EP4225235A1 - Appareil et procédés d'injection rétinienne - Google Patents

Appareil et procédés d'injection rétinienne

Info

Publication number
EP4225235A1
EP4225235A1 EP21878692.9A EP21878692A EP4225235A1 EP 4225235 A1 EP4225235 A1 EP 4225235A1 EP 21878692 A EP21878692 A EP 21878692A EP 4225235 A1 EP4225235 A1 EP 4225235A1
Authority
EP
European Patent Office
Prior art keywords
hollow conduit
eye
distal end
medicament
end portion
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
EP21878692.9A
Other languages
German (de)
English (en)
Inventor
Chen-rei WAN
Rafael Victor Andino
Clay G. PRICKETT
Jay HOPE
Jordan MYKLEBY
Nicolo Garbin
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.)
Clearside Biomedical Inc
Original Assignee
Clearside Biomedical Inc
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 Clearside Biomedical Inc filed Critical Clearside Biomedical Inc
Publication of EP4225235A1 publication Critical patent/EP4225235A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F9/00Methods or devices for treatment of the eyes; Devices for putting-in contact lenses; Devices to correct squinting; Apparatus to guide the blind; Protective devices for the eyes, carried on the body or in the hand
    • A61F9/0008Introducing ophthalmic products into the ocular cavity or retaining products therein
    • A61F9/0017Introducing ophthalmic products into the ocular cavity or retaining products therein implantable in, or in contact with, the eye, e.g. ocular inserts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/34Trocars; Puncturing needles
    • A61B17/3417Details of tips or shafts, e.g. grooves, expandable, bendable; Multiple coaxial sliding cannulas, e.g. for dilating
    • A61B17/3421Cannulas

Definitions

  • Embodiments described herein relate to systems and methods of retinal injection.
  • Systems and methods described herein can be employed in non-surgical and/or minimally invasive procedures (e.g., in an office setting, without surgical imaging, etc.).
  • Retinal injection has several uses in the medical industry, including injection of pharmaceutical agents.
  • each eye is different.
  • the total size of an eye may differ from patient to patient.
  • the thicknesses, density, and other characteristics of various tissue layers (e.g., choroid, retina) in the eye may vary from one patient to another. This variability in tissue layer size, for example, can make it difficult to standardize tooling or methods when precisely injecting into a region of the eye.
  • an important industrial advancement to be made is a system and/or a method of effectively targeting a retinal region of an eye.
  • a method includes inserting a distal end portion of a hollow conduit of a medical injector into an eye of a patient until the distal end portion reaches a reference location, the medical injector having a housing from which the hollow conduit distally extends and confirming positioning of the distal end portion of the hollow conduit in the reference location. After the confirming, the method further includes extending the distal end portion of the hollow conduit distally relative to the housing and from the reference location such that the distal end portion enters an injection region of the eye. With the distal end portion of the hollow conduit disposed in the injection region of the eye, medicament is conveyed into the injection region via the hollow conduit.
  • the reference location can be a suprachoroidal space (SCS) of the eye.
  • SCS suprachoroidal space
  • the injection region can be between the SCS and a boundary layer between a retinal layer and vitreous space of the eye.
  • extending the distal end portion of the hollow conduit includes extending the distal end portion of the hollow conduit between about 50 micrometers and about 5 mm.
  • confirming positioning of the distal end portion of the hollow conduit in the reference location can include sensing a pressure differential within the reference location.
  • confirming positioning of the distal end portion of the hollow conduit in the reference location can include measuring a pressure within the reference location, the pressure meeting a predetermined threshold pressure.
  • confirming positioning of the distal end portion of the hollow conduit in the reference location can include measuring a light emissivity gradient between a sclera of the eye and a retinal pigment epithelium or choroid of the eye.
  • the inserting is performed such that a centerline of the hollow conduit and a surface line tangential to a target surface of the eye can define an angle of entry of between about 0 degrees and about 180 degrees.
  • the medicament can be at least one of a gene therapy, a VEGF, a VEGF inhibitor, a PDGFR inhibitor, a tyrosine kinase inhibitor, a complement inhibitor, an integrin inhibitor, a plasma kallikrein inhibitor, an angiopoietin-2 inhibitor, a Tie-2 agonist, or a combination thereof.
  • the medicament can comprise an integrin inhibitor, axitinib, MK- 409, or aflibercept.
  • the gene therapy can comprise a nucleic acid.
  • the nucleic acid can be DNA or RNA.
  • the nucleic acid can be transcribed to form an anti-sense transcript, wherein the anti-sense transcript inhibits synthesis of an endogenous protein.
  • the nucleic acid can comprise a gene sequence and a promoter sequence.
  • the nucleic acid can be translated to express a protein.
  • the protein can be selected from a VEGF, a VEGF inhibitor, a PDGFR inhibitor, a tyrosine kinase inhibitor, a complement inhibitor, an integrin inhibitor, a plasma kallikrein inhibitor, an angiopoietin-2 inhibitor, a Tie-2 agonist, or a combination thereof.
  • the gene therapy comprises a nucleic acid and a non-viral nanoparticle or a viral vector.
  • a method comprises inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye of a patient, the hollow conduit being in fluidic communication with a medicament container, the medicament container including a medicament, monitoring pressure in at least one of the hollow conduit or the medicament container to detect a change in pressure, and confirming positioning of the distal end portion of the hollow conduit in a first region of the eye, the first region being proximal to the retina of the eye, based on the detection of the change in pressure.
  • the method can include inserting the distal end portion of the hollow conduit a second distance into a region between the SCS and a boundary layer between a retinal layer and vitreous space of the eye, and with the distal end portion of the hollow conduit into the retina of the eye, injecting at least a portion of the medicament into the region between the SCS and the boundary layer between the retinal layer and vitreous space via the hollow conduit.
  • the second distance can be between about 50 micrometers and about 5 mm.
  • the change in pressure can be a drop in pressure. In some embodiments, the change in pressure meets a predefined threshold pressure drop of at least about 5 mmHg.
  • the first region of the eye is in the SCS, between the sclera and the choroid of the eye.
  • a method comprises inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye of a patient, the hollow conduit being in fluidic communication with a medicament container, the medicament container including a medicament, monitoring light emissivity in the anatomical location at the distal end portion of the hollow conduit, and confirming positioning of the distal end portion of the hollow conduit in a first region of the eye, the first region being proximal to the retina of the eye, based on a light emissivity gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • the method can include inserting the distal end portion of the hollow conduit a second distance into a region between the SCS and a boundary layer between a retinal layer and vitreous space of the eye, and with the distal end portion of the hollow conduit into the retina of the eye, injecting at least a portion of the medicament into the region between the SCS and the boundary layer between the retinal layer and vitreous space via the hollow conduit.
  • the second distance can be between about 50 micrometers and about 5 mm.
  • the method can include monitoring pressure in at least one of the hollow conduit or the medicament container to detect a change in pressure.
  • inserting the distal end portion of the hollow conduit the second distance into the region between the SCS and the boundary layer between the retinal layer and vitreous space of the eye can be manual.
  • inserting the distal end portion of the hollow conduit the second distance into the region between the SCS and the boundary layer between the retinal layer and vitreous space of the eye can be automatic.
  • inserting the distal end portion of the hollow conduit the second distance into the region between the SCS and the boundary layer between the retinal layer and vitreous space of the eye is via a motor or mechanical assistance.
  • an apparatus can include a housing, the housing including an actuator and a hollow conduit, the hollow conduit having a distal end, the actuator configured to advance the hollow conduit from a first position to a second position relative to the housing, a measurement device configured to measure a physical property at the distal end of the hollow conduit, and an injection controller including an inner volume fluidically coupled to the hollow conduit, the injection controller configured to deliver a medicament from the inner volume to the distal end of the hollow conduit.
  • the measurement device can include a pressure measurement device configured to measure a pressure at the distal end of the hollow conduit.
  • the pressure measurement device can be a manometer in fluidic communication with the hollow conduit.
  • the measurement device includes a light emissivity measurement device.
  • the light emissivity measurement device can include an emitter, a receiver, and a fiber optic cable, the fiber optic cable running from the distal end of the hollow conduit to the receiver.
  • the pressure device is a manometer
  • the apparatus can further include a puncture cap coupled to the housing, the puncture cap configured to be punctured by a spike to create a fluidic coupling between the manometer and the inner volume of the injection controller.
  • the manometer can be fluidically coupled to the inner volume of the injection controller via one or more pressure tubes.
  • the distal end of the hollow conduit can be beveled, such that the hollow conduit includes a beveled surface.
  • a centerline of the hollow conduit and a line extending distally from the beveled surface can be between about 30 degrees and about 60 degrees.
  • an apparatus includes a housing, the housing including a vacuum space and hollow conduit, the hollow conduit having a distal end.
  • the apparatus includes a vacuum handle, the vacuum handle configured to reduce pressure in the vacuum space and pull layers of an eye toward the distal end of the hollow conduit and secure the layers of the eye to prevent lateral movements of a retina and/or a choroid relative to a sclera.
  • the apparatus includes a measurement device configured to measure a physical property at the distal end of the hollow conduit and an injection controller including an inner volume fluidically coupled to the hollow conduit, the injection controller configured to deliver a medicament from the inner volume to the distal end of the hollow conduit.
  • an apparatus in some aspacts, includes a housing with a pressurized space and hollow conduit, the hollow conduit having a distal end.
  • the apparatus includes a measurement device configured to measure a physical property at the distal end of the hollow conduit.
  • the apparatus further includes an injection controller including an inner volume fluidically coupled to the hollow conduit, the injection controller configured to deliver a medicament from the inner volume to the distal end of the hollow conduit.
  • the pressurized space is in physical contact with the injection controller and is configured to maintain a constant pressure while advancing the injection controller.
  • an apparatus in some aspects, includes a housing, the housing including an actuator and a hollow conduit, the hollow conduit having a distal end, the actuator configured to advance the hollow conduit from a first position to a second position relative to the housing.
  • the apparatus includes a measurement device configured to measure a physical property at the distal end of the hollow conduit.
  • the apparatus includes an injection controller with an inner volume, the inner volume configured to be fluidically coupled to the hollow conduit upon advancement of the hollow conduit via the actuator, the injection controller configured to deliver a medicament from the inner volume to the distal end of the hollow conduit.
  • a method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye of a patient, the hollow conduit being in fluidic communication with a medicament container, the medicament container including a medicament.
  • the method includes monitoring impedance in an anatomical location within the eye at the distal end portion of the hollow conduit.
  • the method includes confirming positioning of the distal end portion of the hollow conduit in a first region of the eye, the first region being proximal to the retina of the eye, based on an electrical impedance gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • the method After confirming disposal of the distal end portion of the hollow conduit in the first region of the eye, the method includes inserting the distal end portion of the hollow conduit a second distance into a region between an SCS and a boundary layer between a retinal layer and vitreous space of the eye. With the distal end portion of the hollow conduit into the retina of the eye, the method includes injecting at least a portion of the medicament into the region between the SCS and the boundary layer between the retinal layer and vitreous space via the hollow conduit.
  • a method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye of a patient, the hollow conduit being in fluidic communication with a medicament container, the medicament container including a medicament.
  • the method includes monitoring acoustic wave measurements in an anatomical location within the eye at the distal end portion of the hollow conduit and confirming positioning of the distal end portion of the hollow conduit in a first region of the eye, the first region being proximal to the retina of the eye, based on an acoustic wave gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • the method After confirming disposal of the the distal end portion of the hollow conduit in the first region of the eye, the method includes inserting the distal end portion of the hollow conduit a second distance into a region between an SCS and a boundary layer between a retinal layer and vitreous space of the eye. With the distal end portion of the hollow conduit in the retina of the eye, the method includes injecting at least a portion of the medicament into the region between the SCS and the boundary layer between the retinal layer and vitreous space via the hollow conduit.
  • a method includes applying a sealing surface on a distal end of an injection apparatus to at least one of a surface of a sclera of an eye or a conjunctiva of the eye, and pushing a first container into a second container to pressurize a liquid within the second container, the second container in fluidic communication with a catheter.
  • the method further includes advancing the catheter through the sealing surface, such that a distal end of the catheter at least partially penetrates an SCS of the eye, the pressurizing of the liquid within the second container causing at least a portion of the liquid to flow from the second container and into the SCS via the catheter due to a reduced backpressure upon entry of the distal end of the catheter into the SCS.
  • the method further includes advancing a hollow conduit, such that a distal end portion of the hollow conduit is distal to a distal end of the catheter, and the distal end of the hollow conduit enters a retina of the eye, the hollow conduit in fluidic communication with a medicament container, the medicament container including a medicament disposed therein.
  • the method includes injecting at least a portion of the medicament into the retina via the hollow conduit.
  • an apparatus can include a housing, a fluid container at least partially disposed in the housing, the fluid container configured to contain a liquid and configured to move indepdendently of the housing, and a medicament container at least partially disposed in the fluid container, the medicament container configured to contain a medicament, the medicament container configured to increase a pressure in the fluid container upon an application of force on the medicament container when the medicament container contains the medicament.
  • the apparatus further includes an outer hollow conduit fixedly coupled to a distal end of the fluid container, the outer hollow conduit fluidically coupled to the fluid container and an inner hollow conduit fixedly coupled to a distal end of the medicament container, the inner hollow conduit fluidically coupled to a volume defined by the medicament container, the inner hollow conduit having a distal end configured to move distally through a lumen defined by the outer hollow conduit upon distal movement by the medicament container.
  • the apparatus further includes an injection controller at least partially disposed in the medicament container, the injection controller configured to push medicament through the inner hollow conduit and move independent of the medicament container.
  • an apparatus can include a sheath having an edge capable of piercing a sclera of an eye, a catheter slidably disposed within a lumen of the sheath and configured to extend beyond a distal end of the sheath, a balloon fixed to a distal end of the catheter, the balloon configured to inflate when a distal end of the catheter extends into at least two adjacent ocular tissue layers, a medicament container configured to contain a medicament, and a hollow conduit slidably coupled to the catheter, the hollow conduit configured to move independently of the catheter.
  • the hollow conduit has a distal end portion, the distal end portion configured to extend beyond the balloon the hollow conduit is fluidically coupled to the medicament container and configured to deliver the medicament to a target region of the eye via the distal end portion of the hollow conduit, the target region distal to the balloon.
  • a method includes at least partially penetrating a sclera of an eye with a distal end of a sheath and advancing a catheter such that a distal end of the catheter enters an SCS of an eye, the catheter disposed in the sheath and moving independently of the sheath.
  • the method further includes inflating a balloon disposed at a distal end of the catheter so as to create a force against a choroid, advancing a hollow conduit such that a distal end of the hollow conduit extends into a retina, the hollow conduit disposed in the catheter and moving independently of the catheter, and injecting a medicament into the retina via the hollow conduit.
  • a method includes inserting an outer sleeve into an eye, the outer sleeve having an inner lumen, inserting a cannula into the eye through the inner lumen of the outer sleeve, the cannula having a distal end with a balloon disposed thereon, inflating the balloon to create tension between two adjacent layers of the eye to secure the balloon therebetween, advancing distally a hollow conduit through the cannula, such that a distal end of the hollow conduit enters a target region of the eye distal to the balloon, the hollow conduit being in fluidic communication with a medicament container, the medicament container including a medicament. With the inflated balloon secured between the two adjacent layers of the eye, the method includes injecting medicament from the medicament container into the target region via the hollow conduit.
  • an apparatus includes a housing, a catheter at least partially disposed in the housing and configured to slidably move within the housing, a hollow conduit fixedly coupled to the catheter and at least partially disposed in the catheter, a medicament container fluidically coupled to the hollow conduit and configured to dispense a medicament to a target region in an eye, and a balloon coupled to a distal end of the catheter.
  • the hollow conduit extends beyond the distal end of the catheter and the balloon, and wherein the balloon is configured to expand in a region proximal to a distal end of the hollow conduit, creating a tension between adjacent layers of the eye.
  • a method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into a first location within an eye, inserting a distal end portion of a catheter of the medical injector a second distance into the eye, such that a balloon coupled to the distal end portion of the catheter is disposed within a region defined between a sclera of the eye and a retina of the eye, the second distance being less than the first distance, and inflating the balloon to at least one of create or expand a space within the region, such that the distal end portion of the hollow conduit changes from the first location to a second location within the eye.
  • the method includes injecting via the hollow conduit a medicament ento the region.
  • FIG. l is a cross-sectional view of an illustration of a human eye.
  • FIG. 2 is a cross-sectional view of portion of the human eye of FIG. 1, taken along the line 2-2.
  • FIGS. 3, 4A, and 4B are cross-sectional views of a portion of the human eye of FIG. 1 taken along the line 3-3, illustrating the suprachoroidal space without the presence of a fluid, the suprachoroidal space with the presence of a fluid, and the retina with the presence of fluid, respectively.
  • FIG. 5 is a schematic illustration of an injection apparatus, according to an embodiment.
  • FIG. 6 is an illustration of an injection apparatus, according to an embodiment.
  • FIGS. 7A-7B are illustrations of an injection apparatus, according to an embodiment.
  • FIG. 8 is an illustration of an injection apparatus, according to an embodiment.
  • FIG. 9 is an illustration of an injection apparatus, according to an embodiment.
  • FIG. 10 is an illustration of an injection apparatus, according to an embodiment.
  • FIG. 11 is a schematic illustration of an injection apparatus, according to an embodiment.
  • FIGS. 12A-12F illustrate a method and apparatus for perfroming an injection into the retinal tissue, according to an embodiment.
  • FIGS. 13A-13O illustrate methods and apparatus for performing an injection into the retinal tissue, according to various embodiments.
  • FIGS. 14A-14D illustrate a method and apparatus for performing an injection into the retinal tissue, according to an embodiment.
  • FIGS. 15A-15E illustrate an injection cover and the placement of the injection cover on the eye, according to an embodiment.
  • FIGS. 16A-16E are illustrations an injection apparatus and a method of operating the injection apparatus, according to an embodiment.
  • FIGS. 17A-17B are illustrations of an injection apparatus, according to an embodiment.
  • FIGS. 18A-18C are illustrations of an injection apparatus, according to an embodiment.
  • FIGS. 19A-19C are illustrations of an injection apparatus and a method of operating the injection apparatus, according to an embodiment.
  • FIG. 20 shows a flow diagram of a method for performing an injection into the retinal tissue, according to an embodiment.
  • FIG. 21 shows a flow diagram of a method for performing an injection into the retinal tissue, according to an embodiment.
  • FIG. 22 shows a flow diagram of a method for performing an injection into the retinal tissue, according to an embodiment.
  • FIG. 23 shows a flow diagram of a method for performing an injection into the retinal tissue, according to an embodiment.
  • FIG. 24 shows a flow diagram of a method for performing an injection into a target region of the eye, according to an embodiment.
  • FIG. 25 shows a flow diagram of a method for performing an injection into a target region of the eye, according to an embodiment.
  • FIG. 26 is a schematic illustration of an injection apparatus, according to an embodiment.
  • FIG. 27 illustrates an injection apparatus, according to an embodiment.
  • FIGS. 28A-28F illustrate methods and apparatus for performing an injection into ocular tissue, according to various embodiments.
  • FIG. 29 shows a flow diagram of a method for performing an injection into a target region of the eye, according to an embodiment.
  • Embodiments described herein relate to systems and methods of retinal injection. These can include systems, devices, and methods to assist in the insertion of a delivery member (e.g., a needle, microneedle, a solid microneedle, a trocar, a catheter, a hollow trephine, cannula, microneedle patch, hollow microneedle, or hollow conduit) into the eye, or more specifically, a retinal space of the eye or a retinal tissue.
  • a delivery member e.g., a needle, microneedle, a solid microneedle, a trocar, a catheter, a hollow trephine, cannula, microneedle patch, hollow microneedle, or hollow conduit
  • systems, devices, and methods described herein are for insertion of a delivery member into a region proximal to the retinal space of the eye or retinal tissue (e.g., between the choroid and the retina, or within the choroid adjacent its distal edge such that inj ectate injected therein could travel towards and into the retina).
  • Human eyes can vary widely in overall size, as well as thickness of various tissue layers.
  • an injectate e.g., a medicament
  • a tissue layer proximal to the vitreous body i.e., pushing the delivery member through a few layers of tissue
  • distal to the vitreous body i.e., pushing the delivery member through the layers of tissue proximal to the vitreous body, the vitreous body, and a few layers of tissue distal to the vitreous body
  • sclera thickness in the human eye can range anywhere from about 350 pm to about 700 pm.
  • Using landmarks in the anatomy of the eye can aid in locating a reference location.
  • Embodiments described herein can include methods and apparatus for detecting a reference location in an eye and injecting an inj ectate into a retinal tissue.
  • the reference location can be in the SCS.
  • the reference location can be the RPE. In some embodiments, the reference location can be in a region between a suprachoroidal space (SCS) and a retinal pigment epithelium (RPE) layer. In some embodiments, the reference location can be between the sclera and the choroid. In some embodiments, the detection of the reference location can be via a pressure change. In some embodiments, the detection of the reference location can be via an emissivity measurement. For example, light can be shined through a pupil and emissivity can be measured, based on how much of the incident light is detected at a distal end of the delivery member. In some embodiments, acoustic measurements can be made to measure tissue water contents. Each tissue in the human eye has a different water content, so water content measurements would change at each interface between layers. In some embodiments, impedance can be measured at various locations to locate the reference location, as electrical resistivity changes from layer to layer.
  • Embodiments described herein can also include devices for measuring the aforementioned properties and delivering inj ectate.
  • the use of the aforementioned properties to locate the reference location and the different tissues of the eye can substantially reduce costs and patient discomfort in the medicament delivery process.
  • Examples of ocular delivery devices are described in U.S. Patent No. 9,539,139 entitled, “Apparatus and Methods for Ocular Injection,” filed May 2, 2014 (“the ‘ 139 patent”), the disclosure of which is incorporated herein by reference in its entirety.
  • a member is intended to mean a single member or a combination of members
  • a material is intended to mean one or more materials, or a combination thereof.
  • proximal and distal refer to the direction closer to and away from, respectively, an operator (e.g., surgeon, physician, nurse, technician, etc.) who would insert the medical device into the patient, with the tip-end (i.e., distal end) of the device inserted inside a patient’s body first.
  • an operator e.g., surgeon, physician, nurse, technician, etc.
  • the tip-end i.e., distal end
  • the end of a delivery member or hollow conduit described herein first inserted inside the patient’s body would be the distal end
  • the opposite end of the microneedle e.g., the end of the medical device being manipulated by the operator
  • the proximal end of the microneedle e.g., the end of the medical device being manipulated by the operator
  • a “set” can refer to multiple features or a singular feature with multiple parts.
  • the set of walls can be considered as one wall with distinct portions, or the set of walls can be considered as multiple walls.
  • the terms “about” and “approximately” generally mean plus or minus 10% of the value stated. For example, about 0.5 would include 0.45 and 0.55, about 10 would include 9 to 11, about 1000 would include 900 to 1100.
  • the terms “delivery member”, “puncture member”, and “puncturing member” are used interchangeably to refer to an article configured to pierce tissue layers and deliver a substance to a target tissue layer, for example, a needle, a microneedle, a solid microneedle, a trocar, a catheter, a hollow trephine, or any other suitable delivery device.
  • the terms “medicament container”, and “medicament containment chamber” are used interchangeably to refer to an article configured to contain a volume of a substance, for example, a medicament.
  • fluid-tight is understood to encompass both a hermetic seal (i.e., a seal that is gas-impervious) as well as a seal that is liquid-impervious.
  • a hermetic seal i.e., a seal that is gas-impervious
  • liquid-impervious is intended to convey that, while total fluid imperviousness is desirable, some minimal leakage due to manufacturing tolerances, or other practical considerations (such as, for example, the pressure applied to the seal and/or within the fluid), can occur even in a “substantially fluid-tight” seal.
  • a “substantially fluid-tight” seal includes a seal that prevents the passage of a fluid (including gases, liquids and/or slurries) therethrough when the seal is maintained at a constant position and at fluid pressures of less than about 5 psig, less than about 10 psig, less than about 20 psig, less than about 30 psig, less than about 50 psig, less than about 75 psig, less than about 100 psig and all values in between.
  • a fluid including gases, liquids and/or slurries
  • a “substantially liquid-tight” seal includes a seal that prevents the passage of a liquid (e.g., a liquid medicament) therethrough when the seal is maintained at a constant position and is exposed to liquid pressures of less than about 5 psig, less than about 10 psig, less than about 20 psig, less than about 30 psig, less than about 50 psig, less than about 75 psig, less than about 100 psig and all values in between.
  • a liquid e.g., a liquid medicament
  • FIGS. 1-4B are include various views of a human eye 10 (with FIGS. 2-4B being cross-sectional views). While specific regions are identified, those skilled in the art will recognize that the proceeding identified regions do not constitute the entirety of the eye 10, rather the identified regions are presented as a simplified example suitable for the discussion of the embodiments herein.
  • the eye 10 includes both an anterior segment 12 (the portion of the eye in front of and including the lens) and a posterior segment 14 (the portion of the eye behind the lens).
  • the anterior segment 12 is bounded by the cornea 16 and the lens 18, while the posterior segment 14 is bounded by the sclera 20 and the lens 18.
  • the anterior segment 12 is further subdivided into the anterior chamber 22, between the iris 24 and the cornea 16, and the posterior chamber 26, between the lens 18 and the iris 24.
  • the cornea 16 and the sclera 20 collectively form a limbus 38 at the point at which they meet.
  • the exposed portion of the sclera 20 on the anterior segment 12 of the eye is protected by a clear membrane referred to as the conjunctiva 45 (see e.g., FIG. 3).
  • Underlying the sclera 20 is the choroid 28 and the retina 27, collectively referred to as retinachoroidal tissue.
  • the retinal pigment epithelium (RPE) 29 is the most outer layer of the retina 27, depicted adjacent to the choroid 28.
  • a vitreous humour 30 (also referred to as the “vitreous” or “vitreous space”) is disposed between a ciliary body 32 (including a ciliary muscle and a ciliary process) and the retina 27.
  • the anterior portion of the retina 27 forms an ora serrata 34.
  • the loose connective tissue, or potential space, between the choroid 28 and the sclera 20 is referred to as the suprachoroid or suprachoroidal space (SCS).
  • FIG. 2 illustrates the cornea 16, which is composed of the epithelium 40, the Bowman’s layer 41, the stroma 42, the Descemet’s membrane 43, and the endothelium 44.
  • FIG. 3 illustrates the sclera 20 with surrounding Tenon’s Capsule 46 or conjunctiva 45, suprachoroidal space 36, choroid 28, and retina 27, substantially without fluid and/or tissue separation in the suprachoroidal space 36 (i.e., the in this configuration, the space is “potential” suprachoroidal space).
  • the sclera 20 has a thickness between about 350 pm and 700 pm.
  • FIG. 3 illustrates sections of the retina 27 including RPE 29, rod and cone outer segments 31, rod and cone nuclei 33, bipolar cells 35, ganglion cells 37, and nerve fibers to the optic nerve 39.
  • FIG. 3 also includes the vitreous humour 30.
  • FIG. 4A illustrates the sclera 20 with the surrounding Tenon’s Capsule 46 or the conjunctiva 45, suprachoroidal space 36, choroid 28, RPE 29, retina 27, with fluid 50 in the SCS 36.
  • FIG. 4B illustrates the sclera 20 with the surrounding Tenon’s Capsule 46 or the conjunctiva 45, suprachoroidal space 36, choroid 28, RPE 29, retina 27, with fluid 51 in the retina 27 (between the RPE 29 and the rod and cone outer segments 31).
  • the term “suprachoroidal space,” or SCS which is synonymous with suprachoroid, or suprachoroidia, describes the space (or volume) and/or potential space (or potential volume) in the region of the eye 10 disposed between the sclera 20 and choroid 28.
  • This region primarily is composed of closely packed layers of long pigmented processes derived from each of the two adjacent tissues; however, a space can develop in this region because of fluid or other material buildup in the suprachoroidal space and the adjacent tissues.
  • the suprachoroidal space can be expanded by fluid buildup because of some disease state in the eye or because of some trauma or surgical intervention.
  • the fluid buildup is intentionally created by the delivery, injection and/or infusion of a drug formulation into the suprachoroid to create and/or expand further the suprachoroidal space 36 (i.e., by disposing a drug formulation therein).
  • This volume may serve as a pathway for uveoscleral outflow (i.e., a natural process of the eye moving fluid from one region of the eye to the other through) and may become a space in instances of choroidal detachment from the sclera.
  • the term “retina” or “retinal space” can include the space (or volume) and/or potential space (or potential volume) between the RPE 29, and (1) the interface between the nerve fibers to the optic nerve 39 and the vitreous body 30, or (2) the photo receptor layer 31.
  • the space between the choroid 28 and the retina 27 can include the space (or volume) and/or potential space (or potential volume) on either side adjacent to the RPE 29 or space within the RPE 29 itself.
  • the dashed line in FIG. 1 represents the equator of the eye 10.
  • the insertion site of any of the microneedles and/or methods described herein is between the equator and the limbus 38 (i.e., in the anterior portion 12 of the eye 10).
  • the insertion site is between about two millimeters and 10 millimeters (mm) posterior to the limbus 38.
  • the insertion site of the microneedle is at about the equator of the eye 10.
  • the insertion site is posterior the equator of the eye 10.
  • FIG. 5 is a schematic illustration of an injection apparatus 100, according to an embodiment.
  • the injection apparatus 100 includes an injection housing 110.
  • the injection housing 110 contains a hollow conduit 120 and an actuator 130 coupled to the hollow conduit 120.
  • the injection apparatus 100 further includes a measurement device 140, a medicament container 150, and an injection controller 160.
  • the measurement device 140 is coupled to the injection housing 110 and measures a parameter at a distal end of the hollow conduit 120.
  • the injection controller 160 controls delivery of injectate (e.g., medicament) from the medicament container 150 to the hollow conduit 120 and out of the distal end of the hollow conduit 120.
  • injectate e.g., medicament
  • the injection housing 110 can be composed of a rigid material. In some embodiments, the injection housing 110 can be composed of a flexible material. In some embodiments, the injection housing 110 can be composed of a polymer, glass, a metal, a tubing material (e.g., Tygon®), or any combination thereof.
  • the injection housing 110 can have a volume of at least about 1 ml, at least about 2 ml, at least about 3 ml, at least about 4 ml, at least about 5 ml, at least about 6 ml, at least about 7 ml, at least about 8 ml, at least about 9 ml, at least about 10 ml, at least about 20 ml, at least about 30 ml, at least about 40 ml, at least about 50 ml, at least about 60 ml, at least about 70 ml, at least about 80 ml, at least about 90 ml, at least about 100 ml, at least about 200 ml, at least about 300 ml, or at least about 400 ml.
  • the injection housing 110 can have a volume of no more than about 500 ml, no more than about 400 ml, no more than about 300 ml, no more than about 200 ml, no more than about 100 ml, no more than about 90 ml, no more than about 80 ml, no more than about 70 ml, no more than about 60 ml, no more than about 50 ml, no more than about 40 ml, no more than about 30 ml, no more than about 20 ml, no more than about 10 ml, no more than about 9 ml, no more than about 8 ml, no more than about 7 ml, no more than about 6 ml, no more than about 5 ml, no more than about 4 ml, no more than about 3 ml, or no more than about 2 ml.
  • Combinations of the above-referenced volumes of the injection housing 110 are also possible (e.g., at least about 1 ml and no more than about 500 ml or at least about 20 ml and no more than about 100 ml), inclusive of all values and ranges therebetween.
  • the injection housing 110 can have a volume of about 1 ml, about 2 ml, about 3 ml, about 4 ml, about 5 ml, about 6 ml, about 7 ml, about 8 ml, about 9 ml, about 10 ml, about 20 ml, about 30 ml, about 40 ml, about 50 ml, about 60 ml, about 70 ml, about 80 ml, about 90 ml, about 100 ml, about 200 ml, about 300 ml, about 400 ml, or about 500 ml.
  • the hollow conduit 120 can be adjustable to move such that the distal end of the hollow conduit 120 moves or extends from a first position relative to the housing 110 to a second position (different from the first position) relative to the housing 110.
  • the distal end of the hollow conduit 120 can protrude from the housing 110 a first distance while in the first position and a second distance while in the second position.
  • the distal end of the hollow conduit 120 can be flush with the housing 110.
  • the first distance and/or the second distance can be at least about 0.5 mm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, at least about 9.5 mm, at least about 10 mm, at least about 11 mm, at least about 12 mm, at least about 13 mm, at least about 14 mm, at least about 15 mm, at least about 16 mm, at least about 17 mm, at least about 18 mm, or at least about 19 mm.
  • the first distance and/or the second distance can be no more than about 20 mm, no more than about 19 mm, no more than about 18 mm, no more than about 17 mm, no more than about 16 mm, no more than about 15 mm, no more than about 14 mm, no more than about 13 mm, no more than about 12 mm, no more than about 11 mm, no more than about 10 mm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, no more than about 1.5 mm, or no more than about 1 mm
  • Combinations of the above-referenced values for the first distance and/or the second distance are also possible (e.g., at least about 0.5 mm and no more than about 20 mm or at least about 2 mm and no more than about 10 mm), inclusive of all values and ranges therebetween.
  • the first distance and/or the second distance can be about 0.5 mm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about 7.5 mm, about 8 mm, about 8.5 mm, about 9 mm, about 9.5 mm, about 10 mm, about 11 mm, about 12 mm, about 13 mm, about 14 mm, about 15 mm, about 16 mm, about 17 mm, about 18 mm, about 19 mm, or about 20 mm.
  • the hollow conduit 120 can be fluidically coupled to the medicament container 150.
  • the injection apparatus 100 can be operable in a first configuration, wherein the hollow conduit 120 is fluidically isolated from the medicament container 150 and a second configuration, wherein the hollow conduit 120 is fluidically coupled to the medicament container 150.
  • a seal can be broken by the user, bringing the hollow conduit 120 and the medicament container 150 into fluidic communication with one another.
  • the hollow conduit 120 can be a needle, a microneedle (e.g., 25 gauge, 27 gauge, 30 gauge, or smaller), a solid microneedle, a trocar, a catheter, a hollow trephine, or any other suitable delivery device.
  • the hollow conduit 120 can be a solid microneedle that can be inserted into a portion of the eye and left in the eye.
  • the solid microneedle can include medicament that can be delivered into the eye over an extended period of time (e.g., about 1 day, about 5 days, about 10 days, about 15 days, about 20 days, about 25 days, about 30 days, about 60 days, about 90 days, about 120 days, about 150 days, about 180 days, about 6 months, or about 1 year, inclusive of all values and ranges therebetween).
  • the distal end of the hollow conduit 120 can be tapered, as described in the ‘ 139 patent.
  • the hollow conduit 120 can be a flexible conduit.
  • the hollow conduit 120 also has a proximal end (i.e., an end closer to the user).
  • the proximal end of the hollow conduit 120 can be disposed in the housing 110.
  • the proximal end of the hollow conduit 120 can be disposed in the medicament container 150.
  • the hollow conduit 120 can move independently of the actuator 130. In other words, the hollow conduit 120 can move from a first position to a second position relative to the housing 110 while the actuator 130 stays in place relative to the housing 110. In some embodiments, the hollow conduit 120 and the actuator 130 can move together, relative to the housing 110.
  • the hollow conduit 120 and the actuator 130 can move the same distance or substantially the same distance relative to the housing 110.
  • the hollow conduit 120 can move a first distance relative to the housing 110 while the actuator 130 moves a second distance relative to the housing 110.
  • the first distance can be larger than the second distance.
  • the first distance can be smaller than the second distance.
  • the hollow conduit 120 can have a beveled end.
  • the opening at the distal end of the hollow conduit 120 can have a first side adjacent to the tip of the bevel and a second side adjacent to the heel of the bevel.
  • the first side can be distal to the second side by a bevel distance. Tuning the bevel distance can aid in proper disposal of medicament. If the bevel distance is too small, this can cause the effective opening at the distal end of the hollow conduit 120 to be small enough, such that application of a small pressure is not sufficient to dispense medicament.
  • the bevel distance can be at least about 1 pm, at least about 2 pm, at least about 3 pm, at least about 4 pm, at least about 5 pm, at least about 6 pm, at least about 7 pm, at least about 8 pm, at least about 9 pm, at least about 10 pm, at least about 20 pm, at least about 30 pm, at least about 40 pm, at least about 50 pm, at least about 60 pm, at least about 70 pm, at least about 80 pm, at least about 90 pm, at least about 100 pm, at least about 200 pm, at least about 300 pm, at least about 400 pm, at least about 500 pm, at least about 600 pm, at least about 700 pm, at least about 800 pm, or at least about 900 pm.
  • the bevel distance can be no more than about 1 mm, no more than about 900 pm, no more than about 800 pm, no more than about 700 pm, no more than about 600 pm, no more than about 500 pm, no more than about 400 pm, no more than about 300 pm, no more than about 200 pm, no more than about 100 pm, no more than about 90 pm, no more than about 80 pm, no more than about 70 pm, no more than about 60 pm, no more than about 50 pm, no more than about 40 pm, no more than about 30 pm, no more than about 20 pm, no more than about 10 pm, no more than about 9 pm, no more than about 8 pm, no more than about 7 pm, no more than about 6 pm, no more than about 5 pm, no more than about 4 pm, no more than about 3 pm, or no more than about 2 pm.
  • the bevel distance can be about 1 pm, about 2 pm, about 3 pm, about 4 pm, about 5 pm, about 6 pm, about 7 pm, about 8 pm, about 9 pm, about 10 pm, about 20 pm, about 30 pm, about 40 pm, about 50 pm, about 60 pm, about 70 pm, about 80 pm, about 90 pm, about 100 pm, about 200 pm, about 300 pm, about 400 pm, about 500 pm, about 600 pm, about 700 pm, about 800 pm, about 900 pm, or about 1 mm.
  • the hollow conduit 120 can have a beveled end with a bevel angle (i.e., an angle created between a centerline of the hollow conduit 120 and a line extending distally from the beveled surface at the distal end of the hollow conduit 120) of at least about 5 degrees, at least about 10 degrees, at least about 15 degrees, at least about 20 degrees, at least about 25 degrees, at least about 30 degrees, at least about 35 degrees, at least about 40 degrees, at least about 45 degrees, at least about 50 degrees, at least about 55 degrees, at least about 60 degrees, at least about 65 degrees, at least about 70 degrees, at least about 75 degrees, at least about 80 degrees, or at least about 85 degrees.
  • a bevel angle i.e., an angle created between a centerline of the hollow conduit 120 and a line extending distally from the beveled surface at the distal end of the hollow conduit 120
  • the bevel angle can be no more than about 90 degrees, no more than about 85 degrees, no more than about 80 degrees, no more than about 75 degrees, no more than about 70 degrees, no more than about 65 degrees, no more than about 60 degrees, no more than about 55 degrees, no more than about 50 degrees, no more than about 45 degrees, no more than about 40 degrees, no more than about 35 degrees, no more than about 30 degrees, no more than about 25 degrees, no more than about 20 degrees, no more than about 15 degrees, or no more than about 10 degrees. Combinations of the above-referenced bevel angles are also possible (e.g., at least about 5 degrees and no more than about 90 degrees or at least about 45 degrees and no more than about 75 degrees), inclusive of all values and ranges therebetween.
  • the bevel angle can be about 5 degrees, about 10 degrees, about 15 degrees, about 20 degrees, about 25 degrees, about 30 degrees, about 35 degrees, about 40 degrees, about 45 degrees, about 50 degrees, about 55 degrees, about 60 degrees, about 65 degrees, about 70 degrees, about 75 degrees, about 80 degrees, or about 85 degrees.
  • a bevel at the distal end of the hollow conduit 120 can make the hollow conduit 120 sharper, such that it can more easily penetrate the sclera without causing additional deformation to the sclera.
  • the distal end of the hollow conduit 120 can be unbeveled.
  • the actuator 130 can be coupled to an inner wall of the housing 110. In some embodiments, the actuator 130 can be coupled to an inner wall of the medicament container 150. In some embodiments, the actuator 130 can move within the housing 110 and/or the medicament container 150 from a first position to a second position. In other words, the actuator 130 can move a distance relative to the housing 110 and/or the medicament container 150.
  • the actuator 130 can move a distance relative to the housing 110 and/or the medicament container 150 of at least about 0.5 mm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, at least about 9.5 mm, at least about 10 mm, at least about 11 mm, at least about 12 mm, at least about 13 mm, at least about 14 mm, at least about 15 mm, at least about 16 mm, at least about 17 mm, at least about 18 mm, or at least about 19 mm.
  • the actuator 130 can move a distance relative to the housing 110 and/or the medicament container 150 of no more than about 20 mm, no more than about 19 mm, no more than about 18 mm, no more than about 17 mm, no more than about 16 mm, no more than about 15 mm, no more than about 14 mm, no more than about 13 mm, no more than about 12 mm, no more than about 11 mm, no more than about 10 mm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, no more than about 1.5 mm, no more
  • Combinations of the above-referenced values for the distance moved by the actuator 130 relative to the housing 110 and/or the medicament container 150 are also possible (e.g., at least about 0.5 mm and no more than about 20 mm or at least about 2 mm and no more than about 10 mm), inclusive of all values and ranges therebetween.
  • the actuator 130 can move a distance relative to the housing 110 and/or the medicament container 150 of about 0.5 mm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about 7.5 mm, about 8 mm, about 8.5 mm, about 9 mm, about 9.5 mm, about 10 mm, about 11 mm, about 12 mm, about 13 mm, about 14 mm, about 15 mm, about 16 mm, about 17 mm, about 18 mm, about 19 mm, or about 20 mm.
  • the actuator 130 can be operated automatically. In some embodiments, the actuator 130 can be activated manually. In some embodiments, the actuator 130 can be connected to a micrometer. In some embodiments, the actuator 130 can be activated by the manual adjustment of a micrometer. In some embodiments, the actuator 130 can be operated by mechanical assistance. In some embodiments, the actuator 130 can be operated manually with mechanical assistance. In some embodiments, the actuator 130 can be operated automatically with mechanical assistance. In some embodiments, the actuator 130 can be operated by a motor. In some embodiments, the actuator 130 can be operated by a piezomotor. In some embodiments, the apparatus 100 can include an adapter (not shown) to act as an interface between the hollow conduit 120 and the actuator 130.
  • the adapter can be a press fit adapter to ensure a tight fit between the hollow conduit 120 and the actuator 130.
  • the press fit adapter can aid in preventing proximal and distal movements of the hollow conduit 120.
  • the press fit adapter can also aid in preventing wobbling (i.e., side-to-side movements) of the hollow conduit 120.
  • the measurement device 140 measures a parameter at the distal end and/or near the distal end of the hollow conduit 120 to detect the SCS, or any other desired reference location.
  • the parameter can be a physical parameter.
  • the parameter can include pressure, emissivity, acoustic vibrations, and/or impedance.
  • the measurement device 140 can measure the parameter at a measurement point, the measurement point proximal to the distal end of the hollow conduit 120.
  • the measurement point can be proximal to the distal end of the hollow conduit 120 by at least about 0.5 mm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, or at least about 9.5 mm.
  • the measurement point can be proximal to the distal end of the hollow conduit 120 by no more than about 10 mm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, no more than about 1.5 mm, or no more than about 1 mm.
  • Combinations of the above-referenced distances between the distal end of the hollow conduit 120 and the measurement point’s location, proximal to the distal end of the hollow conduit 120 are also possible (e.g., at least about 0.5 mm and no more than about 10 mm), inclusive of all values and ranges therebetween.
  • the measurement point can be proximal to the distal end of the hollow conduit 120 by about 0.5 mm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about 7.5 mm, about 8 mm, about 8.5 mm, about 9 mm, about 9.5 mm, or about 10 mm.
  • the measurement device 140 can include a manometer, a barometer, a pressure tube, a piezometer, a bourdon gauge, a diaphragm pressure gauge, a micro manometer, or any combination thereof.
  • the measurement device 140 can be fluidically coupled to the distal end of the hollow conduit 120.
  • the measurement device 140 can include a digital pressure transducer.
  • the measurement device 140 when measuring pressure, can be rated for accuracy to within about 0.1 mmHg, about 0.2 mmHg, about 0.3 mmHg, about 0.4 mmHg, about 0.5 mmHg, about 0.6 mmHg, about 0.7 mmHg, about 0.8 mmHg, about 0.9 mmHg, about 1 mmHg, about 2 mmHg, about 3 mmHg, about 4 mmHg, about 5 mmHg, about 6 mmHg, about 7 mmHg, about 8 mmHg, about 9 mmHg, or about 10 mmHg, inclusive of all values and ranges therebetween.
  • the measurement device 140 can measure light emissivity at and/or near the distal end of the hollow conduit 120.
  • the measurement device 140 can include a fiber optic cable with an emitter and receiver.
  • the emitter can emit light through the pupil and the fiber optic cable can absorb an amount of light at the distal end of the hollow conduit 120, transferring the light to the receiver.
  • the emissivity can then be calculated as a fraction of the light intensity at the receiver to the intensity of the light emitted from the emitter.
  • Each layer of the eye absorbs and reflects light differently.
  • the measurement device 140 can measure an acoustic wave gradient at and/or near the distal end of the hollow conduit 120.
  • the measurement device 140 can include an acoustic wave emitter and acoustic wave receiver.
  • the tissue layers in the eye have differing water contents. Acoustic waves can be emitted from the sound emitter near the distal end of the hollow conduit 120 and interfaces between tissue layers can be detected based on how quickly the acoustic waves return to the acoustic wave receiver.
  • the measurement device 140 can measure an electrical impedance gradient at and/or near the distal end of the hollow conduit 120.
  • the measurement device 140 can include an ohmmeter.
  • the measurement device 140 can measure an ionic current impedance gradient at and/or near the distal end of the hollow conduit 120.
  • the measurement device 140 can measure a resistivity gradient at and/or near the distal end of the hollow conduit 120. Tissue layers in the eye can have slightly different impedance or resistivity values, and detection of these gradients can aid in determining reference location in the eye.
  • the medicament container 150 can include a reservoir that can contain an inj ectate or a medicament. As shown, the medicament container 150 is external to the injection housing 110. In some embodiments, the housing 110 can act as the medicament container 150. In some embodiments, the medicament container 150 can be an additional vessel inside the housing 110. In some embodiments, the injection apparatus 100 can include multiple medicament containers 150. For example, a first vial containing a medicament can be provided separately and attached externally to the housing 110 and medicament can transfer from the first vial to a second vial housed within the housing 110. In some embodiments, the medicament container 150 can include a syringe.
  • the medicament container 150 can have a volume of at least about 0.1 mL, at least about 0.2 mL, at least about 0.3 mL, at least about 0.4 mL, at least about 0.5 mL, at least about 0.6 mL, at least about 0.7 mL, at least about 0.8 mL, at least about 0.9 mL, at least about 1 mL, at least about 2 mL, at least about 3 mL, at least about 4 mL, at least about 5 mL, at least about 6 mL, at least about 7 mL, at least about 8 mL, at least about 9 mL, at least about 10 mL, at least about 20 mL, at least about 30 mL, at least about 40 mL, at least about 50 mL, at least about 60 mL, at least about 70 mL, at least about 80 mL, at least about 90 mL, at least about 100 mL, at least about 200 mL
  • the medicament container 150 can have a volume of no more than about 500 mL, no more than about 400 mL, no more than about 300 mL, no more than about 200 mL, no more than about 100 mL, no more than about 90 mL, no more than about 80 mL, no more than about 70 mL, no more than about 60 mL, no more than about 80 mL, no more than about 70 mL, no more than about 60 mL, no more than about 50 mL, no more than about 40 mL, no more than about 90 mL, no more than about 80 mL, no more than about 70 mL, no more than about 60 mL, no more than about 50 mL, no more than about 40 mL, no more than about 30 mL, no more than about 20 mL, no more than about 10 mL, no more than about 9 mL, no more than about 8 mL, no more than about 7 mL, no more than about 6
  • Combinations of the above-referenced volumes of the medicament container are also possible (e.g., at least about 1 mL and no more than about 500 mL or at least about 50 mL and no more than about 100 mL), inclusive of all values and ranges therebetween.
  • the medicament container 150 can have a volume of about 0.1 mL, about 0.2 mL, about 0.3 mL, about 0.4 mL, about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1 mL, about 2 mL, about 3 mL, about 4 mL, about 5 mL, about 6 mL, about 7 mL, about 8 mL, about 9 mL, about 10 mL, about 20 mL, about 30 mL, about 40 mL, about 50 mL, about 60 mL, about 70 mL, about 80 mL, about 90 mL, about 100 mL, about 200 mL, about 300 mL, about 400 mL, or about 500 mL.
  • the injection controller 160 can be automatic. In some embodiments, the injection controller 160 can be manual. In some embodiments, the injection controller 160 can include a syringe plunger. In some embodiments, the injection controller 160 can be disposed in the medicament container 150. As shown, the injection controller 160 is external to the housing 110. In some embodiments, the injection controller 160 can be disposed in the housing 110.
  • the housing 110 can be brought to a location close to the eye, such that the distal end of the hollow conduit 120 is near the outer surface of the eye.
  • the distal end of the hollow conduit 120 is then advanced a first distance relative to the outer surface of the eye into the reference location in the eye.
  • the actuator 130 can advance the distal end of the hollow conduit 120 to the reference location.
  • the reference location is detected based on measurements done by the measurement device 140. In some emboduiments, the reference location is detected based on a sudden change in pressure detected by the measurement device 140. In some embodiments, the reference location is detected based on a change in emissivity detected by the measurement device 140.
  • the actuator 130 can advance the hollow conduit 120 a second distance into the eye, relative to the outer surface of the eye, such that the distal end of the hollow conduit 120 reaches the injection region.
  • the injection region can be between the SCS and a boundary layer between a retinal layer and vitreous space of the eye.
  • the injection region can be between two layers of retinal tissue (e.g., retinal pigment epithelium and photoreceptor).
  • the second distance can be between about 50 pm and about 1 cm from the reference location.
  • the second distance can be at least about 50 pm, at least about 100 pm, at least about 150 pm, at least about 200 pm, at least about 250 pm, at least about 300 pm, at least about 350 pm, at least about 400 pm, at least about 450 pm, at least about 500 pm, at least about 550 pm, at least about 600 pm, at least about 650 pm, at least about 700 pm, at least about 750 pm, at least about 800 pm, at least about 850 pm, at least about 900 pm, at least about 950 pm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm,
  • the second distance can be no more than about 1 cm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, no more than about 1.5 mm, no more than about 1 mm, no more than about 950 pm, no more than about 900 pm, no more than about 850 pm, no more than about 800 pm, no more than about 750 pm, no more than about 700 pm, no more than about 650 pm, no more than about 600 pm, no more than about 550 pm, no more than about 500 pm, no more than about 450 pm, no more than
  • Combinations of the above-referenced values of the second distance from the reference location are also possible (e.g., at least about 50 pm and no more than about 1 cm or at least about 100 pm and no more than about 500 pm), inclusive of all values and ranges therebetween.
  • the second distance can be about 50 gm, about 100 gm, about 150 gm, about 200 pm, about 250 gm, about 300 gm, about 350 gm, about 400 gm, about 450 gm, about 500 gm, about 550 gm, about 600 gm, about 650 gm, about 700 gm, about 750 gm, about 800 gm, about 850 pm, about 900 gm, about 950 gm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about 7.5 mm, about 8 mm, about 8.5 mm, about 9 mm, about 9.5 mm, or about 1 cm from the reference location.
  • the placement of the distal end of the hollow conduit 120 into the injection region can be done with a high confidence level, due to the proximity of the injection region to the reference location.
  • the hollow conduit 120 can be manually advanced the second distance into the eye once the reference location is detected.
  • the inj ection controller 160 propels inj ectate from the medicament container 150 to the inj ection region via the hollow conduit 120.
  • the hollow conduit 120 can be removed from the eye.
  • a kit that includes an injection apparatus for delivering a medicament to an injection region of a target tissue can include all or parts of the concepts described herein.
  • a kit can include an injection apparatus (e.g., the injection apparatus 100, or any other injection apparatus described herein), a housing that can include, for example, a hollow conduit (e.g., the hollow conduit 120 or any other hollow conduit described herein) and an actuator (e.g., the actuator 130 or any other actuator described herein), a measurement device (e.g., the measurement device 140, or any other measurement device described herein), a medicament container (e.g., the medicament container 150 or any other medicament container described herein), an injection controller (e.g., the injection controller 160 or any other injection controller described herein), and/or any other device or apparatus configured to facilitate delivery of the medicament to the target tissue, for example, the eye.
  • an injection apparatus e.g., the injection apparatus 100, or any other injection apparatus described herein
  • a housing can include, for example, a hollow conduit (e
  • FIG. 6 is an illustration of an injection apparatus 200, according to an embodiment.
  • the injection apparatus 200 includes a housing 210, a hollow conduit 220, a pressure measurement device 240, a medicament container 250, and an injection controller 260.
  • the injection apparatus 200 also includes an actuator (not shown), contained within the housing 210.
  • the pressure measurement device 240 is fluidically coupled to the housing 210 via a pressure tube 210.
  • the housing 210, the hollow conduit 220, the actuator, the pressure measurement device 240, the medicament container 250, and the injection controller 260 can be the same or substantially similar to the housing 110, the hollow conduit 120, the actuator 130, the pressure measurement device 140, the medicament container 150, and the injection controller 160, respectively, as described above with reference to FIG. 5.
  • certain aspects of the housing 210, the hollow conduit 220, the actuator, the pressure measurement device 240, the medicament container 250, and the injection controller 260 are not described in greater detail herein.
  • the housing 210 can include a disposable cartridge 211 loaded into a reusable sheath 212.
  • the disposable cartridge 211 can act as an adapter between the hollow conduit 220 and the housing 210.
  • the disposable cartridge 211 can be loaded into the reusable sheath 212 via threading.
  • the disposable cartridge 211 can be loaded into the reusable sheath 212 via a latch coupling.
  • the disposable cartridge 211 can be loaded into the reusable sheath 212 via a coupling.
  • the coupling can be a latch coupling.
  • the disposable cartridge 211 can be removable by squeezing coupled ridges such that the disposable cartridge 211 can be removed from the reusable sheath 212 (i.e., a “childproof’ lock).
  • the hollow conduit 220 can be disposable. In some embodiments, the hollow conduit 220 and the disposable cartridge 211 can be permanently or irreversibly coupled together (e.g., via an adhesive). In some embodiments, the hollow conduit 220 can be independently removable from the disposable cartridge 211, such that the hollow conduit 220 can be removed and replaced more often than the disposable cartridge 211.
  • the pressure measurement device 240 is fluidically coupled to a distal end of the hollow conduit 220. This fluidic coupling is via the pressure tube 242. In some embodiments, the pressure tube 242 can be primed with fluid, such as the inj ectate.
  • the pressure tube 242 can be primed with fluid prior to the insertion of the distal end of the hollow conduit 220 into the human eye.
  • the injection controller 260 can push inj ectate through an injection tube 251 to prime the pressure tube 242.
  • the injection tube 251 is fluidically coupled to the pressure tube 242.
  • the pressure measurement device 240 and the pressure tube 242 can act as a manometer after the pressure tube 242 is primed with fluid. Based on the fluid coupling between the distal end of the hollow conduit 220 and the pressure measurement device 240, the pressure at the distal end of the hollow conduit 220 can be determined via the reading of the pressure measurement device 240.
  • the pressure measurement device 240 can include a digital pressure transducer.
  • the medicament container 250 and the injection controller 260 can be tethered or physically coupled to the housing 210. In some embodiments, the medicament container 250 and the injection controller 260 can be tethered or physically coupled to the pressure measurement device 240. In some embodiments, the injection controller 260 can be manually operated. Manual operation of the injection controller 260 can enable doctor- or operator- controlled injection and tactile feedback of speed and pressure. In some embodiments, the injection controller 260 can be automatically actuated.
  • the housing 210 can be brought to a location close to the eye, such that the distal end of the hollow conduit 220 is near the outer surface of the eye.
  • the distal end of the hollow conduit 220 is then advanced a first distance into the reference location in the eye.
  • the actuator can advance the distal end of the hollow conduit 220 to the reference location.
  • the reference location is detected based on pressure measurements done by the pressure measurement device 240, which is fluidically coupled to the distal end of the hollow conduit 220 via the pressure tube 242.
  • the actuator can advance the hollow conduit 220 a second distance into the eye, such that the distal end of the hollow conduit 220 reaches the injection region.
  • the injection controller 260 propels inj ectate from the medicament container 250 to the injection region via the injection tube 251 and the hollow conduit 220. After the injection, the hollow conduit 220 can be removed from the eye.
  • FIGS 7 A and 7B show an illustration of an injection apparatus 300, according to an embodiment.
  • FIG. 7 A shows a full view of the injection apparatus 300 in close proximity to a human eye 10
  • FIG. 7B shows an interface between portions of the injection apparatus 300 and the human eye 10 in greater detail.
  • the injection apparatus 300 includes a hollow conduit 320, an adapter 322, a puncture membrane 325, a conjunctiva depressor 326, an actuator 330, a pressure measurement device 340 with a pressure tube 342, a relief valve 344, and a T-adapter 345, a medicament container 350 with an injection tube 351, and an injection controller 360.
  • the hollow conduit 320, the actuator 330, the pressure measurement device 340, the pressure tube 342, the medicament container 350, the injection tube 351, and the injection controller 360 can be the same or substantially similar to the hollow conduit 220, the actuator, the pressure measurement device 240, the pressure tube 242, the medicament container 250, the injection tube 251, and the injection controller 260, respectively, as described above with reference to FIG. 6.
  • certain aspects of the hollow conduit 320, the actuator 330, the pressure measurement device 340, the pressure tube 342, the medicament container 350, the injection tube 351, and the injection controller 360 are not described in greater detail herein.
  • the adapter 322 can act as a sheath to hold the hollow conduit 320.
  • the adapter 322 can be press fit to ensure a tight fit between the hollow conduit 320 and the T-adapter 345.
  • an outer surface of the adapter 322 can form a seal with an inner surface of the T-adapter 345.
  • the adapter 322 can be composed of a polymer.
  • the adapter 322 can be composed of a rigid material.
  • the hollow conduit 320 can be fixed in place in the adapter 322 and fluidically coupled to the pressure line 342.
  • the puncture membrane 325 can aid in sealing an interface between the hollow conduit 320 and the eye 10. This can aid in preventing leakage of fluid from the hollow conduit 320 or from the adapter 322.
  • the puncture membrane 325 can be composed of an elastomer material.
  • the puncture membrane 325 can have a thickness of at least about 100 pm, at least about 200 pm, at least about 300 pm, at least about 400 pm, at least about 500 pm, at least about 600 pm, at least about 700 pm, at least about 800 pm, at least about 900 pm, at least about 1 mm, at least about 2 mm, at least about 3 mm, or at least about 4 mm.
  • the puncture membrane 325 can have a thickness of no more than about 5 mm, no more than about 4 mm, no more than about 3 mm, no more than about 2 mm, no more than about 1 mm, no more than about 900 pm, no more than about 800 pm, no more than about 700 pm, no more than about 600 pm, no more than about 500 pm, no more than about 400 pm, no more than about 300 pm, or no more than about 200 pm. Combinations of the above-referenced thicknesses of the puncture membrane 325 are also possible (e.g., at least about 100 pm and no more than about 5 mm or at least about 1 mm and no more than about 3 mm), inclusive of all values and ranges therebetween.
  • the puncture membrane 325 can have a thickness of about 100 pm, about 200 pm, about 300 pm, about 400 pm, about 500 pm, about 600 pm, about 700 pm, about 800 pm, about 900 pm, about 1 mm, about 2 mm, about 3 mm, about 4 mm, or about 5 mm.
  • the conjunctiva depressor 326 can be opened to spread the patient’s conjunctiva prior to insertion of the hollow conduit 320 into the eye.
  • the conjunctiva depressor 326 can include a top portion that moves upward and a bottom portion that moves downward.
  • the conjunctiva depressor 326 can open to create a gap of at least about 100 pm, at least about 200 pm, at least about 300 pm, at least about 400 pm, at least about 500 pm, at least about 600 pm, at least about 700 pm, at least about 800 pm, at least about 900 pm, at least about 1 mm, at least about 2 mm, at least about 3 mm, at least about 4 mm, at least about 5 mm, at least about 6 mm, at least about 7 mm, at least about 8 mm, at least about 9 mm, at least about 1 cm, or at least about 2 cm.
  • the conjunctiva depressor 326 can open to create a gap of no more than about 3 cm, no more than about 2 cm, no more than about 1 cm, no more than about 9 mm, no more than about 8 mm, no more than about 7 mm, no more than about 6 mm, no more than about 5 mm, no more than about 4 mm, no more than about 3 mm, no more than about 2 mm, no more than about 1 mm, no more than about 900 pm, no more than about 800 pm, no more than about 700 pm, no more than about 600 pm, no more than about 500 pm, no more than about 400 pm, no more than about 300 pm, or no more than about 200 pm.
  • the conjunctiva depressor 326 can open to create a gap of about 100 pm, about 200 pm, about 300 pm, about 400 pm, about 500 pm, about 600 pm, about 700 pm, about 800 pm, about 900 pm, about 1 mm, about 2 mm, about 3 mm, about 4 mm, about 5 mm, about 6 mm, about 7 mm, about 8 mm, about 9 mm, about
  • the actuator 330 is coupled to the T-adapter 345. In some embodiments, a portion of the actuator 330 can be further advanced into the T-adapter 345 to make contact with the adapter 322. A portion of the actuator 330 can be yet further advanced to advance the hollow conduit 320. In some embodiments, the actuator 330 can be a micrometer. In some embodiments, the actuator 330 can be manually operated. In some embodiments, the actuator 330 can be operated by hand. In some embodiments, the actuator 330 can be automatically operated.
  • the actuator 330 can have a lateral range of motion (i.e., right-to- left motion in FIG. 7B) of at least about 5 mm, at least about 6 mm, at least about 7 mm, at least about 8 mm, at least about 9 mm, at least about 1 cm, at least about 2 cm, at least about 3 cm, or at least about 4 cm. In some embodiments, the actuator 330 can have a lateral range of motion of no more than about 5 cm, no more than about 4 cm, no more than about 3 cm, no more than about
  • the actuator 330 can have a lateral range of motion of about 5 mm, about 6 mm, about 7 mm, about 8 mm, about 9 mm, at least about 1 cm, about 2 cm, about 3 cm, about 4 cm, or about 5 cm.
  • the pressure measurement device 340 acts as a manometer.
  • the T-adapter 345 couples the pressure tube 342, the adapter 322, and the actuator 330.
  • a relief valve 344 is also included in the pressure measurement device 340 for venting or sealing the pressure tube 342.
  • the medicament container 350 is a syringe that includes inj ectate 352.
  • the inj ectate 352 can be medicament.
  • the inj ectate 352 can include triamcinolone acetonide, a vascular endothelial growth factor (VEGF), a VEGF inhibitor, or a combination thereof.
  • VEGF vascular endothelial growth factor
  • the inj ectate 352 can include any of the medicaments disclosed in the ‘ 139 patent.
  • the injection controller 360 is a syringe plunger.
  • the injection controller 360 can include a sealing device (e.g., an O-ring and/or a gasket) disposed around a perimeter of the injection controller 360 to create a seal with an inner wall of the medicament container 350.
  • the medicament comprises an anti-inflammatory compound, a tyrosine kinase inhibitor, an integrin inhibitor, a complement inhibitor, a VEGF inhibitor, and/or a gene therapy.
  • the medicament comprises an avP3 inhibitor, an avP5 inhibitor, an avpi inhibitor, an a4pi inhibitor, or an a4p7 inhibitor.
  • the medicament comprises axitinib, MK-409, or aflibercept.
  • the medicament comprises a gene therapy.
  • the gene therapy comprises a nucleic acid.
  • the nucleic acid is DNA.
  • the nucleic acid is RNA.
  • the nucleic acid is transcribed to form transcripts.
  • at least one transcript is an anti-sense transcript.
  • the anti-sense transcript inhibits the synthesis of an endogenous protein.
  • the anti-sense transcript inhibits an endogenous protein with a dominant-negative mutation.
  • the nucleic acid molecule comprises a gene sequence and a promoter sequence. In embodiments, the nucleic acid is less than 10 kb, less than 20 kb, or less than 30 kb.
  • the nucleic acid is translated to a protein.
  • the nucleic acid encodes a protein selected from the group consisting of a cytokine, chemokine, a growth factor, an anti-angiogenesis factor, and an antibody or antibody fragment or construct.
  • the nucleic acid encodes a wild-type form of a protein, where a mutant form of the protein causes an ocular disease.
  • the nucleic acid encodes a protein selected from the group consisting of ABCA4, GUCY2D, RPE65, ELOVL4, CHM, CRB1, MY07A, CDH23, CLRN1, ADGRV1, CIB2, HARS, PCDH15, USH2A, USH1C, USH1G, GPR98, WHRN, CEP290, MERTK, RPGR, Rodospin, PDE6B, VEGF-A, an anti-complement protein, an anti-VEGF protein, an anti-TNFa protein, and rod-derived cone viability factor.
  • a protein selected from the group consisting of ABCA4, GUCY2D, RPE65, ELOVL4, CHM, CRB1, MY07A, CDH23, CLRN1, ADGRV1, CIB2, HARS, PCDH15, USH2A, USH1C, USH1G, GPR98, WHRN, CEP290, MERTK, RPGR, Rodospin, P
  • the gene therapy is delivered in a non-viral nanoparticle or a viral vector.
  • non-viral nanoparticle is a lipid-based nanoparticle.
  • the nanoparticle is a polymer-based nanoparticle.
  • the nanoparticle is a peptide or protein-based nanoparticle.
  • the nanoparticle is an inorganic nanoparticle.
  • the nanoparticle is a carbon-based nanoparticle.
  • the nanoparticle size ranges from about 5 nm to about 500 nm.
  • the viral vector is an adenovirus, (Ad), adenoassociated virus (AAV), or lentivirus.
  • Ad adenovirus
  • AAV adenoassociated virus
  • the viral vector is a self-complementary AAV (scAAV) or helperdependent adenovirus (HD-Ad).
  • AAV vectors suitable for use in the methods and compositions herein include, but are not limited to, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, and AAV9.
  • the VEGF antagonist is selected from the group consisting of aflibercept, ziv-aflibercept, bevacizumab, sonepcizumab, VEGF sticky trap, an anti-VEGF Fab, RGX-314, cabozantinib, foretinib, vandetanib, nintedanib, regorafenib, cediranib, ranibizumab, lapatinib, sunitinib, sorafenib, plitidepsin, verteporfm, bucillamine, axitinib, pazopanib, fluocinolone acetonide, AL8326, 2C3 antibody, AT001 antibody, XtendVEGF antibody, HuMax- VEGF antibody, R3 antibody, AT001/r84 antibody, HyBEV, ANG3070, APX003 antibody, APX004 antibody, ponatinib, BDM-
  • the patient can be positioned such that the eye 10 is mounted in position adjacent to the injection apparatus 300.
  • the conjunctiva depressor 326 can separate conjunctiva for proper sealing when inserting the hollow conduit 320.
  • the injection controller 360 then pumps injectate 352 through the injection tube 351 to prime the pressure tube 342.
  • the relief valve 344 can then be opened to vent the pressure line 342, thereby applying back pressure to the distal end of the hollow conduit 320.
  • the actuator 330 then pushes the adapter 322 and the hollow conduit 320 forward until a pressure change is observed in the pressure measurement device 340, indicating that the reference location has been located. Once the reference location has been located, the relief valve 344 can be closed, stopping injection of injectate 352 into the SCS.
  • the actuator 330 can then advance the adapter 322 and the hollow conduit 320 an additional distance (e.g., between about 50 pm and about 2 mm or between about 100 pm and about 150 pm) to advance into the retinal tissue.
  • additional distance e.g., between about 50 pm and about 2 mm or between about 100 pm and about 150 pm
  • inj ectate can be injected into the retinal layer via the injection controller 360.
  • FIG. 8 is an illustration of an injection apparatus 400 that uses light emissivity measurements to locate a reference location, according to an embodiment.
  • FIG. 8 is a cross-sectional view of the apparatus 400.
  • the apparatus 400 can have a cylindrical shape or a substantially cylindrical shape.
  • the injection apparatus 400 includes a housing 410, a hollow conduit 420, an adapter 422, an actuator 430, a light emissivity measurement device 440, injectate 452, and an injection controller 460.
  • the housing 410, the hollow conduit 420, the adapter 422, the actuator 430, the injectate 452, and the injection controller 460 can be the same or substantially similar to the housing 310, the hollow conduit 320, the adapter 322, the actuator 330, the injectate 352, and the injection controller 360, respectively, as described above with reference to FIGS 7 A and 7B.
  • certain aspects of the housing 410, the hollow conduit 420, the adapter 422, the actuator 430, the injectate 452, and the injection controller 460 are not described in greater detail herein.
  • the injectate 452 is disposed directly in the housing 410.
  • the injection apparatus 400 does not include a second vessel to contain the injectate 452.
  • the injectate 452 can be disposed in a second structure external to the housing 410.
  • the housing 410 contains the hollow conduit 420, the adapter 422, the actuator 430, the injectate 452, and the injection controller 460.
  • the adapter 422 can form a fluid-tight seal with the actuator 430.
  • the adapter 422 can have a sufficient length in the longitudinal direction (i.e., the vertical direction in FIG. 8), such that the adapter 422 can move in the vertical direction while still maintaining a seal with the actuator 430.
  • the adapter 422 can have a longitudinal length longer than the longitudinal length of the actuator 430. [00148] In some embodiments, the adapter 422 can have a longitudinal length of at least about 1 mm, at least about 2 mm, at least about 3 mm, at least about 4 mm, at least about 5 mm, at least about 6 mm, at least about 7 mm, at least about 8 mm, at least about 9 mm, at least about 1 cm, at least about 2 cm, at least about 3 cm, or at least about 4 cm.
  • the adapter can have a longitudinal length of no more than about 5 cm, no more than about 4 cm, no more than about 3 cm, no more than about 2 cm, no more than about 1 cm, no more than about 9 mm, no more than about 8 mm, no more than about 7 mm, no more than about 6 mm, no more than about 5 mm, no more than about 4 mm, no more than about 3 mm, or no more than about 2 mm.
  • Combinations of the above-referenced longitudinal length values for the adapter 422 are also possible (e.g., at least about 1 mm and no more than about 5 cm or at least about 5 mm and no more than about 1 cm), inclusive of all values and ranges therebetween.
  • the adapter 422 can have a longitudinal length of about 1 mm, about 2 mm, about 3 mm, about 4 mm, about 5 mm, about 6 mm, about 7 mm, about 8 mm, about 9 mm, about 1 cm, about 2 cm, about 3 cm, about 4 cm, or about 5 cm.
  • the actuator 430 can have a longitudinal length of at least about 500 pm, at least about 600 pm, at least about 700 pm, at least about 800 pm, at least about 900 pm, at least about 1 mm, at least about 2 mm, at least about 3 mm, at least about 4 mm, at least about 5 mm, at least about 6 mm, at least about 7 mm, at least about 8 mm, at least about 9 mm, or at least about 1 cm.
  • the adapter can have a longitudinal length of no more than about 2 cm, no more than about 1 cm, no more than about 9 mm, no more than about 8 mm, no more than about 7 mm, no more than about 6 mm, no more than about 5 mm, no more than about 4 mm, no more than about 3 mm, or no more than about 2 mm.
  • Combinations of the above-referenced longitudinal length values for the adapter 422 are also possible (e.g., at least about 500 pm and no more than about 2 cm or at least about 5 mm and no more than about 1 cm), inclusive of all values and ranges therebetween.
  • the adapter 422 can have a longitudinal length of about 500 pm, about 600 pm, about 700 pm, about 800 pm, about 900 pm about 1 mm, about 2 mm, about 3 mm, about 4 mm, about 5 mm, about 6 mm, about 7 mm, about 8 mm, about 9 mm, about 1 cm, or about 2 cm.
  • the actuator 430 can include a piezo motor.
  • the actuator 430 can be operable to move the hollow conduit 420 and the adapter 422 relative to the housing 410 while the actuator 430 remains stationary relative to the housing 410.
  • the actuator 430 can move relative to the housing 410 along an inner wall of the housing 410.
  • the actuator 430 can have a longitudinal range of motion of at least about 1 mm, at least about 2 mm, at least about 3 mm, at least about 4 mm, at least about
  • the actuator 430 can have a longitudinal range of motion of no more than about 5 cm, no more than about 4 cm, no more than about 3 cm, no more than about 2 cm, no more than about 1 cm, no more than about 9 mm, no more than about 8 mm, no more than about 7 mm, no more than about
  • the actuator 430 can have a longitudinal range of motion of about 1 mm, about 2 mm, about 3 mm, about 4 mm, about 5 mm, about 6 mm, about 7 mm, about 8 mm, about 9 mm, about 1 cm, about 2 cm, about 3 cm, about 4 cm, or about 5 cm.
  • the emissivity measurement device 440 includes an emitter 441 that shines light through the pupil via a fiber optic wire 447.
  • the fiber optic wire 447 is physically coupled the distal end of the hollow conduit 420 and to a receiver 444.
  • the emissivity measurement device 440 further includes a printed circuit board 448 to control the operation of the emissivity measurement device 440.
  • the printed circuit board 448 can include drivers and/or firmware to control the emissivity measurement device 440. Based on the intensity of light emitted from the emitter 441 and the intensity of light detected in the receiver 444, emissivity can be calculated.
  • the fiber optic wire 447 can run through the hollow conduit 420.
  • the fiber optic wire 447 can be affixed to the outside of the hollow conduit 420.
  • the emitter 441 can emit light at an intensity of at least about 5 lumens, at least about 10 lumens, at least about 15 lumens, at least about 20 lumens, at least about 25 lumens, at least about 30 lumens, at least about 35 lumens, at least about 40 lumens, or at least about 45 lumens.
  • the emitter 441 can emit light at intensity of no more than about 50 lumens, no more than about 45 lumens, no more than about 40 lumens, no more than about 35 lumens, no more than about 30 lumens, no more than about 25 lumens, no more than about 20 lumens, no more than about 15 lumens, or no more than about 10 lumens.
  • Combinations of the above-referenced intensities of light emitted from the emitter 441 are also possible (e.g., at least about 5 lumens and no more than about 50 lumens or at least about 10 lumens and no more than about 20 lumens), inclusive of all values and ranges therebetween.
  • the emitter 441 can emit light at an intensity of about 5 lumens, about 10 lumens, about 15 lumens, about 20 lumens, about 25 lumens, about 30 lumens, about 35 lumens, about 40 lumens, about 45 lumens, or about 50 lumens.
  • the injection controller 460 is disposed directly in the housing 410.
  • the injection controller 460 can move longitudinally within the housing 410 to push the inj ectate 452 through the hollow conduit 420.
  • a sealing member can be disposed around the perimeter of the injection controller 460 to create a seal between the inner surface of the housing 410 and the perimeter of the injection controller 460.
  • the injection controller 460 can be a plunger.
  • the housing 410 can be brought to a location close to the eye, such that the distal end of the hollow conduit 420 is near the outer surface of the eye.
  • the distal end of the hollow conduit 420 is then advanced a first distance into the reference location in the eye.
  • the actuator 430 can advance the distal end of the hollow conduit 420 to the reference location.
  • the reference location is detected based on emissivity measurements done by the emissivity measurement device 440, which operates as described above.
  • the actuator 430 can advance the hollow conduit 420 and/or the adapter 422 a second distance into the eye, such that the distal end of the hollow conduit 420 reaches the injection region.
  • the injection controller 460 propels injectate 452 from the housing 410 to the injection region via the hollow conduit 420.
  • the hollow conduit 420 can be removed from the eye.
  • FIG. 9 is an illustration of an injection apparatus 500, according to an embodiment.
  • the injection apparatus 500 includes a housing 510, a hollow conduit 520, an actuator 530, a pressure line 542, a printed circuit board 548, a medicament container 550, and an injectate 552.
  • the housing 510, hollow conduit 520, the actuator 530, the pressure line 542, and the medicament container 550 can be the same or substantially similar to the housing 210, the hollow conduit 220, the actuator, the pressure line 242, and the medicament container 250, respectively, as described above with reference to FIG. 6.
  • certain aspects of the housing 510, hollow conduit 520, the actuator 530, the pressure line 542, and the medicament container 550 are not described in greater detail herein.
  • the actuator 530 is operable to advance the medicament container 550 and the hollow conduit 520 through the housing 510. Upon actuation, the actuator 530 can propel the medicament container 550 into the void space VS, thereby advancing the hollow conduit 520 and the distal end of the hollow conduit 520.
  • the actuator 530 can include a piezo motor. In some embodiments, the actuator 530 can propel the medicament container 550 relative to the housing 510, while the actuator 530 remains stationary with reference to the housing 510.
  • the medicament container 550 is a separate vessel from the housing 510 disposed in the housing 510.
  • the medicament container 550 can be external to the housing 510.
  • the medicament container 550 includes a puncture cap 558.
  • the puncture cap 558 can be punctured by a puncture spike 559. After the puncture cap 558 is punctured, fluidic communication is established between the distal end of the hollow conduit 520 and the pressure line 542.
  • injectate 552 can be released into the pressure line 542 to prime the pressure line 542, such that a manometer can be formed.
  • the puncture cap 558 can ensure that the injectate 552 is sealed and undisturbed until the medicament container 550 is loaded into the housing 510.
  • the printed circuit board 548 can control pressure transduction within the pressure line 542 and/or the medicament container 550.
  • the injection apparatus 500 can include an emissivity measurement device (not shown).
  • the emissivity measurement device can be the same or substantially similar to the emissivity measurement device 440 described above, with reference to FIG. 8.
  • the printed circuit board 548 can control the emissivity measurement device.
  • the housing 510 can be brought to a location close to the eye, such that the distal end of the hollow conduit 520 is near the outer surface of the eye.
  • the distal end of the hollow conduit 520 is then advanced a first distance into the reference location in the eye.
  • the actuator 530 can advance the distal end of the hollow conduit 520 to the reference location.
  • the puncture spike 559 can pierce the puncture cap 558 to create a fluidic coupling between the pressure line 542 and the distal end of the hollow conduit 520.
  • the reference location is detected based on pressure measurements in the pressure line 542.
  • the actuator 530 can advance the hollow conduit 520 a second distance into the eye, such that the distal end of the hollow conduit 520 reaches the injection region.
  • an injection controller (not shown) propels injectate 552 from the housing 510 to the injection region via the hollow conduit 520. After the injection, the hollow conduit 520 can be removed from the eye.
  • FIG. 10 is an illustration of an injection apparatus 600, according to an embodiment.
  • the injection apparatus 600 includes a housing 610, a hollow conduit 620, an actuator 630, a manual actuator dial 631, a pressure line 642, a medicament container 650, and a control line 661.
  • the housing 610, the hollow conduit 620, the actuator 630, the pressure line 642, and the medicament container 650 can be the same or substantially similar to the housing 210, the hollow conduit 220, the actuator, the pressure line 242, and the medicament container 250, respectively, as described above with reference to FIG. 6.
  • the injection apparatus 600 can include both an automatic actuator and a manual actuator.
  • the actuator 630 can include a piezo motor.
  • the manual actuator dial 631 can control a micrometer based actuator.
  • the micrometer-based actuator can be a second actuator. The use of two different types of actuators can allow for both automatic and manual advancement of the hollow conduit 620, depending on the preference of the person administering the injection.
  • the medicament container 650 is a separate vessel from the housing 610. Inj ectate can flow from the medicament container 650 to the hollow conduit 620 via a medicament line 653.
  • the pressure line 642 can act as a portion of a manometer, the same or substantially similar to the pressure line 542 described above, with reference to FIG. 9.
  • the control line 661 can control operation of the actuator 630. In some embodiments, the control line 661 can control inputs into a motor in the actuator 630.
  • the housing 610 can be brought to a location close to the eye, such that the distal end of the hollow conduit 620 is near the outer surface of the eye.
  • the distal end of the hollow conduit 620 is then advanced a first distance into the reference location in the eye.
  • the actuator 630 can advance the distal end of the hollow conduit 620 to the reference location.
  • the reference location is detected based on pressure measurements in the pressure line 642. Once the reference location is detected, the actuator 630 can advance the hollow conduit 620 a second distance into the eye, such that the distal end of the hollow conduit 620 reaches the injection region.
  • an injection controller (not shown) propels inj ectate from the medicament container 650 to the injection region via the medicament line 653 and the hollow conduit 520. After the injection, the hollow conduit 620 can be removed from the eye.
  • FIG. 11 is a schematic illustration of an injection apparatus 700, according to an embodiment.
  • the injection apparatus 700 can be configured to locate a first region (e.g., an SCS) of a target tissue (e.g., an eye), and use the location of that first region to deliver a medicament to a second region (e.g., a retina or subretinal space (SRS)) of the target tissue.
  • a first region e.g., an SCS
  • a target tissue e.g., an eye
  • a second region e.g., a retina or subretinal space (SRS)
  • the injection apparatus 700 includes a housing 710 configured to be engaged with a hollow conduit 720 and a catheter 770.
  • the housing 710 includes a fluid container 713 disposed therein.
  • the injection apparatus 700 further includes a medicament container 750 contacting and slidably movable within the housing 710.
  • An injection controller 760 is slidably engaged with the medicament container 750 and controls injection of medicament via the hollow conduit 720.
  • medicament in the medicament container 750 can be pushed out of the injection apparatus 700 via the movement of the injection controller 760 from a first position to a second position relative to the housing 710.
  • the housing 710, the hollow conduit 720, the medicament container 750, and the injection controller 760 can be the same or substantially similar to the housing 110, the hollow conduit 120, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • certain aspects of the housing 710, the hollow conduit 720, the medicament container 750, and the injection controller 760 are not described in greater detail herein.
  • the fluid container 713 is contained in the housing 710 and can contain a liquid.
  • the fluid container 713 is fluidically coupled to the catheter 770 such that the liquid can exit the fluid container 713 via the catheter 770.
  • the fluid container 713 is slidably coupled to the housing 710 such that the fluid container 713 can be moved into and out of the housing 710.
  • the medicament container 750 is slidably coupled to the fluid container 713, such that the medicament container 750 can be moved into and out of the fluid container 713. Movement of the medicament container 750 into the fluid container 713 applies a pressure to the liquid in the fluid container 713, causing the liquid to be pushed through the catheter 770 and out of the injection apparatus 700.
  • the hollow conduit 720 has a smaller diameter than the catheter 770 such that the hollow conduit 720 can slidably move into and out of the catheter 770.
  • the hollow conduit 720 has a length such that a distal end of the hollow conduit 720 extends beyond a distal end of the catheter 770 when the medicament container 750 is advanced toward a distal end of the fluid container 713.
  • the medicament container 750 is fluidically coupled to the hollow conduit 720 such that medicament can flow from the medicament container 750 to outside the injection apparatus 700 via the hollow conduit 720.
  • the injection controller 760 is slidably coupled to the medicament container 750, such that advancement of the injection controller 760 applies a pressure to the medicament in the medicament container 750, causing the medicament to flow out of the injection apparatus 700 via the hollow conduit 720.
  • the injection apparatus 700 is moved distally toward a target surface (i.e. a conjunctiva and/or sclera of an eye) when the fluid container 713 is in a first configuration (or position) within the housing 710.
  • a target surface i.e. a conjunctiva and/or sclera of an eye
  • the fluid container 713 is a first distance from a distal end of the housing 710.
  • the distal end of the housing 710 can contact the target surface to form a seal with the target surface.
  • a distal tip of the catheter 770 is inserted into the sclera via movement of the fluid container 713 from the first configuration to a second configuration.
  • the fluid container 713 is a second distance from the distal end of the housing 710, the second distance less than the first distance.
  • movement of the fluid container 713 from the first configuration to the second configuration can be manual (e.g., via use of hands/fingers).
  • a user can manually push the fluid container 713 (e.g., via a flange or plunger) in a distal direction relative to the housing 710.
  • movement of the fluid container 713 from the first configuration to the second configuration can be automated.
  • initial insertion of the catheter 770 into the target surface can include inserting the catheter 770 into the target surface until an outer surface (e.g., distal-most end) of the housing 710 contacts the target surface.
  • the contact between the outer surface of the housing 710 and the target surface can deform the target surface.
  • the contact between the outer surface of the housing 710 and the target surface can form a substantially fluid-tight seal with the conjunctiva around the insertion site of the catheter 770.
  • insertion of the distal end of the catheter 770 into the target surface can be at an angle relative to a line tangent to the target surface (i.e., an angle of entry).
  • the angle of entry can be at least about 0 degrees, at least about 1 degree, at least about 2 degrees, at least about 3 degrees, at least about 4 degrees, at least about 5 degrees, at least about 10 degrees, at least about 15 degrees, at least about 20 degrees, at least about 25 degrees, at least about 30 degrees, at least about 35 degrees, at least about 40 degrees, at least about 45 degrees, at least about 50 degrees, at least about 55 degrees, at least about 60 degrees, at least about 65 degrees, at least about 70 degrees, at least about 75 degrees, at least about 80 degrees, at least about 85 degrees, at least about 90 degrees, at least about 95 degrees, at least about 100 degrees, at least about 105 degrees, at least about 110 degrees, at least about 115 degrees, at least about 120 degrees, at least about 125 degrees, at least about 130 degrees, at least about 135 degrees, at least about 140 degrees, at least about 145 degrees, at least about 150 degrees, at least about 155 degrees, at least about 160 degrees, at least about 165 degrees, at least about 170 degrees, at
  • the angle of entry can be no more than about 180 degrees, no more than about 179 degrees, no more than about 178 degrees, no more than about 177 degrees, no more than about 176 degrees, no more than about 175 degrees, no more than about 170 degrees, no more than about 165 degrees, no more than about 160 degrees, no more than about 155 degrees, no more than about 150 degrees, no more than about 145 degrees, no more than about 140 degrees, no more than about 135 degrees, no more than about 130 degrees, no more than about 125 degrees, no more than about 120 degrees, no more than about 115 degrees, no more than about 110 degrees, no more than about 105 degrees, no more than about 100 degrees, no more than about 95 degrees, no more than about 90 degrees, no more than about 85 degrees, no more than about 80 degrees, no more than about 75 degrees, no more than about 70 degrees, no more than about 65 degrees, no more than about 60 degrees, no more than about 55 degrees, no more than about 50 degrees, no more than about 45 degrees, no more than about 40 degrees,
  • angles of entries are also possible (e.g., at least about 0 degrees and no more than about 180 degrees), inclusive of all values and ranges therebetween.
  • the angle of entry can be about 0 degrees, about 1 degree, about 2 degrees, about 3 degrees, about 4 degrees, about 5 degrees, about 10 degrees, about 15 degrees, about 20 degrees, about 25 degrees, about 30 degrees, about 35 degrees, about 40 degrees, about 45 degrees, about 50 degrees, about 55 degrees, about 60 degrees, about 65 degrees, about 70 degrees, about 75 degrees, about 80 degrees, about 85 degrees, about 90 degrees, about 95 degrees, about 100 degrees, about 105 degrees, about 110 degrees, about 115 degrees, about 120 degrees, about 125 degrees, about 130 degrees, about 135 degrees, about 140 degrees, about 145 degrees, about 150 degrees, about 155 degrees, about 160 degrees, about 165 degrees, about 170 degrees, about 175 degrees, about 176 degrees, about 177 degrees, about 178 degrees, about 179 degrees, or about 180 degrees.
  • the housing 710 can include a mechanism to limit and/or prevent proximal movement of the distal tip of the catheter 770 relative to the housing 710.
  • the housing 710 can include a shoulder (not shown) that engages a corresponding shoulder (not shown) of the catheter 770 to prevent proximal movement of the catheter 770 relative to the housing 710.
  • the injection apparatus 700 can be moved distally to complete the initial insertion of the catheter 770 into the target surface.
  • the user can manually apply a force to a portion of the medicament container 750 and/or the housing 710.
  • the initial insertion operation can be a manual operation that does not rely on any stored energy device (e.g., springs, compressed gas containers, or the like) to produce the force to move the medicament container 750 and/or the housing 710.
  • the injection apparatus 700 is advanced distally such that the distal end of the catheter 770 is in contact with the sclera and/or the choroid.
  • the high densities of the sclera or choroid relative to the retina can provide a resistance to movement of the catheter 770 relative to the target surface and a resistance to flow of the liquid from inside the fluid container 713.
  • at least a portion of the liquid from inside the fluid container 713 can be conveyed through the catheter 770 via movement of the medicament container 750 from a first position relative to the fluid container 713 and/or the housing 710 to a second position relative to the fluid container 713 and/or the housing 710, the second position distal to the first position.
  • advancement of the medicament container 750 can simultaneously convey the liquid from the fluid container 713 to a target region via the catheter 770, as a distal end of the medicament container 750 can press on the liquid to push the liquid through the catheter 770.
  • the medicament container 750 is a second distance from the distal end of the housing 710, the second distance less than the first distance.
  • the liquid within the fluid container 713 can be pressurized (e.g., via a spring springing mechanism).
  • the forces acting on the liquid in the fluid container 713 can become unbalanced when the distal end of the catheter 770 recaches a region of lower resistance (e.g., the SCS), causing the medicament container 750 to move in a distal direction relative to the housing 710.
  • the medicament container 750 then moves to a second configuration (or position) relative to the housing 710.
  • the medicament container 750 can move in a distal direction (thereby injecting the liquid into a target region) via a loss-of-resistance mechanism.
  • the medicament container 750 can be subject to an outside force (e.g., a spring or a springing mechanism), but the high density of the sclera and/or choroid relative to the SCS can create a backpressure and prevent movement of the medicament container 750 relative to the housing 710.
  • the medicament container 750 can advance distally relative to the fluid container 713 and the housing 710, thereby releasing liquid from the fluid container 713.
  • the distal movement of the medicament container 750 relative to the housing 710 and the fluid container 713 can be manually actuated (e.g., via use of hands/fingers).
  • a user can manually push the medicament container 750 (e.g., via a flange) in a distal direction relative to the housing 710 and the fluid container 713.
  • the medicament container 750 can move relative to the housing 710 in response to the entry of the distal end of the injection apparatus 700 into the less dense region (e.g., the SCS).
  • the medicament container 750 can move relative to the housing 710 in response to the aforementioned loss-of-resistance mechanism.
  • the movement of the medicament container 750 relative to the housing 710 can cause the liquid to be deployed from the fluid container 713 via the catheter 770. This deployment of the liquid can thereby inform the user that the distal end of the catheter 770 has entered the SCS.
  • the hollow conduit 720 can be advanced, such that a distal end of the hollow conduit 720 reaches a desired injection region.
  • the hollow conduit 720 is coupled to the medicament container 750 and the distal movement of the medicament container 750 causes the hollow conduit 720 to advance in a distal direction, such that a distal end of the hollow conduit 720 extends beyond the distal end of the catheter 770.
  • the distal end of the hollow conduit 720 can enter a target tissue for medicament injection at a location distal to the catheter 770 (e.g., the SRS).
  • the injection controller 760 can be actuated to inject medicament into the target tissue.
  • the user can manually apply a force (e.g., via use of hands/fingers) to a portion of the medicament container 760 to induce injection of the medicament into the target tissue.
  • a force e.g., via use of hands/fingers
  • the SCS can be used as a reference location to find the target tissue (e.g., the SRS) for medicament injection.
  • the medicament container 750 and the hollow conduit 720 can be advanced, such that the distal end of the hollow conduit 720 is advanced beyond the distal end of the catheter by about 50 pm, about 60 pm, about 70 pm, about 80 pm, about 90 pm, about 100 pm, about 110 pm, about 120 pm, about 130 pm, about 140 pm, about 150 pm, about 160 pm, about 170 pm, about 180 pm, about 190 pm, or about 200 pm, inclusive of all values and ranges therebetween.
  • the distal end of the hollow conduit 720 reaches the target tissue, and injection of the medicament from the medicament container 750 occurs.
  • FIGS. 12A-12F illustrate an injection apparatus 800 and a method of using the injection apparatus 800, according to an embodiment.
  • the injection apparatus 800 includes a housing 810, a fluid container 813, a fluid container hub 814, a handle 815, a sealing surface 816, a seal 817, a fluid container flange 819, a hollow conduit 820, a medicament container 850, a medicament container hub 851, a medicament container flage 853, adapters 855, 856, an injection controller 860, and an outer conduit 870.
  • the outer conduit 870 is a catheter.
  • the outer conduit 870 can can be an outer conduit, such that the hollow conduit 820 is an inner conduit that moves within the outer conduit.
  • the outer conduit 870 can be a multi-lumen conduit.
  • the outer conduit 870 can have multiple lumens, through which inner conduits (e.g., the inner conduit 820) can move. This can allow for delivery of a first liquid and/or medicament to a first location in the eye and a delivery of a second liquid and/or medicament to a second location in the eye.
  • the second location can be deeper in the eye than the first location (e.g., the second location can be in the SRS and the first location can be in the SCS).
  • FIG. 12A shows a cross-sectional view of the injection apparatus 800 while FIG. 12B shows an exploded view of the injection apparatus 800.
  • the housing 810, the fluid container 813, the hollow conduit 820, the medicament container 850, and the injection controller 860 can be the same or substantially similar to the housing 710, the fluid container 713, the hollow conduit 720, the medicament container 750, and the injection controller 760, as described above with reference to FIG. 11.
  • certain aspects of the housing 810, the fluid container 813, the hollow conduit 820, the medicament container 850, and the injection controller 860 are not described in greater detail herein.
  • the handle 815 can include a flat planar shape extending outwardly from a central axis of the housing 810.
  • the handle 815 can provide a grip for the user to hold while operating the injection apparatus 800.
  • the handle 815 can provide a means for stopping the injection apparatus 800 from rolling (e.g., when placed on a table or platform).
  • the sealing surface 816 provides a surface against which a seal can be formed when the injection apparatus 800 contacts a surface (e.g., conjunctiva or sclera) of the eye.
  • the medicament container flange 853 can provide a grip for the user or a platform for the user to manually move the fluid container 813 relative to the housing 810.
  • the injection controller 860 can provide a grip for the user or a platform for the user to manually move the medicament container 850 relative to the housing 810 and/or the medicament container 850.
  • the adapters 855, 856 provide a key-lock pair to secure the medicament container 850 and the fluid container 813 together.
  • the fluid container 813 is at least partially housed in the housing 810 and can move from a first position (i.e., a proximal position) relative to the housing 810 to a second position (i.e., a distal position) relative to the housing 810.
  • the catheter 870 is fixedly coupled to the fluid container 813. In moving from the first position to the second position, the fluid container 813 can correspondingly cause the catheter 870 to move distally such that a distal end of the catheter 870 extends beyond the sealing surface 816.
  • the medicament container 850 is at least partially housed in the fluid container 813 and can move from a first position (i.e., a proximal position) relative to the fluid container 813 to a second position (i.e., a distal position) relative to the fluid container 813.
  • the hollow conduit 820 is fixedly and fluidically coupled to the medicament container 850. In moving from the first position to the second position, the medicament container 850 can cause the hollow conduit 820 to move distally through a lumen of the catheter 870 such that a distal end of the hollow conduit extends beyond the distal end of the catheter 870.
  • the seal 817 can slide along the interior of the fluid container 813 with the medicament container 850. The seal 817 can prevent fluid from exiting the fluid container 813 through the proximal end of the fluid container 813.
  • Rotation of the different components of the injection apparatus 800 afford versatility of motion of the components of the injection apparatus 800.
  • the medicament container 850 is not fixedly coupled to the housing 810, allowing the medicament container 850 to rotate relative to the housing 810.
  • the medicament container 850 is coupled to the fluid container 813 via the adapters 855, 856.
  • the adapter 855 can be coupled to the fluid container flange 819 and the adapter 856 can be coupled to the outside surface of the medicament container 850.
  • the medicament container 850 is not fixedly coupled to the fluid container 813, allowing the medicament container 850 to rotate relative to the housing 810.
  • rotation of the hollow conduit 820 can aid in advancement of the hollow conduit 820 through target tissue, particularly if the hollow conduit 820 has a beveled (i.e., angled) distal tip.
  • the angle of the distal tip of the hollow conduit 820 can allow the beveled distal tip to act like a blunt tip trocar that pushes tissue away to make room for passage of the hollow conduit 820 through the target tissue easier (as compared to an unbeveled distal tip).
  • the operator can rotate the medicament container 850 (and the hollow conduit 820 fixedly coupled to the medicament container 850), relative to the housing 810 and the fluid container 813, by gripping the medicament container 850 (e.g., by the medicament container flange 853).
  • This rotation of the medicament container 850 and the hollow conduit 820 can be done while the housing 810 and the fluid container 813 are relatively stationary and not rotating (as the medicament containter 850 and the hollow conduit 820 are rotatable relative to the housing 810 and the fluid container 813). Keeping the housing 810 and the fluid container 813 stationary can prevent scoring on the surface of the eye (e.g., by the sealing surface 816). While the distal tip of the hollow conduit 820 is extended beyond the sealing surface 816, rotating the hollow conduit 820 to push the tissue away can be analogous to driving a screw into wood. With each rotation of the hollow conduit 820, the beveled tip of the hollow conduit 820 moves deeper into the tissue of the eye until it reaches a target region.
  • the medicament container 850 can be fixedly coupled to the adapter 855 while the fluid container 813.
  • the distal tip of the hollow conduit can be beveled at an angle of about 5 degrees (relative to an unbeveled tip), about 10 degrees, about 15 degrees, about 20 degrees, about 25 degrees, about 30 degrees, about 35 degrees, about 40 degrees, about 45 degrees, about 50 degrees, about 55 degrees, about 60 degrees, about 65 degrees, about 70 degrees, about 75 degrees, about 80 degrees, about 85 degrees, or about 90 degrees, inclusive of all values and ranges therebetween.
  • the sclera (not shown) can be pre-scored.
  • FIGS. 12C and 12D show the transition of the fluid container 813 from the first position to the second position.
  • the clearance distance can be about 50 pm, about 60 pm, about 70 pm, about 80 pm, about 90 pm, about 100 pm, about 110 pm, about 120 pm, about 130 pm, about 140 pm, about 150 pm, about 160 pm, about 170 pm, about 180 pm, about 190 pm, or about 200 pm, inclusive of all values and ranges therebetween.
  • the fluid container flange 819 is pushed (e.g., by a finger) to seal the sealing surface 816 against the pre-scored sclera hole.
  • the fluid container 813 can be rotatable relative to the housing 810, as the outer diameter of the fluid container 813 can be smaller than the inner diameter of the housing 810.
  • the catheter 870 is fixedly coupled to the fluid container 813. As the fluid container 813 rotates and/or slides relative to the housing 810, the catheter 870 can advance distally relative to the sealing surface 816 such that the distal end of the catheter 870 enters the sclera, the SCS, the choroid, the SRS, and/or the retina.
  • the fluid container 813 can be slidably removed from the housing 810. [00184] The medicament container 850 can then be gripped and pushed distally relative to the fluid container 813 (see FIG. 12E).
  • the medicament container 850 can be moved by pushing on the medicament container flange 853 in a distal direction relative to the fluid container 813.
  • the fluid container 813 can have a liquid (e.g., a saline solution) disposed therein.
  • pushing the medicament container 850 can pressurize the liquid in the fluid container 813.
  • the seal 817 can move with the medicament container 850 as the medicament container 850 is being advanced.
  • the fluid that enters the SCS can separate the distal end of the catheter 870 from the choroid, reducing impact to the chorioid.
  • the fluid can enter the space between the sclera and the choroid and hydrodissect the sclera and the choroid to create and/or expand the SCS.
  • the liquid can flow from the fluid container 813 via a loss-of- resistance mechanism.
  • the liquid can be pressurized in an appropriate pressure range such that the liquid is prevented from flowing through the catheter 870 when the distal end of the catheter is in the sclera and the liquid can flow through the catheter 870 when the distal end of the catheter 870 reaches a region (e.g., the SCS) of low pressure (e.g., relative to the sclera).
  • the liquid in the fluid container 813 can be pressurized (e.g., via a force applied by relative distal movement of the medicament container 850.
  • the forces acting on the seal 817 can become unbalanced once the distal end opening of the catheter 870 reaches a low-density region (e.g., the SCS) relative to the sclera, causing the medicament container 850, the seal 817, and the catheter 870 to advance from the first position to the second position, thereby injecting the liquid into the SCS via the catheter 870.
  • a low-density region e.g., the SCS
  • the hollow conduit 820 is fixedly coupled to a distal end of the medicament container 850.
  • the medicament container hub 851 positions and secures the hollow conduit 820 on the medicament container 850.
  • the hollow conduit 820 can advance with the medicament container 850, such that the hollow conduit 820 moves through the catheter 870 and the distal end of the hollow conduit 820 extends beyond the distal end of the catheter 870 and into the injection site (e.g., the SRS).
  • the injection site e.g., the SRS
  • the liquid can move the choroid and the choroid can relax against a distal end of the hollow conduit 820.
  • the distal end opening of the hollow conduit 820 enters the SRS.
  • Medicament can then be delivered to the SRS by pushing on the injection controller 860. More specifically, pushing on the injection controller 860 can cause the medicament to exit the medicament container 850 via the hollow conduit 820 and enter the SRS.
  • FIGS. 13A-13O illustrate methods and apparatus for performing an injection into the retinal tissue, according to various embodiments.
  • Layers of the eye 10 depicted in FIGS. I SABO include the sclera 20, the choroid 28, and the retina 27.
  • FIGS. 13A-13G show a method of spreading the sclera 20 apart from the choroid 28 for injection of medicament
  • FIGS. 13H- 13N show a method of spreading the chodoid 28 apart from the retina 27 for injection of medicament.
  • a trocar 972 delivers an outer sleeve 970 to the eye 10.
  • FIG. 13 A shows the trocar 972 in the process of delivering the outer sleeve 970 to the eye 10.
  • FIG. 13 A shows the trocar 972 in the process of delivering the outer sleeve 970 to the eye 10.
  • the trocar 972 has penetrated the sclera 20 and placed the outer sleeve 970 in the eye 10.
  • the outer sleeve 970 includes a stop 971. Once the stop 971 makes contact with the outer surface of the eye 10, the trocar 972 and the outer sleeve 970 are prevented from advancing further.
  • a sleeve handle 973 and a trocar handle 975 are also depicted in FIGS. 13A and 13B.
  • the sleeve handle 973 is coupled to the outer sleeve 970 and facilitates easy holding and advancement of the outer sleeve 970.
  • the trocar handle 975 is coupled to the trocar 972 and facilitates easy holding and advancement of the trocar 972.
  • the trocar 972 can include a measurement device that aids in determining proper placement of the outer sleeve 970.
  • the measurement device can include a pressure measurement device.
  • the measurement device can include an emissivity measurement device.
  • the measurement device can include an electrical impedance device.
  • the measurement device can include an acoustic wave measurement device.
  • the outer sleeve 970 can include hash marks to assist in measuring penetration depth of the outer sleeve 970.
  • the outer sleeve 970 can be placed or inserted into the eye 10 without the use of the trocar 972.
  • the outer sleeve 970 can be manually inserted into the eye 10 (e.g., via an operators hand(s)). [00189] Once the outer sleeve 970 has been properly placed in the eye 10, the trocar 972 is removed from the outer sleeve 970 (e.g., by pulling the trocar 972 out of a lumen of the outer sleeve 970), leaving the outer sleeve 970 positioned with the stop 971 flush with the outside surface of the eye 10, as depicted in FIG. 13C. In some embodiments, the outer sleeve 970 can be delivered to the eye 10 without the trocar 972.
  • the outer sleeve 970 can be inserted into the eye 10 by manually pushing the outer sleeve 970 into the eye 10.
  • FIG. 13D depicts insertion of a cannula 978 into the lumen of the outer sleeve 970 with an expandable member 976 disposed onto a distal end of the cannula 978.
  • the expandable member 976 is an object that is able to expand (e.g., via inflation) to push apart two adjacent layers of eye tissue.
  • the cannula 978 is inserted such that the expandable member 976 coupled to the distal end of the cannula 978 contacts an interface between two adjacent layers of eye tissue (e.g., the sclera 20 and the choroid 28) and pushes apart the adjacent layers of the eye tissue.
  • the expandable member 976 is a balloon.
  • the expandable member 976 can include any other expandable member that can be inserted having a first cross-sectional area, and expand to a second, larger cross-sectional area.
  • the expandable member 976 can be or include a stent and/or an inflatable tube.
  • the expandable member 976 can be or include one or more wings, similar to a Malecot catheter, a nephrostomy catheter, an expandable foam, a silicone foam, a slow-recovery foam, a stone retrieval basket, a nitinol cage, a shape memory cage, or any other suitable device that can be inserted and/or delivered into the eye in a relatively small, delivery configuration, and then transitioned to an expanded, relatively large, delivered configuration, as discussed in more detail here.
  • wings similar to a Malecot catheter, a nephrostomy catheter, an expandable foam, a silicone foam, a slow-recovery foam, a stone retrieval basket, a nitinol cage, a shape memory cage, or any other suitable device that can be inserted and/or delivered into the eye in a relatively small, delivery configuration, and then transitioned to an expanded, relatively large, delivered configuration, as discussed in more detail here.
  • the balloon 976 is attached to a cannula 978 and includes an annulus 977 for insertion of a hollow conduit.
  • the outer sleeve 970 can be removed from the eye 10 (e.g., by manually pulling the outer sleeve 970 out of the eye 10), as depicted in FIG. 13E.
  • the balloon 976 can then be inflated, as shown in FIG. 13F.
  • the balloon 976 can be inflated via a conduit (not shown) fluidically coupling an inflation medium container (not shown) and the balloon 976.
  • the conduit can fluidically couple the balloon 976 and the inflation medium container by extending through the length of the cannula 978. Inflation of the balloon 976 can aid in positioning and securing the balloon 976 and canula 978 in a fixed location to prevent the balloon 976 from sliding around. Said another way, the inflation of the balloon 976 can create a tension or a taughtness in the sclera 20, the choroid 28, and the retina 27, such that an anchor point is created. Under tension of the layers of the eye 10, the balloon 976 is prevented from lateral movements.
  • the balloon 976 can include a measurement device that aids in determining proper placement of the balloon 976.
  • the measurement device can include a pressure measurement device.
  • the measurement device can include an emissivity measurement device.
  • the measurement device can include an electrical impedance device.
  • the measurement device can include an acoustic wave measurement device.
  • the cannula 978 can include hash marks to assist in measuring the penetration depth of the balloon 976.
  • the balloon 976 can be placed on the end of a placement shaft that includes hash marks to assist in measuring the penetration depth of the balloon 976.
  • the balloon 976 can be placed independent of the trocar 972 and can include measurement marks along the cannula 978.
  • FIG. 13G depicts the insertion of a hollow conduit 920 into the retina 27 of the eye 10.
  • the hollow conduit 920 is inserted through the annulus 977.
  • the creation of the anchor point enables precise injection, as the hollow conduit 920 is able to enter the retina 27 via the annulus 977 through a stable reference point, rather than a reference point that slides around laterally.
  • the hollow conduit 920 can be the same or substantially similar to the hollow conduit 120, as described above with reference to FIG. 5.
  • the balloon 976 can be placed such that a distal end of the balloon 976 is at a reference location, such that the hollow conduit 920 can be inserted to move just past the reference location to a desired injection location.
  • the hollow conduit 920 can include a measurement device disposed on a distal end of the hollow conduit 920.
  • the measurement device can include a pressure measurement device.
  • the measurement device can include an emissivity measurement device.
  • the measurement device can include an electrical impedance device.
  • the measurement device can include an acoustic wave measurement device.
  • insertion of the hollow conduit 920 into the retina 27 can be such that the distal tip of the hollow conduit 920 extends beyond the balloon 976 by at least about 0.5 mm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, or at least about
  • insertion of the hollow conduit 920 into the retina 27 can be such that the distal tip of the hollow conduit 920 extends beyond the balloon 976 by no more than about 1 cm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, no more than about 1.5 mm, or no more than about 1 mm.
  • insertion of the hollow conduit 920 into the retina 27 can be such that the distal tip of the hollow conduit 920 extends beyond the balloon 976 by about 0.5 mm, about 1 mm, about 1.5 mm, about 2 mm, about
  • FIGS. 13H-13N show a method of spreading the choroid 28 apart from the retina 27 for injection of medicament.
  • FIG. 13H shows the trocar 972 in the process of delivering the outer sleeve 970 to the eye 10.
  • the trocar 972 has penetrated the sclera 20 and the choroid 28 and placed the outer sleeve 970 in the eye 10.
  • the stop 971 makes contact with the outer surface of the eye 10
  • the outer sleeve 970 is placed in a desired location and depth in the eye via the trocar 972.
  • the outer sleeve 970 can be placed such that the distal end of the outer sleeve 970 straddles the boundary between the sclera 20 and the choroid 28.
  • the outer sleeve 970 can be placed such that the distal end of the outer sleeve 970 is proximal to the boundary between the sclera 20 and the choroid 28 by about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, or about 300 pm, inclusive of all values and ranges therebetween. As shown in FIG.
  • the outer sleeve 970 is placed such that the distal end of the outer sleeve 970 straddles the boundary between the choroid 28 and the retina 27.
  • the outer sleeve 970 can be placed such that the distal end of the outer sleeve 970 is proximal to the boundary between the choroid 28 and the retina 27 by about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, or about 300 pm, inclusive of all values and ranges therebetween.
  • FIG. 13 J depicts insertion of the cannula 978 into the outer sleeve 970, with the balloon 976 coupled to the distal end of the cannula 978.
  • the cannula 978 is inserted into the outer sleeve 970 such that the balloon 976 extends beyond the distal end of the outer sleeve 970.
  • the balloon 976 includes an annulus 977 through which the hollow conduit 920 can pass.
  • the outer sleeve 970 can be removed from the eye 10, as depicted in FIG. 13L.
  • the outer sleeve 970 can be removed from the eye 10 by manually pulling the outer sleeve 970 from the eye 10.
  • the outer sleeve 970 can be removed from the eye 10 by machine, or an automated process.
  • the balloon 976 can then be inflated in a region at or near the interface of the choroid 28 and the retina 27, as shown in FIG.
  • Inflation of the balloon 976 can aid in anchoring the distal end of the cannula 978 between the choroid 28 and the retina 27, such that a medicament can be delivered to or in the vicinity of the anchor point created by the inflation of the balloon 976.
  • the inflated balloon 976 is securely placed at the anchor point due to back-pressure from the adjacent layers of the eye 10 (i.e., the choroid 28 and the retina 27).
  • the back pressure from the choroid 28 and the retina 27 prevents the balloon 976 from sliding around.
  • the inflation of the balloon 976 can create a tension or a taughtness at the interface between the choroid 28 and the retina 27 that prevents sliding or lateral movements of the balloon 976 within the layers of the eye 10.
  • the hollow conduit 920 can be inserted into the retina 27, as shown in FIG. 13N.
  • the hollow conduit 920 is inserted through the annulus 977, and medicament is injected into the retina 27 via the hollow conduit 920.
  • the medicament can be injected from a medicament container (not shown) fluidically coupled to the hollow conduit 920.
  • the medicament container can include a syringe barrel.
  • the injection can be intiated via an injection controller (e.g., a plunger).
  • the hollow conduit 920 can be part of an injection apparatus (e.g., the injection apparatus 700, as described above with reference to FIG. 11).
  • the hollow conduit 920 can be fluidically coupled to a medicament container (e.g., the medicament container 750), and medicament can be deployed from the medicament container via an injection controller (e.g., the injection controller 760).
  • the insertion of the hollow conduit 920 into the retina 27 of the eye 10 can involve manually advancing the hollow conduit 920 through the lumen of the cannula 978 and the annulus 977 of the balloon 976.
  • the hollow conduit 920 can be automatically inserted through the cannula 978 and the annulus 977.
  • FIG. 130 shows a perspective view of the ballon 976 inflated between the sclera 20 and the choroid 28.
  • the balloon 976 is expanded in the SCS.
  • the balloon 976 can have a spherical or substantially spherical shape.
  • the balloon 976 can have a torus or donut shape, such that the hollow conduit 920 and/or the cannula 978 can fit through the annulus 977 in the middle of the balloon 976.
  • the balloon 976 can have a spheroid shape, an ellipsoid shape, an ellipsoid of revolution shape, or a rotational ellipsoid shape.
  • the balloon 976 can be illuminated for ease of monitoring.
  • a fiber optic cable can be incorporated into the cannula 978 and/or the conduit fluidically coupled to the balloon 976.
  • the balloon 976 can include a radio-opaque material (e.g., iodine, barium, tantalum, bismuth, gold, etc.) that can be visualized via any suitable imaging modality, such as, for example, X-ray.
  • the balloon 976 can be illuminated upon inflation.
  • the balloon 976 can be illuminated upon administration of medicament through the hollow conduit 920.
  • the balloon 976 can be filled with a compressible medium (e.g., a gas). In some embodiments, the balloon 976 can be filled with nitrogen. In some embodiments, the balloon 976 can be filled with air. In some embodiments, the balloon 976 can be filled with an incompressible medium (e.g., a liquid). In some embodiments, the balloon 976 can be filled with water. In some embodiments, the balloon 976 can be filled with a saline solution. In some embodiments, the medium used to fill the balloon 976 can be selected to minimize poisoning danger to the patient if the balloon 976 ruptures. Filling the balloon 976 with air, for example, can have the andvantage of reusability and ease of priming. In some instances, filling the balloon 976 with water or saline without gas can improve safety, as embolisms can be prevented.
  • a compressible medium e.g., a gas
  • the balloon 976 can be filled with nitrogen.
  • the balloon 976 can be filled with air.
  • inflation of the balloon 976 can cause deformation of the eye 10.
  • the eye 10 can change from a substantially spherical shape to a spherical shape with a protrusion based on the inflation of the balloon 976. Reshaping and deforming of the eye 10 can be a visual indication of the effectiveness of the inflation of the balloon 976.
  • the outer surface of the eye 10 can be described by a first set of spherical coordinates prior to inflation of the balloon 976 and a second set of spherical coordinates after inflation of the balloon 976.
  • the first set of spherical coordinates and the second set of spherical coordinates both use a common point inside the eye 10 as the origin in the spherical coordinate system.
  • the maximum r- value of the surface of the eye 10 in the second set of spherical coordinates can be greater than the maximum r-value of the surface of the eye 10 in the first set of spherical coordinates by at least about 50 pm, at least about 100 pm, at least about 150 pm, at least about 200 pm, at least about 250 pm, at least about 300 pm, at least about 350 pm, at least about 400 pm, at least about 450 pm, at least about 500 pm, at least about 550 pm, at least about 600 pm, at least about 650 pm, at least about 700 pm, at least about 750 pm, at least about 800 pm, at least about 850 pm, at least about 900 pm, at least about 950 pm, at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm,
  • Combinations of the above-referenced differences between the maximum value of the second set of spherical coordinates and the maximum value of the first set of spherical coordinates are also possible (e.g., at least about 50 pm and no more than about 1 cm or at least about 300 pm and no more than about 1 mm), inclusive of all values and ranges therebetween.
  • the maximum r-value in the second set of spherical coordinates can be greater than the maximum r-value in the first set of spherical coordinates by about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, about 500 pm, about 550 pm, about 600 pm, about 650 pm, about 700 pm, about 750 pm, about 800 pm, about 850 pm, about 900 pm, about 950 pm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about
  • the balloon 976 can have an interior capacity of at least about 100 pL, at least about 200 pL, at least about 300 pL, at least about 400 pL, at least about 500 pL, at least about 600 pL, at least about 700 pL, at least about 800 pL, at least about 900 pL, at least about 1 mL, at least about 1.1 mL, at least about 1.2 mL, at least about 1.3 mL, at least about 1.4 mL, at least about 1.5 mL, at least about 1.6 mL, at least about 1.7 mL, at least about 1.8 mL, at least about 1.9 mL, at least about 2 mL, at least about 2.1 mL, at least about 2.2 mL, at least about
  • the balloon 976 can have an interior capacity of no more than about 5 mL, no more than about 4.5 mL, no more than about 4 mL, no more than about 3.5 mL, no more than about 3 mL, no more than about 2.5 mL, no more than about 2.4 mL, no more than about 2.3 mL, no more than about 2.2 mL, no more than about 2.1 mL, no more than about 2 mL, no more than about 1.9 mL, no more than about 1.8 mL, no more than about 1.7 mL, no more than about 1.6 mL, no more than about 1.5 mL, no more than about 1.4 mL, no more than about 1.3 mL, no more than about 1.2 m
  • the balloon 976 can have an interior capacity of about 100 pL, about 200 pL, about 300 pL, about 400 pL, about 500 pL, about 600 pL, about 700 pL, about 800 pL, about 900 pL, about 1 mL, about 1.1 mL, about 1.2 mL, about
  • the flexibility of the hollow conduit 920 can be optimized to conform to the SCS, SRS, or any other desired space inside the eye 10, thereby allowing and/or enhancing the ability for and/or effectiveness of the inj ectate to hydrodissect and find, create, expand, etc. the SCS and/or SRS.
  • the flexibility of the hollow conduit 920 can allow the hollow conduit 920 to rest at an interface between two adjacent tissue layers that do not strongly adhere to each other (e.g., the sclera 20 and the choroid 28 or the choroid 28 and the retina 27). Hydrodissection then creates and/or expands the potential space (e.g., the SCS orthe SRS).
  • the hollow conduit 920 can have a specific stiffness of about l*10 6 m 2 /s 2 , about 2*10 6 m 2 /s 2 , about 3*10 6 m 2 /s 2 , about 4*10 6 m 2 /s 2 , about 5*10 6 m 2 /s 2 , about 6*10 6 m 2 /s 2 , about 7*10 6 m 2 /s 2 , about 8*10 6 m 2 /s 2 , about 9*10 6 m 2 /s 2 , about l*10 7 m 2 /s 2 , about 2*10 7 m 2 /s 2 , about 3*10 7 m 2 /s 2 , about 4*10 7 m 2 /s 2 , about 5*10 7 m 2 /s 2 , about 6*10 7 m 2 /s 2 , about 7*10 7 m 2 /s 2 , about 8*10 7 m 2 /s
  • the flexibility of the hollow conduit 920 can be modified by bonding with an adhesive (e.g., superglue). In some embodiments, the flexibility of the hollow conduit 920 can be modified by bonding with a UV-cured adhesive. In some embodiments, the hollow conduit 920 can be absent of a pre-curvature, such that the hollow conduit 920 can take on the curvature of the retina 27.
  • the hollow conduit 920 can have a Young’s modulus of at least about 100 MPa, at least about 200 MPa, at least about 300 MPa, at least about 400 MPa, at least about 500 MPa, at least about 600 MPa, at least about 700 MPa, at least about 800 MPa, at least about 900 MPa, at least about 1 GPa, at least about 2 GPa, at least about 3 GPa, at least about 4 GPa, at least about 5 GPa, at least about 6 GPa, at least about 7 GPa, at least about 8 GPa, or at least about 9 GPa.
  • the hollow conduit 920 can have a Young’s modulus of no more than about 10 GPa, no more than about 9 GPa, no more than about 8 GPa, no more than about 7 GPa, no more than about 6 GPa, no more than about 5 GPa, no more than about 4 GPa, no more than about 3 GPa, no more than about 2 GPa, no more than about 1 GPa, no more than about 900 MPa, no more than about 800 MPa, no more than about 700 MPa, no more than about 600 MPa, no more than about 500 MPa, no more than about 400 MPa, no more than about 300 MPa, or no more than about 200 MPa.
  • Combinations of the above-referenced values of the Young’s modulus of the hollow conduit 920 are also possible (e.g., at least about 100 MPa and no more than about 10 GPa or at least about 500 MPa and no more than about 5 GPa), inclusive of all values and ranges therebetween.
  • the hollow conduit 920 can have a Young’s modulus of about 100 MPa, about 200 MPa, about 300 MPa, about 400 MPa, about 500 MPa, about 600 MPa, about 700 MPa, about 800 MPa, about 900 MPa, about 1 GPa, about 2 GPa, about 3 GPa, about 4 GPa, about 5 GPa, about 6 GPa, about 7 GPa, about 8 GPa, about 9 GPa, or about 10 GPa.
  • inj ectate administration can be monitored in real time.
  • monitoring inj ectate administration can include illuminating the hollow conduit 920 and/or the balloon 976 during administration.
  • inj ectate administration can include incorporation of inj ectate material that can be excited via an external stimulation.
  • the external stimulation can include fluorescent excitation, infrared (IR) excitation, ultraviolet (UV) excitation, or any other suitable excitation method or combiantions thereof.
  • FIGS. 14A-14D show an injection apparatus 1000 and a method of using the injection apparatus 1000, according to an embodiment.
  • Layers of the eye 10 depicted in FIGS. 14A-14D include the sclera 20, the choroid 28, and the retina 27.
  • the injection apparatus 1000 includes a hollow conduit 1020, an outer sleeve 1070 (also referred to as a sheath 1070), a catheter 1074, and a balloon 1076.
  • the hollow conduit 1020, the outer sleeve 1070, and the balloon 1076 can be the same or substantially similar to the hollow conduit 920, the outer sleeve 970, and the balloon 976, as described above with reference to FIGS. 13A-13O.
  • certain aspects of the hollow conduit 1020, the outer sleeve 1070, and the balloon 1076 are not described in greater detail herein.
  • FIG. 14A shows the injection apparatus 1000 approaching the eye 10.
  • the sheath 1070 of the injection apparatus 1000 has begun to penetrate the sclera 20.
  • the sheath 1070 punctures the sclera 20 to access the SCS.
  • the sheath 1070 can include a needle.
  • the sheath 1070 can include a hypotube needle.
  • the sheath 1070 can include a trocar.
  • the sheath 1070 can include a hypotube trocar.
  • the sheath 1070 can include a 17Ga needle, an 18Ga neele, a 19Ga needle, a 20Ga needle, a 21Ga needle, a 22Ga needle, a 23 Ga needle, a 24Ga needle, a 25Ga needle, or any other suitable size needle.
  • the sheath 1070 can have a length of at least about 500 pm, at least about 600 pm, at least about 700 pm, at least about 800 pm, at least about 900 pm, at least about 1 mm, at least about 1.1 mm, at least about 1.2 mm, at least about 1.3 mm, or at least about 1.4 mm.
  • the sheath 1070 can have a length of no more than about 1.5 mm, no more than about 1.4 mm, no more than about 1.3 mm, no more than about 1.2 mm, no more than about 1.1 mm, no more than about 1 mm, no more than about 900 gm, no more than about 800 gm, no more than about 700 gm, or no more than about 600 gm. Combinations of the above-referenced lengths of the sheath 1070 are also possible (e.g., at least about 500 pm and no more than about 1.5 mm or at least about 900 pm and no more than about 1.1 mm), inclusive of all values and ranges therebetween.
  • the sheath 1070 can have a length of about 500 pm, about 600 pm, about 700 pm, about 800 pm, about 900 pm, about 1 mm, about 1.1 mm, about 1.2 mm, about 1.3 mm, about 1.4 mm, or about 1.5 mm.
  • the catheter 1074 is deployed from the sheath 1070, such that a distal end of the catheter 1070 is between the sclera 20 and the choroid 28.
  • the balloon 1076 is inflated to secure the distal end of the catheter between the sclera 20 and the choroid 28. Inflation of the balloon 1076 can aid in preventing the balloon 1076 and the distal end of the catheter 1074 from moving around. In other words, inflation of the balloon 1076 can create tension on the choroid 28.
  • the catheter 1074 can be a multilumen balloon catheter.
  • the catheter 1074 can be a IFr, a 1.1 Fr, a 1.2 Fr, a 1.3 Fr, a 1.4 Fr, or a 1.5 Fr sized multilumen balloon catheter, includsive of all sizes and size ranges therebetween.
  • the catheter 1074 can include a 72D Pebax® catheter, a 70D Pebax® catheter, a 63D Pebax® catheter, a 55D Pebax® catheter, a 40D Pebax® catheter, a 35D Pebax® catheter, or a 25D Pebax® catheter.
  • the balloon 1076 can be composed of urethane. In some embodiments, the balloon 1076 can have a material thickness of about 10 pm, about 15 pm, about 20 pm, about 25 pm, about 30 pm, about 35 pm, about 40 pm, about 45 pm, about 50 pm, inclusive of all values and ranges therebetween.
  • a separation distance between the sclera 20 and the choroid 28 created by the inflation of the balloon 1076 can be about 0.5 mm, about 0.6 mm, about 0.7 mm, about 0.8 mm, about 0.9 mm, about 1 mm, about 1.1 mm, about 1.2 mm, about 1.3 mm, about 1.4 mm, about 1.5 mm, about 1.6 mm, about 1.7 mm, about 1.8 mm, about 1.9 mm, or about 2 mm, inclusive of all values and ranges therebetween.
  • the hollow conduit 1020 is deployed from the catheter 1074 and inserted into the retina 27 of the eye 10.
  • the hollow conduit 1020 can include a needle.
  • the hollow conduit 1020 can include a microneedle. In some embodiments, the hollow conduit 1020 can include a 35Ga microneedle, a 36Ga microneedle, a 37Ga microneedle, a 38Ga microneedle, a 39Ga microneedle, a 40Ga needle, a 41Ga needle, a 42Ga needle, a 43 Ga needle, a 44Ga needle, a 45Ga needle, or any other suitable size microneedle. In some embodiments, the hollow conduit 1020 can be composed of a polymer, a metal, a polyamide, or any other suitable material.
  • the balloon 1076 is inserted and inflated between the sclera 20 and the choroid 28.
  • the balloon 1076 can be inserted and inflated between the choroid 28 and the retina 27.
  • Location of the insertion and inflation of the balloon 1076 can be selected based on the desired injection location. For example, if the desired injection location is in the SRS or in the retinal tissue, the balloon 1076 can be inserted and inflated between the choroid 28 and the retina 27. If the desired injection location is in the SCS or in the choroid, the balloon 1076 can be inserted and inflated between the sclera 20 and the choroid 28.
  • FIGS. 15A-15E illustrate an injection cover 1180 and the placement of the injection cover 1180 on the eye 10, according to an embodiment.
  • the injection cover 1180 includes an adhesive backing 1181 and an injection port 1182.
  • the injection cover 1180 can include an iris hole 1183, such that placement of the injection cover 1180 on the eye 10 does not place the injection cover 1180 in contact with the iris.
  • the adhesive backing can be removed from the injection cover 1180 and the injection cover 1180 can be placed on the eye 10. After removal of the adhesive backing 1181, the injection cover 1180 can be placed on the eye 10.
  • FIG. 15C shows a top view of the injection cover 1180 placed on the eye 10.
  • FIG. 15D shows a cross-sectional view of the injection cover 1180 placed on the eye 10.
  • FIG. 15E shows a cross-sectional view of the injection cover 1180 placed on the eye 10 with a medicament container 1150 configured to inject a medicament into the eye 10 via an injection controller 1160 and a hollow conduit 1120.
  • the hollow conduit 1120, the medicament container 1150, and the injection controller 1160 can be the same or substantially similar to the hollow conduit 120, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • certain aspects of the hollow conduit 1120, the medicament container 1150, and the injection controller 1160 are not described in greater detail herein.
  • the injection port 1182 can form an angle with the outside surface of the eye 10 to guide the angle of entry of the hollow conduit 1120. In some embodiments, the injection port 1182 can form an angle with a line L extending perpendicular from the surface of the eye 10. In some embodiments, the angle formed between the line L and the injection port 1182 can be at least about 0 degrees, at least about 5 degrees, at least about 10 degrees, at least about 15 degrees, at least about 20 degrees, at least about 25 degrees, at least about 30 degrees, at least about 35 degrees, at least about 40 degrees, at least about 45 degrees, at least about 50 degrees, or at least about 55 degrees.
  • the angle formed between the line L and the injection port 882 can be no more than about 60 degrees, no more than about 55 degrees, no more than about 50 degrees, no more than about 45 degrees, no more than about 40 degrees, no more than about 35 degrees, no more than about 30 degrees, no more than about 25 degrees, no more than about 20 degrees, no more than about 15 degrees, no more than about 10 degrees, or no more than about 5 degrees.
  • Combinations of the above-referenced angles formed between the line L and the injection port 1182 are also possible (e.g., at least about 0 degrees and no more than about 60 degrees or at least about 10 degrees and no more than about 45 degrees), inclusive of all values and ranges therebetween.
  • the angle formed between the line L and the injection port 1182 can be about 0 degrees, about 5 degrees, about 10 degrees, about 15 degrees, about 20 degrees, about 25 degrees, about 30 degrees, about 35 degrees, about 40 degrees, about 45 degrees, about 50 degrees, about 55 degrees, or about 60 degrees.
  • FIGS. 16A-16E are illustrations of an injection apparatus 1200 and a method of operating the injection apparatus 1200, according to an embodiment.
  • the injection apparatus 1200 includes a housing 1210, a vacuum handle 1213, a vacuum space 1214, a flange 1215, a hollow conduit 1220, a medicament container 1250, and an injection controller 1260.
  • the housing 1210, the hollow conduit 1220, the medicament container 1250, and the injection controller 1260 can be the same or substantially similar to the housing 110, the hollow conduit 120, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • certain aspects of the housing 1210, the hollow conduit 1220, the medicament container 1250, and the injection controller 1260 are not described in greater detail herein.
  • FIG. 16A shows the injection apparatus 1200 spaced apart from the eye 10.
  • FIG. 16B shows the injection apparatus 1200 contacting the outer surface of the eye.
  • the flange 1215 forms a seal with the outer surface of the eye 10.
  • the vacuum handle 1213 is pulled to reduce the pressure in the vacuum space 1214. This reduction in pressure in the vacuum space 1214 draws the sclera 20, the choroid 28, and the retina 27 toward the injection apparatus 1200.
  • the vacuum created aids in keeping the sclera 20, the choroid 28, and the retina 27 in a fixed position relative to the injection apparatus 1200, such that the injection apparatus 1200 does not slide along the surface of the eye 10 during injection of the medicament.
  • the injection controller 1260 can be pressed to deploy the medicament into the desired space in the eye 10.
  • the injection controller 1260 can be pressed to deploy the medicament and the vacuum handle 1213 can be pulled to reduce the pressure in the vacuum space 1214 simultaneously.
  • the vacuum handle 1213 can be pulled to reduce the pressure in the vacuum space 1214 during a first time period and the injection controller 1260 can be pressed to deploy the medicament during a second time period.
  • the first time period can be completely different from the second time period (i.e., no overlap).
  • the first time period can have some overlap with the second time period. In some embodiments, there can be signigicant overlap between the first time period and the second time period, such that they occur simultaneously.
  • the injection controller 1260 can act as an actuator to advance the hollow conduit 1220 toward the eye 10. In some embodiments, the injection controller 1260 can both advance the hollow conduit 1220 toward the eye 10 and inject the medicament into the eye 10.
  • the vacuum pulled in the vacuum space 1214 can result in a pressure reduction in the vacuum space 1214 of at least about 0 bar, at least about 0.1 bar, at least about 0.2 bar, at least about 0.3 bar, at least about 0.4 bar, at least about 0.5 bar, at least about 0.6 bar, at least about 0.7 bar, at least about 0.8 bar, at least about 0.9 bar, or at least about 1 bar.
  • the vacuum pulled in the vacuum space 1214 can result in a pressure reduction in the vacuum space of no more than about 1 bar, no more than about 0.9 bar, no more than about 0.8 bar, no more than about 0.7 bar, no more than about 0.6 bar, no more than about 0.5 bar, no more than about 0.4 bar, no more than about 0.3 bar, no more than about 0.2 bar, or no more than about 0.1 bar.
  • Combinations of the above-referenced pressure reductions in the vacuum space are also possible (e.g., at least about 0 bar and no more than about 1 bar or at least about 0.3 bar and no more than about 0.7 bar), inclusive of all values and ranges therebetween.
  • the vacuum pulled in the vacuum space 914 can result in a pressure reduction in the vacuum space 914 of about 0 bar, about 0.1 bar, about 0.2 bar, about 0.3 bar, about 0.4 bar, about 0.5 bar, about 0.6 bar, about 0.7 bar, about 0.8 bar, about 0.9 bar, or about 1 bar.
  • the hollow conduit 1220 can include a measurement disposed on the distal end of the hollow conduit 1220.
  • the measurement device can include a pressure measurement device.
  • the measurement device can include an emissivity measurement device.
  • the measurement device can include an electrical impedance device.
  • the measurement device can include an acoustic wave measurement device.
  • FIGS. 17A-17B show an injection apparatus 1300, according to an embodiment.
  • the injection apparatus 1300 includes a housing 1310, a pressure chamber 1316, a hollow conduit 1320, a medicament container 1350, an injection controller 1360, and a trocar 1372.
  • the housing 1310, the hollow conduit 1320, the medicament container 1350, and the injection controller 1360 can be the same or substantially similar to the housing 110, the hollow conduit 120, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • the trocar 1372 can be the same or substantially similar to the trocar 972, as described above with reference to FIGS. 13 A-13E.
  • the injection apparatus 1300 is placed near the surface of the eye and the the trocar 1372 can aid in placement of any guiding apparatus on the eye, as shown in FIG. 17 A.
  • the trocar 1372 can be removed, as shown in FIG. 17B.
  • the pressure chamber 1316 can aid in advancement of the medicament container 1350 and the hollow conduit 1320, such that a distal end portion of the hollow conduit 1320 is placed at a desired location in the eye.
  • the medicament can be injected into the eye.
  • the injection controller 1360 can move relative to the medicament container 1350 to inject the medicament into the desired location within the eye.
  • the pressure chamber 1316 can apply a force to the injection controller 1360, such that the injection controller 1360 deploys the medicament via the hollow conduit 1320 when the distal end of the hollow conduit 1320 reaches an area of low resistance (e.g., when the distal end of the hollow conduit 1320 passes from the sclera to the SCS). This loss of resistance mechanism is described in greater detail in the ‘ 139 patent.
  • the pressure chamber 1316 can be maintained at a constant pressure. In some embodiments, the pressure chamber 1316 can be maintained at constant pressure via an internal pumping mechanism. In some embodiments, the pressure chamber 1316 can be pressurized via manual actuation (e.g., via a mechanical hand pump). In some embodiments, a mechanical pump that pressurizes the pressure chamber 1316 can include a one-way valve to restrict flow of gas out of the pressure chamber. In some embodiments, the pressure chamber 1316 can be pressurized via an automatic mechanical pump. In some embodiments, the pressure chamber can be pressurized via a fluidic coupling with a high pressure gas (e.g., a CO2 cartridge).
  • a high pressure gas e.g., a CO2 cartridge
  • the pressure chamber 1316 can be maintained at a pressure of at least about 0.1 bar gauge, at least about 0.2 bar gauge, at least about 0.3 bar gauge, at least about 0.4 bar gauge, at least about 0.5 bar gauge, at least about 0.6 bar gauge, at least about 0.7 bar gauge, at least about 0.8 bar gauge, at least about 0.9 bar gauge, at least about 1 bar gauge, at least about 2 bar gauge, at least about 3 bar gauge, at least about 4 bar gauge, at least about 5 bar gauge, at least about 6 bar gauge, at least about 7 bar gauge, at least about 8 bar gauge, or at least about 9 bar gauge.
  • the pressure chamber 1316 can be maintained at a pressure of no more than about 9 bar gauge, no more than about 8 bar gauge, no more than about 7 bar gauge, no more than about 6 bar gauge, no more than about 5 bar gauge, no more than about 4 bar gauge, no more than about 3 bar gauge, no more than about 2 bar gauge, no more than about 1 bar gauge, no more than about 0.9 bar gauge, no more than about 0.8 bar gauge, no more than about 0.7 bar gauge, no more than about 0.6 bar gauge, no more than about 0.5 bar gauge, no more than about 0.4 bar gauge, no more than about 0.3 bar gauge, no more than about 0.2 bar gauge, or no more than about 0.1 bar gauge.
  • Combinations of the above-referenced pressures in the pressure chamber 1316 are also possible (e.g., at least about 0.1 bar gauge and no more than about 10 bar gauge or at least about 0.3 bar gauge and no more than abou 1 bar gauge), inclusive of all values and ranges therebetween.
  • the pressure chamber 1316 can be maintained at a pressure of about 0.1 bar gauge, about 0.2 bar gauge, about 0.3 bar gauge, about 0.4 bar gauge, about 0.5 bar gauge, about 0.6 bar gauge, about 0.7 bar gauge, about 0.8 bar gauge, about 0.9 bar gauge, about 1 bar gauge, about 2 bar gauge, about 3 bar gauge, about 4 bar gauge, about 5 bar gauge, about 6 bar gauge, about 7 bar gauge, about 8 bar gauge, about 9 bar gauge, or about 10 bar gauge.
  • FIGS. 18A-18C show an injection apparatus 1400, according to an embodiment.
  • FIG. 18A shows the injection apparatus 1400 before the injection begins.
  • FIG. 18B shows a cross section of a portion of a hollow conduit 1420 of the injection apparatus 1400.
  • FIG. 18C shows the injection apparatus 1400 after the injection has begun.
  • the injection apparatus 1400 includes a housing 1410, the hollow conduit 1420, a sheath 1423, springs 1425a, 1425b (collectively referred to as springs 1425), an actuator contact surface 1426, a deployment chamber 1428, an actuator 1430, trap doors 1431a, 1431b (collectively referred to as trap doors 1431), a medicament container 1450, a void space 1451, injection controllers 1460a, 1460b, 1460c (collectively referred to as injection controllers 1460).
  • the hollow conduit 1420 includes entry ridges 1427a, 1427b (collectively referred to as entry ridges 1427).
  • the housing 1410, the hollow conduit 1420, the actuator 1430, the medicament container 1450, and the injection controllers 1460 can be the same or substantially similar to the housing 110, the hollow conduit 120, the actuator 130, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • certain aspects of the housing 1410, the hollow conduit 1420, the actuator 1430, the medicament container 1450, and the injection controllers 1460 are not described in greater detail herein.
  • the sheath 1423 can provide a protective shell for the hollow conduit 1420 to protect the hollow conduit 1420 from damage.
  • the sheath 1423 can provide a more rigid body than the hollow conduit 1420 to penetrate tougher layers of the eye (e.g., sclera). Once the sclera has been penetrated, the hollow conduit 1420 can be advanced to reach the desired injection site.
  • the entry ridges 1427 can create a dual-lumen cannula for movement of medicament through the hollow conduit 1420.
  • the entry ridge 1427b can provide a flow path for the medicament while the entry ridge 1427a can provide a flow path for gas.
  • the entry ridge 1427a can provide a flow path for gas when the trap door 143 la is open.
  • the entry ridge 1427b can provide a flow path for medicament when the trap door 143 lb is open.
  • the actuator 1430 has an elliptical shape, such that the actuator 1430 can rotate to make contact with and push the actuator contact surface 1426 (i.e., compressing the springs 1425). Pushing the actuator contact surface 1426 can advance the hollow conduit 1420 within the eye.
  • the rotation of the actuator 1430 can simultaneously move the trap doors 1431.
  • the trap door 1431b contains the medicament while the trap door 1431a can contain the void space 1451.
  • rotation of the actuator 1430 can shift the trap door 1431b that contains the medicament to an open position.
  • the rotation of the actuator 1430 can shift the trap door 1431a that can contain the void space 1451 to a closed position, such that the trap door 143 la forms a seal with a wall of the housing and contains the void space 1451. Forming the seal with the wall of the housing 1410 can prevent gas from flowing into the void space 1451, thereby maintaining a high pressure in the medicament container 1450.
  • the medicament can flow into the hollow conduit 1420 via the deployment chamber 1428.
  • the actuator contact surface 1426 can include one or more holes to fluidically couple the medicament container 1450 to the deployment chamber 1428 and allow flow of medicament into the deployment chamber 1428.
  • the actuator contact surface 1426 and the deployment chamber 1428 can act as a female Luer-Lock® fitting. Movement of the medicament can be facilitated by movement of the injection controller 1460c through the medicament container 1450. As shown, the injection controller 1460a is physically coupled to the injection controller 1460b and the injection controller 1460c. In some embodiments, the injection controller 1460a can control the movement of the injection controller 1460c and the injection controller 1460b.
  • the dual-chamber configuration of the injection apparatus 1400 can have several advantages. In some embodiments, force applied to the injection controller 1460a can increase the pressure in the void space 1451 and provide pressure at the distal end of the hollow conduit 1420 to create space in the target tissue (e.g., suprachoroidal space, retinal space).
  • the actuator 1430 can rotate to open the trap door 143 lb while closing the trap door 1431a, and the medicament can flow into the space created.
  • the injection apparatus 1400 can include a measurement device (not shown).
  • the measurement device can have the same or substantially similar properties to the measurement device 140, as described above with reference to FIG. 5.
  • FIGS. 19A-19C show an injection apparatus 1500 and a method of using the injection apparatus 1500, according to an embodiment.
  • the injection apparatus 1500 includes a housing 1510, a vacuum handle 1513, a vacuum space 1514, a vacuum nozzle 1515, a hollow conduit 1520, an actuator 1530, and actuator springs 1531a, 1531b (collectively referred to as actuator springs 1531).
  • the housing 1510, the hollow conduit 1520, and the actuator 1530 can be the same or substantially similar to the housing 110, the hollow conduit 120, and the actuator 130, as described above with reference to FIG. 5.
  • the vacuum handle 1513 and the vacuum space 1514 can be the same or substantially similar to the vacuum handle 1213 and the vacuum space 1214, as described above with reference to FIGS. 16A- 16E.
  • the injection apparatus 1500 can include an injection controller (not shown).
  • the injection controller can be the same or substantially similar to the injection controller 160, as described above with reference to FIG. 5.
  • FIG. 19A shows the injection apparatus 1500 spaced apart from the eye 10.
  • the injection apparatus 1500 is contacting the eye 10, and the vacuum handle 1513 is engaged to pull a portion of the sclera 20 through the vacuum nozzle 1515.
  • the hollow conduit 1520 has been advanced such that a distal end of the hollow conduit 1520 is at a desired injection site.
  • the injection apparatus 1500 is placed such that the vacuum nozzle 1515 contacts the eye 10.
  • the vacuum handle 1513 is then pulled to lower the pressure in the vacuum space 1514. Pulling the vacuum on a portion of the outer surface of the eye 10 can keep the layers of the eye 10 secured during injection.
  • the actuator 1530 then advances the medicament container 1550 and the hollow conduit 1520 such that the distal end of the hollow conduit 1520 is at a desired injection site in the eye 10. As shown, advancement of the medicament container 1550 and the hollow conduit 1520 is performed against a force imposed by the actuator springs 1531.
  • the actuator 1530 can be a micrometer. Afer the distal end of the hollow conduit 1520 is at the desired injection site in the eye 10, the medicament is deployed via the hollow conduit 1520.
  • FIG. 20 shows a flow diagram of a method 1600 for performing an injection into the retinal tissue, according to an embodiment.
  • the method 1600 includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye.
  • the hollow conduit is in fluidic communication with a medicament container, and the medicament container includes a medicament.
  • insertion of the distal tip of the hollow conduit can be automatic.
  • insertion of the distal tip of the hollow conduit can be manual.
  • the medicament can include one or more of the medicaments listed in the ‘ 139 patent.
  • the first distance can be sufficient to reach the SCS.
  • the first distance can be at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, at least about 9.5 mm, at least about 1 cm, at least about 1.5 cm, at least about 2 cm, at least about 2.5 cm, at least about 3 cm, at least about 3.5 cm, at least about 4 cm, or at least about 4.5 cm.
  • the first distance can be no more than about 5 cm, no more than about 4.5 cm, no more than about 4 cm, no more than about 3.5 cm, no more than about 3 cm, no more than about 2.5 cm, no more than about 2 cm, no more than about 1.5 cm, no more than about 1 cm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, or no more than about 1.5 mm.
  • Combinations of the above-referenced values for the first distance are also possible (e.g., at least about 1 mm and no more than about 5 cm or at least about 5 mm and no more than about
  • the first distance can be about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about
  • the insertion can be performed such that a centerline of the hollow conduit and a surface line tangential to a target surface of the eye defines an angle of entry. If the angle of entry is too small or zero degrees, this can potentially deform the sclera and put the retina under tension. In some embodiments, the beveling of the distal end of the hollow conduit and the angle of entry can be selected to minimize deformation of the sclera and tension upon the retina.
  • the angle of entry can be at least about 30 degrees, at least about 35 degrees, at least about 40 degrees, at least about 45 degrees, at least about 50 degrees, at least about 55 degrees, at least about 60 degrees, at least about 65 degrees, at least about 70 degrees, at least about 75 degrees, at least about 80 degrees, at least about 85 degrees, at least about 90 degrees, at least about 95 degrees, at least about 100 degrees, at least about 105 degrees, at least about 110 degrees, at least about 115 degrees, at least about 120 degrees, at least about 125 degrees, at least about 130 degrees, at least about 135 degrees, at least about 140 degrees, or at least about 145 degrees.
  • the angle of entry can be no more than about 150 degrees, no more than about 145 degrees, no more than about 140 degrees, no more than about 135 degrees, no more than about 130 degrees, no more than about 125 degrees, no more than about 120 degrees, no more than about 115 degrees, no more than about 110 degrees, no more than about 105 degrees, no more than about 100 degrees, no more than about 95 degrees, no more than about 90 degrees, no more than about 85 degrees, no more than about 80 degrees, no more than about 75 degrees, no more than about 70 degrees, no more than about 65 degrees, no more than about 60 degrees, no more than about 55 degrees, no more than about 50 degrees, no more than about 45 degrees, no more than about 40 degrees, or no more than about 35 degrees.
  • angles of entries are also possible (e.g., at least about 30 degrees and no more than about 150 degrees), inclusive of all values and ranges therebetween.
  • the angle of entry can be about 30 degrees, about 35 degrees, about 40 degrees, about 45 degrees, about 50 degrees, about 55 degrees, about 60 degrees, about 65 degrees, about 70 degrees, about 75 degrees, about 80 degrees, about 85 degrees, about 90 degrees, about 95 degrees, about 100 degrees, about 105 degrees, about 110 degrees, about 115 degrees, about 120 degrees, about 125 degrees, about 130 degrees, about 135 degrees, about 140 degrees, about 145 degrees, or about 150 degrees.
  • an angle control device can be placed on the outer surface of the eye to guide the movement of the hollow conduit and guide the angle of entry.
  • the method 1600 further includes monitoring pressure in the hollow conduit and the medicament container at step 1602.
  • the pressure in the hollow conduit can be monitored to detect a change (e.g., drop) in pressure in the hollow conduit.
  • a seal between the hollow conduit and the medicament container can be broken prior to monitoring the pressure in the hollow conduit and the medicament container.
  • monitoring the pressure in the hollow conduit and the medicament container can be via a manometer, a digital pressure transducer, a barometer, a pressure tube, a piezometer, a bourdon gauge, a diaphragm pressure gauge, a micromanometer, or any other suitable pressure measurement device, or combinations thereof.
  • the method 1600 further includes confirming disposal or positioning of the distal end portion of the hollow conduit in a reference location of the eye at step 1603.
  • the reference location is proximal to the retina of the eye, and can be detected based on the detected change in pressure.
  • the reference location can be an interface between a choroid tissue and the RPE.
  • the reference location can be in the RPE.
  • the reference location can be in the SCS.
  • the reduction in pressure can be at least about 5 mmHg, at least about 10 mmHg, at least about 20 mmHg, at least about 30 mmHg, at least about 40 mmHg, at least about 50 mmHg, at least about 60 mmHg, at least about 70 mmHg, at least about 80 mmHg, at least about 90 mmHg, at least about 100 mmHg, at least about 125 mmHg, at least about 150 mmHg, at least about 175 mmHg, at least about 200 mmHg, at least about 225 mmHg, at least about 250 mmHg, at least about 275 mmHg, at least about 300 mmHg, at least about 325 mmHg, at least about 350 mmHg, at least about 375 mmHg, at least about 400 mmHg, at least about 425 mmHg, at least about 450 mmHg, at least about 475 mmHg, at least about 400
  • the reduction in pressure can be no more than about 1,000 mmHg, no more than about 975 mmHg, no more than about 950 mmHg, no more than about 925 mmHg, no more than about 900 mmHg, no more than about 875 mmHg, no more than about 850 mmHg, no more than about 825 mmHg, no more than about 800 mmHg, no more than about 775 mmHg, no more than about 760 mmHg, no more than about 750 mmHg, no more than about 725 mmHg, no more than about 700 mmHg, no more than about 675 mmHg, no more than about 650 mmHg, no more than about 625 mmHg, no more than about 600 mmHg, no more than about 575 mmHg, no more than about 550 mmHg, no more than about 525 mmHg, no more than about 500 mmHg, no more than about 475
  • Combinations of the above-referenced values of the reduction in pressure are also possible (e.g., at least about 5 mmHg and no more than about 1,000 mmHg or at least about 50 mmHg and no more than about 500 mmHg), inclusive of all values and ranges therebetween.
  • the reduction in pressure can be about 5 mmHg, about 10 mmHg, about 20 mmHg, about 30 mmHg, about 40 mmHg, about 50 mmHg, about 60 mmHg, about 70 mmHg, about 80 mmHg, about 90 mmHg, about 100 mmHg, about 125 mmHg, about 150 mmHg, about 175 mmHg, about 200 mmHg, about 225 mmHg, about 250 mmHg, about 275 mmHg, about 300 mmHg, about 325 mmHg, about 350 mmHg, about 375 mmHg, about 400 mmHg, about 425 mmHg, about 450 mmHg, about 475 mmHg, about 500 mmHg, about 525 mmHg, about 550 mmHg, about 575 mmHg, about 600 mmHg, about 625 mmHg, about 650 mmHg
  • the reference location can be identified based on a gauge pressure value in the hollow conduit and the medicament container (i.e., the difference between the pressure in the hollow conduit and medicament container and atmospheric pressure). In some embodiments, the reference location can be identified upon the gauge pressure reaching a threshold pressure value.
  • the threshold pressure value can be at least about 5 mmHg gauge, at least about 10 mmHg gauge, at least about 20 mmHg gauge, at least about 30 mmHg gauge, at least about 40 mmHg gauge, at least about 50 mmHg gauge, at least about 60 mmHg gauge, at least about 70 mmHg gauge, at least about 80 mmHg gauge, at least about 90 mmHg gauge, at least about 100 mmHg gauge, at least about 125 mmHg gauge, at least about 150 mmHg gauge, at least about 175 mmHg gauge, at least about 200 mmHg gauge, at least about 225 mmHg gauge, at least about 250 mmHg gauge, at least about 275 mmHg gauge, at least about 300 mmHg gauge, at least about 325 mmHg gauge, at least about 350 mmHg gauge, at least about 375 mmHg gauge, at least about 400 mmHg gauge, at least about 425 mmHg gauge, at
  • the threshold pressure value can be no more than about 900 mmHg gauge, no more than about 875 mmHg gauge, no more than about 850 mmHg gauge, no more than about 825 mmHg gauge, no more than about 800 mmHg gauge, no more than about 775 mmHg gauge, no more than about 760 mmHg gauge, no more than about 750 mmHg gauge, no more than about 725 mmHg gauge, no more than about 700 mmHg gauge, no more than about 675 mmHg gauge, no more than about 650 mmHg gauge, no more than about 625 mmHg gauge, no more than about 600 mmHg gauge, no more than about 575 mmHg gauge, no more than about 550 mmHg gauge, no more than about 525 mmHg gauge, no more than about 500 mmHg gauge, no more than about 475 mmHg gauge, no more than about 450 mmHg gauge, no more than about 425 mm
  • the reference location can be identified based on a gauge pressure value in the hollow conduit and the medicament container (i.e., the difference between the pressure in the hollow conduit and medicament container and atmospheric pressure). In some embodiments, the reference location can be identified upon the gauge pressure reaching a threshold pressure value.
  • the threshold pressure value can be about 5 mmHg gauge, about 10 mmHg gauge, about 20 mmHg gauge, about 30 mmHg gauge, about 40 mmHg gauge, about 50 mmHg gauge, about 60 mmHg gauge, about 70 mmHg gauge, about 80 mmHg gauge, about 90 mmHg gauge, about 100 mmHg gauge, about 125 mmHg gauge, about 150 mmHg gauge, about 175 mmHg gauge, about 200 mmHg gauge, about 225 mmHg gauge, about 250 mmHg gauge, about 275 mmHg gauge, about 300 mmHg gauge, about 325 mmHg gauge, about 350 mmHg gauge, about 375 mmHg gauge, about 400 mmHg gauge, about 425 mmHg gauge, about 450 mmHg gauge, about 475 mmHg gauge, about 500 mmHg gauge, about 525 mmHg gauge, about 550 mmHg gauge, about
  • the method 1600 includes further inserting the distal end portion of the hollow conduit a second distance into an injection region of the eye at step 1604.
  • the second distance can be sufficient for the distal tip of the hollow conduit to reach the injection region.
  • the second distance can be sufficient for the distal tip of the hollow conduit to reach a region between the SCS and a boundary layer between a retinal layer and vitreous space.
  • the second distance can be at least about 50 pm, at least about 75 pm, at least about 100 pm, at least about 125 pm, at least about 150 pm, at least about 175 pm, at least about 200 pm, at least about 225 pm, at least about 250 pm, at least about 275 pm, at least about 300 pm, at least about 350 pm, at least about 400 pm, at least about 450 pm, at least about 500 pm, at least about 550 pm, at least about 600 pm, at least about 650 pm, at least about 700 pm, at least about 750 pm, at least about 800 pm, at least about 850 pm, at least about 900 pm, at least about 950 pm, at least about 1 mm, at least about 1.1 mm, at least about 1.2 mm, at least about 1.3 mm, at least about 1.4 mm, at least about 1.5 mm, at least about 1.6 mm, at least about 1.7 mm, at least about 1.8 mm, or at least about 1.9 mm.
  • the second distance can be no more than about 2 mm, no more than about 1.9 mm, no more than about 1.8 mm, no more than about 1.7 mm, no more than about 1.6 mm, no more than about 1.5 mm, no more than about 1.4 mm, no more than about 1.3 mm, no more than about 1.2 mm, no more than about 1.1 mm, no more than about 1 mm, no more than about 950 pm, no more than about 900 pm, no more than about 850 pm, no more than about 800 pm, no more than about 750 pm, no more than about 700 pm, no more than about 650 pm, no more than about 600 pm, no more than about 550 pm, no more than about 500 pm, no more than about 450 pm, no more than about 400 pm, no more than about 350 pm, no more than about 300 pm, no more than about 275 pm, no more than about 250 pm, no more than about 225 pm, no more than about 200 pm, no more than about 175 pm, no more than about 150 pm,
  • the second distance can be about 50 pm, about 75 pm, about 100 pm, about 125 pm, about 150 pm, about 175 pm, about 200 pm, about 225 pm, about 250 pm, about 275 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, about 500 pm, about 550 pm, about 600 pm, about 650 pm, about 700 pm, about 750 pm, about 800 pm, about 850 pm, about 900 pm, about 950 pm, about 1 mm, about 1.1 mm, about 1.2 mm, about 1.3 mm, about 1.4 mm, about 1.5 mm, about 1.6 mm, about 1.7 mm, about 1.8 mm, about 1.9 mm, or about 2 mm.
  • the method 1600 further includes injecting at least a portion of the medicament into the injection region at step 1605.
  • the injection region can be in the retina. In some embodiments, the injection region can be within a region between the SCS and the boundary layer between the retinal layer and vitreous space. In some embodiments, the injection region can be between the choroid and the boundary layer between the retinal tissue and the vitreous body. In some embodiments, the injection region can be between the RPE and the boundary layer between the retinal tissue and the vitreous body. In some embodiments, the injection region can be between the rods and cones of the retina and the boundary layer between the retinal tissue and the vitreous body.
  • the injection region can be between the bipolar cells of the retina and the boundary layer between the retinal tissue and the vitreous body. In some embodiments, the injection region can be between the ganglion cells of the retina and the boundary layer between the retinal tissue and the vitreous body. In some embodiments, the injection region can be between the SCS and the ganglion cells. In some embodiments, the injection region can be between the choroid and the ganglion cells. In some embodiments, the injection region can be between the RPE and the ganglion cells. In some embodiments, the injection region can be between the rods and cones of the retina and the ganglion cells. In some embodiments, the injection region can be between the bipolar cells of the retina and the ganglion cells.
  • the injection region can be between the SCS and the bipolar cells of the retina. In some embodiments, the injection region can be between the choroid and the bipolar cells of the retina. In some embodiments, the injection region can be between the rods and cones of the retina and the bipolar cells of the retina. In some embodiments, the injection region can be between the SCS and the rods and cones of the retina. In some embodiments, the injection region can be between the choroid and the rods and cones of the retina. In some embodiments, the injection region can be between the RPE and the rods and cones of the retina. In some embodiments, the injection region can be between the RPE and the rod and cone outer segments of the retina.
  • the injection region can be between the RPE and the rod and cone nuclei of the retina. In some embodiments, the injection region can be between the SCS and the RPE. In some embodiments, the injection region can include the choroid and the retina (i.e., the injection region can straddle the RPE).
  • all or substantially all of the medicament can be injected.
  • a portion of the medicament can be injected.
  • the portion of the medicament injected can be at least about 1%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 95%.
  • the portion of the medicament injected can be no more than about 99%, no more than about 95%, no more than about 90%, no more than about 85%, no more than about 80%, no more than about 75%, no more than about 70%, no more than about 65%, no more than about 60%, no more than about 55%, no more than about 50%, no more than about 45%, no more than about 40%, no more than about 35%, no more than about 30%, no more than about 25%, no more than about 20%, no more than about 15%, or no more than about 10%.
  • the portion of the medicament injected can be about 1%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 99%.
  • a volume of the medicament can be injected.
  • the volume can be at least about 10 pL, at least about 20 pL, at least about 30 pL, at least about 40 pL, at least about 50 pL, at least about 60 pL, at least about 70 pL, at least about 80 pL, at least about 90 pL, at least about 100 pL, at least about 200 pL, at least about 300 pL, at least about 400 pL, at least about 500 pL, at least about 600 pL, at least about 700 pL, at least about 800 pL, at least about 900 pL, at least about 1 mL, at least about 2 mL, at least about 3 mL, or at least about 4 mL.
  • the volume can be no more than about 5 mL, no more than about 4 mL, no more than about 3 mL, no more than about 2 mL, no more than about 1 mL, no more than about 900 pL, no more than about 800 pL, no more than about 700 pL, no more than about 600 pL, no more than about 500 pL, no more than about 400 pL, no more than about 300 pL, no more than about 200 pL, no more than about 100 pL, no more than about 90 pL, no more than about 80 pL, no more than about 70 pL, no more than about 60 pL, no more than about 50 pL, no more than about 40 pL, no more than about 30 pL, or no more than about 20 pL.
  • the volume can be about 10 pL, about 20 pL, about 30 pL, about 40 pL, about 50 pL, about 60 pL, about 70 pL, about 80 pL, about 90 pL, about 100 pL, about 200 pL, about 300 pL, about 400 pL, about 500 pL, about 600 pL, about 700 pL, about 800 pL, about 900 pL, about 1 mL, about 2 mL, about 3 mL, about 4 mL, or about 5 mL.
  • the method 1600 can further include removing the hollow conduit from the eye at step 1606.
  • the removal can be manual.
  • the removal can be automatic.
  • the removal can be actuated (e.g., via a piezo motor).
  • FIG. 21 shows a flow diagram of a method 1700 for performing an injection into the retinal tissue using light emissivity measurements, according to an embodiment.
  • the method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye at step 1701.
  • the method 1700 further includes monitoring a light emissivity gradient between the sclera and the RPE at step 1702, confirming disposal of the distal end portion of the hollow conduit in a reference location of the eye, based on a measured light emissivity gradient at step 1703, further inserting the distal tip of the needle a second distance into the eye at step 1704, injecting at least a portion of the medicament into the retina at step 1705, and removing the hollow conduit from the eye at step 1706.
  • step 1701, step 1704, step 1705, and step 1706 can be the same or substantially similar to step 1601, step 1604, step 1605, and step 1606, respectively, as described above with reference to FIG. 20. Thus, certain elements of step 1701, step 1704, step 1705, and step 1706 are not described in greater detail herein.
  • step 1702 can include a constant measurement of emissivity at the distal end of the hollow conduit. In some embodiments, step 1702 can include a periodic measurement of emissivity at the distal end of the hollow conduit. In some embodiments, the measurement of emissivity at the distal end of the hollow conduit can be implemented throughout the execution of the method 1700. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the SCS.
  • the measurement can begin as soon as the distal end of the hollow conduit enters the SCS. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the RPE. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the RPE.
  • the reference location identified in step 1703 can be in the RPE. In some embodiments, locating the RPE can be reliably done via light emissivity measurements. In some embodiments, the reference location can be an interface between a choroid tissue and a retinal pigment epithelium. In some embodiments, the reference location can be in the SCS. In some embodiments, identifying the reference location at step 1703 can be based on a light emissivity gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • the reference location can be identified based on a single light emissivity value, such as the fraction of light emitted through the pupil (or light emitted from the medical injector) that reaches the distal end of the hollow conduit.
  • the emissivity value can be at least about 0.05, at least about 0.1, at least about 0.15, at least about 0.2, at least about 0.25, at least about 0.3, at least about 0.35, at least about 0.4, at least about 0.45, at least about 0.5, at least about 0.55, at least about 0.60, at least about 0.65, at least about 0.7, at least about 0.75, at least about 0.80, at least about 0.85, at least about 0.90, or at least about 0.95.
  • the emissivity value can be no more than about 0.99, no more than about 0.95, no more than about 0.9, no more than about 0.85, no more than about 0.8, no more than about 0.75, no more than about 0.7, no more than about 0.65, no more than about 0.6, no more than about 0.55, no more than about 0.5, no more than about 0.45, no more than about 0.4, no more than about 0.35, no more than about 0.3, no more than about 0.25, no more than about 0.2, no more than about 0.15, or no more than about 0.1.
  • the above referenced emissivity values are also possible (e.g., at least about 0.05 and no more than about 0.99 or at least about 0.5 and no more than about 0.75), inclusive of all values and ranges therebetween.
  • the emissivity value can be about 0.05, about 0.1, about 0.15, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.60, about 0.65, about 0.7, about 0.75, about 0.80, about 0.85, about 0.90, about 0.95, or about 0.99.
  • the reference location can be identified by a sudden change in emissivity while inserting the distal end of the hollow conduit into the eye.
  • the change in emissivity can be a change value per pm of insertion depth into the eye.
  • the change in emissivity indicating reaching the reference location can be at least about 0.0001/pm, at least about 0.0005/pm, at least about 0.001/pm, at least about 0.005/pm, at least about 0.01/pm, at least about 0.02/pm, at least about 0.03/pm, at least about 0.04/pm, at least about 0.05/pm, at least about 0.06/pm, at least about 0.07/pm, at least about 0.08/pm, or at least about 0.09/pm.
  • the change in emissivity indicating reaching the reference location can be no more than about 0.1/pm, no more than about 0.09/pm, no more than about 0.08/pm, no more than about 0.07/pm, no more than about 0.06/pm, no more than about 0.05/pm, no more than about 0.04/pm, no more than about 0.03/pm, no more than about 0.02/pm, no more than about 0.01/pm, no more than about 0.005/pm, no more than about 0.001/pm, or no more than about 0.0005/pm.
  • the change in emissivity indicating reaching the reference location can be about 0.0001/pm, about 0.0005/pm, about 0.001/pm, about 0.005/pm, about 0.01/pm, about 0.02/pm, about 0.03/pm, about 0.04/pm, about 0.05/pm, about 0.06/pm, about 0.07/pm, about 0.08/pm, about 0.09/pm, or about 0.1/pm.
  • FIG. 22 shows a flow diagram of a method 1800 for performing an injection into the retinal tissue, according to an embodiment.
  • the method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye at step 1801.
  • the method 1800 further includes monitoring an acoustic wave gradient between the sclera and the RPE at step 1802, confirming disposal of the distal end portion of the hollow conduit in a reference location of the eye, based on a measured acoustic wave gradient at step 1803, further inserting the distal tip of the needle a second distance into the eye at step 1804, injecting at least a portion of the medicament into the retina at step 1805, and removing the hollow conduit from the eye at step 1806.
  • step 1801, step 1804, step 1805, and step 1806 can be the same or substantially similar to step 1601, step 1604, step 1605, and step 1606, respectively, as described above with reference to FIG. 20. Thus, certain elements of step 1801, step 1804, step 1805, and step 1806 are not described in greater detail herein.
  • step 1802 can include a constant measurement of emissivity at the distal end of the hollow conduit.
  • step 1802 can include a periodic acoustic wave measurement at the distal end of the hollow conduit.
  • the acoustic wave measurement at the distal end of the hollow conduit can be implemented throughout the execution of the method 1800.
  • the measurement can begin as soon as the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the SCS. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the SCS. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the RPE. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the RPE.
  • the reference location identified in step 1803 can be in the RPE. In some embodiments, locating the RPE can be reliably done via acoustic wave measurements. In some embodiments, the reference location can be an interface between a choroid tissue and a retinal pigment epithelium. In some embodiments, the reference location can be in the SCS. In some embodiments, identifying the reference location at step 1803 can be based on an acoustic wave gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • the reference location can be identified based on a single threshold acoustic wave measurement. In some embodiments, the reference location can be identified by a sudden acoustic wave change while inserting the distal end of the hollow conduit into the eye. In some embodiments, the acoustic wave change can be a change value per pm of insertion depth into the eye.
  • FIG. 23 shows a flow diagram of a method 1900 for performing an injection into the retinal tissue, according to an embodiment.
  • the method includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye at step 1901.
  • the method 1900 further includes monitoring an electrical impedance gradient between the sclera and the RPE at step 1902, confirming disposal of the distal end portion of the hollow conduit in a reference location of the eye, based on a measured electrical impedance gradient at step 1903, further inserting the distal tip of the needle a second distance into the eye at step 1904, injecting at least a portion of the medicament into the retina at step 1905, and removing the hollow conduit from the eye at step 1906.
  • step 1901, step 1904, step 1905, and step 1906 can be the same or substantially similar to step 1601, step 1604, step 1605, and step 1606, respectively, as described above with reference to FIG. 20. Thus, certain elements of step 1901, step 1904, step 1905, and step 1906 are not described in greater detail herein.
  • step 1902 can include a constant measurement of electrical impedance at the distal end of the hollow conduit. In some embodiments, step 1902 can include a periodic electrical impedance measurement at the distal end of the hollow conduit. In some embodiments, the electrical impedance measurement at the distal end of the hollow conduit can be implemented throughout the execution of the method 1900. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the sclera. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the SCS.
  • the measurement can begin as soon as the distal end of the hollow conduit enters the SCS. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the choroid. In some embodiments, the measurement can begin before the distal end of the hollow conduit enters the RPE. In some embodiments, the measurement can begin as soon as the distal end of the hollow conduit enters the RPE.
  • the reference location identified in step 1903 can be in the RPE. In some embodiments, locating the RPE can be reliably done via electrical impedance measurements. In some embodiments, the reference location can be an interface between a choroid tissue and a retinal pigment epithelium. In some embodiments, the reference location can be in the SCS. In some embodiments, identifying the reference location at step 1903 can be based on an electrical impedance gradient between a sclera of the eye and a retinal pigment epithelium of the eye.
  • FIG. 24 shows a flow diagram of a method 2000 for performing an injection into the retinal tissue, according to an embodiment.
  • the method 2000 includes forming a seal between a distal end of a medicament container and an outer surface of an eye at step 2001 and inserting a distal end portion of a catheter of the medical injector a first distance into the eye at step 2002.
  • the method 2000 then optionally includes injecting a liquid into a target tissue via the catheter at step 2003.
  • the method 2000 further includes advancing a hollow conduit through the catheter, such that a distal end of the hollow conduit enters the target tissue, the hollow conduit in fluidic communication with a medicament container, the medicament container including a medicament at step 2004 and injecting at least a portion of the medicament into the target tissue at step 2005.
  • the method 2000 optionally includes removing the hollow conduit from the eye in step 2006.
  • step 2001 can include scoring the sclera of the eye.
  • forming the seal between the distal end of the medical injector and the outer surface of the eye can include advancing a medicament container such that a distal end of the medicament container contacts the sclera.
  • the catheter inserted in step 2002 can be in fluidic communication with a fluid container.
  • the fluid container can include a liquid.
  • Step 2003 optionally includes injecting the liquid into a target tissue via the catheter.
  • the liquid can include a saline solution a carboxymethyl cellulose (CMC) solution, and/or a hyaluronic acid (HA) solution.
  • CMC carboxymethyl cellulose
  • HA hyaluronic acid
  • injection of the liquid into the target tissue can occur simultaneously to advancement of a medicament container.
  • advancement of the medicament container can also cause the liquid to be injected into the target tissue.
  • advancement of the medicament container can be manual.
  • the medicament container can advance automatically (e.g., via a loss-of-resistance mechanism).
  • Step 2004 includes advancing a hollow conduit through the catheter.
  • the hollow conduit can include a needle.
  • the hollow conduit is in fluidic communication with a medicament container and the medicament container includes a medicament.
  • the hollow conduit can include a microneedle.
  • Step 2005 includes injecting at least a portion of the medicament into the target tissue.
  • step 2005 can include manual injection of the medicament into the target tissue.
  • step 2005 can include automatic injection of the medicament into the target tissue (i.e., via a loss-of-resistance mechanism).
  • Step 2006 includes optionally removing the hollow conduit from the eye.
  • FIG. 25 shows a flow diagram of a method 2100 for performing an injection into the target tissue, according to an embodiment.
  • the method 2100 includes delivering an outer sleeve to a desired region of an eye via a trocar at step 2101.
  • the method 2100 includes removing the trocar from the outer sleeve at step 2102, inserting a cannula into the outer sleeve at step 2103.
  • the cannula has a distal end with a balloon disposed thereon.
  • the method 2100 optionally includes removing the outer sleeve from the eye at step 2104.
  • the method 2100 includes inflating the balloon to create a tension between two adjacent layers of the eye at step 2105.
  • the method 2100 includes advancing a hollow conduit through the cannula at step 2106, such that a distal end of the hollow conduit enters a target tissue, the hollow conduit in fluidic communication with a medicament container.
  • the medicament container includes a medicament.
  • the method 2100 includes injecting at least a portion of the medicament into the target tissue at step 2107, optionally removing the hollow conduit from the eye at step 2108, and optionally removing the cannula from the eye at step 2109.
  • Step 2101 includes delivering an outer sleeve to an eye via a trocar.
  • the trocar can have a sharp distal surface that can penetrate the sclera of the eye.
  • the trocar can be positioned in a lumen of the outer sleeve.
  • the outer sleeve can be situated around the outside of the trocar.
  • the outer sleeve can have a sharp distal surface that can penetrate the sclera of the eye.
  • the outer sleeve can include a stop to stop the outer sleeve from advancing any further once the distal end of the outer sleeves reaches a desired depth inside the eye.
  • Step 2102 includes removing the trocar from the outer sleeve. Upon the removal of the trocar, the outer sleeve remains disposed in the eye. In some embodiments, removal of the trocar from the outer sleeve can include pulling the trocar manually from the eye. In some embodiments, removal of the trocar from the outer sleeve can be automated.
  • Step 2103 includes inserting a cannula into the outer sleeve.
  • the cannula can be inserted into the lumen of the outer sleeve.
  • the distal end of the cannula has a balloon disposed thereon.
  • the cannula can be inserted into the outer sleeve such that balloon is between the sclera and the retina.
  • the cannula can be inserted into the outer sleeve such that balloon is between the sclera and the choroid.
  • the cannula can be inserted into the outer sleeve such that balloon is between the choroid and the retina.
  • the cannula can include a conduit therein, the conduit fluidically coupled to the balloon. The conduit can allow for flow of fluid (e.g., gas, liquid) to inflate the balloon.
  • Step 2104 is optional and includes removing the outer sleeve from the eye.
  • removing the outer sleeve can be via manual pulling of the outer sleeve.
  • removing the outer sleeve can be automated.
  • Step 2105 includes inflating the balloon to create a tension between between two adjacent layers of the eye.
  • inflating the balloon can create a tension between the sclera and the choroid.
  • inflating the balloon can create a tension between the choroid and the retina. The tension can aid in securing the placement of the distal end of the balloon at the desired location in the eye (e.g., between the choroid and the retina or between the sclera and the choroid) and preventing lateral movements of the eye.
  • the inflation and securement of the balloon creates a stable anchor point, and the medicament can be injected into a location at a specified distance distal to the stable anchor point (e.g., about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, or about 500 pm distal to the stable anchor point, inclusive of all values and ranges therebetween).
  • a specified distance distal to the stable anchor point e.g., about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, or about 500 pm distal to the stable anchor point, inclusive of all values and ranges therebetween.
  • Step 2106 includes advancing a hollow conduit through the cannula, such that a distal end of the hollow conduit enters the target tissue.
  • the hollow conduit is in fluidic communication with a medicament container and the medicament container includes a medicament.
  • the hollow conduit can be a part of an injection apparatus.
  • the hollow conduit can be advanced such that a distal end of the hollow conduit is in the retinal tissue of the eye.
  • the hollow conduit can be advanced such tha the distal end of the hollow conduit is in the choroidal tissue of the eye.
  • Step 2107 includes injecting at least a portion of the medicament into the target tissue.
  • the injecting can be via pushing on an injection controller.
  • Step 2108 is optional and includes removing the hollow conduit from the eye.
  • removing the hollow conduit from the eye can be done by manually pulling the hollow conduit from the cannula.
  • removing the hollow conduit from the eye can be automated.
  • step 2107 can include automatic injection of the medicament into the target tissue (i.e., via a loss-of-resistance mechanism).
  • Step 2109 is optional and includes removing the cannula from the eye.
  • removing the cannula can be done by manually pulling the cannula from the eye.
  • the removal of the cannula can be automated.
  • the balloon can be deflated prior to removing the cannula from the eye.
  • FIG. 26 is a schematic illustration of an injection apparatus 2200, according to an embodiment.
  • the injection apparatus 2200 includes a housing 2210, a hollow conduit 2220, a medicament container 2250, an inflation medium container 2255, an injection controller 2260, an inflation controller 2265, a catheter 2270, and a balloon 2276.
  • the hollow conduit 2220, the catheter 2270, and the balloon 2276 can be the same or substantially similar to, in form and/or in function, the hollow conduit 920, the outer sleeve 970, and the balloon 976, as described above with reference to FIGS. 13 A-13O.
  • the medicament container 2250 and the injection controller 2260 can be the same or substantially similar to, in form and/or in function, the housing 110, the medicament container 150, and the injection controller 160, as described above with reference to FIG. 5.
  • certain aspects of the housing 2210, the hollow conduit 2220, the medicament container 2250, the injection controller 2260, the catheter 2270, and the balloon 2276 are not described in greater detail herein.
  • the balloon 2276 can be disposed at a distal end of the housing 2210. In some embodiments, the balloon 2276 can be disposed at a distal end of the catheter 2270.
  • the inflation medium container 2255 is fluidically coupled to the balloon 2276, such that the inflation medium container 2255 dispenses an inflation medium to inflate the balloon 2276.
  • the inflation controller 2265 moves the inflation medium from the inflation medium container 2255 to the balloon 2276.
  • the inflation controller 2265 can include a plunger.
  • the inflation medium can be transferred to the balloon 2276 manually (e.g., by pushing on the inflation controller 2265).
  • the inflation medium can be transferred to the balloon 2276 automatically.
  • the inflation medium container 2255 can be fluidically coupled to the balloon 2276 via a conduit.
  • the inflation medium can be a gas.
  • the inflation medium can be a liquid.
  • the inflation medium can have any of the properties described above with reference to FIGS.13 A-13O.
  • the balloon 2276 can have an annular shape, such that the hollow conduit 2220 can advance through the annulus of the balloon 2276.
  • FIG. 27 is an illustration of an injection apparatus 2300, according to an embodiment.
  • the injection apparatus 2300 includes a housing 2310, a handle 2319 coupled to the housing 2310, a hollow conduit 2320, a medicament container 2350, a medicament conduit 2351, an inflation medium container 2355, an inflation medium conduit 2356, a conduit union 2357, a conduit casing 2358, an injection controller 2360, an inflation controller 2265, a catheter 2370, and a balloon 2376.
  • the housing 2310, the hollow conduit 2320, the medicament container 2350, the inflation medium container 2355, the injection controller 2360, the inflation controller 2365, the catheter 2370, and the balloon 2376 can be the same or substantially similar to, in form and/or in function, the housing 2210, the hollow conduit 2220, the medicament container 2250, the inflation medium container 2255, the injection controller 2260, the inflation controller 2265, the catheter 2270, and the balloon 2276, as described above with reference to FIG. 26.
  • certain aspects of the housing 2310, the hollow conduit 2320, the medicament container 2350, the inflation medium container 2355, the injection controller 2360, the inflation controller 2365, the catheter 2370, and the balloon 2376 are not described in greater detail herein.
  • the handle 2319 is an optional feature that allows the user to grip the housing 2310 more easily.
  • the medicament conduit 2351 fluidically couples the medicament container 2350 to the hollow conduit 2320.
  • the inflation medium conduit 2356 fluidically couples the inflation medium container 2355 to the balloon 2376.
  • the conduit union 2357 joins the medicament conduit 2351 and the inflation medium conduit 2356 in the conduit casing 2358, without fluidically couplng the the medicament conduit 2351 and the inflation medium conduit 2356.
  • the conduit casing 2358 can include a tube that connects the conduit union 2357 and the housing 2310 (e.g., via the handle 2319).
  • the medicament conduit 2351 and the inflation medium conduit 2356 can separate from each other, as the medicament conduit 2351 joins with the hollow conduit 2320.
  • the inflation medium conduit 2356 can run lengthwise along an interior of the housing 2310 and couple to the balloon 2376. In some embodiments, the inflation medium conduit 2356 can run lengthwise along the interior of the catheter 2370 and couple with the balloon 2376.
  • FIGS. 28A-28F illustrate methods and apparatus for performing an injection into ocular tissue, according to various embodiments.
  • an injection apparatus 2400 includes a housing 2410, a hollow conduit 2420, a catheter 2470, and a balloon 2476.
  • the injection apparatus 2400, the housing 2410, the hollow conduit 2420, the catheter 2470, and the balloon 2476 can be the same or substantially similar to, in form and/or in function, the injection apparatus 2300, the housing 2310, the hollow conduit 2320, the catheter 2370, and the balloon 2376, as described above with reference to FIG. 27, and/or other embodiments described herein.
  • the injection apparatus 2400 can also include the additional components of the injection apparatus 2300 (e.g., the medicament container 2350, the injection medium container 2355, etc.) not shown in FIGS. 28A-28F.
  • FIGS. 28A-28C show a process of injecting a medicament into the SCS or the choroid 28, while FIGS. 28D-28F show a process of injecting the medicament into the SRS or the retina 27.
  • FIGS. 28A shows the injection apparatus 2400 approaching the eye 10.
  • the catheter 2470 and the hollow conduit 2420 have advanced such that the balloon 2476 is at an interface between the sclera 20 and the choroid 28. As shown, the catheter 2470 is coupled to the housing 2410.
  • the catheter 2470 can be fixedly coupled to the housing 2410, such that the catheter 2470 moves with the housing 2410. In some embodiments, the catheter 2470 can be slidably coupled to the housing 2410, such that the catheter 2470 can move independently of the housing 2410. As shown, the hollow conduit 2420 is fixedly coupled to the catheter 2470. In some embodiments, the hollow conduit 2420 can be slidably coupled to the catheter 2470. As shown, the advancement of the catheter and the hollow conduit 2420 has caused the hollow conduit 2420 to advance into the choroid 28.
  • the position of the opening of the hollow conduit 2420 relative to the sclera 20 and the choroid 28 can be measured via any of the aforementioned methods (e.g., pressure, emissivity).
  • the opening of the hollow conduit is positioned at the distal end of the hollow conduit 2420.
  • the opening of the hollow conduit 2420 can be proximal to the distal end of the hollow conduit 2420. The distal end of the hollow conduit 2420 can then beinserted slightly further into the eye 10, based on the distance between the balloon 2476 and the distal end of the hollow conduit 2420.
  • the opening of the hollow conduit 2420, while the hollow conduit 2420 is fixedly coupled to the catheter 2470, can extend beyond the distal end of the uninflated balloon 2476 by about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, about 500 pm, about 550 pm, about 600 pm, about 650 pm, about 700 pm, about 750 pm, about 800 pm, about 850 pm, about 900 pm, about 950 pm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, or about 5 mm, inclusive of all values and ranges therebetween.
  • the hollow conduit 2420 can slide, such that the opening of the hollow conduit 2420 extends beyond the distal end of the uniflated balloon 2476 by about 50 pm, about 100 pm, about 150 pm, about 200 pm, about 250 pm, about 300 pm, about 350 pm, about 400 pm, about 450 pm, about 500 pm, about 550 pm, about 600 pm, about 650 pm, about 700 pm, about 750 pm, about 800 pm, about 850 pm, about 900 pm, about 950 pm, about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, or about 5 mm, inclusive of all values and ranges therebetween. [00271] FIG.
  • the balloon 2476 in an inflated state.
  • the balloon 2476 can be inflated by conveying an inflation medium from an inflation medium container (not shown) to the balloon 2476 via a conduit (not shown) that fluidically couples the inflation medium container and the balloon 2476.
  • the inflation of the balloon 2476 has caused the layers of the eye 10 to move relative to the hollow conduit 2420 and has created, expanded, and/or opened up the SCS between the sclera 20 and the choroid 28.
  • the position of the opening of the hollow conduit 2420 has retreated relative to the layers of the eye 10 upon inflation of the balloon 2476.
  • the opening of the hollow conduit 2420 Prior to the inflation of the balloon 2476, the opening of the hollow conduit 2420 is in the choroid 28, and after the inflation of the balloon 2476, the opening of the hollow conduit 2420 has retreated to the SCS. As shown, the opening of the hollow conduit 2420 is disposed in the SCS after the inflation of the balloon 2476. In some embodiments, the opening of the hollow conduit 2420 can be disposed in the choroid 28 after inflation of the balloon 2476. In some embodiments, the hollow conduit 2420 can be flexible, such that the hollow conduit 2420 slides more easily in the interfaces between the layers of the eye 10 and does not get caught on various surfaces of the eye 10.
  • the balloon 2476 can be pressurized prior to entering the SCS (e.g., while in the sclera 20), and the balloon 2476 can expand (e.g., automatically) upon entering the relatively low- pressure region of the SCS.
  • the balloon 2476 can inflate via a loss-of-resistance mechanism, such that the balloon 2476 inflates when it moves from a region with a relatively high resistance to expansion to a region with a relatively low resistance to expansion.
  • inflation of the balloon 2476 can serve as an indicator to the user (e.g., by visual confirmation) that the balloon 2476 as reached the SCS (or in other instances, the SRS, retina, or other target region of the eye).
  • the balloon 2476 in its expanded state, can have a diameter or a dimension perpendicular to the length of the hollow conduit 2420 of at least about 1 mm, at least about 1.5 mm, at least about 2 mm, at least about 2.5 mm, at least about 3 mm, at least about 3.5 mm, at least about 4 mm, at least about 4.5 mm, at least about 5 mm, at least about 5.5 mm, at least about 6 mm, at least about 6.5 mm, at least about 7 mm, at least about 7.5 mm, at least about 8 mm, at least about 8.5 mm, at least about 9 mm, or at least about 9.5 mm.
  • the balloon 2476 in its expanded state, can have a diameter or a dimension perpendicular to the length of the hollow conduit 2420 of no more than about 10 mm, no more than about 9.5 mm, no more than about 9 mm, no more than about 8.5 mm, no more than about 8 mm, no more than about 7.5 mm, no more than about 7 mm, no more than about 6.5 mm, no more than about 6 mm, no more than about 5.5 mm, no more than about 5 mm, no more than about 4.5 mm, no more than about 4 mm, no more than about 3.5 mm, no more than about 3 mm, no more than about 2.5 mm, no more than about 2 mm, or no more than about 1.5 mm.
  • the balloon 2476 in its expanded state, can have a diameter or a dimension perpendicular to the length of the hollow conduit 2420 of about 1 mm, about 1.5 mm, about 2 mm, about 2.5 mm, about 3 mm, about 3.5 mm, about 4 mm, about 4.5 mm, about 5 mm, about 5.5 mm, about 6 mm, about 6.5 mm, about 7 mm, about 7.5 mm, about 8 mm, about 8.5 mm, about 9 mm, about 9.5 mm, or about 10 mm.
  • the injection of medicament into a target region can hydrodissect the sclera 20 and the choroid 28 to create the SCS.
  • the injection of the medicament can create a pressure, such that the sclera 20 and the choroid 28 separate to create the SCS.
  • FIGS. 28D-28F show a process of injecting a medicament into the SRS or the retina 27.
  • FIG. 28D shows the injection apparatus approaching the eye 10.
  • catheter 2470 and the hollow conduit 2420 have advanced relative to the layers of the eye 10, such that the balloon 2476 is at an interface between the choroid 28 and the retina 27 and the opening of the hollow conduit 2420 is in the vitreous space 30.
  • FIG. 28F shows the balloon 2476 in an inflated state.
  • the balloon 2476 can be inflated by conveying an inflation medium from an inflation medium container (not shown) to the balloon 2476 via a conduit (not shown) that fluidically couples the inflation medium container and the balloon 2476.
  • the inflation of the balloon 2476 has caused the layers of the eye 10 to retreat relative to the hollow conduit 2420 and has created, expanded, and/or opened up the SRS between the choroid 28 and the retina 27.
  • the position of the opening of the hollow conduit 2420 has changed relative to the layers of the eye 10 upon inflation of the balloon 2476, i.e., its position has moved proximally away from the vitreous space 30 and into the SRS.
  • the location of the opening of the hollow conduit 2420 after inflation of tha balloon 2476 can be a function of the location of the opening of the hollow conduit 2420 prior to inflation of the balloon 2476 and the volume of the balloon 2476 upon inflation.
  • the opening of the hollow conduit 2420 has moved relative to the eye 10 from the vitreous space 30 to the SRS due to the inflation of the balloon 2476.
  • the opening of the hollow conduit 2420 can move from the vitreous space 30 to the retina 27 due to the inflation of the balloon 2476.
  • the balloon 2476 can be pressurized prior to entering the SRS (e.g., while in the choroid 28), and the balloon 2476 can expand upon entering the relatively low-pressure region of the SRS, similar to as described above with respect to the SCS.
  • FIG. 29 shows a flow diagram of a method 2500 for performing an injection into a target region of the eye, according to an embodiment.
  • the method 2500 includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye at step 2501, inserting a distal end portion of a catheter of the medical injector a second distance into the eye, such that a balloon coupled to the distal end portion of the catheter is between the sclera and the retina at step 2502, inflating the balloon to create a space between two adjacent layers of the eye, such that the position of the distal end portion of the hollow conduit changes relative to the layers of the eye and is disposed in a target region at step 2503, and injecting a medicament into the target region via the hollow conduit at step 2504.
  • the method 2500 optionally includes removing the hollow conduit from they eye at step 2505 and removing the catheter from the eye at step 2506.
  • Step 2501 includes inserting a distal end portion of a hollow conduit of a medical injector a first distance into an eye. Inserting the distal end portion of the hollow conduit the first distance into the eye places the distal end of the hollow conduit in a first location in the eye.
  • the first location can be in the vitreous space.
  • the first location can be in the retina.
  • the first location can be in the SRS.
  • the first location can be in the choroid.
  • the first location can be in the SCS.
  • the first location can be in the sclera.
  • Step 2502 includes inserting a distal end portion of the catheter a second distance into the eye.
  • the second distance can be less than the first distance.
  • the inserting of the distal end portion of the catheter and the inserting of the distal end portion of the hollow conduit can be part of the same motion. In other words, the catheter and the hollow conduit can be coupled to each other such that they advance together.
  • Step 2503 includes inflating the balloon to create a space between two adjacent layers and/or potential space of the eye.
  • the two adjacent layers can include the sclera and the choroid.
  • the two adjacent layers can include the choroid and the retina.
  • the potential space can include the SCS.
  • the potential space can include the SRS.
  • Inflating the balloon causes the distal end of the hollow conduit to change position from the first location to a second location, relative to the layers of the eye. This relative movement of the distal end of the hollow conduit can be caused by deformation of the layers of the eye due to the inflation of the balloon.
  • the second location can be in the retina.
  • the second location can be in the SRS. In some embodiments, the second location can be in the choroid. In some embodiments, the second location can be in the SCS. In some embodiments, the second location can be in the sclera.
  • Step 2504 includes injecting a medicament into the target region via the hollow conduit.
  • the target region can be the second location, as described above with reference to step 2503.
  • the hollow conduit can be fluidically coupled to a medicament container, and at least a portion of the medicament in the medicament container can be delivered to the target region.
  • Step 2505 is optional and includes removing the hollow conduit from the eye.
  • Step 2506 is optional and includes removing the catheter from the eye.
  • removal of the hollow conduit and the catheter from the eye can be accomplished in one motion.
  • the hollow conduit and/or the catheter can be removed from the eye via manual pulling.
  • the hollow conduit and/or the catheter can be removed from the eye via automation.
  • the balloon can be deflated prior to removing the catheter from the eye.
  • step 2504 can include automatic injection of the medicament into the target tissue (i.e., via a loss-of-resistance mechanism).
  • an apparatus can include both a pressure measurement device and a light emissivity device for finding the reference location. In some embodiments, an apparatus can include both a pressure measurement device and an acoustic wave measurement device for finding the reference location. In some embodiments, an apparatus can include both a pressure measurement device and an electrical impedance measurement device for finding the reference location. In some embodiments, an apparatus can include both a pressure measurement device and an acoustic wave measurement device for finding the reference location. In some embodiments, an apparatus can include both a light emissivity device and an electrical impedance measurement device for finding the reference location.
  • an apparatus can include both a light emissivity device and an acoustic wave measurement device for finding the reference location. In some embodiments, an apparatus can include both an electrical impedance measurement and an acoustic wave measurement for finding the reference location. In some embodiments, an apparatus can include a pressure measurement device, a light emissivity device, an acoustic wave measurement device, an electrical impedance device, or any combinations thereof.
  • an angiogenesis inhibitor is delivered via the methods and devices described herein, in one embodiment, is interferon gamma ip, interferon gamma ip (Actimmune®) with pirfenidone, ACUHTR028, aVp5, aminobenzoate potassium, amyloid P, ANG1122, ANG1170, ANG3062, ANG3281, ANG3298, ANG4011, anti-CTGF RNAi, Aplidin, astragalus membranaceus extract with salvia and schisandra chinensis, atherosclerotic plaque blocker, Azol, AZX100, BB3, connective tissue growth factor antibody, CT140, danazol, Esbriet, EXC001, EXC002, EXC003, EXC004, EXC005, F647, FG3019, Fibrocorin, Follistatin, FT011, a galectin-3 inhibitor, GKT13783
  • a VEGF modulator is delivered via one of the devices described herein.
  • the VEGF modulator is a VEGF antagonist.
  • the VEGF modulator is a VEGF-receptor kinase antagonist, an anti-VEGF antibody or fragment thereof, an anti-VEGF receptor antibody, an anti-VEGF aptamer, a small molecule VEGF antagonist, a thiazolidinedione, a quinoline or a designed ankyrin repeat protein (DARPin).
  • DARPin ankyrin repeat protein
  • the VEGF antagonist is an antagonist of a VEGF receptor (VEGFR), i.e., a drug that inhibits, reduces, or modulates the signaling and/or activity of a VEGFR.
  • VEGFR may be a membrane-bound or soluble VEGFR.
  • the VEGFR is VEGFR-1, VEGFR-2 or VEGFR-3.
  • the VEGF antagonist targets the VEGF-C protein.
  • the VEGF modulator is an antagonist of a tyrosine kinase or a tyrosine kinase receptor.
  • the VEGF modulator is a modulator of the VEGF-A protein.
  • the VEGF antagonist is a monoclonal antibody.
  • the monoclonal antibody is a humanized monoclonal antibody.
  • the VEGF modulator is one or more of the following: AL8326, 2C3 antibody, AT001 antibody, HyBEV, bevacizumab (Avastin®), ANG3070, APX003 antibody, APX004 antibody, ponatinib (AP24534), BDM-E, VGX100 antibody (VGX100 CIRCADIAN), VGX200 (c-fos induced growth factor monoclonal antibody), VGX300, COSMIX, DLX903/1008 antibody, ENMD2076, sunitinib malate (Sutent®), INDUS815C, R84 antibody, KD019, NM3, allogenic mesenchymal precursor cells combined with an anti-VEGF antagonist (e.g., anti-VEGF antibody), MGCD265, MG516, VEGF-Receptor kinase inhibitor, MP0260, NT503, anti- DLL4/VEGF bispecific antibody, PAN9080
  • an anti-VEGF antagonist
  • an immunosuppressive agent is delivered via one of the devices described herein.
  • the immunosuppressive agent is a glucocorticoid, cytokine inhibitor, cytostatic, alkylating agent, anti-metabolite, folic acid analogue, cytotoxic antibiotic, interferon, opioid, T-cell receptor directed antibody or an IL-2 receptor directed antibody.
  • the immunosuppressive agent is an anti-metabolite and the antimetabolite is a purine analog, pyrimidine analogue, folic acid analogue or a protein synthesis inhibitor.
  • the immunosuppressive agent is an interleukin-2 inhibitor (e.g., basiliximab or daclizumab).
  • interleukin-2 inhibitor e.g., basiliximab or daclizumab.
  • Other immunosuppressive agents amenable for use with the methods and formulations described herein include, but are not limited to cyclophosphamide, nitrosourea, methotrexate, azathioprine, mercaptopurine, fluorouracil, dactinomycin, anthracycline, mitomycin C, bleomycin, mithramycin, muromonab-CD3, cyclosporine, tacrolimus, sirolimus or mycophenolate.
  • the drug formulation comprises an effective amount mycophenolate.
  • the drug formulation delivered via one of the devices described herein comprises an effective amount of vascular permeability inhibitor.
  • the vascular permeability inhibitor is a vascular endothelial growth factor (VEGF) antagonist or an angiotensin converting enzyme (ACE) inhibitor.
  • VEGF vascular endothelial growth factor
  • ACE angiotensin converting enzyme
  • the vascular permeability inhibitor is an angiotensin converting enzyme (ACE) inhibitor and the ACE inhibitor is captopril.
  • the drug is a steroid or a non-steroid anti-inflammatory drug (NSAID).
  • the anti-inflammatory drug is an antibody or fragment thereof, an anti-inflammatory peptide(s) or an anti-inflammatory aptamer(s).
  • Steroidal compounds that can be administered via the methods provided herein include hydrocortisone, hydrocorti sone- 17-butyrate, hydrocorti sone- 17-aceponate, hydrocorti sone- 17- buteprate, cortisone, tixocortol pivalate, prednisolone, methylprednisolone, prednisone, triamcinolone, triamcinolone acetonide, mometasone, amcinonide, budesonide, desonide, fluocinonide, halcinonide, bethamethasone, bethamethasone dipropionate, dexamethasone, fluocortolone, hydrocortisone-17-valerate, halometasone, alclometasone dipropionate, predni carb ate, cl obetasone- 17-butyrate, clobetasol-17-propionate, flu
  • NSAIDs that can be administered via the methods provided herein include salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid derivatives, fenamic acid derivatives and cyclooxygenase-2 (COX-2) inhibitors.
  • the methods provided herein are used to deliver one or more of the following NSAIDs to the eye of a patient in need thereof: acetylsalicylic acid, diflunisal, salsalate, ibuprofen, dexibuprofen, naproxen, fenoprofen, keotoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxaprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac or nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam or isoxicam, mefanamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, celecoxib, refecoxib, valdecoxib, parecoxib, lumiracoxib, etoricoxib
  • anti-inflammatory drugs include, but are not limited to: my cophenolate, remicase, nepafenac, 19AV agonist(s), 19GJ agonists, 2MD analogs, 4SC101, 4SC102, 57-57, 5-HT2 receptor antagonist, 64G12, A804598, A967079, AAD2004, AB1010, AB224050, abatacept, etaracizumab (AbegrinTM), Abevac®, AbGnl34, AbGnl68, Abki, ABN912, ABR215062, ABR224050, cyclosporine (Abrammune®), docosanol (behenyl alcohol, Abreva®), ABS15, ABS4, ABS6, ABT122, ABT325, ABT494, ABT874, ABT963, ABXIL8, ABXRB2, AC430, Accenetra, lysozyme chloride (Acdeam®), ACE77
  • the drug is a drug that inhibits, reduces or modulates the signaling and/or activity of PDGF-receptors (PDGFR).
  • the PDGFR modulator is an anti-PDGF aptamer, an anti-PDGF antibody or fragment thereof, an anti-PDGFR antibody or fragment thereof, or a small molecule antagonist.
  • the PDGF antagonist is an antagonist of the PDGFRa or PDGFRp.
  • the PDGF antagonist is the anti-PDGF-P aptamer El 0030, dasatinib, sunitinib, axitinib, sorefenib, imatinib, imatinib mesylate, nintedanib, pazopanib HC1, ponatinib, MK-2461, pazopanib, crenolanib, PP-121, telatinib, imatinib, KRN 633, CP 673451, TSU-68 (orantinib), Ki8751, amuvatinib, tivozanib, masitinib, motesanib diphosphate, dovitinib, dovitinib dilactic acid, FOVISTA, or linifanib (ABT- 869).
  • the PDGF antagonist also has VEGF antagonist activity.
  • VEGF antagonist activity for example, an anti-VEGF/PDGF-B darpin, dasatinib, dovitinib, Ki8751, telatinib, TSU-68 (orantinib) or motesanib diphosphate are known inhibitors of both VEGF and PDGF, and can be used in the methods described herein.
  • Examples of other suitable drugs for use with the devices and methods described herein include, but are not limited to: A0003, A36 peptide, AAV2-sFLT01, ACE041, ACU02, ACU3223, ACU4429, AdPEDF, aflibercept, AG13958, aganirsen, AGN150998, AGN745, AL39324, AL78898A, AL8309B, ALN-VEG01, alprostadil, AMI 101, amyloid beta antibody, anecortave acetate, Anti-VEGFR-2 Alterase, Aptocine, APX003, ARC1905, ARC1905 with Lucentis, ATG3, ATP -binding cassette, sub-family A, member 4 gene, ATXS10, Avastin with Visudyne, AVT101, AVT2, bertilimumab, bevacizumab with verteporfin, bevasiranib sodium, bevasiranib sodium, bevasi
  • the drug is interferon gamma lb (Actimmune®) with pirfenidone, ACUHTR028, AlphaVBeta5, aminobenzoate potassium, amyloid P, ANG1122, ANG1170, ANG3062, ANG3281, ANG3298, ANG4011, Anti-CTGF RNAi, Aplidin, astragalus membranaceus extract with salvia and schisandra chinensis, atherosclerotic plaque blocker, Azol, AZX100, BB3, connective tissue growth factor antibody, CT140, danazol, Esbriet, EXC001, EXC002, EXC003, EXC004, EXC005, F647, FG3019, Fibrocorin, Follistatin, FT011, Galectin-3 inhibitors, GKT137831, GMCT01, GMCT02, GRMD01, GRMD02, GRN510, Heberon Alfa
  • the drug is AKB9778, bevasiranib sodium, Cand5, choline fenofibrate, Cortiject, c-raf 2-methoxy ethyl phosphorothioate oligonucleotide, DEI 09, dexamethasone, DNA damage inducible transcript 4 oligonucleotide, FOV2304, iCo007, KH902, MP0112, NCX434, Optina, Ozurdex, PF4523655, SARI 118, sirolimus, SK0503 or TriLipix.
  • the drug is selected from VEGF modulators, PDGF modulators, anti-inflammatory drugs.
  • drugs that can be administered via IVT include, but are not limited to: A0003, A0006, Acedolone, AdPEDF, aflibercept, AG13958, aganirsen, AGN208397, AKB9778, AL78898A, amyloid P, Angiogenesis Inhibitor Gene Therapy, ARC 1905, Aurocort, bevasiranib sodium, brimonidine, Brimonidine, brimonidine tartrate, bromfenac sodium, Cand5, CERE140, Ciganclor, CLT001, CLT003, CLT004, CLT005, complement component 5 aptamer (pegylated), complement factor D antibody, Cortiject, c-raf 2-methoxy ethyl phosphorothioate oligonucleotide, cyclosporine, triamcinolone, DEI 09,
  • the reference location can be identified based on a single threshold electrical impedance measurement. In some embodiments, the reference location can be identified by a sudden electrical impedance change while inserting the distal end of the hollow conduit into the eye. In some embodiments, the electrical impedance change can be a change value per pm of insertion depth into the eye.
  • Various concepts may be embodied as one or more methods, of which at least one example has been provided.
  • the acts performed as part of the method may be ordered in any suitable way. Accordingly, embodiments may be constructed in which acts are performed in an order different than illustrated, which may include performing some acts simultaneously, even though shown as sequential acts in illustrative embodiments.
  • features may not necessarily be limited to a particular order of execution, but rather, any number of threads, processes, services, servers, and/or the like that may execute serially, asynchronously, concurrently, in parallel, simultaneously, synchronously, and/or the like in a manner consistent with the disclosure. As such, some of these features may be mutually contradictory, in that they cannot be simultaneously present in a single embodiment. Similarly, some features are applicable to one aspect of the innovations, and inapplicable to others.
  • the disclosure may include other innovations not presently described. Applicant reserves all rights in such innovations, including the right to embodiment such innovations, file additional applications, continuations, continuations-in-part, divisionals, and/or the like thereof. As such, it should be understood that advantages, embodiments, examples, functional, features, logical, operational, organizational, structural, topological, and/or other aspects of the disclosure are not to be considered limitations on the disclosure as defined by the embodiments or limitations on equivalents to the embodiments.
  • a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

Landscapes

  • Health & Medical Sciences (AREA)
  • Ophthalmology & Optometry (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)

Abstract

Des modes de réalisation de la présente invention concernent des systèmes et des procédés d'injection rétininienne. Selon certains aspects, un procédé comprend l'insertion d'une partie d'extrémité distale d'un conduit creux d'un injecteur médical dans un oeil d'un patient jusqu'à ce que la partie d'extrémité distale atteigne un emplacement de référence, l'injecteur médical ayant un boîtier à partir duquel le conduit creux s'étend distalement et confirme le placement de la partie d'extrémité distale du conduit creux dans l'emplacement de référence. Après confirmation, le procédé comprend en outre le prolongement distal de la partie d'extrémité distale du conduit creux par rapport au boîtier et depuis l'emplacement de référence de sorte que la partie d'extrémité distale pénètre dans une région d'injection de l'oeil. Lorsque la partie d'extrémité distale du conduit creux est placée dans la région d'injection de l'oeil, le médicament est transporté dans la région d'injection à travers le conduit creux.
EP21878692.9A 2020-10-09 2021-10-11 Appareil et procédés d'injection rétinienne Pending EP4225235A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202063089829P 2020-10-09 2020-10-09
US202063115654P 2020-11-19 2020-11-19
PCT/US2021/054395 WO2022076938A1 (fr) 2020-10-09 2021-10-11 Appareil et procédés d'injection rétinienne

Publications (1)

Publication Number Publication Date
EP4225235A1 true EP4225235A1 (fr) 2023-08-16

Family

ID=81126176

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21878692.9A Pending EP4225235A1 (fr) 2020-10-09 2021-10-11 Appareil et procédés d'injection rétinienne

Country Status (2)

Country Link
EP (1) EP4225235A1 (fr)
WO (1) WO2022076938A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070202186A1 (en) 2006-02-22 2007-08-30 Iscience Interventional Corporation Apparatus and formulations for suprachoroidal drug delivery
WO2024015652A1 (fr) * 2022-07-15 2024-01-18 Clearside Biomedical, Inc. Systèmes, dispositifs et procédés pour injections oculaires sans aiguille

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8343106B2 (en) * 2009-12-23 2013-01-01 Alcon Research, Ltd. Ophthalmic valved trocar vent
BR112015027762A2 (pt) * 2013-05-03 2017-08-29 Clearside Biomedical Inc Aparelho e métodos para injeção ocular
CN112998944B (zh) * 2015-06-03 2023-08-22 阿奎西斯公司 外路眼内分流器放置

Also Published As

Publication number Publication date
WO2022076938A1 (fr) 2022-04-14

Similar Documents

Publication Publication Date Title
US10722396B2 (en) Apparatus and methods for ocular injection
US9956114B2 (en) Variable diameter cannula and methods for controlling insertion depth for medicament delivery
US10390901B2 (en) Ocular injection kit, packaging, and methods of use
US10188550B2 (en) Apparatus and methods for drug delivery using multiple reservoirs
IL305537A (en) Devices and methods for adjusting the insertion depth of a drug administration needle
EP4225235A1 (fr) Appareil et procédés d'injection rétinienne
EP3554435A1 (fr) Systèmes et procédés d'administration de médicaments à un tissu rétinien
US20230363941A1 (en) Apparatus and methods of retinal injection
US20220280386A1 (en) Methods and apparatus for extracting fluid from container
WO2024015652A1 (fr) Systèmes, dispositifs et procédés pour injections oculaires sans aiguille

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230504

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)