide (e.g., FLAG, hemagglutinin(HA), poly-histidine, glutathione-S-transferase (GST), or HALO tags).
  • the SPP1 active agent comprises an amino acid sequence having 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% or greater amino acid sequence identity to the following SPP1 amino acid sequence: MRIAVICFCLLGITCAIPVKQADSGSSEEKQLYNKYPDAVATWLNPDPSQKQNLLAPQTLPSKSNESHDH MDDMDDEDDDDHVDSQDS IDSNDSDDVDDTDDSHQSDESHHSDESDELVTDFPTDLPATEVFTPVVPTVD TYDGRGDSVVYGLRSKSKKFRRPDIQYPDATDEDITSHMESEELNGAYKAIPVAQDLNAPSDWDSRGKDS YETSQLDDQSAETHSHKQSRLYKRKANDESNEHSDVIDSQELSKVSREFHSHEFHSHEDMLVVDPKSKEE DKHLKFRI SHELDSASSEVN ( SEQ ID NO : 1 )
  • the SPP1 active agent comprises an amino acid sequence having 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% or greater amino acid sequence identity to the following SPP1 amino acid sequence: MRLAVICFCLFGIASSLPVKVTDSGSSEEKLYSLHPDP IATWLVPDPSQKQNLLAPQNAVSSEEKDDFKQ ETLPSNSNESHDHMDDDDDDDDGDHAESEDSVDSDESDESHHSDESDETVTASTQADTFTPIVPTVDV PNGRGDSLAYGLRSKSRSFQVSDEQYPDATDEDLTSHMKSGESKESLDVIPVAQLLSMPSDQDNNGKGSH ESSQLDEPSLETIIRLEnSKESQESADQSDVIDSQASSKASLEnQSnKFIISIIKDKLVLDPKSKEDDRYLKF RISHELESSSSEVN ( SEQ ID NO : 3 )
  • the SPP1 active agent comprises an amino acid sequence having 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% or greater amino acid sequence identity to the following SPP1 amino acid sequence: LPVKVTDSGSSEEKLYSLHPDP IATWLVPDPSQKQNLLAPQNAVSSEEKDDFKQETLPSNSNESHDHMDD DDDDDDDDGDHAESEDSVDSDESDESHHSDESDETVTASTQADTFTPIVPTVDVPNGRGDSLAYGLRSKS RSFQVSDEQYPDATDEDLTSHMKSGESKESLDVIPVAQLLSMPSDQDNNGKGSHESSQLDEPSLETHRLE HSKESQESADQSDVIDSQASSKASLEHQSHKFHSHKDKLVLDPKSKEDDRYLKFRISHELESSSSEVN ( SEQ ID NO : 4 )
  • the SPP1 active agent is a peptidomimetic that exhibits the desired activity.
  • Peptidomimetics of interest include, without limitation: peptidomimetics incorporating D- amino acids, that may, for example, improve enzymatic stability; peptidomimetics incorporating unnatural amino acids that may, for example, support bio-orthogonal chemistries (e.g., to enable selective cross-linking); -turn mimetics; cyclic peptides that may, for example, improve biological activity; or any combination thereof.
  • the SPP1 active agent is a nucleic acid encoding a SPP1 polypeptide.
  • the nucleic acid sequence may vary.
  • Nucleic acids of interest include those encoding a SPP1 polypeptide provided above.
  • a nucleic acid of the present disclosure may be a sequence of DNA or RNA having an open reading frame that encodes a polypeptide of interest, i.e., a polypeptide coding sequence, and is capable, under appropriate conditions, of being expressed as a polypeptide of interest, e.g., a SPP1 protein or active fragment thereof.
  • sequence similarity between homologues is 20% or higher, such as 25 % or higher, and including 30 %, 35%, 40%, 50%, 60%, 70% or higher, including 75%, 80%, 85%, 90% and 95% or higher.
  • Sequence similarity is calculated based on a reference sequence, which may be a subset of a larger sequence, such as a conserved motif, coding region, flanking region, etc.
  • a reference sequence may be 18 nt long or longer, such as 30 nt long or longer, and may extend to the complete sequence that is being compared.
  • nucleic acids of substantially the same length as nucleic acids mentioned above, where by substantially the same length is meant that any difference in length does not exceed about 20 number %, usually does not exceed about 10 number % and more usually does not exceed about 5 number %; and have sequence identity to any of these sequences of at 90% or greater, such as 95% or greater and including 99% or greater over the entire length of the nucleic acid.
  • the nucleic acids have a sequence that is substantially similar or identical to the above specific sequences.
  • substantially similar is meant that sequence identity is 60% or greater, such as 75% or greater and including 80, 85, 90, or even 95% or greater.
  • Nucleic acids of interest also include nucleic acids that encode the proteins encoded by the above described nucleic acids, but differ in sequence from the above described nucleic acids due to the degeneracy of the genetic code.
  • the SPP1 active agent is a nucleic acid encoding a SPP1 polypeptide
  • the nucleic acid may be incorporated into an expression vector (e.g., a recombinant expression vector) to facilitate delivery of the nucleic acid to the tissue surrounding, and/or disrupted by, the foreign body.
  • an expression vector e.g., a recombinant expression vector
  • the expression vector may be a viral vector or a non-viral vector.
  • eukaryotic promoters include, without limitation, cytomegalovirus (CMV) immediate early promoters, herpes simplex virus (HSV) thymidine kinase promoters, SV40 early and late promoters, Rous-Sarcoma virus (RSV) promoters, p-actin promoters, tubulin promoters, and EF1 a promoters.
  • the eukaryotic promoter may be a cell or tissue-specific promoter. Cell and tissue-specific promoters are well known in the art and described, e.g., in “Tissue-specific Promoters” (www[.]invivogen[.]com/tissue-specific-promoters).
  • the nucleic acid encoding the SPP1 polypeptide may further comprise a ribosome binding site for initiation of translation (e.g., an internal ribosome entry site; IRES) and/or a transcription terminator.
  • IRES internal ribosome entry site
  • the expression vector may be a viral vector.
  • Viral vectors are known in the art and described, for example, in Warnock, James N., Claire Daigre, and Mohamed Al-Rubeai. "Introduction to viral vectors.” Viral vectors for gene therapy: methods and protocols (2011 ): 1-25, the entirety of which is incorporated herein by reference.
  • Suitable viral vectors for use in the present methods and compositions include, without limitation, adenovirus vectors (see, e.g., WO 94/12649, WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655), adeno-associated virus (AAV) vectors (see, e.g., Ali et al., Hum Gene Ther 9:81 86, 1998, Flannery et aL, PNAS 94:6916 6921 , 1997), lentiviral and retroviral vectors (e.g., lentivirus, Rous- Sarcoma virus, Murine Leukemia virus, and human immunodeficiency virus related vectors).
  • adenovirus vectors see, e.g., WO 94/12649, WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/119
  • the expression vector may be a non-viral vector.
  • Non-viral vectors are known in the art and described, for example, in Yin, Hao, et al. "Non-viral vectors for gene-based therapy.” Nature Reviews Genetics 15.8 (2014): 541 -555, the entirety of which is incorporated herein by reference.
  • Suitable non-viral vectors for use in the present methods and compositions include, without limitation, lipid-based vectors (such as a liposome or lipid nanoparticle, see, e.g., Liposomes: Methods and Protocols, Volume 1 : Pharmaceutical Nanocarriers: Methods and Protocols, (ed. Weissig). Humana Press, 2009, ISBN 160327359X.
  • polymeric vectors e.g., polymeric poly(L-lysine), poly(lactic acid), and poly(D,L-lactide-co-glycolide) vectors, see, e.g., Functional Polymer Colloids and Microparticles volume 4 Microspheres, microcapsules & liposomes), (eds. Arshady & Guyot). Citus Books, 2002 and Polymers in Drug Delivery, (eds. Uchegbu & Schatzlein). ORC Press, 2006.)
  • the SPP1 active agent is a small molecule that can increase expression of SPP1 or activate SPP1 signaling.
  • Candidate agents comprise functional groups for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups.
  • the candidate agents may include cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Such molecules may be identified, among other ways, by employing screening protocols.
  • an effective amount of a SPP1 active agent refers to an amount of an a SPP1 active agent suitable to reducing foreign body response (FBR) in a subject.
  • an effective amount of a SPP1 active agent comprises a SPP1 active agent ranging from, e.g., 1.5 pg/ml to 5 pg/ml, 1 .5 pg/ml to 10 pg/ml, 1 .5 pg/ml to 15 pg/ml, 1 .5 pg/ml to 50 pg/ml, 1 .5 pg/ml to 100 pg/ml, 1.5 pg/ml to 150 pg/ml, 15 pg/ml to 50 pg/ml, 15 pg/ml to 100 pg/ml, 15 pg/ml to 150 pg/ml, 50 pg/ml to 75 pg/ml, 50 pg/ml to 100 pg/ml, 50 pg/ml to 150 pg/ml, or 150 pg/ml to 300 pg/ml.
  • a SPP1 active agent of the present disclosure may be present in a SPP1 active agent composition.
  • the active agent composition includes the active agent and a vehicle.
  • the vehicle may comprise one or more known pharmaceutically acceptable excipients.
  • the vehicle may comprise a pharmaceutically acceptable salt. Suitable salts for use in a present composition include, without limitation, sodium chloride, sodium citrate, sodium benzoate, sodium phosphates (e.g., phosphate buffered saline), potassium phosphate, potassium citrate, calcium carbonate, and calcium phosphate, or combinations thereof.
  • the vehicle may include a pharmaceutically acceptable sugar or polyol.
  • Suitable sugars or polyols for use in a present composition include, without limitation, sucrose, trehalose, mannose, mannitol, raffinose, lactitol, sorbitol and lactobionic acid, glucose, maltulose, iso-maltulose, lactulose, maltose, lactose, iso-maltose, maltitol, palatinit, stachyose, melezitose, dextran, or combinations thereof.
  • the vehicle may include an amino acid.
  • Suitable amino acids for use in a present composition include, without limitation, glycine, arginine, lysine, and glutamine.
  • the vehicle may include a pharmaceutically acceptable thickening agent.
  • suitable thickening agents for use in a present composition include, without limitation, polysaccharides (e.g., hyaluronic acid, alginate, chitosan, dextran, cellulose), silicones (e.g., polysiloxane, polydimethylsiloxane), and other suitable polymers (e.g., polyethylene glycol).
  • the vehicle is a controlled release vehicle that thus provides for a controlled release composition.
  • controlled release composition is meant any composition comprising an active agent which is formulated to provide a longer duration of exposure to an active agent after administration compared to a corresponding immediate release composition comprising an equivalent amount of an active agent.
  • Controlled release compositions may alternatively be described, without limitation, as “extended release”, “sustained release”, “prolonged release”, “programmed release”, “time release”, or “rate controlled” compositions.
  • a controlled release composition may release an effective amount of a SPP1 active agent over any suitable period of time including e.g., one day or more, 2 days or more, 3 days or more, 4 days or more, 5 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, 21 days or more, 28 days or more, or 30 days or more.
  • a controlled release composition may retain a given amount of SPP1 active agent with a half-life from about 8 days to about 32 days, e.g., from about 8 to 9 days, 10 to 11 days, 12 to 13 days, 14 to 16 days, 17 to 18 days, 19 to 20 days, 21 to 22 days, 23 to 24 days, 25 to 26 days, 27 to 28 days, 29 to 30 days, or 31 to 32 days.
  • a controlled release vehicle may include one or more known pharmaceutically acceptable excipients, including any combination the pharmaceutically acceptable excipients discussed herein.
  • the controlled release vehicle comprises a gel.
  • a gel of the present disclosure may be formed from cross-linking of one or more precursors.
  • the gel precursors may be polymeric or non-polymeric.
  • Crosslinks may be formed by covalent bonds or non-covalent interactions. Examples of non-covalent bonds include ionic bonds or hydrophobic interactions.
  • the gel is a hydrogel (i.e., the interstitial fluid comprising the gel is water).
  • a hydrogel of the present disclosure comprises a polymer.
  • the polymer may be synthetic.
  • the polymer may be naturally occurring.
  • the polymer comprises a polysaccharide.
  • suitable polysaccharides include, without limitation, hyaluronic acid (HA), alginate, chitosan, dextran, cellulose, and natural or synthetic derivatives thereof.
  • the polysaccharide is hydroxypropyl methylcellulose.
  • a hydrogel of the present disclosure comprises a nanoparticle.
  • the nanoparticles comprise a polymer.
  • the polymer may be synthetic. In some cases, the polymer may be naturally occurring.
  • nanoparticle polymers include, without limitation, poly(amidoamine), PEGylated poly(amidoamine), polyethylene glycol) methyl ether methacrylate, polypeptides (e.g., albumin), polysaccharides (e.g., chitosan), poly(lactic acid), poly(lactic-co-glycolic acid), and polyethylene glycol) (i.e., PEG).
  • the polymer is a block copolymer.
  • a block copolymer is a polymer comprised of two or more homopolymer subunits (i.e., “blocks”).
  • the block copolymer is a diblock copolymer (i.e., comprised of two distinct “blocks”).
  • the diblock copolymer comprises polyethylene glycol) and poly(lactic acid).
  • aspects of the methods include implanting a foreign body in a subject in combination with an effective amount of an SPP1 active agent to reduce FBR in a subject.
  • the foreign body is coated with a SPP1 active agent composition of the present disclosure.
  • the foreign body is an implantable device.
  • Suitable implantable devices for use in the methods of the present disclosure include, without limitation, implants for reconstructive and/or cosmetic surgery (e.g., breast implants, calf implants, gastric implants, gluteal implants, penile implants, testicular implants), prostheses, tissue-expanders, surgical mesh implants, electro-stimulatory implants (e.g., pacemakers and neuromodulator implants), biosensors, drug-delivery ports, catheters, orthopedic implants, and vascular and non-vascular stents, among others.
  • implants for reconstructive and/or cosmetic surgery e.g., breast implants, calf implants, gastric implants, gluteal implants, penile implants, testicular implants), prostheses, tissue-expanders, surgical mesh implants, electro-stimulatory implants (e.g., pacemakers and neuromodulator implants), biosensors, drug-delivery ports, catheters, orthopedic implants, and vascular and non-vascular stents, among others.
  • implants for reconstructive and/or cosmetic surgery e.g., breast implants
  • the foreign body may include a suitable amount of the SPP1 active agent composition, such that an amount of an SPP1 active agent suitable for reducing the foreign body response (FBR) in the subject (i.e., an effective amount of a SPP1 active agent).
  • a suitable amount of the SPP1 active agent composition such that an amount of an SPP1 active agent suitable for reducing the foreign body response (FBR) in the subject (i.e., an effective amount of a SPP1 active agent).
  • the SPP1 active agent composition may comprise a SPP1 active agent ranging from, e.g., 1.5 pg/ml to 5 pg/ml, 1.5 pg/ml to 10 pg/ml, 1.5 pg/ml to 15 pg/ml, 1.5 pg/ml to 50 pg/ml, 1.5 pg/ml to 100 pg/ml, 1.5 pg/ml to 150 pg/ml, 15 pg/ml to 50 pg/ml, 15 pg/ml to 100 pg/ml, 15 pg/ml to 150 pg/ml, 50 pg/ml to 75 pg/ml, 50 pg/ml to 100 pg/ml, 50 pg/ml to 150 pg/ml, or 150 pg/ml to 300 pg/ml.
  • a SPP1 active agent ranging from, e.g.,
  • the SPP1 active agent composition may be applied to the foreign body.
  • the foreign body may be coated with the composition. All or a part of the foreign body may be coated with the SPP1 active agent composition, e.g., a controlled release composition.
  • all or a part of the surface of the foreign body may be coated with the SPP1 active agent composition.
  • surface it is meant any part or area of the foreign body that, without a coating, would otherwise be in contact with the surrounding tissue when the foreign body is implanted in a subject.
  • the percent of the surface of the foreign body coated with a SPP1 active agent composition can range from, e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% of the surface area of the foreign body coated with a SPP1 active agent composition.
  • the thickness (e.g., the average thickness) of the SPP1 active agent composition coating the foreign body may range from about 1 pm to about 100 pm, e.g., from about 5 pm to about 95 pm, from about 10 pm to about 90 pm, from about 15 pm to about 85 pm, from about 20 pm to about 80 pm, from about 25 pm to about 75 pm from about 30 pm to about 70 pm, from about 35 pm to about 65 pm, from about 40 pm to about 60 pm, or from about 45 pm to 55 pm across the surface of the foreign body.
  • the thickness (e.g., the average thickness) of the SPP1 active agent composition coating the foreign body may range from about 0.1 mm to about 1 mm, e.g., from about 0.2 mm to 0.9mm, from about 0.3 mm to about 0.8 mm, and from about 0.4mm to about 0.6 mm.
  • the thickness of the coating across the surface of the foreign body may vary from about 50% to about 150% of the average thickness of the coating, e.g., from about 75% to about 125%, from about 80% to about 120%, from about 90% to 110%, or from about 95% to about 105%.
  • the percent area of the foreign body to coated and the thickness of the coating may vary depending on the size and nature of the foreign body and can be readily determined by the person of skill using the common knowledge in the art.
  • aspects of the methods include reducing foreign body response (FBR) in a subject.
  • FBR foreign body response
  • a fibrotic aspect of the FBR includes eventual encapsulation of the implant in a layer of the fibrous tissue that acts as a barrier (i.e., foreign body capsule) between the implant and surrounding tissue.
  • the thickness and composition of the foreign body capsule can impact the function of an implantable device and the health of the surrounding tissue.
  • Fibrous encapsulation of an implanted foreign body i.e., formation of a foreign body capsule
  • Fibrous encapsulation of an implanted foreign body is characterized by increased collagen density, decreased elastin density, and decreased elasticity compared to the surrounding tissue.
  • the methods of the present disclosure result in a reduced foreign body capsule thickness compared to a control (e.g., a foreign body implanted without a SPP1 active agent).
  • a suitable control may include a substantially identical foreign body implanted in combination with a vehicle (e.g., one or more pharmaceutically acceptable excipients) substantially identical to the vehicle in the corresponding SPP1 active agent composition, but lacking the SPP1 active agent.
  • a suitable control can include a substantially identical foreign body implanted in combination with a controlled release vehicle comprising a hydrogel and a phosphate buffered saline (PBS) excipient, the controlled release vehicle and PBS together being equivalent in volume to a corresponding SPP1 active agent composition, substantially identical to the control vehicle composition with the exception that the active agent composition comprises an amount of SPP1 active agent.
  • the capsule thickness is decreased by 10% or more compared to a control, e.g., 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, or 95% or more, as comparted to a suitable control.
  • the methods of the present disclosure may result in decreased expression of known markers of fibrosis, e.g., alpha smooth muscle actin (a-SMA), in the foreign body capsule.
  • the methods of the present disclosure result in reduced a-SMA expression in the foreign body capsule compared to a control.
  • a suitable control may include a substantially identical foreign body implanted in combination with a vehicle (e.g., one or more pharmaceutically acceptable excipients) substantially identical to the vehicle in the corresponding SPP1 active agent composition, but lacking the SPP1 active agent.
  • a-SMA expression is decreased by 10% or more compared to a control, e.g., 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, or 90% or more.
  • the methods of the present disclosure result in increased SPP1 expression in myeloid cells of the foreign body capsule compared to a control.
  • a suitable control may include a substantially identical foreign body implanted in combination with a vehicle (e.g., one or more pharmaceutically acceptable excipients) substantially identical to the vehicle in the corresponding SPP1 active agent composition, but lacking the SPP1 active agent.
  • SPP1 expression is increased by 10% or more compared to a control, e.g., 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, or 100% or more.
  • the methods of the present disclosure result in reduced foreign body capsule collagen density compared to a control.
  • a suitable control may include a substantially identical foreign body implanted in combination with a vehicle (e.g., one or more pharmaceutically acceptable excipients) substantially identical to the vehicle in the corresponding SPP1 active agent composition, but lacking the SPP1 active agent.
  • the capsule collagen density is decreased by 10% or more compared to a control, e.g., 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, or 95% or more.
  • the methods of the present disclosure result in an increased foreign body capsule elastin density compared to a control.
  • the capsule elastin density is increased by 10% or more compared to a control, e.g., 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 95% or more, or 100% or more.
  • the methods of the present disclosure result in increased foreign body capsule elasticity compared to a control.
  • a suitable control may include a substantially identical foreign body implanted in combination with a vehicle (e.g., one or more pharmaceutically acceptable excipients) substantially identical to the vehicle in the corresponding SPP1 active agent composition, but lacking the SPP1 active agent.
  • the elasticity of the foreign body capsule may be quantified by the tensile Young’s elastic modulus of the foreign body capsule. In some cases, the Young’s elastic modulus is decreased by 5% or more compared to a control, e.g., 5% or more, 10% or more, 15% or more, 20% or more, 25% or more, 30% or more, 40% or more, or 50% or more.
  • the methods of the present disclosure may result in all or any sub-combination of the effects described above, compared to a control.
  • the methods of the present disclosure may result in a reduced foreign body capsule thickness, a reduced foreign body capsule collagen density, and an increased foreign body capsule elasticity compared to a control.
  • the methods of the present disclosure may result in reduced a- SMA expression in the foreign body capsule and increased SPP1 expression in myeloid cells of the foreign body capsule compared to a control.
  • the methods of the present disclosure may result in a reduced foreign body capsule thickness, a reduced foreign body capsule collagen density, an increased foreign body capsule elasticity, reduced a-SMA expression in the foreign body capsule, and increased SPP1 expression in myeloid cells of the foreign body capsule compared to a control.
  • Embodiments of the methods of the present invention can be practiced on any suitable subject.
  • a subject of the present invention may be a “mammal” or “mammalian”, where these terms are used broadly to describe organisms which are within the class mammalia, including the orders carnivore e.g., dogs and cats), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys). In some instances, the subjects are humans.
  • the methods may be applied to human subjects of both genders and at any stage of development (/.e., neonates, infant, juvenile, adolescent, adult), where in certain embodiments the human subject is a juvenile, adolescent or adult.
  • the present disclosure provides foreign bodies coated with a SPP1 active agent composition.
  • the foreign body is an implantable device coated with a SPP1 active agent composition.
  • Implantable devices include, without limitation, implants for reconstructive and/or cosmetic surgery (e.g., breast implants, calf implants, gastric implants, gluteal implants, penile implants, testicular implants), prostheses, tissue-expanders, surgical mesh implants, electro-stimulatory implants (e.g., pacemakers and neuromodulator implants), biosensors, drugdelivery ports, catheters, orthopedic implants, and vascular and non-vascular stents, among others.
  • the SPP1 active agent composition may be any SPP1 active agent composition of the present disclosure.
  • the SPP1 active agent composition is a controlled release composition.
  • the controlled release composition may release an effective amount of a SPP1 active agent over any suitable period of time including e.g., one day or more, 2 days or more, 3 days or more, 4 days or more, 5 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, 21 days or more, 28 days or more, or 30 days or more.
  • the controlled release composition may retain an amount of SPP1 active agent with a half-life from about 8 days to about 32 days, e.g., from about 8 to 9 days, 10 to 11 days, 12 to 13 days, 14 to 16 days, 17 to 18 days, 19 to 20 days, 21 to 22 days, 23 to 24 days, 25 to 26 days, 27 to 28 days, 29 to 30 days, or 31 to 32 days.
  • the SPP1 active agent composition comprises a gel.
  • a gel of the present disclosure may be formed from cross-linking of one or more precursors.
  • the gel precursors may be polymeric or non-polymeric.
  • Crosslinks may be formed by covalent bonds or non-covalent interactions. Examples of non-covalent bonds include ionic bonds or hydrophobic interactions.
  • the gel is a hydrogel (i.e., the interstitial fluid comprising the gel is water).
  • a hydrogel of the present disclosure comprises a polymer.
  • the polymer may be synthetic.
  • the polymer may be naturally occurring.
  • the polymer comprises a polysaccharide.
  • Suitable polysaccharides include, without limitation, hyaluronic acid (HA), alginate, chitosan, dextran, cellulose, and natural or synthetic derivatives thereof.
  • the polysaccharide is hydroxypropyl methylcellulose.
  • a hydrogel of the present disclosure comprises a nanoparticle.
  • the nanoparticles comprise a polymer.
  • the polymer is a block copolymer.
  • a block copolymer is a polymer comprised of two or more homopolymer subunits (i.e., “blocks”).
  • the block copolymer is a diblock copolymer (i.e., comprised of two distinct “blocks”).
  • the diblock copolymer comprises polyethylene glycol) and poly (lactic acid).
  • All or a part of the foreign body may be coated with the SPP1 active agent composition, e.g., a controlled release composition.
  • all or a part of the surface of the foreign body may be coated with the SPP1 active agent composition.
  • surface it is meant any part or area of the foreign body that, without a coating, would otherwise be in contact with the surrounding tissue when the foreign body is implanted in a subject.
  • the percent of the surface of the foreign body coated with a SPP1 active agent composition can range from, e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% of the surface area of the foreign body coated with a SPP1 active agent composition.
  • the thickness (e.g., the average thickness) of the SPP1 active agent composition coating the foreign body may range from about 1 pm to about 100 pm, e.g., from about 5 pm to about 95 pm, from about 10 pm to about 90 pm, from about 15 pm to about 85 pm, from about 20 pm to about 80 pm, from about 25 pm to about 75 pm from about 30 pm to about 70 pm, from about 35 pm to about 65 pm, from about 40 pm to about 60 pm, or from about 45 pm to 55 pm across the surface of the foreign body.
  • the thickness (e.g., the average thickness) of the SPP1 active agent composition coating the foreign body may range from about 0.1 mm to about 1 mm, e.g., from about 0.2 mm to 0.9mm, from about 0.3 mm to about 0.8 mm, and from about 0.4mm to about 0.6 mm. In some cases, the thickness of the coating across the surface of the foreign body may vary from about 50% to about 150% of the average thickness of the coating, e.g., from about 75% to about 125%, from about 80% to about 120%, from about 90% to 110%, or from about 95% to about 105%.
  • the percent area of the foreign body to coated and the thickness of the coating may vary depending on the size and nature of the foreign body and can be readily determined by the person of skill using the common knowledge in the art.
  • the percent area of the foreign body to coated and the thickness of the coating may vary depending on the size and nature of the foreign body and can be readily determined by the person of skill using the common knowledge in the art.
  • a foreign body of the present disclosure may be coated with an SPP1 active agent composition, or a precursor thereof, using any suitable method known in the art.
  • a precursor of an SPP1 active agent composition includes compositions wherein the SPP1 active agent carried by a precursor vehicle that may undergo a physical or chemical transformation to arrive at the final vehicle compound (e.g., a gel precursor which can polymerize into a gel upon mixing with one or more additional gel precursors and/or addition of a physical or chemical stimulus).
  • the method for coating a foreign body with an SPP1 active agent may vary depending on the size and shape of the foreign body. In some cases, the SPP1 active agent composition may be applied using drop-coating methods.
  • a defined volume of the SPP1 active agent composition is applied to the surface of the foreign body (e.g., with a pipette) and the composition is allowed to adhere, dry, and/or polymerize on the surface (e.g., in the cases where the SPP1 active agent composition comprises a hydrogel).
  • Drop-coating methods may be used to, e.g., coat defined areas of a foreign body.
  • the SPP1 active agent composition may be applied using knife-coating methods, also known as spread-coating, bar-coating, or blade-coating methods.
  • a fixed knife or blade, or other spreader
  • the substrate to be coated is moved underneath at a defined distance.
  • the substrate to be coated is fixed and the knife (or blade, or other spreader) is moved over the substrate to be coated at a defined distance.
  • the knife or blade, or other spreader
  • the knife can spread the SPP1 active agent composition over the substrate to be coated with a defined thickness set by the distance between the substrate and the knife.
  • the SPP1 active agent composition is then allowed to adhere, dry, and/or polymerize on the surface (e.g., in the cases where the SPP1 active agent composition comprises a hydrogel). Knife-coating may be used, e.g., when the foreign body has a flexible surface without a defined surface pattern.
  • the SPP1 active agent composition may be applied using spray-coating methods.
  • the SPP1 active agent composition in spraycoating methods, is aerosolized or atomized through a suitable spray nozzle by pressurized air or gas (e.g., nitrogen or argon) and then transferred via the atomized spray onto the object to be coated. Once transferred to the surface of the object, the SPP1 active agent composition is allowed to adhere, dry, and/or polymerize on the surface (e.g., in the cases where the SPP1 active agent composition comprises a hydrogel).
  • Spray-coating methods are contactless and may be useful, e.g., for sensitive substrate surfaces and/or substrate surfaces with complex shapes. In some cases, the SPP1 active agent composition may be applied using dip-coating methods.
  • the object to be coated is immersed, partially or completely, in the SPP1 active agent composition, or precursor thereof, for a set amount of time and then withdrawn to allow the SPP1 active agent composition to adhere, dry, and/or polymerize on the surface (e.g., in the cases where the SPP1 active agent composition comprises a hydrogel).
  • Dip-coating methods may be used, e.g., for depositing layers 1 pm to 100 pm thick and/or for substrate surfaces with complex shapes.
  • the SPP1 active agent composition may be applied using spin-coating methods.
  • spin-coating methods uniform thin layers of a SPP1 active agent composition, or precursor thereof, are spread across a substrate using centrifugal forces, after which the SPP1 active agent composition is adhere, dry, and/or polymerize on the surface (e.g., in the cases where the SPP1 active agent composition comprises a hydrogel).
  • Spin coating may be useful, e.g., for applying coatings to flat surfaces.
  • kits comprising an implantable device and a SPP1 active agent composition, e.g., as described above.
  • Implantable devices include, without limitation, implants for reconstructive and/or cosmetic surgery (e.g., breast implants, calf implants, gastric implants, gluteal implants, penile implants, testicular implants), prostheses, tissue-expanders, surgical mesh implants, electro-stimulatory implants (e.g., pacemakers and neuromodulator implants), biosensors, drug-delivery ports, catheters, orthopedic implants, and vascular and non- vascular stents, among others.
  • the SPP1 active agent composition may be any SPP1 active agent composition of the present disclosure.
  • the kit may include an implantable device pre-coated with an SPP1 active agent composition.
  • the kit may include the SPP1 active agent composition separate from the implantable device.
  • the kit may include one or more SPP1 active agent composition precursor(s), in individual containers, each separate from the implantable device.
  • the kit may include an SPP1 active agent with an associated pharmaceutically acceptable vehicle or carrier in a first container, and one or more additional vehicle precursors (e.g., gel precursors which can polymerize into a gel upon mixing with one or more addition gel precursors) in one or more additional containers.
  • additional vehicle precursors e.g., gel precursors which can polymerize into a gel upon mixing with one or more addition gel precursors
  • the SPP1 active agent composition precursors may then be combined to arrive at an SPP1 active agent composition.
  • the SPP1 active agent composition, or precursors thereof may be in liquid form or may be lyophilized. In some cases, some of the SPP1 active agent composition precursors may be in liquid form while others are
  • the size of the container may depend on the volume of the SPP1 active agent composition, or composition precursor, to be held in the container.
  • the container may be configured to hold an amount of an SPP1 active agent composition, or precursor thereof, ranging from 0.1 mg to 1000 mg, such as from 0.1 mg to 900 mg, such as from 0.1 mg to 800 mg, such as from 0.1 mg to 700 mg, such as from 0.1 mg to 600 mg, such as from 0.1 mg to 500 mg, such as from 0.1 mg to 400 mg, or 0.1 mg to 300 mg, or 0.1 mg to 200 mg, or 0.1 mg to 100 mg, 0.1 mg to 90 mg, or 0.1 mg to 80 mg, or 0.1 mg to 70 mg, or 0.1 mg to 60 mg, or 0.1 mg to 50 mg, or 0.1 mg to 40 mg, or 0.1 mg to 30 mg, or 0.1 mg to 25 mg, or 0.1 mg to 20 mg, or 0.1 mg to 15 mg, or 0.1 mg to 10 mg, or 0.1 mg to 5 mg, or 0.1 mg
  • the container is configured to hold an amount of an SPP1 active agent composition, or precursor thereof, ranging from 0.1 g to 10 g, or 0.1 g to 5 g, or 0.1 g to 1 g, or 0.1 g to 0.5 g.
  • the container may be configured to hold a volume (e.g., a volume of a liquid) ranging from 0.1 ml to 1000 ml, such as from 0.1 ml to 900 ml, or 0.1 ml to 800 ml, or 0.1 ml to 700 ml, or 0.1 ml to 600 ml, or 0.1 ml to 500 ml, or 0.1 ml to 400 ml, or 0.1 ml to 300 ml, or 0.1 ml to 200 ml, or 0.1 ml to 100 ml, or 0.1 ml to 50 ml, or 0.1 ml to 25 ml, or 0.1 ml to 10 ml, or 0.1 ml to 5 ml, or 0.1 ml to 1 ml, or 0.1 ml to 0.5 ml.
  • the container is configured to hold a volume (e.g., a volume of a liquid SPP1
  • Suitable containers for the compositions include, for example, bottles, vials, syringes, and test tubes.
  • Containers can be formed from a variety of suitable materials, including glass or plastic.
  • the container may be composed of glass, such as, but not limited to, silicate glass, borosilicate glass, sodium borosilicate glass (e.g., PYREXTM), fused quartz glass, fused silica glass, and the like.
  • suitable materials for the containers include plastics, such as, but not limited to, polypropylene, polymethylpentene, polytetrafluoroethylene (PTFE), perfluoroethers (PFE), fluorinated ethylene propylene (FEP), perfluoroalkoxy alkanes (PFA), polyethylene terephthalate (PET), polyethylene (PE), polyetheretherketone (PEEK), and the like.
  • plastics such as, but not limited to, polypropylene, polymethylpentene, polytetrafluoroethylene (PTFE), perfluoroethers (PFE), fluorinated ethylene propylene (FEP), perfluoroalkoxy alkanes (PFA), polyethylene terephthalate (PET), polyethylene (PE), polyetheretherketone (PEEK), and the like.
  • the container may be sealed. That is, the container may include a seal that substantially prevents the contents of the container from exiting the container.
  • the seal of the container may also substantially prevent other substances from entering the container.
  • the seal may be a water-tight seal that substantially prevents liquids from entering or exiting the container, or may be an air-tight seal that substantially prevents gases from entering or exiting the container.
  • the seal is a removable or breakable seal, such that the contents of the container may be exposed to the surrounding environment when so desired, e.g., if it is desired to remove a portion of the contents of the container.
  • the seal is made of a resilient material to provide a barrier (e.g., a water-tight and/or air-tight seal) for retaining a sample in the container.
  • a barrier e.g., a water-tight and/or air-tight seal
  • Particular types of seals include, but are not limited to, films, such as polymer films, caps, etc., depending on the type of container.
  • Suitable materials for the seal include, for example, rubber or polymer seals, such as, but not limited to, silicone rubber, natural rubber, styrene butadiene rubber, ethylene-propylene copolymers, polychloroprene, polyacrylate, polybutadiene, polyurethane, styrene butadiene, and the like, and combinations thereof.
  • a container may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the seal is a septum pierceable by a needle, syringe, or cannula.
  • the seal may also provide convenient access to a sample in the container, as well as a protective barrier that overlies the opening of the container.
  • the seal is a removable seal, such as a threaded or snap-on cap or other suitable sealing element that can be applied to the opening of the container. For instance, a threaded cap can be screwed over the opening before or after a sample has been added to the container.
  • a subject kit may further include, in some embodiments, instructions for practicing one or more of the subject methods described herein.
  • a subject kit may include instructions for preparing a SPP1 active agent composition (e.g., a SPP1 active agent composition comprising a SPP1 active agent and a hydrogel) from one or more composition precursors (e.g., a SPP1 active agent and one or more gel precursors).
  • the subject kit may include instructions for applying the SPP1 active agent composition to the implantable device of the kit (e.g., instructions for coating all or a part of the device with the SPP1 active agent composition).
  • these instructions may be present in printed information on a suitable medium or substrate, e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, and the like.
  • a suitable medium or substrate e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, and the like.
  • a computer readable medium e.g., diskette, compact disk (CD), DVD, flash drive, and the like, on which the information has been recorded.
  • Yet another form of these instructions that may be present is a website address which may be used via the internet to access the information at a removed site.
  • FBR foreign body response
  • ADM acellular dermal matrix
  • scRNA-seq single-cell RNA sequencing
  • SPP1 Osteopontin-1
  • Acellular dermal matrix (ADM), a decellularized tissue with an intact extracellular matrix (ECM), is thought to attenuate FBR by providing a biomimetic scaffold for native tissue incorporation. Rigorous molecular characterization of this phenomenon presents an opportunity to not only revolutionize prosthetic-based medicine, but also highjack a conventionally inevitable biologic process.
  • Implant-based breast reconstruction is used as a model for understanding how ADM modulates FBR.
  • ADM capsule biologically active with significant differences seen in capsule architecture, microenvironment protein signaling, and transcriptional activity in capsule specimens exposed to ADM
  • Native capsule Native capsule not exposed to ADM
  • ADM capsule Native capsule not exposed to ADM
  • ADM capsule Native capsule not exposed to ADM
  • ADM alters foreign body capsule phenotype and decreases FBR
  • Capsule specimens were collected from patients undergoing expander-based breast reconstruction. During this two-stage procedure, patients first receive a tissue expander and then underwent exchange of the expander with a permanent implant following tissue expansion. Expanders were partially wrapped in ADM at the time of placement. As a result, at the time of expander-implant exchange, patients had both capsule that developed adjacent to the expander (“Native capsule”) and capsule adjacent to ADM (“ADM capsule”). These capsule specimens were retrieved from the same expander, allowing for patient-matched capsule specimens of both native and ADM capsule.
  • Paired capsule specimens were obtained from 13 patients, with a mean age of 51 .5 years (SD 11 .2, range 35-68) and mean time to implant exchange of 7 months (SD 3.9, range 4-15, Table 1).
  • Capsule specimens were sampled at a minimum distance of 2 cm distal from Native and ADM capsule overlap to avoid Native and ADM capsule overlap, and minimize the possibility of introducing tissue overlap as a confounding variable (Fig. 1A), and subsequently processed for histology and a multiplexed immunoassay (Luminex).
  • capsules were taken consistently from the posterior side of the expander pocket to minimize differences in FBR capsule due to variability in overlying tissue composition and resultant mechanical force disparities.
  • elastin participates in wound healing by providing mechanical elasticity, decreasing wound contracture, and ultimately assisting in improved regeneration of the dermis.
  • elastin is known to confer elasticity to ECM, thus indicating that ADM capsules are more elastic than Native capsules.
  • This increased elastin expression may provide insights into the reduced rates of pathologic capsular contracture with ADM application.
  • Quantitative analysis of ECM ultrastructure revealed that the ECM of Native and ADM capsule exhibit notable differences.
  • UMAP Uniform manifold approximation and projection mapping of 294 quantified ECM ultrastructure parameters illustrated considerable divergence between ADM capsule and Native capsule ultrastructures (Fig. 1 E). Additionally, elastin staining of the Native and ADM capsules supported differences in the ECM ultrastructure with ADM use, with increased normalized mean elastin density found in ADM capsules (Fig. 1 E, 2.0 vs. 0.10, **** P ⁇ 0.0001). Normalized elastin density via van Gieson’s staining, similarly to normalized collagen density, represents a ratio obtained by staining ADM alone with van Gieson’s stain (Fig.
  • ADM capsule was performed. This revealed Native capsules to have a higher Young’s Elastic Modulus, although it was not statistically significant (Fig. 7F). Collectively, these data suggest that the divergence in cellular signaling may lead to the phenotypic differences in capsule formation in ADM and Native capsules.
  • Spp1 was also the highest differentially expressed gene between ADM and Native capsule monocytes/macrophages (fold change of 3.70, adjusted p-value 6.83 x 10 33 ) (Fig. 2F, bottom) Further interrogation using pathway and Gene Ontology (GO) analysis revealed enrichment for ‘collagen-containing ECM formation’, ‘autophagy regulation’, and ‘foam cell differentiation’ in cluster 2 (Fig. 9A).
  • cluster 1 was enriched in Native capsule, and notable for gene expression terms for ‘prostaglandin signaling’ and ‘VEGF signaling’.
  • Cluster 0 and cluster 3 showed upregulation of more inflammatory terms including ‘IL-1 signaling’ and ‘antigen presentation’ (Fig. 9B).
  • cluster 2 Further examination using pathway analysis revealed enrichment for ‘HIF-1 Signaling’ in cluster 2 cells, a pathway that is known to be associated with tissue protection and adaptation. Interestingly, this cluster also showed enrichment for ‘Lipoprotein particle binding.’ Lipoproteins have previously been shown to have a modulating effect on inflammatory and fibrotic disease, and the presence of this term for cluster 2 is thus demonstrative of a decreased fibrotic signature in ADM capsules (Fig. 19A, right top and bottom rows). In contrast, cluster 1 was enriched in Native capsule, and notable for ‘complement and coagulation cascades,’ and ‘phagosome’ pathways, as well as terms including ‘complement’ and ‘neutrophil degranulation’, all demonstrative of increased inflammatory signature (Fig. 19A, left top and bottom rows).
  • Cluster 0 and cluster 3 showed upregulation of more inflammatory terms including ‘IL-1 signaling’ and ‘antigen presentation’ (Fig. 19B). Further subcluster analysis revealed similar changes to SPP1 expression (Figs. 20A-20B)
  • CellChat a computational tool to investigate cell-cell communication, showed elevated interactions between diverse cell types in ADM capsules compared to Native capsules (Fig. 10A). Cell-cell signaling pathways also differed in ADM versus Native capsules (Fig. 10A).
  • WNT Wingless and lnt-1
  • NCAM neural cell adhesion molecule 1
  • BMP bone morphogenetic protein
  • SPP1 signaling between macrophages/monocytes, fibroblasts, and endothelial cells is illustrated by the increased thickness in the brown, blue, and indigo vectors in the plot. Immunostaining further revealed significantly greater expression of SPP1 in ADM specimens compared to Native capsules in the Integrin Alpha X (CD1 1C + )- expressing cells (Fig. 2H) (** P ⁇ 0.01 ), and decreased expression of collagen 1 (COL1 ) associated with increased SPP1 expression in ADM capsules (Fig.lOC).
  • CD1 1c was selected as a marker for monocytes and macrophages as its expression was specific to those two cell populations in comparison to the pan-myeloid marker CD11b, which was also expressed in some T cells, and fibroblasts (Figs. 21A-21 B).
  • SPP1 is an integrin-binding glycophosphoprotein with multiple roles in bone mineralization and tumorigenesis. In wound healing, loss of function of SPP1 in mice has been shown to delay wound closure and cause defective macrophage infiltration. Given the parallels between fibrotic wound healing and FBR, it was chosen to investigate this gene further.
  • lipid deposition profiles on the surface of biomaterials can influence FBR, with anti-fibrotic materials modifying lipid deposition and facilitating evasion of the immune response.
  • clusters 0 and 4 more prevalent by population in Native capsules, were enriched for ECM processes including ‘contraction’ and ‘actin cytoskeleton’, as well as immune signaling, including ‘interleukin 17 (IL-17) signaling’ and ‘nuclear factor kappa beta (NF-kappaB)’ (Fig. 12A).
  • IL-17 interleukin 17
  • NF-kappaB nuclear factor kappa beta
  • CO-Detection by indexing (CODEX) demonstrates that cell-cell crosstalk differs in ADM and Native Capsules
  • CO-Detection by indEXing was utilized, an assay in which a panel of at least 37 individual protein markers are sequentially labeled and iteratively imaged via cyclic additions and washouts of dye-labeled oligonucleotide- conjugated antibodies (Figs. 3A-3C).
  • CODEX permits building upon the scRNA-seq analyses described above, to also explore changes to the spatial organization of proteins and examine the spatial cell-cell profiles of ADM and Native capsules.
  • 14 cell clusters were based on CODEX protein expression signatures including cluster of differentiation antigen 4 (CD4) T cells, cluster of differentiation antigen 8 (CD8) T cells, two endothelial clusters, five fibroblast clusters, three myeloid clusters, and two smooth muscle cell clusters (Figs. 3B, 3D).
  • Myeloid cells were defined based on marker expression of CD45, CD68, and CD1 1c. SPP1 expression was greater in ADM compared to Native capsules, while COL1 expression was greater in Native compared to ADM capsules (Fig. 13A). These results parallel the histologic and scRNA-seq data, wherein increased collagen was associated with increased fibrosis in Native capsule, while increased SPP1 was demonstrative of the less fibrotic phenotype in ADM capsule. Given the observations made in monocytes and macrophage subclusters in the scRNA-seq dataset, differential protein expression among myeloid subpopulations in ADM and Native capsule was homed in on, which was spatially resolved at a single-cell level using CODEX.
  • ADM capsules had greater numbers of Myeloid 1 and 3 (Fig. 3E, top and bottom row), while Native had greater numbers of Myeloid 2 clusters (Fig. 3E, middle row, * P ⁇ 0.05).
  • the cross talk between Myeloid 1 and 3 was also significantly greater in ADM versus Native capsules (* P ⁇ 0.05) (Fig. 13B).
  • myeloid cells from ADM and Native capsules differed in their protein expression as well.
  • ADM decreases foreign body response in a mouse model
  • Masson’s trichrome staining revealed significantly increased collagen density measured by mean area of collagen staining in mNative capsules (84.7%, SD 7.2), compared to mADM capsules (65.2%, SD 6.3, **** P ⁇ 0.0001 , Fig. 4C).
  • 294 ECM ultrastructure parameters were measured from Picrosirius red-stained images of mNative and mADM capsules, again demonstrating divergent fibrotic phenotypes between experimental conditions (Fig. 4D), as observed in the human data.
  • a-SMA alpha smooth muscle actin
  • mouse implant FBR model recapitulated the phenotypic characteristics of the patient-derived Native and ADM foreign body capsules, with attenuated fibrosis observed in the presence of ADM.
  • mNative and mADM capsules were subjected to scRNA-seq at POD 28 (Fig. 5A). Similar to human samples, there was cell heterogeneity across all cells in mouse capsules (Fig. 5B), with fibroblasts and myeloid cells encompassing a large proportion of all cells. Examination of fibroblasts revealed 5 transcriptionally unique Seurat-based subclusters, with relative overlap between mNative and mADM conditions, analogous to the human dataset (Figs. 15A-15B).
  • Monocytes derived from mADM specimens showed relatively decreased proportions of subclusters 2 (12.26% mADM vs 22.85% mNative) and 3 (1 1.00% mADM vs 17.61% mNative) and an increased proportion of subcluster 4 compared to monocytes derived from mNative specimens (21.56% mADM vs 4.19% mNative) (Fig. 5D).
  • the top differentially expressed genes from each subcluster are illustrated in Fig. 5E and Fig. 15D, with subclusters 2 and 3 enriched for genes involved in inflammation, including ‘apoptosis’, ‘oxidative stress-related signaling’ pathways, and ‘chemokine/cytokine’ response GO terms.
  • Inflammation is a key trigger for fibrosis, and plays a crucial role in the acute and chronic stages of FBR.
  • These clusters were also elevated in genes associated with mechanotransduction signaling pathways, such as focal adhesion kinase (Fig. 15E).
  • Mechanotransduction signaling is known to be a driver of fibrosis and FBR.
  • Focal adhesion kinase signaling has specifically been implicated in macrophage integrin binding to material surfaces. Formation of focal adhesions promotes fibroblast activation and encourages the development of contractile forces. Contraction and stiff ECM deposition can further activate integrins, ultimately leading to activation of TGF-beta and therefore driving pro-fibrotic programs.
  • Fig. 19A human monocyte/macrophage cluster 1
  • Cluster 4 from mouse monocytes was enriched for genes involved in ECM-related GO terms, as well as with ‘HIF-1 Signaling’ and ‘Vascular Endothelial Growth Factor (VEGF) A-VEGFR2 Signaling’.
  • HIF-1 signaling is known to be associated with tissue protection and adaptation.
  • VEGF signaling is known to play a role in angiogenesis and tissue regeneration.
  • fibrosis often leads to dysfunctional and decreased vasculature, thus upregulation of this transcriptional profile is evidence of a pro-regenerative phenotype for this cluster.
  • These data mirror those from human monocyte/macrophage cluster 2, which was enriched in ADM capsules and were evidence of a decreased fibrotic signature in ADM.
  • the remaining subclusters showed relatively similar proportions between mADM and mNative conditions, with respective pathways and GO terms illustrated in Fig. 16.
  • Interrogation of Spp1 expression revealed subcluster 4 to be one of 3 subclusters expressing this gene (Fig. 5F, left).
  • Spp1 was found to be associated with ‘IL-18 signaling’, ‘toll-like receptor regulation’, ‘FGF signaling’, ‘osteopontin signaling’, and ‘focal adhesion signaling’ pathways upon EnrichR analysis.
  • Spp1 expression exhibited increased expression in mADM specimens relative to mNative specimens, reflecting findings from the human data (Fig. 5F, right).
  • Increased Spp1 signaling was also noted between fibroblasts and myeloid cells on CellChat in ADM specimens (Fig. 5G), paralleling cell-cell interaction inference observed in the human scRNA-seq dataset.
  • immunostaining showed significantly enhanced SPP1 and CD11 C expression (Fig.
  • CD11c was selected as a marker for monocytes as its expression was specific to this cell population in comparison to the pan-myeloid marker CD11 b, which also showed more elevated expression in granulocytes, T cells and macrophages (Figs. 25B-25C).
  • Spp1 knockout mice exhibit disorganized remodeling of the ECM and defective macrophage infiltration in the context of wound healing, compared to wounds in wild-type mice. Histologically, the KO mice exhibit more cell debris and relatively homogeneous and smaller collagen fibrils at the site of wound healing, but this has been shown to have no effect on the tensile strength of healing incisional wounds.
  • FBR capsule formation would be expected to occur as it would in wild-type mice, though it may appear histologically distinct.
  • FBR capsule formation was compared under 3 conditions in Spp1 KO mice: (1 ) implant only (KO mNative); (2) ADM covering the implant (KO mADM); and (3) implant together with recombinant SPP1 (R&D Systems, Inc., Minneapolis, MN) formulated in a sustained-release hydrogel (KO +SPP1 ); (4) ADM covering implant with hydrogel-delivered recombinant SPP1 (KO mADM +SPP1); and (5) ADM covering implant with a non-drug-loaded empty hydrogel (KO mADM -SPP1 ) (Fig. 6A and 27 A).
  • This polymer-nanoparticle (PNP) hydrogel developed by Appel and colleagues, is easily injectable and rapidly self-heals, limiting cargo burst release, and has excellent biocompatibility in several species (rodents, sheep, and rabbits).
  • the PNP hydrogel has previously been shown to facilitate slow release of a variety of cargos, including cells, small molecules, and proteins such as the receptor-binding domain of the SARS-CoV-2 spike protein, which is of comparable molecular size to SPP1.
  • SPP1 -formulated PNP hydrogel confirmed sustained release of SPP1 over the course of a month at physiologic temperature, with a half-life of 16 days (Fig. 17A). As softer materials have been shown to elicit less severe FBR, to avoid introducing a confounding variable with application of the gel, it was specifically injected as a sustained- released drug depot adjacent to the implant.
  • KO +SPP1 capsules measured a mean of 20.4 pm (SD 4.3), and were significantly thinner compared to KO mNative capsules (mean 47.5 pm, SD 7.9; **** P ⁇ 0.0001 ) or KO mADM capsules (mean 48.9 SD 12.9; **** P ⁇ 0.0001 ) (Fig. 6B).
  • Immunostaining showed increased SPP1 expression in CD11 c co-expressing cells (identified as myeloid cells) and decreased fibrotic marker expression (COL1A1 and aSMA) in KO +SPP1 capsules relative to KO mNative and KO mADM capsules (Fig. 6D and 17C, * P ⁇ 0.05, P ⁇ 0.001).
  • KO mADM +SPP1 (mean 19.3 pm, SD 3.567) capsules were significantly thinner than KO mADM -SPP1 capsules (mean 47.84 pm, SD 11 .95; **** P ⁇ 0.0001 ) (Fig. 27B).
  • collagen density was significantly lower in KO mADM +SPP1 capsules (mean 44.85%, SD 19.09) relative to KO mADM -SPP1 capsules (mean 93.92%, SD 3.971 ; **** P ⁇ 0.0001 ) (Fig. 27C).
  • KO mADM +SPP1 capsule thickness and collagen density paralleled those of KO +SPP1 capsules (Figs.
  • KO +mADM -SPP1 capsule thickness and collagen density paralleled those of KO -SPP1 and KO mADM capsules Figs. 6B- 6C
  • KO mADM +SPP1 displayed similar architectural properties to KO +SPP1 capsule specimens (Fig. 17B, 27D).
  • Immunostaining showed decreased fibrotic marker expression (aSMA and COL1) in KO mADM +SPP1 capsules (Figs. 27E-27F, ** P ⁇ 0.01 ), as well as increased SPP1 expression in CD11c co-expressing cells (Fig. 27F).
  • Mean mNative +SPP1 capsule thickness was 30.9 pm (SD 4.9), compared to 50.0 pm (SD 6.7; **** P ⁇ 0.0001 ) in mNative -SPP1 capsules and 26.2 pm (SD 2.7; ** P ⁇ 0.01 ) in mADM capsules (Fig. 6F).
  • Immunostaining confirmed decreased a-SMA and COL1 expression in mNative +SPP1 compared to mADM and mNative -SPP1 capsules (Fig. 6H and Fig. 17D). Immunostaining also showed increased SPP1 expression in CD11c-expressing cells (identified as myeloid cells) in mNative +SPP1 and mADM relative to mNative -SPP1 capsules (Fig. 17D and Fig. 27E). Finally, ECM ultrastructural analysis of 294 unique parameters on picrosirius-red stained capsule specimens revealed overlap of capsule ultrastructure of mADM and mNative +SPP1 capsules, and divergence from mNative -SPP1 capsule (Fig. 17E). Collectively, these data support that SPP1 release could substitute for ADM and is sufficient to restore the fibrotic capsule phenotype in the murine FBR model.
  • SPP1 resuspended in PBS was injected subcutaneously every other day for 28 days in an otherwise healthy, wildtype mouse. As observed on H&E, no visible toxicity was observed with all expected skin architecture and associated appendages present (Fig. 28A). To ensure that the hydrogel on its own did not influence capsule formation, an additional group of wild-type C57BL/6 mice received implants and injections of SPP1 resuspended in PBS every other day for 28 days (mNative +SPP1 PBS) (Fig. 28B).
  • SPP1 is well known to interact via two separate pathways. Firstly, it has been shown that SPP1 can bind to integrin av 3 and activate integrin-mediated signaling. Secondly, SPP1 can activate downstream gene expression via CD44. Investigation of expression of Integrin Subunit Alpha V(JTGV5) and Integrin Subunit Beta 3 (ITB3), genes encoding for subunits of integrin av[33, suggested expression of only one of the two subunits and only in Seurat cluster 2 of human monocytes/macrophages (29A). In contrast, CD44 showed expression across all subclusters of human monocytes/macrophages (29B), and was confirmed at a protein level via immunostaining (29C).
  • MMP9 a known direct downstream target of SPP1-CD44 signaling
  • SPP9-CD44 signaling was expressed at significantly higher levels in ADM capsule monocytes/macrophages relative to Native capsule monocytes/macrophages (log fold change 1 .57, adjusted P-value 6 x 10' 10 ) (29D).
  • Staining via IF confirmed increased protein expression of MMP9 (* P ⁇ 0.05) (29E).
  • SPP1 is an integrin-binding glycophosphoprotein that has been shown to play significant roles in cancer, bone homeostasis, and metabolism. In addition, SPP1 has been shown to also bind and mediate signals via CD44 in hair follicle stem cells. Associated with tumor progression, invasion, and metastasis, elevated expression levels of SPP1 have been connected with poor prognosis; yet, how it relates to gene mutation and immune cell infiltration is not well understood.
  • these findings utilize clinical and mouse models to establish molecular pathways that provide therapeutic targets to overcome FBR.
  • ADM may be altering the implant microenvironment and changing the resulting fibrotic capsule.
  • PYK2 is an adhesion kinase in macrophages, localized in podosomes and activated by beta(2)-integrin ligation.
  • CD44-ICD matrix metalloproteinase 9
  • Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates. Nat Mater 16, 671-680 (2017).
  • a 3 x 1 cm area of native and ADM capsules were obtained and divided into three smaller portions for (i) histological analysis, (ii) Fluorescence activated cell sorting (FACS) and transcriptomic analysis, and (iii) proteomic analysis.
  • FACS Fluorescence activated cell sorting
  • Intra-operatively capsule specimens were obtained from the native capsule and the ADM capsule. ADM capsule was removed with ADM attached, as it was well integrated into the surrounding tissues. If the ADM was not incorporated, the patient’s samples were excluded from analysis. An additional three capsule specimens were collected who underwent tissue expander implant exchange without ADM-wrap. Patient demographic information is presented in Table 3.
  • ADM Acellular dermal matrix
  • Implants were fabricated in a modified method as previously reported by Lin et. al. Briefly, hemispherical 0.2 mL implants were created by casting Sylgard® 184 (Sigma-Aldrich) polydimethylsiloxane (PDMS) in a 10:1 ratio of base-agent to curing agent in a prefabricated mold. The PDMS solution was then temperature treated at 65 °C for 15 minutes and the reaction was allowed to continue for an additional 15 minutes at room temperature before removing the implants from the mold. After fabrication, the molds were placed in 100% Ethanol to allow unreacted materials to leach out and sterilize the implants before storing them in sterile containers.
  • Sylgard® 184 Sigma-Aldrich polydimethylsiloxane
  • the hydrogel was formulated as previously described. Briefly, the hydrogel was formulated with final concentrations of 2 wt% (percentage by weight) hydroxypropylmethylcellulose (HPMC)-Ci2 and 10 wt% polyethylene glycol)-block-poly(lactic acid) (PEG-PLA) nanoparticles (NPs). HPMC-Ci2 was synthesized as previously described, dissolved in PBS at 6 wt%, and loaded into a 1 mL luer-lock syringe.
  • HPMC-Ci2 hydroxypropylmethylcellulose
  • PEG-PLA polyethylene glycol)-block-poly(lactic acid)
  • a 20 wt% solution of NPs in PBS was diluted with additional PBS, or PBS containing recombinant Osteopontin protein (SPP1 ) (R&D Systems, Inc., Minneapolis, MN) for a final dose of 15 pg per 100 pL and loaded into a separate 1 mL luer-lock syringe.
  • the two syringes were connected with a female-female luer-lock elbow with care to avoid air at the interface of HPMC-C12 and the NP solution.
  • the two solutions were mixed for 1 min or until a homogenous hydrogel was formed. After mixing, the elbow was removed and a luer lock cap was placed and the material was stored at 4°C for up to 7 days prior to surgery.
  • SPP1 -AF647 was concentrated to 1 mg/mL and incorporated into hydrogel at 15 pg per 100 pL for the in vitro release assay.
  • Glass capillary tubes were cut to 4 inches, sealed on one end with epoxy, and allowed to cure for at least 24 hours.
  • a PDMS implant alone or PDMS implant covered with ADM was then placed in the subcutaneous pocket. Mice were divided into different experimental groups, each receiving one implant. For mice that received the ADM alone, a 1 x 1 cm square of ADM was placed in the subcutaneous pocket. The wound was closed without tension using 6 interrupted nylon monofilament 4-0 sutures (DynarexTM, Orangeburg, NY). The incision was dressed using KrazyglueTM (Elmer’s Products, Inc., Westerville, OH). Incisions were inspected daily. For mice that received the hydrogel with recombinant SPP1 (Osteopontin), 100 pL of the gel suspension was injected adjacent to the implant once the incision was closed and glued. Mice were hydrated with 1 mL of PBS at the end of the procedure.
  • SPP1 Hydrogel with recombinant SPP1
  • the capsule surrounding the implant was harvested with dissecting scissors under 2.5X Loupe magnification and processed for subsequent analysis. Mice were euthanized by carbon dioxide (CO2) narcosis and cervical dislocation. The superior surface of the capsule was meticulously dissected following the edge of the implant, with overlying skin intact. Harvested capsule was sent for histology. Additionally, capsule used for fluorescence- activated cell sorting (FACS) was meticulously removed from the overlying skin using 2.5X Loupe magnification and mechanically digested using dissecting scissors to finely mince each specimen. Harvested capsule for use in histology and immunofluorescent (IF) staining was placed in tissue embedding cassettes.
  • FACS fluorescence- activated cell sorting
  • ADM and PDMS
  • ADM and PDMS samples were tested using an Instron 5565 using a 100 N load cell. Materials were cut into tapered 5mm by 10mm pieces. Tissue pieces were subsequently anchored between grips. The tissue was slowly separated (1% increase per second) until failure (defined by a clear drop in measured stress as tension increased). Young’s Modulus was determined by taking the slope of the linear portion of the stress-strain curve.
  • ThermoFisher ScientificTM Waltham, MA was used to dehydrate samples in a gradient of alcohols. Tissue was then embedded using ThermoFisher Histostar Tissue Embedding station. Paraffin blocks were trimmed as necessary and cut into 8 pm-thick sections. Paraffin ribbons were placed in a water bath at 40°C and mounted onto Superfrost/Plus adhesive slides (ThermoFisher ScientificTM, Waltham, MA). Sections were baked at 50°C overnight.
  • Hematoxylin and eosin (Cat:H-3502; Vector Laboratories, Burlingame, California), Masson’s Trichrome (ab150686; Abeam®, Waltham, MA), Picro-sirius Red (ab150681 ; Abeam®, Waltham, MA) and Modified Verhoeff Van Gieson stain (Abeam®, Waltham, MA) with standard protocols were used. Cryosection samples were first dehydrated using a slide rack, submerged into 1% PBS for 10 minutes, followed by 30% ethanol (EtOH), 50% EtOH, 70% EtOH, 95% EtOH, and 100% EtOH for 15 minutes each.
  • EtOH 30% ethanol
  • Paraffin sections were hydrated prior to staining by placement in xylene for 20 minutes, followed by 10 minutes each of 100% EtOH, 95% EtOH, 70% EtOH, 50% EtOH, and 30% EtOH. Slides were then submerged in running tap water for 10 minutes.
  • the digest was quenched with FACS buffer (ThermoFisher ScientificTM, Waltham, MA) containing 2X FBS, 1X Penicillin-Streptomycin, 1 X Poloxamer 188), then centrifuged at 1250rpm for 5 min at 4 °C, resuspended in FACS buffer, and filtered through 100, 70, and 40 pm cell strainers (Falcon cell strainer, ThermoFisher ScientificTM, Waltham, MA). Histopaque was performed using Histopaque-11 19 (Sigma-Aldrich, St. Louis, MO), per the manufacturer’s protocol. Cells were counted and resuspended in FACS buffer. Primary antibodies were applied, and cells were stained in the dark with gentle agitation for 30 min. Cells were then washed thoroughly in FACS buffer.
  • FACS buffer ThermoFisher ScientificTM, Waltham, MA
  • FACS buffer ThermoFisher ScientificTM, Waltham, MA
  • Histopaque was performed using
  • Single-cell Barcoding, Library Preparation, and Sequencing (Human Samples): Individual human native and ADM capsule dissociated tissue cellular suspensions were tagged with hashtag oligos (HTOs) per the manufacturer’s protocol and then pooled. Cells were counted and filtered just prior to loading into the 10X machine. Single cells were barcoded using the 10x Chromium Single Cell platform, and cDNA libraries were prepared according to the manufacturer’s protocol (Single Cell 3’ v3, 10x Genomics, USA). In brief, cell suspensions, reverse transcription master mix and partitioning oil were loaded on a single-cell chip, then run on the Chromium Controller. Reverse Transcription was performed within the droplets at 53 °C for 45 min.
  • HTOs hashtag oligos
  • cDNA was amplified for a 12 cycles total on a BioRad C1000 Touch thermocycler. cDNA size selection was performed using SpriSelect beads (Beckman Coulter, USA) and a ratio of SpriSelect reagent volume to sample volume of 0.6. cDNA was analyzed on an Agilent Bioanalyzer High Sensitivity DNA chip for qualitative control purposes. cDNA was fragmented using the proprietary fragmentation enzyme blend for 5 min at 32 °C, followed by end repair and A-tailing at 65 °C for 30 min. cDNA were double-sided size selected using SpriSelect beads. Sequencing adaptors were ligated to the cDNA at 20 °C for 15 min.
  • cDNA was amplified using a sample-specific index oligo as primer, followed by another round of double-sided size selection using SpriSelect beads. Final libraries were analyzed on an Agilent Bioanalyzer High Sensitivity DNA chip for qualitative control purposes. cDNA libraries were sequenced on a NextSeq 500 Illumina platform aiming for 50,000 reads per cell.
  • UMIs Unique molecular identifiers
  • Raw UMI counts were normalized with a scale factor of 10,000 UMIs per cell and subsequently natural log transformed with a pseudocount of 1 using the R package Seurat (version 4.0.5).
  • HTOs for human samples were_demultiplexed using Seurat’s implementation HTODemux. Briefly, k-medoid_clustering is performed on the normalized HTO values, after which a ‘negative’ HTO distribution is calculated. For each HTO, the cluster with the lowest average value is treated as the negative group and a negative binomial distribution is fit to this cluster.
  • each cell is classified as positive or negative for each HTO.
  • Cells that are positive for more than one HTOs are annotated as doublets and removed. Cells that are not positive for any HTO are also removed.
  • Aggregated data were then evaluated using uniform manifold approximation and projection (UMAP) analysis over the first 15 principal components. Louvain-based clustering analysis was chosen for the scRNA-seq data as it is the most used graph-based clustering method for single-cell RNA-seq data and is used to assist users in identifying different cell types or cell subpopulations within a given dataset. It is commonly used in part due to its scalability for large single-cell RNA-seq analysis datasets.
  • Seurat was made use of for the scRNA-seq analyses, which applies the Louvain algorithm as the default clustering method.
  • Cell annotations were ascribed using SingleR (version 3.11 ) against the Blueprint + ENCODE reference database for human cells.
  • Base calls were converted to reads using the Cell Ranger (10X Genomics; version 3.1 ) implementation mkfastq and then aligned against the Cell Ranger mml 0 reference genome, using Cell Ranger’s count function with SC3Pv3 chemistry and 5,000 expected cells per sample, as previously described.
  • a maximum percent mitochondrial RNA cutoff of 15% and 7,500 maximum unique genes and 85,000 maximum RNA counts were used.
  • UM Is Unique molecular identifiers
  • UMAP uniform manifold approximation and projection
  • Louvain-based clustering analysis was chosen for the scRNA-seq data as it is the most used graph-based clustering method for single-cell RNAseq data and is used to assist users in identifying different cell types or cell subpopulations within a given dataset. It is commonly used in part due to its scalability for large single-cell RNA-seq analysis datasets.
  • Seurat was made use of for the scRNA-seq analyses, which applies the Louvain algorithm as the default clustering method.
  • Cell annotations were ascribed using SingleR (version 3.11 ) against the Mouse-RNAseq reference dataset, available at https://rdrr(.)io/qithub/dviraran/SinqleR/man/mouse(.)rnaseq(.)html.
  • Cell-type marker lists were generated using Seurat’s native FindMarkers function with a log fold change threshold of 0.25 using the ROC test to assign predictive power to each gene.
  • the 200 most highly ranked genes from this analysis for each cluster were used to perform gene set enrichment analysis in a programmatic fashion using EnrichR (version 2.1).
  • EnrichR version 2.1
  • the recently developed CellChat platform was applied. This was implemented using the scRNA-seq Seurat object in R, in conjunction with the standalone CellChat Shiny App for its Cell-Cell Communication Atlas Explorer. Cells were binned according to the SingleR-defined cell type classifications. Default parameterizations were used throughout, and Secreted Signaling, ECM- Receptor, and Cell-Cell Contact relationships were considered.
  • CODEX Co-Detection by Indexing
  • the CODEX was visualized using Akoya Biosciences Multiplex Analysis Viewer (MAV) in Imaged.
  • MAV Multiplex Analysis Viewer
  • the resulting .fcs files were then concatenated in FlowJo and imported into the Monocle3 and STvEA R packages for further analysis.
  • the processed UMAP manifold was analyzed through Monocle3 with a post-manifold threshold of >10,000 cells per cluster. Analysis of the protein staining patterns was then used to assign cell types.
  • Table 2 List of protein markers with their associated barcodes for the CODEX experiment performed in this study.
  • Table 4 List of protein markers with their associated barcodes for the CODEX experiment performed in this study and additional information.
  • CDH1 E-Cadherin
  • CD68 Cluster of Differentiation 68
  • IL6 Interleukin
  • MGP Matrix Gia Protein
  • CD45 Protein Tyrosine Phosphatase Receptor Type C
  • CD11c Integrin Subunit Alpha X
  • CD8 Cluster of Differentiation 8
  • IL1 B Interleukin 1 Beta
  • CD20 Membrane Spanning 4-Domain A1
  • HLA-DR Major Histocompatibility Complex Class II
  • S100A4 S100 Calcium Binding Protein A4
  • FN1 Fibronectin 1
  • CD26 Dipeptidylpeptiidase IV
  • PDGFRa Platelet Derived Growth Factor Receptor Alpha
  • COLIV Collagen Type IV
  • aSMA Alpha Smooth Muscle Actin
  • Ki67 Marker of Proliferation Ki-67
  • VIM Vimentin
  • PANCK Pan-Cytokeratin
  • FAK Focal Adhesion Kinase
  • MYH11 Myosin Heavy Chain 11
  • ADIPOQ Adiponectin, C1Q and Collagen Domain
  • TAGLN Transgelin
  • PDGFRB Platelet Dervied Growth Factor Receptor Beta
  • DES Desmin
  • PIEZO1 Piezo Type Mechanosensitive Ion Channel Component 1
  • PIEZO2 Piezo Type Mechanosensitive Ion Channel Component 2
  • PDPN Podoplanin
  • COL1 Collagen Type I
  • SPP1 Osteopontin.
  • a range includes each individual member.
  • a group having 1-3 articles refers to groups having 1 , 2, or 3 articles.
  • a group having 1 -5 articles refers to groups having 1 , 2, 3, 4, or 5 articles, and so forth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dermatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Inorganic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Transplantation (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne des procédés de réduction de la réponse à un corps étranger (FBR) chez un sujet. Des aspects des procédés comprennent l'implantation d'un corps étranger, par exemple, un dispositif implantable, chez le sujet en combinaison avec une quantité efficace d'un agent actif SPP1, par exemple, un polypeptide SPP1, pour réduire la FBR chez le sujet. L'invention concerne également des dispositifs implantables revêtus d'une composition d'agent actif SPP1, par exemple, une composition à libération contrôlée, et des kits comprenant un dispositif implantable et une composition d'agent actif SPP1.
PCT/US2024/044222 2023-08-29 2024-08-28 Procédés et compositions pour réduire la réponse à un corps étranger Pending WO2025049603A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363535205P 2023-08-29 2023-08-29
US63/535,205 2023-08-29

Publications (1)

Publication Number Publication Date
WO2025049603A1 true WO2025049603A1 (fr) 2025-03-06

Family

ID=94820388

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/044222 Pending WO2025049603A1 (fr) 2023-08-29 2024-08-28 Procédés et compositions pour réduire la réponse à un corps étranger

Country Status (1)

Country Link
WO (1) WO2025049603A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048577A1 (en) * 2000-08-01 2002-04-25 University Of Washington Methods and devices to modulate the wound response
US20130236502A1 (en) * 2009-11-25 2013-09-12 Healionics Corporation Implantable medical devices having microporous surface layers and method for reducing foreign body response to the same
US20170246347A1 (en) * 2015-11-01 2017-08-31 Massachusetts Institute Of Technology Materials with improved properties
US20200305770A1 (en) * 2010-10-06 2020-10-01 Profusa, Inc. Tissue-integrating sensors
US20220000789A1 (en) * 2018-09-27 2022-01-06 Sigilon Therapeutics, Inc. Implantable devices for cell therapy and related methods

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048577A1 (en) * 2000-08-01 2002-04-25 University Of Washington Methods and devices to modulate the wound response
US20130236502A1 (en) * 2009-11-25 2013-09-12 Healionics Corporation Implantable medical devices having microporous surface layers and method for reducing foreign body response to the same
US20200305770A1 (en) * 2010-10-06 2020-10-01 Profusa, Inc. Tissue-integrating sensors
US20170246347A1 (en) * 2015-11-01 2017-08-31 Massachusetts Institute Of Technology Materials with improved properties
US20220000789A1 (en) * 2018-09-27 2022-01-06 Sigilon Therapeutics, Inc. Implantable devices for cell therapy and related methods

Similar Documents

Publication Publication Date Title
Rijal et al. A versatile 3D tissue matrix scaffold system for tumor modeling and drug screening
Jansen et al. Zwitterionic PEG-PC hydrogels modulate the foreign body response in a modulus-dependent manner
Moulisová et al. Engineered microenvironments for synergistic VEGF–Integrin signalling during vascularization
Piccoli et al. Decellularized colorectal cancer matrix as bioactive microenvironment for in vitro 3D cancer research
Heymann et al. Polypropylene mesh implantation for hernia repair causes myeloid cell–driven persistent inflammation
Freiman et al. Adipose-derived endothelial and mesenchymal stem cells enhance vascular network formation on three-dimensional constructs in vitro
Hinz et al. Alpha-smooth muscle actin expression upregulates fibroblast contractile activity
Sundarakrishnan et al. Bioengineered in vitro tissue model of fibroblast activation for modeling pulmonary fibrosis
Shimizu et al. Epithelial morphogenesis in hydra requires de novo expression of extracellular matrix components and matrix metalloproteinases
US20190201586A1 (en) Three-Dimensional Tissue Matrix Scaffold System
Schmid et al. A new printable alginate/hyaluronic acid/gelatin hydrogel suitable for biofabrication of in vitro and in vivo metastatic melanoma models
López-Martínez et al. A natural xenogeneic endometrial extracellular matrix hydrogel toward improving current human in vitro models and future in vivo applications
Hewawasam et al. Chemical modification of human decellularized extracellular matrix for incorporation into phototunable hybrid-hydrogel models of tissue fibrosis
Dempsey et al. Functional insights from the proteomic inventory of ovine forestomach matrix
Schweller et al. Stiffness of protease sensitive and cell adhesive PEG hydrogels promotes neovascularization in vivo
Pilipchuk et al. Influence of crosslinking on the stiffness and degradation of dermis‐derived hydrogels
Kim et al. Angiogenic effects of collagen/mesoporous nanoparticle composite scaffold delivering VEGF165
Padmanabhan et al. Allometrically scaling tissue forces drive pathological foreign-body responses to implants via Rac2-activated myeloid cells
Kim et al. In vivo biodegradation and biocompatibility of PEG/sebacic acid‐based hydrogels using a cage implant system
Duong et al. Photo‐Responsive Decellularized Small Intestine Submucosa Hydrogels
Vigier et al. Exploring the Extracellular Matrix to Create
Gray et al. Biocompatibility of common implantable sensor materials in a tumor xenograft model
Frati et al. Reinforced alginate/gelatin sponges functionalized by avidin/biotin-binding strategy: a novel cardiac patch
Kohn-Polster et al. Dual-component gelatinous peptide/reactive oligomer formulations as conduit material and luminal filler for peripheral nerve regeneration
Clift et al. Evaluation of therapeutic collagen-based biomaterials in the infarcted mouse heart by extracellular matrix targeted MALDI imaging mass spectrometry

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24860945

Country of ref document: EP

Kind code of ref document: A1