INJECTABLE SILK FIBROIN PARTICLES AND USES THEREOF

The inventions provided herein relate to compositions, methods, delivery devices and kits for repairing or augmenting a tissue in a subject. The compositions described herein are injectable such that they can be placed in a tissue to be treated with a minimally-invasive procedure (e.g., by injection) and/or be molded flexibly into a tissue void of any shape and/or size. In some embodiments, the composition described herein comprises a plurality of silk fibroin particles, which can retain their original volume within the tissue for a period of time. The compositions can be used as a filler to replace a tissue void, e.g., for tissue repair and/or augmentation, or as a scaffold to support tissue regeneration and/or reconstruction. In some embodiments, the compositions described herein can be used for soft tissue repair or augmentation.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C § 119(e) of U.S. Provisional Application No. 61/557,603 filed Nov. 9, 2011, the content of which is incorporated herein by reference in its entirety.

GOVERNMENT SUPPORT

This invention was made with government support under Grant No. EB002520 awarded by the National Institutes of Health and W81XWH-08-2-0032 awarded by the US Army. The government has certain rights in the invention.

TECHNICAL FIELD OF DISCLOSURE

The inventions provided herein generally relate to silk fibroin-based materials for biomedical applications, e.g., in soft tissue repair, augmentation and/or reconstruction.

BACKGROUND

The restoration of soft tissue defects from trauma, surgical excision or congenital defects should start with a strategy that will maintain tissue size and shape to near normal dimensions for extended time frames. Current clinical strategies include free fat transfers and artificial fillers. In the case of breast cancer patients receiving mastectomies, silicone shells filled with saline or silicone are used to replace the void. This leaves the patient with an unnatural look and feel, and the risk of capsular contracture resulting in a revision surgery. The fat grafting and artificial filler options fail to retain volume over time. Thus, the fat grafting and artificial filler options can require a second surgical site, have avascular necrosis and generally do not regenerate the original tissue.

Bovine and human collagen have gained widespread use as injectable materials for soft tissue augmentation and filling. Collagen, the principal extracellular structural protein of the animal body, has been used as an implant material to replace or augment connective tissue, such as skin, tendon, cartilage and bone. Additionally, collagen has been injected or implanted into the human body for cosmetic purposes for a number of years. However, the use of collagen in soft tissue augmentation and/or filling could be costly and it does not have a long lasting effect, e.g., the results often only last for about 3 months.

Hyaluronic acid (HA) is a glycosaminoglycan that is naturally found in the human body and is widely distributed throughout connective, epithelial, and neural tissues. Compositions of non-crosslinked hyaluronic acid tend to degrade within a few months after injection and thus require fairly frequent reinjection to maintain their soft tissue augmenting effect. More recently, compositions of cross-linked hyaluronic acid have been used for soft tissue augmentation. However, such cross-linked compositions contain fairly large particles, around approximately 2 mm each, of hyaluronic acid suspended in a gel. While the larger particles could have a longer lasting effect, the larger particle size can make the injection more challenging and create an unpleasant experience to a recipient.

In summary, the major disadvantages of the current strategies for soft tissue regeneration, repair and/or augmentation include a large amount of tissues required for grafting large tissue defects; donor site morbidity, possibility of second surgical site, avascular necrosis; loss of shape and/or size of the scaffolds over time; material mismatch with native tissue; and failure to regenerate tissue. Accordingly, there is a strong need to develop a strategy or a scaffold that can be administered with a minimally invasive procedure and will provide sustained retention of volume restoration for at least 3 months or longer, e.g., for at least 6 months or at least one year, while the body gradually remodels and regenerates the site into near-normal tissue structure and function.

SUMMARY

Embodiments of various aspects described herein are based on, at least in part, engineering silk fibroin scaffolds in an injectable format, e.g., silk fibroin particles, which can retain at least a portion of the original volume within a tissue to be repaired or augmented for a period of time. For example, such silk fibroin particles can be placed with a minimally invasive procedure (e.g., injection) into a subject's tissue to be repaired or augmented as a filler to replace a void, e.g., for tissue augmentation or repair, or as a scaffold, e.g., for tissue regeneration or reconstruction.

Accordingly, one aspect provided herein is an injectable composition for use in repairing or augmenting a tissue in a subject, comprising a plurality of silk fibroin particles, wherein at least a portion of the silk fibroin particles retain at least a portion (e.g., at least about 50%) of their original volume within the tissue to be repaired or augmented for a period of time (e.g., at least about 6 weeks).

Another aspect provided herein relates to a method for repairing or augmenting a tissue in a subject. The method includes placing in the tissue to be repaired or augmented a composition comprising a plurality of silk fibroin particles, wherein at least a portion of the silk fibroin particles retain at least a portion of its original volume (e.g., at least about 50% or more) within the tissue for a period of time (e.g., at least about 6 weeks or longer). In one embodiment, the composition is placed into the tissue to be repaired or augmented by injection.

In certain embodiments of the compositions and methods provided herein, the silk fibroin particles can exclude an amphiphilic peptide. In other embodiments, the silk fibroin particles can include an amphiphilic peptide. An exemplary amphiphilic peptide, for example, can comprise a RGD motif.

In some embodiments of the compositions and methods provided herein, at least a portion of the silk fibroin particles can retain at least about 50% of their original volume, including at least about 60%, at least about 70%, at least about 80% or more, of their original volume within the tissue for a period of time.

In some embodiments of the composition and method provided herein, at least a portion of the silk fibroin particles can retain at least a portion of its original volume for at least about 6 weeks, at least about 3 months, at least about 6 months or longer.

Volume retention of the silk fibroin particles can be, in part, controlled by modulating the degradation and/or solubility properties of the silk fibroin particles. In such embodiments, at least a portion of the silk fibroin particles can be adapted to degrade no more than 50% of their original volume, for example, including no more than 30%, no more than 10%, of their original volume, in at least about 6 weeks, including at least about 3 months, 6 months or longer.

Depending on the defect size of the tissue and/or desired properties of the silk fibroin particles, the silk fibroin particles can be adapted to be any size. In some embodiments, the silk fibroin particles can have a size suitable for injection into a tissue. For example, the silk fibroin particles provided herein can have a size of about 500 nm to about 5000 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 2000 μm. In some embodiments, the silk fibroin particles can have a size of about 10 μm to about 1500 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 1000 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 500 μm. In some embodiments, the silk fibroin particles can have a size of about 3 μm to about 425 μm. In some embodiments, the silk fibroin particles can have a size of about 500 μm to about 1200 μm. In some embodiments, the silk fibroin particles can have a size of about 800 μm to about 1000 μm.

The silk fibroin particles can be adapted to mimic the structural morphology of native tissues and/or to deliver an active agent to a local area of a tissue. For example, the silk fibroin particles can be porous. In some embodiments, the porosity of the silk fibroin particles can be adapted to mimic the structural morphology and/or gradient of cellular densities found in native tissue. In some embodiments, the porosity of the silk fibroin particles can be adapted to deliver an active agent to a tissue in a pre-determined release profile. In some embodiments, the porosity of the silk fibroin particles can be adapted to retain at least a portion of their original volume for a period of time. For example, the silk fibroin porous particles can have a porosity of at least about 1%, e.g., including at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 50%, at least about 70%, at least about 80%, at least about 90% or higher. The pore size of such porous silk fibroin particles can range from about 10 nm to about 2000 μm, from about 50 nm to about 1500 μm, from about 0.5 μm to about 1500 μm, from about 1 μm to about 1000 μm, or from about 1 μm to about 500 μm. In some embodiments, the pore size of the porous silk fibroin particles can range from about 3 μm to about 500 μm. In some embodiments, the pore size of the porous silk fibroin particles can range from about 8 μm to about 1000 μm. In some embodiments, the silk fibroin particles need not be porous.

The silk fibroin particles, in one embodiment, can be fabricated by reducing a solid-state silk fibroin into particles. For example, a solid-state silk fibroin can be reduced into particles by a mechanical means, for example, but not limited to, micronizing, milling, pulverizing, crushing, grinding, cutting, and any combinations thereof. A solid-state silk fibroin can be made by any methods known in the art. To produce a porous silk fibroin structure, porogen leaching method or any other art-recognized can be used. In some embodiments, the silk fibroin particles can have no optical properties, for example, but not limited to, diffraction. In such embodiments, the silk fibroin particles can be produced from a solid-state silk fibroin without any optical elements imprinted on or added thereto.

The injectable composition described herein comprising the silk fibroin particles can further comprise at least one active agent. In some embodiments, the silk fibroin particles of the composition described herein can further comprise at least one active agent. Non-limiting examples of the active agents can include biologically active agents, cosmetically active agents, cell attachment agents, a dermal filler material, and any combinations thereof. In some embodiments, the active agent can be a therapeutic agent. In some embodiments, the active agent can be a cosmetically active agent. In some embodiments, the active agent can be a dermal filler material.

In some embodiments, the injectable composition or the composition comprising a plurality of silk fibroin particles can further comprise at least one cell, e.g., a stem cell. In some embodiments, the cell can be obtained from a biological fluid or concentrate, such as lipoaspirate, bone marrow aspirate or any combinations thereof.

Accordingly, in some embodiments, the injectable composition or the composition comprising a plurality of silk fibroin particles can further comprise a biological fluid or concentrate, such as lipoaspirate, bone marrow aspirate or any combinations thereof. In one embodiment, the injectable composition or the composition comprising a plurality of silk fibroin particles can further comprise a lipoaspirate. In these embodiments, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 1:38 to about 12:19, or about 1:19 to about 10:19, or about 2:19 to about 8:19. In one embodiment, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 3:19. In another embodiment, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 6:19.

In some embodiments, the composition described herein can further comprise a hydrogel, e.g., in a form of separate hydrogel particles, and/or distributed within silk fibroin particles.

Various embodiments of the composition described herein can be injected into a tissue to be repaired or augmented by any known methods in the art, e.g., subcutaneously, submuscularly, or intramuscularly. When injected in a tissue, some embodiments of the composition can be at least partially dry. Alternatively, the composition can be at least partially hydrated, e.g., the composition can further comprise a pharmaceutically-acceptable carrier, e.g., a buffered solution, when injected in a tissue. In some embodiments, the silk fibroin particles in the composition are small enough such that the silk fibroin particles can be delivered through a needle or a catheter without any prior compression before being introduced into a tissue to be repaired or augment.

The tissue to be repaired or augmented by the composition and/or the method described herein can be a soft tissue. Exemplary examples of a soft tissue include, but are not limited to, a tendon, a ligament, skin, a breast tissue, a fibrous tissue, a connective tissue, a muscle, and any combinations thereof. In certain embodiments, the soft tissue is skin. In other embodiments, the soft tissue is a breast tissue.

A delivery device comprising one embodiment of an injectable composition and/or silk fibroin particles is also provided herein. A delivery device can include any conventional injection device (e.g., a syringe) and/or any administration device that is minimally invasive. Accordingly, in some embodiments, provided herein relate to syringes comprising one embodiment of an injectable composition. The syringe can further comprise a needle, a cannula, and/or a catheter. In some embodiments, the delivery device (e.g., a syringe) can further comprise an injection carrier, e.g., a buffered solution. In some embodiments, the delivery device (e.g., a syringe) can further comprise a local anesthetic.

In some embodiments of any aspects described herein, the compositions and/or delivery devices can be stored or transported at a temperature about 0° C. and about 60° C., e.g., between about 10° C. and about 60° C. or between about 15° C. and about 60° C. At such temperatures, the bioactivity of active agents embedded or distributed inside the silk fibroin particles can be stabilized for a period of time.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows an exemplary method of using one or more embodiments of the injectable compositions described herein. The porous silk fibroin scaffold bits (e.g., formed by reducing a solid-state porous silk fibroin into particles or bits) can be mixed with lipoaspirate as a carrier, optionally containing adipose-derived stem cells (ASCs), to form an exemplary injectable composition. The injectable compositions can then be injected into a subject, e.g., an animal model.

FIG. 2 shows images of one or more embodiments of the injectable compositions described herein. The silk fibroin scaffold particles can be of any size, for example, from submicrons to about 2 mm. The left panel shows that the silk fibroin scaffold particles can have a size of about 3 μm to about 425 μm, while the right panel shows that the silk fibroin scaffold particles can have a size of about 0.8 mm to about 1 mm.

FIG. 3 shows a set of hematoxylin and eosin images of injectable silk fibroin porous particles mixed with various ratios of lipoaspirate at the 6-week post-injection. The silk fibroin porous particles (with a pore size of about 3 μm to about 500 μm) as shown in the first row of the images were produced by micronizing a porous silk fibroin scaffold with a pore size of about 300 microns to about 500 microns, while the ones shown in the second and third rows of the images (silk fibroin particles with a pore size of about 8 μm to about 1000 μm) were produced from a silk fibroin scaffold with a pore size of about 850 microns to about 1000 microns. The term “dose” as used in FIG. 3 refers to the amount of the silk fibroin particles injected in a subject, e.g., an animal model. In some embodiments, the dose can be indicated by the final volume ratio of silk fibroin particles to lipoaspirate. For example, the final volume ratios can range from about 3:19 to about 6:19.

DETAILED DESCRIPTION OF THE INVENTION

Described herein are methods, compositions, delivery devices, and kits for repairing or augmenting a tissue in a subject. In accordance with embodiments of various aspects described herein, an injectable format of silk fibroin scaffolds (e.g., silk fibroin particles) can be placed (e.g., by injection) into a tissue to be repaired or augmented and retain at least a portion of their original volume (e.g., at least about 50% of their original volume) within the tissue to be repaired or augmented for a period of time (e.g., at least about 6 weeks). Such injectable silk fibroin particles can be introduced into a defect site with a minimally-invasive procedure, while enabling a skilled practitioner to flexibly mold the injectable silk fibroin particles to fit into a defect of any shape and/or size.

Silk Fibroin Particles

Silk fibroin particles described herein can retain their original volume upon administration to a tissue (e.g., by injection) to be repaired or augmented for a period of time.

By “original volume” in reference to the silk fibroin particles described herein is generally meant the volume of silk fibroin particles as measured immediately before the silk fibroin particles are placed into a tissue to be repaired or augmented, or the corresponding increase in tissue volume as measured immediately after the silk fibroin particles are placed in a tissue to be repaired or augmented. For example, the original volume of silk fibroin particles can be measured, for example, within about 20 minutes, before or after the silk fibroin particles are placed into a tissue to be repaired or augmented. In some instances, the original volume of the silk fibroin particles can be measured, for example, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes, before or after the silk fibroin particles are placed into a tissue to be repaired or augmented. In some embodiments, the volume of the silk fibroin particles prior to placing into a tissue can refer to the volume of the silk fibroin particles in a dried state. In alternative embodiments, the volume of the silk fibroin particles prior to placing into a tissue can refer to the volume of silk fibroin particles in a hydrated state. In other embodiments, the volume of the silk fibroin particles prior to placing in a tissue can refer to the volume of silk fibroin particles suspended in a fluid or a carrier. In some embodiments, the volume of the silk fibroin particles prior to placing in a tissue can refer to the injection volume of the mixture comprising the silk fibroin particles.

As used herein, the term “retain” refers to maintaining the volume (e.g., size and/or shape) of at least a portion of the silk fibroin particles described herein over a period of time. In some embodiments, at least a portion of the silk fibroin particles can retain over a period of time at least about 20% of their original volume, including, for example, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% of their original volume or higher. In some embodiments, at least a portion of the silk fibroin particles can retain over a period of time at least about 1%, at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20% of their original volume or higher. In some embodiments, at least a portion of the silk fibroin particles can retain 100% of their original volume, e.g., no detectable changes in the volume, within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can retain at least about 1% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can retain at least about 50% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can retain at least about 60% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can retain at least about 70% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can retain at least about 80% of their original volume within the tissue to be repaired or augmented for a period of time. The volume of the silk fibroin particles placed into a tissue can be determined or indicated by a change in at least one of the tissue properties, e.g., tissue volume, tissue elasticity, and/or tissue hardness. In some embodiments, the volume of the silk fibroin particles placed into a tissue can be determined from explants, e.g., weight measurements and/or volume displacement. In one embodiment, the volume of the silk fibroin particles placed into a tissue can be monitored and/or measured by imaging.

The silk fibroin particles can retain at least a portion of their original volume for any period of time, e.g., weeks, months, or years. In some embodiments, the silk fibroin particles can retain, e.g., at least about 50% of their original volume (including e.g., at least about 60%, at least about 70%, at least about 80%, or higher, of their original volume) for at least about 6 weeks, at least about 7 weeks, at least about 8 weeks, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about 1 year, at least about 2 years, at least 3 years, at least about 4 years, at least 5 years or longer. In certain embodiments, the silk fibroin particles can retain, e.g., at least about 70% of their original volume or higher, for at least about 3 months or longer. In other embodiments, there can be no significant changes in the volume of the silk fibroin particles after placed into a tissue to be repaired or augmented for at least about 3 months or longer. In some embodiments, the silk fibroin particles can retain, e.g., at least about 70% of their original volume or higher, for at least about 6 months or longer (including, e.g., at least about 9 months, at least about 12 months, at least about 18 months or longer). In other embodiments, there can be no significant changes in the volume of the silk fibroin particles after placed into a tissue to be repaired or augmented for at least about 6 months or longer. In particular embodiments, the silk fibroin particles can retain at least about 20% of their original volume or higher for at least about 1 year or longer (including, e.g., at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years or longer). In some embodiments, the silk fibroin particles can retain at least about 50% of their original volume or higher for at least about 1 year or longer (including, e.g., at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years or longer). In one embodiment, at least a portion of the silk fibroin particles can retain at least about 1%, at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 40% of their original volume or more, within the tissue to be repaired or augmented for at least about 6 weeks (including, e.g., at least about 3 months, at least about 6 months, or longer).

The volume retention of the silk fibroin particles can also be characterized by, e.g., degradation of the silk fibroin particles. Generally, the slower the silk fibroin particles degrade, the longer the silk fibroin particles can retain their original volume in a tissue. Accordingly, some embodiments provided herein are directed to injectable compositions for use in repairing or augmenting a tissue in a subject, the compositions comprising a plurality of silk fibroin particles, wherein at least a portion of the silk fibroin particles are adapted to degrade within the tissue to be repaired or augmented over a period of time.

As used in reference to the silk fibroin particles described herein, the term “degrade” or “degradation” refers to a decrease in volume or size of the silk fibroin particles. The degradation of the silk fibroin particles can occur via cleavage of the silk fibroin particles into smaller fragments and/or dissolution of the silk fibroin particles or fragments thereof. In some embodiments, at least a portion of the silk fibroin particles can be adapted to degrade no more than 80% of their original volume, including, for example, no more than 70%, no more than 60%, no more than 50%, no more than 40%, no more than 30%, no more than 20%, no more than 10% of their original volume or lower. In some embodiments, at least a portion of the silk fibroin particles can exhibit no significant degradation (e.g., no detectable changes in the volume) within the tissue to be repaired or augmented. In one embodiment, at least a portion of the silk fibroin particles can be adapted to degrade no more than 50% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can be adapted to degrade no more than 40% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can be adapted to degrade no more than 30% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can be adapted to degrade no more than 20% of their original volume within the tissue to be repaired or augmented for a period of time. In one embodiment, at least a portion of the silk fibroin particles can be adapted to degrade no more than 10% of their original volume within the tissue to be repaired or augmented for a period of time.

The silk fibroin particles can be adapted to degrade at any rate. In some embodiments, the silk fibroin particles can be adapted to degrade at least a portion of their original volume over any period of time, e.g., weeks, months, or years. In some embodiments, the silk fibroin particles can be adapted to degrade, e.g., no more than 50% of their original volume (including e.g., no more than 40%, no more than 30%, no more than 20% or lower, of their original volume) in at least about 6 weeks, at least about 7 weeks, at least about 8 weeks, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about 1 year, at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years or longer. In certain embodiments, the silk fibroin particles can be adapted to degrade, e.g., no more than 30% of their original volume or lower, in at least about 3 months or longer. In other embodiments, there can be no significant degradation (i.e., no detectable changes in the volume of the silk fibroin particles) after placed into a tissue to be repaired or augmented for at least about 3 months or longer. In some embodiments, the silk fibroin particles can be adapted to degrade, e.g., no more than 30% of their original volume or lower, in at least about 6 months or longer (including, e.g., at least about 9 months, at least about 12 months, at least about 18 months or longer). In other embodiments, there can be no significant degradation (i.e., no detectable changes in the volume of the silk fibroin particles) after placed into a tissue to be repaired or augmented for at least about 6 months or longer. In particular embodiments, the silk fibroin particles can be adapted to degrade no more than 80% of their original volume or lower in at least about 1 year or longer (including, for example, at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years or longer). In some embodiments, the silk fibroin particles can be adapted to degrade no more than 50% of their original volume or lower in at least about 1 year or longer.

The same or similar formulation of the silk fibroin particles or injectable compositions can manifest different responses in a subject. By way of example only, the volume retention or degradation rate of the silk fibroin particles in a tissue can vary from one subject to another, e.g., because of different tissue microenvironment such as species and/or levels of various proteins or enzymes (e.g., proteolytic enzymes) present in the tissue.

In some embodiments, the silk fibroin particles can be adapted to maintain a constant volume retention rate and/or degradation rate over a period of time. In some embodiments, the silk fibroin particles can be adapted to have a volume retention rate or degradation rate varying with time. For example, the silk fibroin particles can be coated with a polymeric material, e.g., silk fibroin of a different concentration and/or a different biodegradable and biocompatible polymer. Such coating can possess a different function and/or a different degradation rate from that of the silk fibroin particle core. By way of example only, the coating of the silk fibroin particle can contain at least one active agent and be adapted to degrade at a different rate (e.g., at a faster rate) from that of the silk fibroin particle core. Thus, upon placing the silk fibroin particles in a tissue, the coating of the silk fibroin particles can be adapted to degrade faster, e.g., to release the active agent for relieving the pain and/or promoting the wound healing, while the core of the silk fibroin particles can retain their volume for a longer period of time.

Silk fibroin is a particularly appealing biopolymer candidate to be used for various embodiments described herein, e.g., because of its versatile processing e.g., all-aqueous processing (Sofia et al., 54 J. Biomed. Mater. Res. 139 (2001); Perry et al., 20 Adv. Mater. 3070-72 (2008)), relatively easy functionalization (Murphy et al., 29 Biomat. 2829-38 (2008)), and biocompatibility (Santin et al., 46 J. Biomed. Mater. Res. 382-9 (1999)). For example, silk has been approved by U.S. Food and Drug Administration as a tissue engineering scaffold in human implants. See Altman et al., 24 Biomaterials: 401 (2003).

As used herein, the term “silk fibroin” includes silkworm fibroin and insect or spider silk protein. See e.g., Lucas et al., 13 Adv. Protein Chem. 107 (1958). Any type of silk fibroin can be used in different embodiments described herein. Silk fibroin produced by silkworms, such as Bombyx mori, is the most common and represents an earth-friendly, renewable resource. For instance, silk fibroin used in a silk film may be attained by extracting sericin from the cocoons of B. mori. Organic silkworm cocoons are also commercially available. There are many different silks, however, including spider silk (e.g., obtained from Nephila clavipes), transgenic silks, genetically engineered silks, such as silks from bacteria, yeast, mammalian cells, transgenic animals, or transgenic plants (see, e.g., WO 97/08315; U.S. Pat. No. 5,245,012), and variants thereof, that can be used.

In various embodiments, the silk fibroin can be modified for different applications and/or desired mechanical or chemical properties (e.g., to facilitate formation of a gradient of active agent in silk fibroin particles). One of skill in the art can select appropriate methods to modify silk fibroins, e.g., depending on the side groups of the silk fibroins, desired reactivity of the silk fibroin and/or desired charge density on the silk fibroin. In one embodiment, modification of silk fibroin can use the amino acid side chain chemistry, such as chemical modifications through covalent bonding, or modifications through charge-charge interaction. Exemplary chemical modification methods include, but are not limited to, carbodiimide coupling reaction (see, e.g. U.S. Patent Application. No. US 2007/0212730), diazonium coupling reaction (see, e.g., U.S. Patent Application No. US 2009/0232963), avidin-biotin interaction (see, e.g., International Application No.: WO 2011/011347) and pegylation with a chemically active or activated derivatives of the PEG polymer (see, e.g., International Application No. WO 2010/057142). Silk fibroin can also be modified through gene modification to alter functionalities of the silk protein (see, e.g., International Application No. WO 2011/006133). For instance, the silk fibroin can be genetically modified, which can provide for further modification of the silk such as the inclusion of a fusion polypeptide comprising a fibrous protein domain and a mineralization domain, which can be used to form an organic-inorganic composite. See WO 2006/076711. Additionally, the silk fibroin matrix can be combined with a chemical, such as glycerol, that, e.g., affects flexibility of the matrix. See, e.g., WO 2010/042798, Modified Silk films Containing Glycerol.

As used herein, the phrase “silk fibroin particles” generally refer to particles comprising silk fibroin. In some embodiments, the phrase “silk fibroin particles” refers to particles in which silk fibroin constitutes at least about 30% of the total composition, including at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95% or higher, of the total composition. In certain embodiments, the silk fibroin particles can be substantially formed from silk fibroin. In various embodiments, the silk fibroin particles can be substantially formed from silk fibroin comprising at least one active agent.

The silk fibroin particles described herein can be adapted to be any shape, e.g., a spherical shape, polygonal-shaped, elliptical-shaped. As used in reference to silk fibroin particles, the term “particle” as used herein refers to a particle of any shape, e.g., but not limited to, a spherical shape, a polygonal shape, or an elliptical shape. The particle size can vary with a number of factors including, without limitations, the size of the tissue to be repaired or augmented and/or desired properties of the silk fibroin particles, e.g., volume retention or degradation profile. In some embodiments, the particle size can range from about 500 nm to about 5000 μm, about 1 μm to about 2000 μm, about 10 μm to about 1500 μm, about 20 μm to about 1000 μm, about 50 μm to about 750 μm, or about 100 μm to about 500 μm. In certain embodiments, the silk fibroin particles provided herein can have a size of about 500 nm to about 5000 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 2000 μm. In some embodiments, the silk fibroin particles can have a size of about 10 μm to about 1500 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 1000 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 500 μm. In some embodiments, the silk fibroin particles can have a size of about 3 μm to about 425 μm. In some embodiments, the silk fibroin particles can have a size of about 500 μm to about 1200 μm. In some embodiments, the silk fibroin particles can have a size of about 800 μm to about 1200 μm. In some embodiments, the silk fibroin particles can have a size of about 1 μm to about 5 μm. In some embodiments, the silk fibroin particles can have a size of about 5 μm to about 20 μm. In some embodiments, the silk fibroin particles can have a size of about 20 μm to about 50 μm. In some embodiments, the silk fibroin particles can have a size of about 50 μm to about 100 μm. In some embodiments, the silk fibroin particles can have a size of about 100 μm to about 250 μm. In some embodiments, the silk fibroin particles can have a size of about 500 μm to about 750 μm. In some embodiments, the silk fibroin particles can have a size of about 750 μm to about 1000 μm. In some embodiments, the silk fibroin particles can have a size of about 1000 μm to about 1200 μm. In some embodiments, the silk fibroin particles can have a size less than 1 μm. In some embodiments, the silk fibroin particles can be inherently so small than they can be delivered through a needle and/or a catheter without any prior compression. In such embodiments, the silk fibroin particles can have a size smaller than the inner diameter of a needle and/or a catheter so that the silk fibroin particles need no prior compression before injection into a tissue through a needle and/or a catheter.

In some embodiments, the silk fibroin particles can exhibit a distribution of particle sizes around the indicated “size.” In such embodiments, the term “particle size” as used herein refers to the mode of a size distribution of silk fibroin particles, i.e., the value that occurs most frequently in the size distribution. Methods for measuring the particle size are known to a skilled artisan, e.g., by dynamic light scattering (such as photocorrelation spectroscopy, laser diffraction, low-angle laser light scattering (LALLS), and medium-angle laser light scattering (MALLS)), light obscuration methods (such as Coulter analysis method), or other techniques (such as rheology, and light or electron microscopy).

The silk fibroin particles can be produced from aqueous-based or organic solvent-based silk fibroin solutions. In some embodiments, the silk fibroin particles produced from organic solvent-based silk fibroin solution can retain their original volume for a longer period of time than the aqueous-based silk fibroin particles. The aqueous- or organic solvent-based silk fibroin solution used for making silk fibroin particles described herein can be prepared using any techniques known in the art. The concentration of silk fibroin in solutions used for soft tissue repair or augmentation can be suited to the particular volume retention requirement, e.g., if higher concentrations of silk fibroin solutions can be used when longer volume retention of the silk fibroin particles is desired when injected into the tissue to be repaired or augmented. In some embodiments, the silk fibroin solution for making the silk fibroin particles described herein can vary from about 4% (w/v) to about 30% (w/v), inclusive, or about 4% (w/v) to about 20% (w/v), inclusive. In some embodiments, the silk fibroin solution can vary from about 6% (w/v) to about 20% (w/v). In some embodiments, the silk fibroin solution can vary from about 6% (w/v) to about 17% (w/v). Suitable processes for preparing silk fibroin solution are disclosed, for example, in U.S. Pat. No. 7,635,755; and International Application Nos: WO/2005/012606; and WO/2008/127401. A micro-filtration step can be used herein. For example, the prepared silk fibroin solution can be processed further, e.g., by centrifugation and/or syringe based micro-filtration before further processing into silk fibroin particles described herein.

In some embodiments, the silk fibroin can be also mixed with other biocompatible and/or biodegradable polymers to form mixed polymer particles comprising silk fibroin. One or more biocompatible and/or biodegradable polymers (e.g., two or more biocompatible polymers) can be added to the silk fibroin solution. The biocompatible polymer that can be used herein include, but are not limited to, polyethylene oxide (PEO), polyethylene glycol (PEG), collagen, fibronectin, keratin, polyaspartic acid, polylysine, alginate, chitosan, chitin, hyaluronic acid, pectin, polycaprolactone, polylactic acid, polyglycolic acid, polyhydroxyalkanoates, dextrans, polyanhydrides, polymer, PLA-PGA, polyanhydride, polyorthoester, polycaprolactone, polyfumarate, collagen, chitosan, alginate, hyaluronic acid and other biocompatible and/or biodegradable polymers. See, e.g., International Application Nos.: WO 04/062697; WO 05/012606.

In some embodiments, at least one active agent described herein can be added to the silk fibroin solution before further processing into silk fibroin particles described herein. In some embodiments, the active agent can be dispersed homogeneously or heterogeneously within the silk fibroin, dispersed in a gradient, e.g., using the carbodiimide-mediated modification method described in the U.S. Patent Application No. US 2007/0212730.

In some embodiments, the silk fibroin particles can be first formed and then contacted with (e.g., dipped into) at least one active agent such that the open surface of the particles can be coated with at least one active agent.

In some embodiments, the silk fibroin particles described herein can be reduced from a solid-state silk fibroin by a mechanical means. Exemplary mechanical means to obtain silk fibroin particles include micronizing, milling, pulverizing, crushing, grinding, freeze-drying or any combination thereof. Methods of forming a solid-state silk fibroin from a silk fibroin solution are well known to a skilled artisan, e.g., using a solvent-based or an aqueous-based silk fibroin solution. See, e.g., Wang Y. et al. (2008) 29 Biomaterials 3415, U.S. Pat. No. 7,635,755; and International Application Nos: WO/2005/012606; and WO/2008/127401.

In some embodiments, the silk fibroin particles described herein can comprise porous structures, e.g., to mimic the structural morphology of a native tissue, to modulate the degradation rate/volume retention rate of the silk fibroin particles, and/or to module release profile of an active agent embedded therein, if any. As used herein, the terms “porous” and “porosity” are generally used to describe a structure having a connected network of pores or void spaces (which can, for example, be openings, interstitial spaces or other channels) throughout its volume. The term “porosity” is a measure of void spaces in a material, and is a fraction of volume of voids over the total volume, as a percentage between 0 and 100% (or between 0 and 1).

In some embodiments, the porous silk fibroin particles can be configured to have any porosity, depending on the desired properties. For example, in some embodiments, the porous silk fibroin particles can have a porosity of at least about 1%, at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or higher. In some embodiments, the porosity can range from about 50% to about 99%, about 70% to about 99%, or from about 80% to about 98%. The pore size and total porosity values can be quantified using conventional methods and models known to those of skill in the art. For example, the pore size and porosity can be measured by standardized techniques, such as mercury porosimetry and nitrogen adsorption. One of ordinary skill in the art can determine the optimal porosity of the silk fibroin particles used for various purposes. For example, the porosity and/or pore size of the silk fibroin particles can be optimized based on the desired degradation rate or volume retention rate of the silk fibroin particles, release profiles of an active agent from the silk fibroin particles, and/or the structural morphology of the tissue to be repaired or augmented.

The pores can be adapted to have any shape, e.g., circular, elliptical, or polygonal. The porous silk fibroin particles can be adapted to have a pore size of about 10 nm to about 2000 μm, from about 50 nm to about 1500 μm, from about 0.5 μm to about 1500 μm, from about 1 μm to about 1500 μm, about 2 μm to about 1500 μm, from about 1 μm to about 1000 μm, from about 3 μm to 1000 μm, from about 1 μm to about 500 μm, or from about 3 μm to about 500 μm. In some embodiments, the pore size of the porous silk fibroin particles can range from about 3 μm to about 500 μm. In some embodiments, the pore size of the porous silk fibroin particles can range from about 8 μm to about 1000 μm. In some embodiments, the silk fibroin particles need not be porous. In such embodiments, the pore size of the silk fibroin particles can be less than 10 nm or non-detectable. The term “pore size” as used herein refers to a dimension of a pore. In some embodiments, the pore size can refer to the longest dimension of a pore, e.g., a diameter of a pore having a circular cross section, or the length of the longest cross-sectional chord that can be constructed across a pore having a non-circular cross-section. In other embodiments, the pore size can refer the shortest dimension of a pore.

Methods for generating porous structures within silk fibroin matrix, e.g., freeze-drying, salt-leaching, and gas foaming methods, are well known in the art and have been described in, e.g., U.S. Pat. No. 7,842,780; and US patent application Nos: US 2010/0279112; and US 2010/0279112, the contents of which are incorporated herein by reference in their entirety.

In some embodiments, porous silk fibroin particles can be produced by a porogen-leaching method (e.g., salt-leaching method). See, e.g., U.S. Pat. No. 7,842,780; and US 2010/0279112. In some embodiments, the porous silk fibroin particles can be reduced from a solid-state porous silk fibroin by a mechanical means as discussed earlier. By way of example only, the silk fibroin solution can be placed into a non-stick container (e.g., a Teflon-coated container) containing water-soluble particles, or porogens that are insoluble in organic solvents. Alternatively, the porogens can be mixed with the silk fibroin solution prior to placement in the container. The diameter of the particles (porogens) can vary in accordance with the pre-determined pore size. Examples of water-soluble porogens can be used herein include, NaCl, alkali metals, alkali earth metal halides, phosphates, and sulfates, sugar crystals, water-soluble microspheres, polysaccharides and protein microspheres. The dried silk fibroin matrix can then be immersed in water or other solvent in which the particles, or porogens are soluble but silk fibroin is insoluble, to remove the particles (porogens), resulting in a porous solid-state silk fibroin described herein. The porous solid-state silk fibroin can then be reduced into the porous silk fibroin particles described herein, e.g., by a mechanical means such as micronizing, milling, pulverizing, crushing, grinding, freeze-drying or any combination thereof.

Using a porogen-leaching method (e.g., salt-leaching method), the silk fibroin particles can be created by, for example, but not limited to, micronizing a solid-state silk fibroin with pores corresponding to a porogen size ranging from, e.g., about 300 microns to about 500 microns and/or about 850 microns to about 1000 microns. In some embodiments, the pore size range need not be affected by a micronization process. Depending on how the solid-state silk fibroin is cut (or micronized), the resultant silk fibroin particles can have a pore size corresponding to any portion of the porogen size range. In some embodiments, the pores of the silk fibroin particles need not be intact pores (e.g., having a pore size to be a portion of the porogen size range, e.g., at least about 1%, at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 98% or higher, of the porogen size range). In some embodiments, the pores of the silk fibroin particles can be intact (e.g., having a pore size substantially same as the porogen size). In some embodiments, the intact pores of the silk fibroin particles can remain essentially the same as the size of porogens used, e.g., between about 300 microns and 500 microns, or between about 850 microns and about 1000 microns. In some embodiments, the smaller silk fibroin particles need not maintain intact pores, and thus the pores can be much smaller than the size of the porogens, for example, at least about 1%, at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 98% or higher, of the porogen size range. In some embodiments, the silk fibroin particles need not be porous. In various embodiments, the silk fibroin particles can comprise intact pores, partial pores, or a combination thereof.

In alternative embodiments, porous silk fibroin particles can be produced by freeze-drying method. See, e.g., U.S. Pat. No. 7,842,780 and US 2010/0279112. In such embodiments, the silk fibroin solution placed in a non-stick container can be frozen at sub-zero temperatures, e.g., from about −80° C. to about −20° C., for at least about 12 hours, at least about 24 hours, or longer, followed by lyophilization. In one embodiment, the silk fibroin solution can be frozen from one direction. In some embodiments, the silk fibroin solution can contain no salt. In some embodiments, alcohol such as 15%-25% of methanol or propanol can be added to the silk fibroin solution. The porous solid-state silk fibroin can then be reduced into the porous silk fibroin particles described herein, e.g., by a mechanical means such as micronizing, milling, pulverizing, crushing, grinding, freeze-drying or any combination thereof. In some embodiments, the porous solid-state silk fibroin is not produced by freeze-drying method as described herein.

In some embodiments, silk fibroin particles or a solid-state silk fibroin described herein can be subjected to a post-treatment that will affect at least one silk fibroin property. For example, post-treatment of silk fibroin particles or a solid-state silk fibroin can affect silk fibroin properties including β-sheet content, solubility, active agent loading capacity, degradation time, drug permeability, or any combinations thereof. Silk post-processing options include controlled slow drying (Lu et al., 10 Biomacromolecules 1032 (2009)), water annealing (Jin et al., Water-Stable Silk Films with Reduced β-Sheet Content, 15 Adv. Funct. Mats. 1241 (2005)), stretching (Demura & Asakura, Immobilization of glucose oxidase with Bombyx mori silk fibroin by only stretching treatment and its application to glucose sensor, 33 Biotech & Bioengin. 598 (1989)), compressing, and solvent immersion, including methanol (Hofmann et al., 2006), ethanol (Miyairi et al., 1978), glutaraldehyde (Acharya et al., 2008) and I-ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC) (Bayraktar et al., 2005).

In some embodiments, post-treatment of the solid-state silk fibroin or silk fibroin particles, e.g., water-annealing or solvent immersion, can allow controlling the release of an active agent from the silk fibroin particles. In some embodiments, post-treatment of the solid-state silk fibroin or silk fibroin particles, e.g., water-annealing or solvent immersion, can enable modulating the degradation or solubility properties of the silk fibroin particles described herein. In some embodiments, post-treatment of the solid-state silk fibroin or silk fibroin particles, e.g., water-annealing or solvent immersion, can enable modulating the volume retention properties of the silk fibroin particles described herein.

In some embodiments, the silk fibroin particles described herein can be coated with at least one layer of a biocompatible and/or biodegradable polymer described herein, e.g., to modulate the degradation and/or volume retention properties of the silk fibroin particles upon injection into a tissue to be treated and/or to modulate the rate of active agents released from the silk fibroin particles. In such embodiments, the biocompatible and/or biodegradable polymer can comprise at least one active agent.

In some embodiments, the silk fibroin particles described herein can be coated with cell adhesion molecules, e.g., but not limited to, fibronectin, vitronectin, laminin, collagen, any art-recognized extracellular matrix molecules, and any combinations thereof.

In some embodiments, the silk fibroin particles described herein can be sterilized. Sterilization methods for biomedical devices are well known in the art, including, but not limited to, gamma or ultraviolet radiation, autoclaving (e.g., heat/steam); alcohol sterilization (e.g., ethanol and methanol); and gas sterilization (e.g., ethylene oxide sterilization).

Further, the silk fibroin particles described herein can take advantage of the many techniques developed to functionalize silk fibroin (e.g., active agents such as dyes and sensors). See, e.g., U.S. Pat. No. 6,287,340, Bioengineered anterior cruciate ligament; WO 2004/000915, Silk Biomaterials & Methods of Use Thereof: WO 2004/001103, Silk Biomaterials & Methods of Use Thereof; WO 2004/062697, Silk Fibroin Materials & Use Thereof; WO 2005/000483, Method for Forming inorganic Coatings: WO 2005/012606, Concentrated Aqueous Silk Fibroin Solution & Use Thereof; WO 2011/005381, Vortex-Induced Silk fibroin Gelation for Encapsulation & Delivery; WO 2005/123114, Silk-Based Drug Delivery System; WO 2006/076711, Fibrous Protein Fusions & Uses Thereof in the Formation of Advanced Organic/Inorganic Composite Materials; U.S. Application Pub. No. 2007/0212730, Covalently immobilized protein gradients in three-dimensional porous scaffolds; WO 2006/042287, Method for Producing Biomaterial Scaffolds; WO 2007/016524, Method for Stepwise Deposition of Silk Fibroin Coatings; WO 2008/085904. Biodegradable Electronic Devices; WO 2008/118133, Silk Microspheres for Encapsulation & Controlled Release; WO 2008/108838, Microfluidic Devices & Methods for Fabricating Same WO 2008/127404, Nanopatterned Biopolymer Device & Method of Manufacturing Same; WO 2008/118211, Biopolymer Photonic Crystals & Method of Manufacturing Same; WO 2008/127402, Biopolymer Sensor & Method of Manufacturing Same; WO 2008/127403, Biopolymer Optofluidic Device & Method of Manufacturing the Same; WO 2008/127401, Biopolymer Optical Wave Guide & Method of Manufacturing Same; WO 2008/140562, Biopolymer Sensor & Method of Manufacturing Same; WO 2008/127405, Microfluidic Device with Cylindrical Microchannel & Method for Fabricating Same; WO 2008/106485, Tissue-Engineered Silk Organs; WO 2008/140562, Electroactive Biopolymer Optical & Electro-Optical Devices & Method of Manufacturing Same: WO 2008/150861, Method for Silk Fibroin Gelation Using Sonication; WO 2007/103442, Biocompatible Scaffolds & Adipose-Derived Stem Cells; WO 2009/155397, Edible Holographic Silk Products; WO 2009/100280,3-Dimensional Silk Hydroxyapatite Compositions; WO 2009/061823, Fabrication of Silk Fibroin Photonic Structures by Nanocontact Imprinting; WO 2009/126689, System & Method for Making Biomaterial Structures.

In an alternative embodiment, the silk fibroin particles can include plasmonic nanoparticles to form photothermal elements. This approach takes advantage of the superior doping characteristics of silk fibroin. Thermal therapy has been shown to aid in the transdermal delivery of various agents, see Park et al., Effect of Heat on Skin Permeability, 359 intl. J. Pharm. 94 (2008). In one embodiment, short bursts of heat on very limited areas can be used to maximize permeability with minimal harmful effects on surrounding tissues. Thus, plasmonic particle-doped silk fibroin particles can add specificity to thermal therapy by focusing light to locally generate heat only via the particles. In some embodiments, the silk fibroin particles can include photothermal agents such as gold nanoparticles.

In some embodiments, the silk fibroin particles can include an amphiphilic peptide. In other embodiments, the silk fibroin particles can exclude an amphiphilic peptide. “Amphiphilic peptides” possess both hydrophilic and hydrophobic properties. Amphiphilic molecules can generally interact with biological membranes by insertion of the hydrophobic part into the lipid membrane, while exposing the hydrophilic part to the aqueous environment. In some embodiment, the amphiphilic peptide can comprise a RGD motif. An example of an amphiphilic peptide is a 23RGD peptide having an amino acid sequence: HOOC-Gly-ArgGly-Asp-Ile-Pro-Ala-Ser-Ser-Lys-Gly-Gly-Gly-Gly-SerArg-Leu-Leu-Leu-Leu-Leu-Leu-Arg-NH2. Other examples of amphiphilic peptides include the ones disclosed in the U.S. patent app. No.: US 2011/0008406.

Injectable Compositions Comprising Silk Fibroin Particles

In another aspect, provided herein is an injectable composition for use in repairing or augmenting a tissue in a subject comprising a plurality of silk fibroin particles described herein, wherein at least a portion of the silk fibroin particles retain their original volume (e.g., at least about 50% or higher) within the tissue to be repaired or augmented for a period of time (e.g., at least about 6 weeks or longer).

As used herein, the term “injectable composition” generally refers to a composition that can be delivered or administered into a tissue with a minimally invasive procedure. The term “minimally invasive procedure” refers to a procedure that is carried out by entering a subject's body through the skin or through a body cavity or an anatomical opening, but with the smallest damage possible (e.g., a small incision, injection). In some embodiments, the injectable composition can be administered or delivered into a tissue by injection. In some embodiments, the injectable composition can be delivered into a tissue through a small incision on the skin followed by insertion of a needle, a cannula, and/or tubing, e.g., a catheter. Without wishing to be limited, the injectable composition can be administered or placed into a tissue by surgery, e.g., implantation.

In some embodiments, the injectable compositions can comprise at least one active agent described herein. In some embodiments, the injectable compositions can comprise at least one cell. The term “cells” used herein refers to any cell, prokaryotic or eukaryotic, including plant, yeast, worm, insect and mammalian. In some embodiments, the cells can be mammalian cells. Mammalian cells include, without limitation: primate, human and a cell from any animal of interest, including without limitation; mouse, hamster, rabbit, dog, cat, domestic animals, such as equine, bovine, murine, ovine, canine, feline, etc. The cells can be a wide variety of tissue types without limitation such as; hematopoietic, neural, mesenchymal, cutaneous, mucosal, stromal, muscle spleen, reticuloendothelial, epithelial, endothelial, hepatic, kidney, gastrointestinal, pulmonary, T-cells etc. Stem cells, pluripotent stem cells (iPSCs), embryonic stem (ES) cells, ES-derived cells and stem cell progenitors are also included, including without limitation, hematopoietic, neural, stromal, muscle, cardiovascular, hepatic, pulmonary, and gastrointestinal stem cells and adipose-derived stem cells. In one embodiment, the cells are adipose-derived stem cells. In some embodiments, the cells can be ex vivo or cultured cells, e.g. in vitro. For example, for ex vivo cells, cells can be obtained from a subject, where the subject is healthy and/or affected with a disease.

Cells can be obtained, as a non-limiting example, by biopsy or other surgical means known to those skilled in the art. In some embodiments, adipose cells can be harvested from a subject by conventional liposuction or aspiration techniques. In such embodiments, the cells can be derived from a lipoaspirate. In other embodiments, the cells can be derived from a bone-marrow aspirate. Depending on the types of tissues to be repaired or augmented, cells can be derived from any biological fluid or concentrate, e.g., a lipoaspirate or a bone-marrow aspirate.

Accordingly, in some embodiments, the injectable composition or the silk fibroin particles can be directly delivered with a biological fluid or concentrate, e.g., a lipoaspirate or a bone-marrow aspirate. In some embodiments, the injectable composition or the composition comprising a plurality of silk fibroin particles can further comprise a biological fluid or concentrate, such as lipoaspirate, bone marrow aspirate, or any combinations thereof. In one embodiment, the injectable composition or the composition comprising a plurality of silk fibroin particles can further comprise a lipoaspirate.

In these embodiments, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 1:38 to about 12:19, or about 1:19 to about 10:19, or about 2:19 to about 8:19. In one embodiment, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 3:19. In another embodiment, the composition or the injectable composition can comprise silk fibroin particles and a biological fluid or concentrate (e.g., a lipoaspirate or a bone-marrow aspirate) in a volume ratio of about 6:19.

Cells can be obtained from donors (allogenic) or from recipients (autologous). Cells can also be of established cell culture lines, or even cells that have undergone genetic engineering. Additionally, cells can be collected from a multitude of hosts including but not limited to human autograft tissues, transgenic mammals, or bacterial cultures (possibly for use as a probiotic treatment). In certain embodiments, the injectable compositions and/or silk fibroin particles can comprise human stem cells such as, e.g., mesenchymal stem cells, pluripotent stem cells (iPSCs), synovial derived stem cells, embryonic stem cells, adult stem cells, umbilical cord blood cells, umbilical Wharton's jelly cells, osteocytes, fibroblasts, neuronal cells, lipocytes, adipocytes, bone marrow cells, assorted immunocytes, precursor cells derived from adipose tissue, bone marrow derived progenitor cells, peripheral blood progenitor cells, stem cells isolated from adult tissue and genetically transformed cells or combinations of the above cells; or differentiated cells such as, e.g., muscle cells, adipose cells.

Stem cells can be obtained with minimally invasive procedures from bone marrow, adipose tissue, or other sources in the body, are highly expandable in culture, and can be readily induced to differentiate into adipose tissue forming cells after exposure to a well-established adipogenic inducing supplement. Cells can be added to the injectable compositions and/or silk fibroin particles described herein and cultured in vitro for a period of time prior to administration to a region of the body, or added to injectable compositions and/or silk fibroin particles described herein and administered into a region of the body. The cells can be seeded on the silk fibroin particles for a short period of time (less than 1 day) just prior to administration, or cultured for a longer (more than 1 day) period to allow for cell proliferation and extracellular matrix synthesis within the seeded matrix prior to administration.

When utilized as a source of stem cells, adipose tissue can be obtained by any method known to a person of ordinary skill in the art. For example, adipose tissue can be removed from an individual by suction-assisted lipoplasty, ultrasound-assisted lipoplasty, and excisional lipectomy. In addition, the procedures can include a combination of such procedures. Suction assisted lipoplasty can be desirable to remove the adipose tissue from an individual as it provides a minimally invasive method of collecting tissue with minimal potential for stem cell damage that can be associated with other techniques, such as ultrasound assisted lipoplasty. The adipose tissue should be collected in a manner that preserves the viability of the cellular component and that minimizes the likelihood of contamination of the tissue with potentially infectious organisms, such as bacteria and/or viruses.

In some embodiments, preparation of the cell population can require depletion of the mature fat-laden adipocyte component of adipose tissue. This is typically achieved by a series of washing and disaggregation steps in which the tissue is first rinsed to reduce the presence of free lipids (released from ruptured adipocytes) and peripheral blood elements (released from blood vessels severed during tissue harvest), and then disaggregated to free intact adipocytes and other cell populations from the connective tissue matrix. Disaggregation can be achieved using any conventional techniques or methods, including mechanical force (mincing or shear forces), enzymatic digestion with single or combinatorial proteolytic enzymes, such as collagenase, trypsin, lipase, liberase HI and pepsin, or a combination of mechanical and enzymatic methods. For example, the cellular component of the intact tissue fragments can be disaggregated by methods using collagenase-mediated dissociation of adipose tissue, similar to the methods for collecting microvascular endothelial cells in adipose tissue, as known to those of skill in the art. Additional methods using collagenase that can be used are also known to those of skill in the art. Furthermore, methods can employ a combination of enzymes, such as a combination of collagenase and trypsin or a combination of an enzyme, such as trypsin, and mechanical dissociation.

The cell population (processed lipoaspirate) can then be obtained from the disaggregated tissue fragments by reducing the presence of mature adipocytes. Separation of the cells can be achieved by buoyant density sedimentation, centrifugation, elutriation, differential adherence to and elution from solid phase moieties, antibody-mediated selection, differences in electrical charge; immunomagnetic beads, fluorescence activated cell sorting (FACS), or other means.

Following disaggregation the active cell population can be washed/rinsed to remove additives and/or by-products of the disaggregation process (e.g., collagenase and newly released free lipid). The active cell population could then be concentrated by centrifugation. In one embodiment, the cells are concentrated and the collagenase removed by passing the cell population through a continuous flow spinning membrane system or the like, such as, for example, the system disclosed in U.S. Pat. Nos. 5,034,135; and 5,234,608, which are incorporated by reference herein.

In addition to the foregoing, there are many post-wash methods that can be applied for further purifying the cell population. These include both positive selection (selecting the target cells), negative selection (selective removal of unwanted cells), or combinations thereof. In another embodiment the cell pellet could be resuspended, layered over (or under) a fluid material formed into a continuous or discontinuous density gradient and placed in a centrifuge for separation of cell populations on the basis of cell density. In a similar embodiment, continuous flow approaches such as apheresis and elutriation (with or without countercurrent) could be used. Adherence to plastic followed by a short period of cell expansion has also been applied in bone marrow-derived adult stem cell populations. This approach uses culture conditions to preferentially expand one population while other populations are either maintained (and thereby reduced by dilution with the growing selected cells) or lost due to absence of required growth conditions. The cells that have been concentrated, cultured and/or expanded can be incorporated into the silk fibroin particles and/or injectable compositions described herein.

In one embodiment, stem cells are harvested, the harvested cells are contacted with an adipogenic medium for a time sufficient to induce differentiation into adipocytes, and the adipocytes are loaded onto a biocompatible matrix which is implanted. In additional embodiments, at least some of the stem cells can be differentiated into adipocytes so that a mixture of both cell types is initially present that changes over time to substantially only adipocytes, with stem cells being present in small to undetectable quantities. Adipose tissue is fabricated in vivo by the stem cells or prepared ex vivo by the stem cells.

A number of different cell types or combinations thereof can be employed in the injectable compositions, depending upon the types of tissues to be repaired or augmented. These cell types include, but are not limited to: smooth muscle cells, skeletal muscle cells, cardiac muscle cells, epithelial cells, endothelial cells, urothelial cells, fibroblasts, myoblasts, chondrocytes, chondroblasts, osteoblasts, osteoclasts, keratinocytes, hepatocytes, bile duct cells, pancreatic islet cells, thyroid, parathyroid, adrenal, hypothalamic, pituitary, ovarian, testicular, salivary gland cells, adipocytes, and precursor cells. By way of example only, smooth muscle cells and endothelial cells can be employed when the injectable compositions are used to repair or augment muscular and/or vascular tissues, such as vascular, esophageal, intestinal, rectal, or ureteral tissues: chondrocytes can be included in injectable compositions for cartilaginous tissues; fibroblasts can be included in injectable compositions intended to replace and/or enhance any of the wide variety of tissue types (e.g., skin) that contains extracellular matrix, e.g., collagen; adipocytes can be included in injectable compositions intended to repair or augment any of the wide variety of adipose tissues. In general, any cells that are found in the natural tissue can be included in the injectable compositions used for corresponding tissue. In addition, progenitor cells, such as myoblasts or stem cells, can be included to produce their corresponding differentiated cell types.

In some embodiments, the injectable compositions can further comprise a pharmaceutically acceptable carrier. As used herein, the term “pharmaceutically acceptable carrier” refers to a pharmaceutically-acceptable material, composition or vehicle for administration of the silk fibroin particles, and optionally an active agent. Pharmaceutically acceptable carriers include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, and isotonic and absorption delaying agents, which are compatible with the silk fibroin particles and the activity of the active agent, if any, and are physiologically acceptable to the subject. The pharmaceutical formulations suitable for injection include sterile aqueous solutions or dispersions. The carrier can be a solvent or dispersing medium containing, for example, water, cell culture medium, buffers (e.g., phosphate buffered saline), polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof. In some embodiments, the pharmaceutical carrier can be a buffered solution (e.g. PBS).

Additionally, various additives which enhance the stability, sterility, and isotonicity of the injectable compositions, including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. In many cases, it may be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. The injectable compositions can also contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, colors, and the like, depending upon the preparation desired. Standard texts, such as “REMINGTON'S PHARMACEUTICAL SCIENCE”, 17th edition, 1985, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.

Viscosity of the injectable compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent. In one embodiment, methylcellulose is used because it is readily and economically available and is easy to work with. Other suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like. The preferred concentration of the thickener will depend upon the agent selected, and the desired viscosity for injection. The important point is to use an amount which will achieve the selected viscosity, e.g., addition of such thickening agents into some embodiments of the injectable compositions.

Typically, any additives (in addition to the silk fibroin particles described herein and/or additional active agents) can be present in an amount of 0.001 to 50 wt % dry weight or in a buffered solution. In some embodiments, the active agent can be present in the order of micrograms to milligrams to grams, such as about 0.0001 to about 5 wt %, about 0.0001 to about 1 wt %, about 0.0001 to about 0.05 wt % or about 0.001 to about 20 wt %, about 0.01 to about 10 wt %, and about 0.05 to about 5 wt %. For any pharmaceutical composition to be administered to a subject in need thereof, it is preferred to determine toxicity, such as by determining the lethal dose (LD) and LD50 in a suitable animal model e.g., rodent such as mouse; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response. Such determinations do not require undue experimentation from the knowledge of the skilled artisan.

Active Agents

In some embodiments, the injectable composition and/or the silk fibroin particles described herein can further comprise at least one active agent. The active agent can be mixed, dispersed, or suspended in the injectable composition, and/or it can be distributed or embedded in the silk fibroin particles. In some embodiments, the active agent can be distributed, embedded or encapsulated in the silk fibroin particles. In some embodiments, the active agent can be coated on surfaces of the silk fibroin particles. In some embodiments, the active agent can be mixed with the silk fibroin particles to form an injectable composition. The term “active agent” can also encompass combinations or mixtures of two or more active agents, as described below. Examples of active agents include, but are not limited to, a biologically active agent (e.g., a therapeutic agent), a cosmetically active agent (e.g., an anti-aging agent), a cell attachment agent (e.g., integrin-binding molecules), and any combinations thereof.

The term “biologically active agent” as used herein refers to any molecule which exerts at least one biological effect in vivo. For example, the biologically active agent can be a therapeutic agent to treat or prevent a disease state or condition in a subject. Examples of biologically active agents include, without limitation, peptides, peptidomimetics, aptamers, antibodies or a portion thereof, antibody-like molecules, nucleic acids (DNA, RNA, siRNA, shRNA), polysaccharides, enzymes, receptor antagonists or agonists, hormones, growth factors, autogenous bone marrow, antibiotics, antimicrobial agents, small molecules and therapeutic agents. The biologically active agents can also include, without limitations, anti-inflammatory agents, anesthetics, active agents that stimulate issue formation, and/or healing and regrowth of natural tissues, and any combinations thereof.

Anti-inflammatory agents can include, but are not limited to, naproxen, sulindac, tolmetin, ketorolac, celecoxib, ibuprofen, diclofenac, acetylsalicylic acid, nabumetone, etodolac, indomethacin, piroxicam, cox-2 inhibitors, ketoprofen, antiplatelet medications, salsalate, valdecoxib, oxaprozin, diflunisal, flurbiprofen, corticosteroids, MMP inhibitors and leukotriene modifiers or combinations thereof.

Agents that increase formation of new tissues and/or stimulates healing or regrowth of native tissue at the site of injection can include, but are not limited to, fibroblast growth factor (FGF), transforming growth factor-beta (TGF-β, platelet-derived growth factor (PDGF), epidermal growth factors (EGFs), connective tissue activated peptides (CTAPs), osteogenic factors including bone morphogenic proteins, heparin, angiotensin II (A-II) and fragments thereof, insulin-like growth factors, tumor necrosis factors, interleukins, colony stimulating factors, erythropoietin, nerve growth factors, interferons, biologically active analogs, fragments, and derivatives of such growth factors, and any combinations thereof.

Anesthetics can include, but are not limited to, those used in caudal, epidural, inhalation, injectable, retrobulbar, and spinal applications, such as bupivacaine, lidocaine, benzocaine, cetacaine, ropivacaine, and tetracaine, or combinations thereof.

In some embodiments, the active agents can be cosmetically active agents. By the term “cosmetically active agent” is meant a compound that has a cosmetic or therapeutic effect on the skin, hair, or nails, e.g., anti-aging agents, anti-free radical agents, lightening agents, whitening agents, depigmenting agents, darkening agents such as self-tanning agents, colorants, anti-acne agents, shine control agents, anti-microbial agents, anti-inflammatory agents, anti-mycotic agents, anti-parasite agents, external analgesics, sun-blocking agents, photoprotectors, antioxidants, keratolytic agents, detergents/surfactants, moisturizers, nutrients, vitamins, energy enhancers, anti-perspiration agents, astringents, deodorants, hair removers, firming agents, anti-callous agents, muscle relaxants, agents for hair, nail, and/or skin conditioning, and any combination thereof.

In one embodiment, the cosmetically active agent can be selected from, but not limited to, the group consisting of hydroxy acids, benzoyl peroxide, sulfur resorcinol, ascorbic acid, D-panthenol, hydroquinone, octyl methoxycinnamate, titanium dioxide, octyl salicylate, homosalate, avobenzone, polyphenolics, carotenoids, free radical scavengers, ceramides, polyunsaturated fatty acids, essential fatty acids, enzymes, enzyme inhibitors, minerals, hormones such as estrogens, steroids such as hydrocortisone, 2-dimethylaminoethanol, copper salts such as copper chloride, coenzyme Q10, lipoic acid, amino acids such a proline and tyrosine, vitamins, lactobionic acid, acetyl-coenzyme A, niacin, riboflavin, thiamin, ribose, electron transporters such as NADH and FADH2, and other botanical extracts such as aloe vera, feverfew, and soy, and derivatives and mixtures thereof. Examples of vitamins include, but are not limited to, vitamin A, vitamin Bs (such as vitamin B3, vitamin B5, and vitamin B12), vitamin C, vitamin K, and vitamin E, and derivatives thereof.

In one embodiment, the cosmetically active agents can be antioxidants. Examples of antioxidants include, but are not limited to, water-soluble antioxidants such as sulfhydryl compounds and their derivatives (e.g., sodium metabisulfite and N-acetyl-cysteine), lipoic acid and dihydrolipoic acid, resveratrol, lactoferrin, ascorbic acid, and ascorbic acid derivatives (e.g., ascorbyl palmitate and ascorbyl polypeptide). Oil-soluble antioxidants suitable for use in the compositions described herein include, but are not limited to, butylated hydroxytoluene, tocopherols (e.g., tocopheryl acetate), tocotrienols, and ubiquinone. Natural extracts containing antioxidants suitable for use in the injectable compositions described herein, include, but not limited to, extracts containing flavonoids and isoflavonoids and their derivatives (e.g., genistein and diadzein), and extracts containing resveratrol. Examples of such natural extracts include grape seed, green tea, pine bark, and propolis. Other examples of antioxidants can be found on pages 1612-13 of the ICI Handbook.

In some embodiments, the active agents can be cell attachment agents. Examples of cell attachment agents include, but are not limited to, hyaluronic acid, collagen, crosslinked hyaluronic acid/collagen, an integrin-binding molecule, chitosan, elastin, fibronectin, vitronectin, laminin, proteoglycans, any derivatives thereof, and any combinations thereof.

In some embodiments, the injectable compositions and/or silk fibroin particles can further comprise at least one additional material for soft tissue augmentation, e.g., dermal filler materials, including, but not limited to, poly(methyl methacrylate) microspheres, hydroxylapatite, poly(L-lactic acid), collagen, elastin, and glycosaminoglycans, hyaluronic acid, commercial dermal filler products such as BOTOX® (from Allergan), DYSPORT®, COSMODERM®, EVOLENCE®, RADIESSE®, RESTYLANE®, JUVEDERM® (from Allergan), SCULPTRA®, PERLANE®, and CAPTIQUE®, and any combinations thereof.

In some embodiments, the injectable composition and/or silk fibroin particles can comprise metallic nanoparticles (e.g., but not limited to, gold nanoparticles), optical molecules (e.g., but not limited to, fluorescent molecules, and/or quantum dots), and any other art-recognized contrast agent, e.g., for biomedical imaging.

In various embodiments, the injectable compositions can be stored or transported dried or hydrated.

When the active agents are embedded in the silk fibroin particles, the bioactivity of the active agents (e.g., at least about 30% of the bioactivity of the active agents) can be stabilized for a period of time (e.g., days, weeks, or months) under specific conditions. Such conditions can include, but are not limited to, a state-changing cycle (e.g., freeze-thaw cycles), temperatures (e.g., above 0° C.), air pressures, humidity, and light exposure. See U.S. Application Ser. No. 61/477,737. Some embodiments of the injectable composition can be stored or transported between about 0° C. and about 60° C., about 10° C. and about 60° C., or about 15° C. and about 60° C. In these embodiments, the injectable compositions can be stored or transported at room temperatures while the bioactivity of the active agents (e.g., at least about 30% of the bioactivity of the active agents) can be stabilized for a period of time, e.g., at least about 3 weeks or longer.

Applications of Injectable Compositions and Silk Fibroin Particles Described Herein

The injectable compositions described herein can be used in a variety of medical uses, including, without limitation, fillers for tissue space, templates for tissue reconstruction or regeneration, scaffolds for cells in tissue engineering applications, or as a vehicle/carrier for drug delivery. A plurality of silk fibroin particles injected into a tissue to be repaired or augmented can act as a scaffold to mimic the extracellular matrices (ECM) of the body, and/or promote tissue regeneration. The scaffold can serve as both a physical support and/or an adhesive template for cells to proliferate therein. In some embodiments, the silk fibroin particles can contain no cells. Yet the silk fibroin particles can be coated with cell attachment agents, e.g., collagen, and/or chemoattractants, e.g., growth factors, that can attract host cells to the silk fibroin particles and support the cell proliferation. In some embodiments, the silk fibroin particles can be seeded with cells prior to administration to a target tissue to be repaired or augmented.

In some embodiments, provided herein are injectable compositions that can be used to fill, volumize, and/or regenerate a tissue in need thereof. The injectable compositions can generally be used for tissue filling or volumizing, soft tissue augmentation, replacement, cosmetic enhancement and/or tissue repair in a subject. Additionally, the injectable compositions can be used for filling of any tissue void or indentation that are either naturally formed (e.g., aging) or created by surgical procedure for removal of tissue (e.g., a dermal cyst or a solid tumor), corticosteroid treatment, immunologic reaction resulting in lipoatrophy, tissue damage resulting from impact injuries or therapeutic treatment (e.g., radiotherapy or chemotherapy). The injectable compositions can also be used to raise scar depressions.

In certain embodiments, the injectable compositions can be used for soft tissue augmentation. As used herein, by the term “augmenting” or “augmentation” is meant increasing, filling in, restoring, enhancing or replacing a tissue. In some embodiments, the tissue can lose its elasticity, firmness, shape and/or volume. In some embodiments, the tissue can be partially or completely lost (e.g., removal of a tissue) or damaged. In those embodiments, the term “augmenting” or “augmentation” can also refer to decreasing, reducing or alleviating at least one symptom or defect in a tissue (for example, but not limited to, loss of elasticity, firmness, shape and/or volume in a tissue; presence of a void or an indentation in a tissue; loss of function in a tissue) by injecting into the tissue with at least one injectable composition described herein. In such embodiments, at least one symptom or defect in a tissue can be decreased, reduced or alleviated by at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80% or higher, as compared to no treatment. In some embodiments, at least one symptom or defect in a tissue can be decreased, reduced or alleviated by at least about 90%, at least about 95%, at least about 97%, or higher, as compared to no treatment. In some embodiments, at least one symptom or defect in a tissue can be decreased, reduced or alleviated by 100% (defect-free or the defect is undetectable by one of skill in the art), as compared to no treatment. In other embodiments, the tissue can be augmented to prevent or delay the onset of defect manifestation in a tissue, e.g., loss of elasticity, firmness, shape and/or volume in a tissue, or signs of wrinkles. As used herein, the phrase “soft tissue augmentation” is generally used in reference to altering a soft tissue structure, including but not limited to, increasing, filling in, restoring, enhancing or replacing a tissue, e.g., to improve the cosmetic or aesthetic appearance of the soft tissue. For example, breast augmentation (also known as breast enlargement, mammoplasty enlargement, augmentation mammoplasty) alters the size and shape of a woman's breasts to improve the cosmetic or aesthetic appearance of the woman. Examples of soft tissue augmentation includes, but is not limited to, dermal tissue augmentation; filling of lines, folds, wrinkles, minor facial depressions, and cleft lips, especially in the face and neck; correction of minor deformities due to aging or disease, including in the hands and feet, fingers and toes; augmentation of the vocal cords or glottis to rehabilitate speech; dermal filling of sleep lines and expression lines; replacement of dermal and subcutaneous tissue lost due to aging; lip augmentation; filling of crow's feet and the orbital groove around the eye; breast augmentation; chin augmentation; augmentation of the cheek and/or nose; bulking agent for periurethral support, filling of indentations in the soft tissue, dermal or subcutaneous, due to, e.g., overzealous liposuction or other trauma; filling of acne or traumatic scars; filling of nasolabial lines, nasoglabellar lines and intraoral lines. In some embodiments, the injectable compositions and/or silk fibroin particles described herein can be used to treat facial lipodystrophies. In some embodiments, the injectable compositions can be used for breast augmentation and/or reconstruction.

In some embodiments, the injectable compositions can be used for soft tissue repair. The term “repair” or “repairing” as used herein, with respect to a tissue, refers to any correction, reinforcement, reconditioning, remedy, regenerating, filling of a tissue that restores volume, shape and/or function of the tissue. In some embodiments “repair” includes full repair and partial repair. For example, the volume, shape and/or function of a tissue to be repaired can be restored by at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80% or higher, as compared to no treatment. In some embodiments, the volume, shape and/or function of a tissue to be repaired can be restored by at least about 90%, at least about 95%, at least about 97%, or higher, as compared to no treatment. In some embodiments, the volume, shape and/or function of a tissue to be repaired can be restored by 100% (defect-free or the defect is undetectable by one of skill in the art), as compared to no treatment. In various embodiments, the injectable compositions can be used to repair any soft tissues discussed earlier, e.g., breast, skin, and any soft tissues amenable for soft tissue augmentation. In some embodiments, the term “repair” or “repairing” are used herein interchangeably with the term “regeneration” or “regenerate” when used in reference to tissue treatment.

In some embodiments, the injectable compositions can be used for soft tissue reconstruction. As used herein, the phrase “soft tissue reconstruction” refers to rebuilding a soft tissue structure that was severely damaged or lost, e.g., by a dramatic accident or surgical removal. For example, breast reconstruction is the rebuilding of a breast, usually in women. Conventional methods of construct a natural-looking breast generally involve using autologous tissue or prosthetic material. In some embodiments, such breast reconstruction can include reformation of a natural-looking areola and nipple, wherein such procedure can involve the use of implants or relocated flaps of the patient's own tissue. In some embodiments, administration of injectable compositions and/or silk fibroin particles into a soft tissue region to be reconstructed can maintain the shape and/or size of the reconstructed soft tissue structure for a period of time, e.g., at least 6 weeks, at least about 2 months, at least about 3 months or longer.

Without wishing to be bound, some embodiments of the injectable compositions can be used for hard tissue (musculoskeletal) augmentation or repair, such as augmentation or repair of bone, cartilage and ligament.

The injectable compositions and silk fibroin particles described herein can also be used for filling a tissue located at or near a prosthetic implant, for example, but not limited to, a conventional breast implant or knee replacement implant. In some embodiments, the injectable compositions and silk fibroin particles can be used to interface between a prosthetic implant and a tissue, e.g., to fill a void between the prosthetic implant and the tissue, and/or to prevent the tissue in direct contact with the prosthetic implant. By way of example only, after placing a prosthetic implant (e.g., a breast implant) in a subject, an injectable composition described herein can be introduced at or adjacent to the implant to fill any void between the implant and the tissue (e.g., breast tissue) and/or “sculpt” the tissue for a more natural look.

In any of the uses described herein, silk fibroin particles could be combined with cells for purposes of a biologically enhanced repair. Cells could be collected from a multitude of hosts including but not limited to human autograft tissues, or transgenic mammals. More specifically, human cells used can comprise cells selected from stem cells (e.g., adipocyte-derived stem cells), osteocytes, fibroblasts, lipocytes, assorted immunocytes, cells from lipoaspirate or any combinations thereof. In some embodiments, the cells can be added after rinsing of the silk fibroin particles themselves. They can be blended into the silk fibroin particles, carrier solution, or mixture of silk fibroin particles and carrier solution prior to injection.

In some embodiments, administering the cells (e.g., stem cells or lipoaspirate) with silk fibroin particles or an injectable composition described herein can enhance or accelerate host integration and/or tissue formation over time. The cells can be mixed with the silk fibroin particles or an injectable composition described herein, or they can be administered prior to, concurrently with, or after the silk fibroin particles or an injectable composition is introduced into a target site. Without wishing to be bound by theory, the cells can secrete pro-angiogenic factors and/or growth factors at the target site. As the tissue regenerates or remodels to fill up a void or repair a defect, the silk fibroin particles can degrade accordingly. In some embodiments, the silk fibroin particles can integrate with the regenerated host tissue.

In addition, active agents such as therapeutic agents, pharmaceuticals, or specific growth factors added to the silk fibroin particles for purposes of improved outcome can be introduced at any or a combination of several points throughout the silk fibroin particle production process. In some embodiments, these factors can be added to silk fibroin solution or the accelerant phase prior to drying and solidification, they can be soaked into the silk fibroin matrix during the accelerant rinsing process, or they can be coated onto the bulk silk fibroin following rinsing. The active-agent containing solid-state silk fibroin can then be reduced into particles using the methods described herein. In some embodiments, a solid-state silk fibroin can be reduced into particles before introducing an active agent into the silk fibroin particles. For example, an active agent can be soaked into the silk fibroin particles, coated onto the silk fibroin particles, or introduced into a carrier fluid before or after blending with the silk fibroin particles.

In some aspects, the injectable composition and silk fibroin particles described herein can be used as tissue space fillers. In one embodiment, the tissue space filler is a dermal filler. The dermal filler can be used to improve skin appearance or condition, including, but not limited to, rehydrating the skin, providing increased elasticity to the skin, reducing skin roughness, making the skin tauter, reducing or eliminating stretch lines or marks, giving the skin better tone, shine, brightness, and/or radiance, reducing or eliminating wrinkles in the skin, providing wrinkle resistance to the skin and replacing loss of soft tissue.

A dermal filler comprising silk fibroin particles can be modulated for particle hardness and opacity through alteration of silk fibroin concentration and formulation method. In one embodiment, a dermal filler can be produced by forming a solid-state silk fibroin from a silk fibroin solution of about 6% (w/v) to about 20% (w/v), followed by reducing the solid-state silk fibroin into silk fibroin particles such that they can be injected into a tissue through a needle or a cannula. The needle or cannula can have an outer diameter of no larger than 3 mm, no larger than 2 mm, no larger than 1 mm, no larger than 0.8 mm, no larger than 0.6 mm, no larger than 0.4 mm, no larger than 0.2 mm or no larger than 0.1 mm. In some embodiments, the needle or cannula gauge can range from 12 to 34, 15 to 34, 20 to 32, or 25 to 30. The size of the needle or cannula can be determined to allow for an appropriate extrusion force of less than 40 N (nominal deliverable injection force for a human hand).

Accordingly, another aspect described herein provides a method of improving a condition and/or appearance of skin in a subject in need thereof. Non-limiting examples of a skin condition or and/or appearance include dehydration, lack of skin elasticity, roughness, lack of skin tautness, skin stretch line and/or marks, skin paleness, and skin wrinkles. The method comprises injecting an injectable composition described herein into a dermal region of the subject, wherein the injection improves the skin condition and/or appearance. For example, improving a skin appearance include, but are not limited to, rehydrating the skin, providing increased elasticity to the skin, reducing skin roughness, making the skin tauter, reducing or eliminating stretch lines or marks, giving the skin better tone, shine, brightness and/or radiance to reduce paleness, reducing or eliminating wrinkles in the skin, and providing wrinkle resistance to the skin.

As used herein, the term “dermal region” refers to the region of skin comprising the epidermal-dermal junction and the dermis including the superficial dermis (papillary region) and the deep dermis (reticular region). The skin is composed of three primary layers: the epidermis, which provides waterproofing and serves as a barrier to infection; the dermis, which serves as a location for the appendages of skin; and the hypodermis (subcutaneous adipose layer). The epidermis contains no blood vessels, and is nourished by diffusion from the dermis. The main type of cells which make up the epidermis include, but are not limited to, keratinocytes, melanocytes, Langerhans cells and Merkels cells.

The dermis is the layer of skin beneath the epidermis that consists of connective tissue and cushions the body from stress and strain. The dermis is tightly connected to the epidermis by a basement membrane. It also harbors many mechanoreceptor/nerve endings that provide the sense of touch and heat. It contains the hair follicles, sweat glands, sebaceous glands, apocrine glands, lymphatic vessels and blood vessels. The blood vessels in the dermis provide nourishment and waste removal from its own cells as well as from the Stratum basale of the epidermis. The dermis is structurally divided into two areas: a superficial area adjacent to the epidermis, called the papillary region, and a deep thicker area known as the reticular region.

The papillary region is composed of loose areolar connective tissue. It is named for its fingerlike projections called papillae that extend toward the epidermis. The papillae provide the dermis with a “bumpy” surface that interdigitates with the epidermis, strengthening the connection between the two layers of skin. The reticular region lies deep in the papillary region and is usually much thicker. It is composed of dense irregular connective tissue, and receives its name from the dense concentration of collagenous, elastic, and reticular fibers that weave throughout it. These protein fibers give the dermis its properties of strength, extensibility, and elasticity. Also located within the reticular region are the roots of the hair, sebaceous glands, sweat glands, receptors, nails, and blood vessels. Stretch marks from pregnancy are also located in the dermis.

The hypodermis is not part of the skin, and lies below the dermis. Its purpose is to attach the skin to underlying bone and muscle as well as supplying it with blood vessels and nerves. It consists of loose connective tissue and elastin. The main cell types are fibroblasts, macrophages and adipocytes (the hypodermis contains 50% of body fat). Fat serves as padding and insulation for the body.

In one embodiment, provided herein is a method of treating skin dehydration, which comprises injecting to a dermal region suffering from skin dehydration one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition rehydrates the skin, thereby treating skin dehydration.

In another embodiment, a method of treating a lack of skin elasticity comprises injecting to a dermal region suffering from a lack of skin elasticity one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition increases the elasticity of the skin, thereby treating a lack of skin elasticity.

In yet another embodiment, a method of treating skin roughness comprises injecting to a dermal region suffering from skin roughness one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition decreases skin roughness, thereby treating skin roughness.

In still another embodiment, a method of treating a lack of skin tautness comprises injecting to a dermal region suffering from a lack of skin tautness one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition makes the skin tauter, thereby treating a lack of skin tautness.

In a further embodiment, a method of treating a skin stretch line or mark comprises injecting to a dermal region suffering from a skin stretch line or mark one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition reduces or eliminates the skin stretch line or mark, thereby treating a skin stretch line or mark.

In another embodiment, a method of treating skin paleness comprises injecting to a dermal region suffering from skin paleness one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition increases skin tone or radiance, thereby treating skin paleness.

In another embodiment, a method of treating skin wrinkles comprises injecting to a dermal region suffering from skin wrinkles one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition reduces or eliminates skin wrinkles, thereby treating skin wrinkles.

In yet another embodiment, a method of treating, preventing or delaying the formation of skin wrinkles comprises injecting to a dermal region susceptible to, or showing signs of wrinkles one or more embodiments of an injectable composition described herein. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally a biological fluid carrier (e.g., a lipoaspirate) and/or an active agent, wherein the injection of the composition makes the skin resistant to skin wrinkles, thereby treating, preventing or delaying the formation of skin wrinkles.

The effective amount and administration schedule of silk fibroin particles injected into a dermal region can be determined by a person of ordinary skill in the art taking into account various factors, including, without limitation, the type of skin condition, the location of the skin condition, the cause of the skin condition, the severity of the skin condition, the degree of relief desired, the duration of relief desired, the particular silk fibroin particle formulation used, the rate of degradation or volume retention of the particular silk fibroin particle formulation used, the pharmacodynamics of the particular silk fibroin particle formulation used, the nature of the other compounds included in the particular silk fibroin particle formulation used, the particular characteristics, history and risk factors of the individual, such as, e.g., age, weight, general health, and any combinations thereof. In some embodiments, the silk fibroin particles can be injected into a dermal region every 3 months, every 6 months, every 9 months, every year, every two years or longer.

In another aspect, the injectable compositions can be used as a dermal filler for dermal bulking to reconstruct or augment a soft tissue body part, such as, e.g., a lip, a breast, a breast part such as the nipple, a muscle, or any other soft body part where adipose and/or connective tissue is used to provide shape, insulation, or other biological function. In fillers used for these applications, the silk fibroin concentration and/or the amount of a carrier (e.g., saline) added to silk fibroin particle mixture can be adjusted for the relevant constraints of a given biological environment. For example, silk fibroin particles for breast augmentation can be adapted for particle hardness and volume retention through alteration of silk fibroin concentration and processing method. For example, about 4% (w/v) to about 8% (w/v) silk fibroin concentration, optionally containing an active agent, e.g., adipose cells such adipose-derived stem cells or cells from lipoaspirate, can be used to produce the silk fibroin particles. Carrier content in the case of saline can be on the order of 0% to 25% (v/v). Other factors such as, e.g., defect type, defect size and needs for a specific depth of injection of the filler, should be also considered.

Without wishing to be bound, while injection is minimally-invasive, other administration method can be also be used, e.g., implantation, when needed, e.g., to repair or argument a large defect area. For example, for dermal injection and lip augmentation, a syringe needle sized 26 g-30 g can be used. In applications involving large quantities of filler, e.g., breast reconstruction or breast augmentation, a larger particle size and a larger bore needle such as 23 g-27 g or smaller needle gauge can be used to administer the filler. In some embodiments, surgery, e.g., implantation, can also be employed to administer large quantities of filler and/or to reach a certain depth of tissues.

Accordingly, yet another aspect described herein provide a method of soft tissue reconstruction, repair, or augmentation, the method comprising administering one or more embodiments of an injectable composition described herein to a soft tissue region of an individual in need thereof. For example, the injectable composition can comprise a plurality of silk fibroin particles, and optionally an active agent and/or a carrier (e.g., a biological fluid carrier such as a lipoaspirate). Administration methods of an injectable composition described herein can be determined by an ordinary artisan. In some embodiments, the administration method can be injection. In some embodiments, the administration method can be surgery, e.g., implantation.

While injectable compositions and/or silk fibroin particles described herein can be directly applied on a target region (e.g., injection or surgery), in some embodiments, an injectable composition and/or silk fibroin particles disclosed herein can also be used to fill an expandable implantable medical device, such as, e.g., an expandable breast implant shell, which is placed in a defect area. In such embodiments, provided herein is a method of soft tissue reconstruction, repair or augmentation, the method comprising placing an implantable medical device into a soft tissue region of an individual at the desired location and expanding the device by filling the device with silk fibroin particles and/or injectable compositions described herein, wherein expansion of the medical device by filling it with silk fibroin particles and/or injectable compositions described herein can reconstruct or augment the soft tissue.

The silk fibroin particles or injectable compositions disclosed herein can be also used in conjunction with interventional radiology embolization procedures for blocking abnormal blood (artery) vessels (e.g., for the purpose of stopping bleeding) or organs (to stop the extra function e.g. embolization of the spleen for hypersplenism) including uterine artery embolization for percutaneous treatment of uterine fibroids. Modulation of silk fibroin particle hardness and volume retention rate can be done through alteration of silk fibroin concentration and processing methods as described earlier.

The silk fibroin particles or injectable compositions disclosed herein can be used to repair void space in a spine, e.g., created by spine disk nucleus removal surgery, to help maintain the normal distance between the adjacent vertebral bodies. In some embodiments, a vertebral disc filler comprising a plurality of silk fibroin particles can be used to repair void space present in the spine, e.g., between vertebral bodies, and/or in a ruptured spine disk. In such embodiments, a silk fibroin concentration of about 4% (w/v) to about 10% (w/v) can be used to fabricate the silk fibroin particles described herein. Accelerant and/or active agents can also be mixed with silk fibroin particles and/or injectable compositions before, during, or after injection into the site of interest.

The silk fibroin particles or injectable compositions disclosed herein can be used to fill up the vitreous cavity to support the eyeball structure and maintain the retina's position. The viscosity of the injectable composition described herein can be adjusted for the viscosity of vitreous fluid in the eye by one of skill in the art.

In some embodiments, the silk fibroin particles and/or injectable compositions can be used as a template for tissue reconstruction or augmentation, e.g., soft tissue reconstruction or augmentation (e.g., breast augmentation), or even for small bone or cartilage defects such as fractures. The administration of silk fibroin particles or injectable compositions described herein can be used to facilitate cartilage/bone cell ingrowth and proliferation and support collagen matrix deposition thus to improve cartilage/bone repair. In another aspect, prior to administration, donor cartilage cells can be seeded or mixed with silk fibroin particles and/or injectable compositions described herein to expand cell population and thus to promote the development of cartilage tissue. In some embodiments, specific growth factors such as TGF-β or bone morphogenic proteins (BMPs) which support cartilage or bone tissue formation, respectively, can be added into silk fibroin particles.

In another embodiment, the silk fibroin particles and/or injectable compositions described herein can be used for facial plastic surgery, such as, e.g., nose reconstruction. The reconstruction strategy discussed above for repairing a cartilage/bone defect can also be applicable for facial plastic surgery.

In some embodiments, the silk fibroin particles and/or injectable compositions described herein can be used as scaffolds to support cell growth for tissue engineering. For example, the silk fibroin particles and/or injectable compositions described herein can be administered into an incision or wound site to promote wound healing or wound disclosure. The methods generally comprise administering an injectable composition or silk fibroin particles described herein, at the wound or incision site and allowing the wound or incision to heal while the silk fibroin particles is eroded or absorbed in the body and is replaced with the individual's own viable tissue. The methods can further comprise seeding the silk fibroin particles or mixing the injectable composition with viable cellular material, either from the individual or from a donor, prior to or during administration.

In another aspect, the injectable composition comprising silk fibroin particles can be used, directly or indirectly, in methods of repairing, augmenting, or reconstructing a tissue in a subject, e.g., augmenting or reconstructing the breast of a human being. In some embodiments, the injectable compositions or silk fibroin particles can be directly placed into a tissue (e.g., a breast tissue) to be repaired or augmented, e.g., by injection. The injectable compositions or silk fibroin particles can be injected into a tissue (e.g., a breast tissue) every 6 months, every year, every 2 years, every 3 years, or longer. In other embodiments, the injectable compositions or silk fibroin particles can be used to enhance support of a conventional tissue implant, e.g., by enhancing support of the lower pole position of a breast implant. In alternative embodiments, the method can generally comprise administering an injectable composition and/or silk fibroin particles near or in proximity to a tissue implant, for example, a conventional breast implant, and seeding the injectable composition and/or silk fibroin particles with viable cellular material prior to or during administration. In yet another embodiment, an injectable composition and/or silk fibroin particles can be used to partially or completely cover a tissue implant (e.g., a breast implant) to provide a beneficial interface with host tissue and to reduce the potential for malpositioning or capsular contracture.

In some embodiments, the silk fibroin particles and/or injectable compositions described herein can be used as fillers to promote or support adipogenesis, e.g., to treat facial lipodystrophies. In such embodiments, the injectable compositions and/or silk fibroin particles can be seeded or mixed with adipose-associated cells, such adipose-derived stem cells or lipoaspirate, prior to or concurrently with the injection to a target area suffering from facial lipodystrophies in a subject. In some embodiments, the silk fibroin particles can be injected every 3 months, every 6 months, every 9 months, every year, every two years or longer, to maintain the treatment.

In still another embodiment, the silk fibroin particles and/or injectable compositions described herein can be used as the scaffold for cells useful for peripheral nerve repair. Silk fibroin particles can be delivered (e.g., via injection) to the location of the nerve defect with or without additional device to aid the connection to the nerve ends. For such purpose, specific growth factors such as nerve growth factor (NGF), which supports nerve regeneration can be added into injectable compositions and/or mixed with silk fibroin particles prior to or during administration. In such embodiments, softer silk fibroin particles, e.g. using a silk fibroin concentration of about 0.5 (w/v) to about 3% (w/v), can be used. Depending on the brain microenvironment, harder silk fibroin particles can also be used. The silk fibroin particles and/or injectable compositions can be infused with or added with appropriate therapeutic factors according to the methods described above.

Any cells described herein can be seeded upon a surface of silk fibroin particles described herein. For example, silk fibroin particles can be submersed in an appropriate growth medium for the cells of interest, and then directly exposed to the cells. The cells are allowed to proliferate on the surface and interstices of the silk fibroin particles. The silk fibroin particles are then removed from the growth medium, washed if necessary, and administered. Alternatively, the cells can be placed in a suitable buffer or liquid growth medium and drawn through silk fibroin particles by using vacuum filtration. Cells can also be admixed with silk fibroin solution prior to forming silk fibroin particles, capturing at least some of the cells within the silk fibroin particles. In another embodiment, the cells of interest can be dispersed into an appropriate solution (e.g., a growth medium or buffer) and then sprayed onto silk fibroin particles. For example, electro-spraying involves subjecting a cell-containing solution with an appropriate viscosity and concentration to an electric field sufficient to produce a spray of small charged droplets of solution that contain cells.

In some embodiments, the silk fibroin particles or injectable compositions comprising at least one active agent can be used as a platform for drug delivery. For example, the silk fibroin particles can be formed with a pharmaceutical agent either entrained in or bound to the particles and then administered into the body (e.g., injection, implantation or even oral administration). In some embodiments, an active agent can be mixed with silk fibroin particles and/or injectable compositions and then administered into the body (e.g., injection, implantation or even oral administration). For extended or sustained release, silk fibroin particles can manipulated, e.g., to modulate its beta-sheet content, for its volume retention and/or degradation rate. To further control the drug release profile, the pharmaceutically-active drug-containing silk fibroin particles can be mixed with an additional silk fibroin gel phase acting as a carrier either with or without a viscosity inducing component, a surfactant, and/or an included lubricant fluid like saline. The therapeutic-bound silk fibroin particles can also be further crosslinked to enhance the stability to extend the release period. In an alternative approach, silk fibroin particles can be mixed with other polymers, for examples, hyaluronic acid, to prolong the release of certain growth factors or cytokines and to stabilize the functionality. Furthermore, the silk fibroin particles and/or injectable compositions can also be used for coating coaxial drug delivery systems, e.g., by spraying.

As used herein, the term “sustained release” refers to the release of a pharmaceutically-active drug over a period of about seven days or more. In aspects of this embodiment, a drug delivery platform comprising the silk fibroin particles and/or injectable compositions releases a pharmaceutically-active drug over a period of, e.g., at least about 7 days after administration, at least about 15 days after administration, at least about 30 days after administration, at least about 45 days after administration, at least about 60 days after administration, at least about 75 days after administration, or at least about 90 days after administration.

As used herein, the term “extended release” refers to the release of a pharmaceutically-active drug over a period of time of less than about seven days. In such embodiments, a drug delivery platform comprising the silk fibroin particles and/or injectable compositions described herein can release a pharmaceutically-active drug over a period of, e.g., about 1 day after administration, about 2 days after administration, about 3 days after administration, about 4 days after administration, about 5 days after administration, or about 6 days after administration.

Depending on the formulation and processing methods of the silk fibroin particles and the associated applications, the injectable compositions or silk fibroin particles can be administered (e.g., by injection) periodically, for example, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, every year, every 2 years or longer.

In some embodiments of any of the applications described herein, the injectable compositions or silk fibroin particles can be at least partially dry when administered in a tissue to be repaired or augmented. In some embodiments, the injectable compositions or silk fibroin particles can be dried (e.g., in the absence of a carrier) when administered in a tissue to be repaired or augmented.

In some embodiments of any of the applications described herein, the injectable compositions or silk fibroin particles can be at least partially hydrated when administered in a tissue to be repaired or augmented. In some embodiments, the injectable compositions or silk fibroin particles can be hydrated (e.g., in the presence of an injection carrier, e.g., a buffered solution and/or lipoaspirate) when administered in a tissue to be repaired or augmented.

In some embodiments of any of the applications described herein, the injectable compositions or silk fibroin particles can be injected subcutaneously, submuscularly, or intramuscularly.

In some embodiments, the methods and/or compositions described herein can be used in the dermal region. In some embodiments, the methods and/or compositions described herein can be used in the epidermal layer, dermal layer, hypodermis layer, or any combinations thereof.

Delivery Devices and Kits Comprising Silk Fibroin Particles

Delivery devices comprising an injectable composition or silk fibroin particles described herein are also provided herein. Delivery devices can be any conventional delivery device used for injection purposes, e.g., a syringe. Accordingly, a further aspect provided herein is a syringe comprising an injectable composition or silk fibroin particles.

In some embodiments, the delivery device (e.g., a syringe) can further comprise a needle. In some embodiments, the delivery device (e.g., a syringe) can further comprise a catheter or a cannula.

In various embodiments, the delivery device (e.g., a syringe) can include an injection carrier, e.g., a buffered solution.

In various embodiments, the delivery device (e.g., a syringe) can include an anesthetic.

Further provided herein is a kit comprising one embodiment of an injectable composition or silk fibroin particles packaged in a syringe with a needle or a cannula. In some embodiments, a local anesthetic can be blended with the injectable composition or silk fibroin particles insides the syringe. In alternative embodiments, a local anesthetic can be packaged in a separate container or in a separate syringe. For example, it is desirable to apply a local anesthetic to a target tissue to be treated prior to further treatment. An exemplary anesthetic includes, but is not limited to, lidocane. Dependent upon application, the kit can include syringes sizes from 0.5 mL to 60 mL, where applications requiring larger volumes (e.g., bone fillers, disc fillers) are supplied in a larger size syringe. Additionally, needle gauge can adjusted according to injection site with an acceptable range of 10 g to 30 g needles. For example, 26 g to 30 g needles can be used for intradermal injections.

In some embodiments, the kit can further comprise a plurality of syringes (each with a needle) containing an injectable composition or silk fibroin particles described herein. Each syringe can be individually packaged.

In some embodiments, the kit can further comprise a container containing a buffered solution or an injection carrier.

In some embodiments, the kit can further comprise at least one additional empty syringe. In some embodiments, the kit can further comprise at least one additional needle. In some embodiments, the kit can further comprise at least one catheter or cannula.

Embodiments of the various aspects described herein can be illustrated by the following numbered paragraphs.

    • 1. A method for repairing or augmenting a tissue in a subject comprising: injecting in the tissue to be repaired or augmented a composition comprising a plurality of silk fibroin particles, wherein at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue for at least about 6 weeks.
    • 2. The method of paragraph 1, wherein said at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue for at least about 3 months.
    • 3. The method of paragraph 2, wherein said at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue for at least about 6 months.
    • 4. The method of any of paragraphs 1-3, wherein said at least a portion of the silk fibroin particles retain at least about 60% of their original volume within the tissue for at least about 6 weeks.
    • 5. The method of paragraph 4, wherein said at least a portion of the silk fibroin particles retain at least about 70% of their original volume within the tissue for at least about 6 weeks.
    • 6. The method of paragraph 5, wherein said at least a portion of the silk fibroin particles retain at least about 80% of their original volume within the tissue for at least about 6 weeks.
    • 7. The method of any of paragraphs 1-6, wherein said at least a portion of the silk fibroin particles retain at least about 70% of their original volume within the tissue for at least 3 months.
    • 8. The method of any of paragraphs 1-7, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 50% of their original volume in at least about 6 weeks.
    • 9. The method of paragraph 8, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 50% of their original volume in at least about 3 months.
    • 10. The method of any of paragraphs 1-9, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 30% of their original volume in at least about 6 weeks.
    • 11. The method of paragraph 10, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 10% of their original volume in at least about 6 weeks.
    • 12. The method of any of paragraphs 1-11, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 30% of their original volume in at least about 3 months.
    • 13. The method of any of paragraphs 1-12, wherein the silk fibroin particles are porous.
    • 14. The method of paragraph 13, wherein the porous silk fibroin particles have a porosity of at least about 1%, at least about 5%, at least or 10%, at least about 15%, or at least about 30%.
    • 15. The method of paragraph 14, wherein the porous silk fibroin particles have a porosity of at least about 50%.
    • 16. The method of paragraph 15, wherein the porous silk fibroin particles have a porosity of at least about 70%.
    • 17. The method of any of paragraphs 13-16, wherein the pores have a size of about 10 nm to about 1000 μm.
    • 18. The method of paragraph 17, wherein the pores have a size of about 1 μm to about 1000 μm.
    • 19. The method of any of paragraphs 1-18, wherein the silk fibroin particles have a size of about 500 nm to about 5000 μm.
    • 20. The method of paragraph 19, wherein the silk fibroin particles have a size of about 1 μm to about 2000 μm.
    • 21. The method of paragraph 20, wherein the silk fibroin particles have a size of about 10 μm to about 1500 μm.
    • 22. The method of any of paragraphs 1-21, wherein the porous silk fibroin particles are reduced from a solid-state porous silk fibroin by a mechanical means.
    • 23. The method of paragraph 22, wherein the mechanical means is selected from the group consisting of micronizing, milling, pulverizing, crushing, grinding, cutting, and any combinations thereof.
    • 24. The method of any of paragraphs 22-23, wherein the solid-state porous silk fibroin is formed by a porogen-leaching method.
    • 25. The method of any of paragraphs 1-24, wherein the composition or the silk fibroin particles further comprise(s) at least one active agent.
    • 26. The method of paragraph 25, wherein the at least one active agent is a biologically active agent, a cosmetically active agent, a cell attachment agent, a contrast agent, or any combinations thereof.
    • 27. The method of paragraph 26, wherein the biologically active agent is selected from the group consisting of a drug, a therapeutic agent, an anesthetic, a cell growth factor, a peptide, a peptidomimetic, an antibody or a portion thereof, an antibody-like molecule, nucleic acid, a polysaccharide, and any combinations thereof.
    • 28. The method of paragraph 26, wherein the cell attachment agent is selected from the group consisting of hyaluronic acid, collagen, crosslinked hyaluronic acid/collagen, an integrin-binding molecule, chitosan, elastin, fibronectin, vitronectin, laminin, proteoglycans, any derivatives thereof, and any combinations thereof.
    • 29. The method of paragraph 26, wherein the cosmetically active agent is selected from the group consisting of an anti-aging agent, an anti-free radical agent, an anti-oxidant, a hydrating agent, a whitening agent, a colorant, a depigmenting agent, a sun-blocking agent, a muscle relaxant, and any combinations thereof.
    • 30. The method of any of paragraphs 1-29, wherein the composition further comprises a cell.
    • 31. The method of paragraph 30, wherein the cell is a stem cell.
    • 32. The method of any of paragraphs 1-31, wherein the composition further comprises a biological fluid or concentrate.
    • 33. The method of paragraph 32, wherein the biological fluid or concentrate is lipoaspirate, bone marrow aspirate, or any combinations thereof.
    • 34. The method of paragraph 32, wherein the biological fluid or concentrate is lipoaspirate.
    • 35. The method of any of paragraphs 32-34, wherein the volume ratio of the silk fibroin particles to the biological fluid or concentrate ranges from about 1:19 to about 12:19.
    • 36. The method of paragraphs 32-34, wherein the volume ratio of the silk fibroin particles to the biological fluid or concentrate ranges from about 3:19 to about 6:19.
    • 37. The method of any of paragraphs 1-36, wherein the composition or the silk fibroin particles further comprise(s) a hydrogel.
    • 38. The method of any of paragraphs 1-37, wherein the composition or the silk fibroin particles further comprise(s) a dermal filler material.
    • 39. The method of paragraph 38, wherein the dermal filler material is selected from the group consisting of poly(methyl methacrylate) microspheres, hydroxylapatite, poly(L-lactic acid), hyaluronic acid, collagen, and any combinations thereof.
    • 40. The method of any of paragraphs 1-39, wherein the composition further comprises a pharmaceutically-acceptable carrier.
    • 41. The method of any of paragraphs 1-40, wherein the injection is performed subcutaneously, submuscularly, or intramuscularly.
    • 42. The method of any of paragraphs 1-41, wherein the injection is performed with a needle with a gauge of about 25-26.
    • 43. The method of any of paragraphs 142, wherein the composition is at least partially dry when injected in the tissue.
    • 44. The method of any of paragraphs 1-43, wherein the composition is at least partially hydrated when injected in the tissue.
    • 45. The method of any of paragraphs 1-44, wherein the tissue is a soft tissue.
    • 46. The method of paragraph 45, wherein the soft tissue is selected from the group consisting of a tendon, a ligament, skin, a breast tissue, a fibrous tissue, a connective tissue, a muscle, and any combinations thereof.
    • 47. The method of paragraph 46, wherein the soft tissue is skin.
    • 48. The method of paragraph 46, wherein the soft tissue is a breast tissue.
    • 49. The method of any of paragraphs 1-48, wherein the subject is a mammalian subject.
    • 50. The method of paragraph 49, wherein the mammalian subject is a human.
    • 51. The method of any of paragraphs 1-50, wherein the composition is stored or transported dried.
    • 52. The method of paragraph 51, wherein the composition is stored or transported at a temperature between about 0° C. and about 60° C.
    • 53. The method of paragraph 52, wherein the composition is stored or transported at a temperature between about 10° C. and about 60° C.
    • 54. The method of paragraph 53, wherein the composition is stored or transported at a temperature between about 15° C. and about 60° C.
    • 55. The method of any of paragraphs 1-54, wherein the silk fibroin particles exclude an amphiphilic peptide.
    • 56. The method of paragraph 55, wherein the amphiphilic peptide comprises a RGD motif.
    • 57. An injectable composition for use in repairing or augmenting a tissue in a subject, comprising a plurality of silk fibroin particles, wherein at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue to be repaired or augmented for at least about 6 weeks.
    • 58. The composition of paragraph 57, wherein the silk fibroin particles exclude an amphiphilic peptide.
    • 59. The composition of paragraph 58, wherein the amphiphilic peptide comprises a RGD motif.
    • 60. The composition of any of paragraphs 57-59, wherein said at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue for at least about 3 months.
    • 61. The composition of any of paragraphs 57-60, wherein said at least a portion of the silk fibroin particles retain at least about 50% of their original volume within the tissue for at least about 6 months.
    • 62. The composition of any of paragraphs 57-61, wherein said at least a portion of the silk fibroin particles retain at least about 60% of their original volume within the tissue for at least about 6 weeks.
    • 63. The composition of paragraph 62, wherein said at least a portion of the silk fibroin particles retain at least about 70% of their original volume within the tissue for at least about 6 weeks.
    • 64. The composition of paragraph 63, wherein said at least a portion of the silk fibroin particles retain at least about 80% of their original volume within the tissue for at least about 6 weeks.
    • 65. The composition of any of paragraphs 57-64, wherein said at least a portion of the silk fibroin particles retain at least about 70% of their original volume within the tissue for at least 3 months.
    • 66. The composition of any of paragraphs 57-65, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 50% of their original volume in at least about 6 weeks.
    • 67. The composition of any of paragraphs 57-66, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 50% of their original volume in at least about 3 months.
    • 68. The composition of any of paragraphs 57-67, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 30% of their original volume in at least about 6 weeks.
    • 69. The composition of any of paragraphs 57-68, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 10% of their original volume in at least about 6 weeks.
    • 70. The composition of any of paragraphs 57-69, wherein said at least a portion of the silk fibroin particles are adapted to degrade no more than 30% of their original volume in at least about 3 months.
    • 71. The composition of any of paragraphs 57-70, wherein the silk fibroin particles are porous.
    • 72. The composition of paragraph 71, wherein the porous silk fibroin particles have a porosity of at least about 1%, at least about 5%, at least about 10%, at least about 15%, or at least about 30%.
    • 73. The composition of paragraph 72, wherein the porous silk fibroin particles have a porosity of at least about 50%.
    • 74. The composition of paragraph 73, wherein the porous silk fibroin particles have a porosity of at least about 70%.
    • 75. The composition of any of paragraphs 71-74, wherein the pores have a size of about 10 nm to about 1000 μm.
    • 76. The composition of paragraph 75, wherein the pores have a size of about 1 μm to about 1000 μm.
    • 77. The composition of any of paragraphs 57-76, wherein the silk fibroin particles have a size of about 500 nm to about 5000 μm.
    • 78. The composition of paragraph 77, wherein the silk fibroin particles have a size of about 1 μm to about 2000 μm.
    • 79. The composition of paragraph 78, wherein the silk fibroin particles have a size of about 10 μm to about 1500 μm.
    • 80. The composition of any of paragraphs 71-79, wherein the porous silk fibroin particles are reduced from a solid-state porous silk fibroin by a mechanical means.
    • 81. The composition of paragraph 80, wherein the mechanical means is selected from the group consisting of micronizing, milling, pulverizing, crushing, grinding, cutting, and any combinations thereof.
    • 82. The composition of paragraph 80 or 81, wherein the solid-state porous silk fibroin porous is formed by a porogen-leaching method.
    • 83. The composition of any of paragraphs 57-82, wherein the injectable composition or the silk fibroin particles further comprise(s) at least one active agent.
    • 84. The composition of paragraph 83, wherein the at least one active agent is a biologically active agent, a cosmetically active agent, a cell attachment agent, a contrast agent, or any combinations thereof.
    • 85. The composition of paragraph 84, wherein the biologically active agent is selected from the group consisting of a therapeutic agent, an anesthetic, a cell growth factor, a peptide, a peptidomimetic, an antibody or a portion thereof, an antibody-like molecule, nucleic acid, a polysaccharide, and any combinations thereof.
    • 86. The composition of paragraph 84, wherein the cell attachment agent is selected from the group consisting of hyaluronic acid, collagen, crosslinked hyaluronic acid/collagen, an integrin-binding molecule, chitosan, elastin, fibronectin, vitronectin, laminin, proteoglycans, any derivatives thereof, and any combinations thereof.
    • 87. The composition of paragraph 84, wherein the cosmetically active agent is selected from the group consisting of an anti-aging agent, an anti-free radical agent, an anti-oxidant, a hydrating agent, a whitening agent, a colorant, a depigmenting agent, a sun-blocking agent, a muscle relaxant, and any combinations thereof.
    • 88. The composition of any of paragraphs 57-87, further comprising a cell.
    • 89. The composition of paragraph 88, wherein the cell is a stem cell.
    • 90. The composition of any of paragraphs 57-89, further comprising a biological fluid or concentrate.
    • 91. The composition of paragraph 90, wherein the biological fluid or concentrate is lipoaspirate, bone marrow aspirate, or any combinations thereof.
    • 92. The composition of paragraph 90, wherein the biological fluid or concentrate is lipoaspirate.
    • 93. The composition of any of paragraphs 90-92, wherein the volume ratio of the silk fibroin particles to the biological fluid or concentrate ranges from about 3:19 to about 6:19.
    • 94. The composition of any of paragraphs 57-93, wherein the injectable composition or the silk fibroin particles further comprise(s) a hydrogel.
    • 95. The composition of any of paragraphs 57-94, wherein the injectable composition or the silk fibroin particles further comprise(s) a dermal filler material.
    • 96. The composition of paragraph 95, wherein the dermal filler material is selected from the group consisting of poly(methyl methacrylate) microspheres, hydroxylapatite, poly(L-lactic acid), hyaluronic acid, collagen, and any combinations thereof.
    • 97. The composition of any of paragraphs 57-96, wherein the injectable composition further comprises a pharmaceutically-acceptable carrier.
    • 98. The composition of any of paragraphs 57-97, wherein the injectable composition is stored or transported dried.
    • 99. The composition of paragraph 98, wherein the injectable composition is stored or transported at a temperature between about 0° C. and about 60° C.
    • 100. The composition of paragraph 99, wherein the injectable composition is stored or transported at a temperature between about 10° C. and about 60° C.
    • 101. The composition of paragraph 100, wherein the injectable composition is stored or transported at a temperature between about 15° C. and about 60° C.
    • 102. A delivery device comprising an injectable composition of any of paragraphs 57-101.
    • 103. The delivery device of paragraph 102, further comprising a syringe.
    • 104. The delivery device of paragraph 103, wherein the syringe further comprises a needle.
    • 105. The delivery device of any of paragraphs 102-104, further comprising a catheter.
    • 106. The delivery device of any of paragraphs 102-105, further comprising an injection carrier.
    • 107. A syringe comprising an injectable composition of any of paragraphs 57-101.
    • 108. The syringe of paragraph 107, further comprising a needle.
    • 109. The syringe of paragraph 107 or 108, further comprising a catheter.
    • 110. The syringe of any of paragraphs 107-109, further comprising an injection carrier.

Some Selected Definitions of Terms

As used herein, a “subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. In certain embodiments of the aspects described herein, the subject is a mammal, e.g., a primate, e.g., a human. A subject can be male or female. Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of tissue repair, regeneration and/or reconstruction. In addition, the methods and compositions described herein can be used to treat domesticated animals and/or pets.

The term “statistically significant” or “significantly” refers to statistical significance and generally means a two standard deviation (2SD) below or above a reference level. The term refers to statistical evidence that there is a difference. It is defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true. The decision is often made using the p-value.

As used herein, the terms “proteins” and “peptides” ar used interchangeably herein to designate a series of amino acid residues connected to the other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues. The terms “protein”, and “peptide”, which are used interchangeably herein, refer to a polymer of protein amino acids, including modified amino acids (e.g., phosphorylated, glycated, etc.) and amino acid analogs, regardless of its size or function. Although “protein” is often used in reference to relatively large polypeptides, and “peptide” is often used in reference to small polypeptides, usage of these terms in the art overlaps and varies. The term “peptide” as used herein refers to peptides, polypeptides, proteins and fragments of proteins, unless otherwise noted. The terms “protein” and “peptide” are used interchangeably herein when referring to a gene product and fragments thereof. Thus, exemplary peptides or proteins include gene products, naturally occurring proteins, homologs, orthologs, paralogs, fragments and other equivalents, variants, fragments, and analogs of the foregoing.

The term “nucleic acids” used herein refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA), polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogs of natural nucleotides, which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences, as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer, et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka, et al., J. Biol. Chem. 260:2605-2608 (1985), and Rossolini, et al., Mol. Cell. Probes 8:91-98 (1994)). The term “nucleic acid” should also be understood to include, as equivalents, derivatives, variants and analogs of either RNA or DNA made from nucleotide analogs, and, single (sense or antisense) and double-stranded polynucleotides.

The term “short interfering RNA” (siRNA), also referred to herein as “small interfering RNA” is defined as an agent which functions to inhibit expression of a target gene, e.g., by RNAi. An siRNA can be chemically synthesized, it can be produced by in vitro transcription, or it can be produced within a host cell. siRNA molecules can also be generated by cleavage of double stranded RNA, where one strand is identical to the message to be inactivated. The term “siRNA” refers to small inhibitory RNA duplexes that induce the RNA interference (RNAi) pathway. These molecules can vary in length (generally 18-30 base pairs) and contain varying degrees of complementarity to their target mRNA in the antisense strand. Some, but not all, siRNA have unpaired overhanging bases on the 5′ or 3′ end of the sense 60 strand and/or the antisense strand. The term “siRNA” includes duplexes of two separate strands, as well as single strands that can form hairpin structures comprising a duplex region.

The term “shRNA” as used herein refers to short hairpin RNA which functions as RNAi and/or siRNA species but differs in that shRNA species are double stranded hairpin-like structure for increased stability. The term “RNAi” as used herein refers to interfering RNA, or RNA interference molecules are nucleic acid molecules or analogues thereof for example RNA-based molecules that inhibit gene expression. RNAi refers to a means of selective post-transcriptional gene silencing. RNAi can result in the destruction of specific mRNA, or prevents the processing or translation of RNA, such as mRNA.

The term “enzymes” as used here refers to a protein molecule that catalyzes chemical reactions of other substances without it being destroyed or substantially altered upon completion of the reactions. The term can include naturally occurring enzymes and bioengineered enzymes or mixtures thereof. Examples of enzyme families include kinases, dehydrogenases, oxidoreductases, GTPases, carboxyl transferases, acyl transferases, decarboxylases, transaminases, racemases, methyl transferases, formyl transferases, and α-ketodecarboxylases.

As used herein, the term “aptamers” means a single-stranded, partially single-stranded, partially double-stranded or double-stranded nucleotide sequence capable of specifically recognizing a selected non-oligonucleotide molecule or group of molecules. In some embodiments, the aptamer recognizes the non-oligonucleotide molecule or group of molecules by a mechanism other than Watson-Crick base pairing or triplex formation. Aptamers can include, without limitation, defined sequence segments and sequences comprising nucleotides, ribonucleotides, deoxyribonucleotides, nucleotide analogs, modified nucleotides and nucleotides comprising backbone modifications, branchpoints and nonnucleotide residues, groups or bridges. Methods for selecting aptamers for binding to a molecule are widely known in the art and easily accessible to one of ordinary skill in the art.

As used herein, the term “antibody” or “antibodies” refers to an intact immunoglobulin or to a monoclonal or polyclonal antigen-binding fragment with the Fc (crystallizable fragment) region or FcRn binding fragment of the Fc region. The term “antibodies” also includes “antibody-like molecules”, such as fragments of the antibodies, e.g., antigen-binding fragments. Antigen-binding fragments can be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. “Antigen-binding fragments” include, inter alia, Fab, Fab′, F(ab′)2, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), single domain antibodies, chimeric antibodies, diabodies, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide. Linear antibodies are also included for the purposes described herein. The terms Fab, Fc, pFc′, F(ab′) 2 and Fv are employed with standard immunological meanings (Klein, Immunology (John Wiley, New York, N.Y., 1982); Clark, W. R. (1986) The Experimental Foundations of Modern Immunology (Wiley & Sons, Inc., New York); and Roitt, I. (1991) Essential Immunology, 7th Ed., (Blackwell Scientific Publications, Oxford)). Antibodies or antigen-binding fragments specific for various antigens are available commercially from vendors such as R&D Systems, BD Biosciences, e-Biosciences and Miltenyi, or can be raised against these cell-surface markers by methods known to those skilled in the art.

As used herein, the term “Complementarity Determining Regions” (CDRs; i.e., CDR1, CDR2, and CDR3) refers to the amino acid residues of an antibody variable domain the presence of which are necessary for antigen binding. Each variable domain typically has three CDR regions identified as CDR1, CDR2 and CDR3. Each complementarity determining region may comprise amino acid residues from a “complementarity determining region” as defined by Kabat (i.e. about residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a “hypervariable loop” (i.e. about residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). In some instances, a complementarity determining region can include amino acids from both a CDR region defined according to Kabat and a hypervariable loop.

The expression “linear antibodies” refers to the antibodies described in Zapata et al., Protein Eng., 8(10):1057-1062 (1995). Briefly, these antibodies comprise a pair of tandem Fd segments (VH1-CH1-VH-CH1) which, together with complementary light chain polypeptides, form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.

The expression “single-chain Fv” or “scFv” antibody fragments, as used herein, is intended to mean antibody fragments that comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. (Plückthun, The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994)).

The term “diabodies,” as used herein, refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) Connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. (EP 404,097; WO 93/11161; Hollinger et ah, Proc. Natl. Acad. Sd. USA, P0:6444-6448 (1993)).

As used herein, the term “small molecules” refers to natural or synthetic molecules including, but not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, aptamers, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e., including heteroorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds.

The term “antibiotics” is used herein to describe a compound or composition which decreases the viability of a microorganism, or which inhibits the growth or reproduction of a microorganism. As used in this disclosure, an antibiotic is further intended to include an antimicrobial, bacteriostatic, or bactericidal agent. Exemplary antibiotics include, but are not limited to, penicillins, cephalosporins, penems, carbapenems, monobactams, aminoglycosides, sulfonamides, macrolides, tetracyclins, lincosides, quinolones, chloramphenicol, vancomycin, metronidazole, rifampin, isoniazid, spectinomycin, trimethoprim, and sulfamethoxazole.

The term “therapeutic agents” is art-recognized and refers to any chemical moiety that is a biologically, physiologically, or pharmacologically active substance that acts locally or systemically in a subject. Examples of therapeutic agents, also referred to as “drugs”, are described in well-known literature references such as the Merck Index, the Physicians Desk Reference, and The Pharmacological Basis of Therapeutics, and they include, without limitation, medicaments; vitamins; mineral supplements; substances used for the treatment, prevention, diagnosis, cure or mitigation of a disease or illness; substances which affect the structure or function of the body; or pro-drugs, which become biologically active or more active after they have been placed in a physiological environment. Various forms of a therapeutic agent may be used which are capable of being released from the subject composition into adjacent tissues or fluids upon administration to a subject. Examples include steroids and esters of steroids (e.g., estrogen, progesterone, testosterone, androsterone, cholesterol, norethindrone, digoxigenin, cholic acid, deoxycholic acid, and chenodeoxycholic acid), boron-containing compounds (e.g., carborane), chemotherapeutic nucleotides, drugs (e.g., antibiotics, antivirals, antifungals), enediynes (e.g., calicheamicins, esperamicins, dynemicin, neocarzinostatin chromophore, and kedarcidin chromophore), heavy metal complexes (e.g., cisplatin), hormone antagonists (e.g., tamoxifen), non-specific (non-antibody) proteins (e.g., sugar oligomers), oligonucleotides (e.g., antisense oligonucleotides that bind to a target nucleic acid sequence (e.g., mRNA sequence)), peptides, proteins, antibodies, photodynamic agents (e.g., rhodamine 123), radionuclides (e.g., I-131, Re-186, Re-188, Y-90, Bi-212, At-211, Sr-89, Ho-166, Sm-153, Cu-67 and Cu-64), toxins (e.g., ricin), and transcription-based pharmaceuticals.

As used herein, the term “hormones” generally refers to naturally or non-naturally occurring hormones, analogues and mimics thereof. In certain embodiments, the term “hormones” refers to any hormones used in therapeutic treatment, e.g., growth hormone treatment. As used herein, “growth hormone” or “GH” refers to growth hormone in native-sequence or in variant form, and from any source, whether natural, synthetic, or recombinant. Examples include human growth hormone (hGH), which is natural or recombinant GH with the human native sequence (somatotropin or somatropin), and recombinant growth hormone (rGH), which refers to any GH or variant produced by means of recombinant DNA technology, including somatrem, somatotropin, and somatropin. In one embodiment, hormones include insulin.

As used herein, a “contrast agent” can be any chemical moiety that is used to increase the degree of difference between the lightest and darkest part of a scan or an imaging, e.g., during medical scan or imaging, relative to a scan performed without the use of a contrast agent. For example, contrast agents can include imaging agents containing radioisotopes such as indium or technetium; dyes containing iodine, gadolinium or cyanine; enzymes such as horse radish peroxidase, GFP, alkaline phosphatase, or β-galactosidase; fluorescent substances such as europium derivatives: luminescent substances such as N-methylacrydium derivatives or the like. In some embodiments, contrast agents can include gold nanoparticles and/or quantum dots.

As used herein, the term “substantially” means a proportion of at least about 60%, or preferably at least about 70% or at least about 80%, or at least about 90%, at least about 95%, at least about 97% or at least about 99% or more, or any integer between 70% and 100%. In some embodiments, the term “substantially” means a proportion of at least about 90%, at least about 95%, at least about 98%, at least about 99% or more, or any integer between 90% and 100%. In some embodiments, the term “substantially” can include 100%.

As used herein, the term “comprising” means that other elements can also be present in addition to the defined elements presented. The use of “comprising” indicates inclusion rather than limitation.

The term “consisting of” refers to the components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.

As used herein the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the invention

Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term “about.” The term “about” when used in connection with percentages may mean±1%.

The singular terms “a,” “an,” and “the” include plural referents unless context clearly indicates otherwise. Similarly, the word “or” is intended to include “and” unless the context clearly indicates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The abbreviation, “e.g.” is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation “e.g.” is synonymous with the term “for example.”

Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Definitions of common terms in diseases and disorders, separation and detection techniques can be found in The Merck Manual of Diagnosis and Therapy, 18th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-18-2); Robert S. Porter et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8).

It should be understood that this invention is not limited to the particular methodology, protocols, and reagents, etc., described herein and as such may vary. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which is defined solely by the claims.

All patents and other publications identified throughout the specification are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents are based on the information available to the applicants and do not constitute any admission as to the correctness of the dates or contents of these documents.

Some embodiments described herein are further illustrated by the following example which should not be construed as limiting.

The contents of all references cited throughout this application, examples, as well as the figures and tables are incorporated herein by reference in their entirety.

EXAMPLES Example 1. Comparison of Injectable Silk Fibroin Scaffold Particles with Other Silk Fibroin Scaffolds or Hydrogels Exemplary Materials and Methods

Materials: All chemicals and solvents used in the Examples for silk scaffold preparation were purchased from Sigma Aldrich (St. Louis, Mo.) unless otherwise noted. Sterile water and saline were purchased from Invitrogen (Carlsbad, Calif.). It should be noted that equivalent materials can be used and purchased from other commercial vendors.

Preparation of silk fibroin solutions: Bombyx mori silkworm cocoons were purchased from Tajimia Shoji Co. (Yokohama, Japan). Coccons were cut into pieces, and boiled in 0.02 M Na2CO1 for about 10-60 minutes, and preferably for about 30 minutes. The resulting silk fibroin fibers were rinsed in distilled water and let dried. The dried silk fibroin fibers were re-solubilized in 9.3 M LiBr at 60° C., for about 1-4 hours, until dissolved. The silk fibroin solution was dialyzed, with a molecular weight cutoff of 3500 Daltons, against distilled water for at least 6 water changes. The aqueous silk fibroin solution was lyophilized till dry and then dissolved in hexa-fluoro-iso-propanol (HFIP) to yield a 17% (w/v) solvent based silk fibroin solution.

Preparation of silk fibroin porous scaffolds: Silk fibroin porous scaffolds were formed from either aqueous or solvent based silk fibroin solutions (e.g., 6%-20% w/v). Porogens (e.g., salts such as sodium chloride NaCl) with a size ranging from 100 microns to 1.2 millimeters were used. Porogens were packed into a Teflon coated container, and silk fibroin solution was poured atop the salt. The container was covered and allowed to sit at room temperature for about 1-3 days. The container was then left uncovered for about 1 day. The container was placed into distilled water to leach out the salt. Alternatively, the scaffold can be placed in a methanol bath or water-annealed for about 1 hour to about 1 day to further induce beta-sheet formation. In some embodiments, the silk fibroin porous scaffolds can be further coated with extracellular matrix molecules, such as laminin, to facilitate cell attachment. In some embodiments, the silk fibroin porous scaffolds can be coated with no extracellular matrix molecules.

Preparation of injectable silk fibroin porous scaffolds: Aqueous or solvent based silk fibroin porous scaffolds were chopped manually or by mechanical means, for example, with a rotating blade, such as the ones in conventional food processors. Chopped scaffolds were passed through sieves of various openings to obtain the desired range of sizes. The chopped scaffolds were allowed to dry and then autoclaved to sterilize. The dry, autoclaved, chopped scaffolds (termed as “silk fibroin scaffold particles” below) are stored at room temperature until use.

Preparation of injectable silk fibroin hydrogels (“silk fibroin vortexed gels”): Aqueous silk fibroin solution (e.g., ˜4% w/v) was first sterilized by being passed through a 0.22 micron filter unit. The sterile silk fibroin solution was then concentrated, for example, to ˜8%, ˜10%, and ˜12% w/v solutions, using centrifugal filter units (Amicon Ultra-15 Centrifugal Filter Unit, Millipore, Billerica, Mass.) according to manufacturers' protocols. To form the injectable silk fibroin hydrogels, a vortexing method was employed (Yucel et al, 2009. Biophys J. 97: 2044). Briefly, the aqueous silk fibroin solutions were vortexed (Vortex-Genie 2, Fisher Scientific, Pittsburgh, Pa.) with varying power and time, depending on the sample volume and concentration, until the clear solution became turbid. The turbid solution was placed in ˜37° C. for about 30 minutes to further induce gelation. After 30 minutes, the hydrogel alone or mixed with lipoaspirate was loaded into a syringe and injected.

Injection methods: Silk fibroin scaffolds were injected via different methods, e.g., subcutaneously, intramuscularly, or submuscularly, in a variety of preparations. Exemplary preparations include, but are not limited to, dry state, dry state mixed with lipoaspirate, hydrated state (e.g., in sterile water or normal saline), or hydrated state mixed with lipoaspirate.

In vivo injections: A female nude rat model was used for assessing the silk fibroin scaffolds and hydrogels described herein. Other mammalian models (e.g., mouse, rabbit, canine, or porcine models) can also be used depending on the applications of the injectable silk fibroin scaffolds and the tissues to be modeled for treatment. Six month old rats were weighed and anesthetized with isoflurane in oxygen prior to injection. A total of about 1 ml was used per injection. Briefly, dry silk fibroin scaffold particles were immersed in saline or lipoaspirate immediately before loading into a syringe. The filled syringe was attached to a cannula no larger than 2 mm inner diameter. Subcutaneous injections were performed above the pectoral muscles. Intramuscular and submuscular injections were performed between the pectoralis major and pectoralis minor muscles or underneath the pectoral muscles, respectively. A fanning subcutaneous injection method was performed in the dorsus of the rat. See, for example, FIG. 1. Injected samples were explanted, and evaluated for weight, volume retention and histological outcomes after 1, 2, 10, 30 days. Volume retention was performed by 2 methods, e.g., scale measurements and volume displacement. In some embodiments, whole silk fibroin porous scaffolds (5 mm diameter×2 mm height) were implanted in the same locations for comparison.

Histology: Explants were cut in half and fixed in 10% formalin overnight at 4° C. One group was embedded in OCT freezing media, cryosectioned into 10 micron sections, and stained with Oil Red O. The remaining half were placed through a series of dehydration steps, embedded in paraffin and cut in 10 micron sections. Three continuous sections were placed on each slide, and stained according to standard histological methods for hematoxylin and eosin (H&E), or processed for immunohistochemistry. After antigen retrieval, the sections were incubated with primary anti-rat CD31, CD68, CD80, CD163, anti-human nucleus antibodies for about 1 hour. The sections were then incubated with secondary biotinylated antibodies raised in the species of the primary antibodies for about 1 hour. The Vector Labs ABC antibody detection kit was used along with a DAB substrate to enhance colorimetric expression.

Results

In some embodiments, silk fibroin vortexed gels, alone or mixed with lipoaspirate, can be resorbed faster than lipoaspirate alone in a 6-week study. Accordingly, some embodiments of the silk fibroin vortexed gels may not be ideal for applications, which require size and/or shape retention for an extended time such as 6 weeks or longer. However, in some embodiments, the volume retention properties of the silk fibroin vortexed gel can be adjusted, for example, by modulating the silk fibroin concentration, and/or vortexing or shearing rate of the silk fibroin solution.

Implanted silk fibroin porous scaffolds can retain their shape and size over an extended period of time. However, the implanted silk fibroin porous scaffolds generally require one or more incisions to be placed. In addition, the implanted silk fibroin porous scaffolds are generally cast to fill a particular anatomical void prior to surgery, and/or they cannot be molded during surgery to fill an irregular shaped void.

Unlike silk fibroin vortexed gels or implanted silk fibroin porous scaffolds, an injectable silk fibroin porous scaffold (e.g., silk fibroin particles described herein) can serve as a therapy that is both minimally invasive and capable of sustaining its size and shape for at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 6 months, or at least about 1 year or longer, while still being an off-the-shelf product. While the injectable silk fibroin scaffolds (e.g., silk fibroin particles described herein) can reduce the administration to a minimally invasive procedure, they can also allow the surgeon to flexibly mold the injectable silk fibroin scaffolds (e.g., in form of particles) into any shape or size defect.

As shown in Table 1, different combinations of various parameters related to scaffold parameters and/or injection methods were used to assess the in vivo application of injectable silk fibroin scaffolds.

TABLE 1 Various combinations of exemplary parameters related to injectable scaffold particle properties and injection methods Pore size of a Particle silk fibroin Injection Hydration Time Diameter (μm) scaffold (μm) carrier state Injection site points Submicron 300-500  No carrier Dry Subcutaneous 1 day 1-5 850-1000 Saline Hydrated Intramuscular 2 day  5-20 Lipoaspirate (saline) Submuscular 10 day 20-50 14 day  50-100 1 month 100-250 500-750  750-1000 1000-2000

In some embodiments, two different methods of processing silk fibroin were employed to alter the degradation rate of silk fibroin. It has been previously shown that silk fibroin porous scaffold implants in a subcutaneous rat model degrade within 3-6 months when prepared with the aqueous method, but remain for at least up to 2 years when prepared with solvent based method (Wang et al. 2008. Biomaterials. 29: 3415). Accordingly, either method can be used to prepare injectable silk fibroin scaffolds, depending on desired properties of the scaffolds, applications and/or tissues to be treated. In some embodiments, aqueous methods for preparing injectable silk fibroin scaffolds can be used. In other embodiments, solvent based methods for preparing injectable silk fibroin scaffolds can be used. In this Example, injectable silk fibroin scaffolds prepared by both aqueous and solvent base methods were studied to evaluate integration after the silk fibroin material is completely gone and long-term integration of the material. Early time points (e.g., 1-2 days) can be used to evaluate signs of acute inflammation, while later time points are evaluated for overall tissue integration, vascularity, volume retention, and scaffold degradation.

Another variable to be assessed was pore size range. Smaller pores, 300-500 microns were compared to larger pores, 850-1000 microns. Further, various diameters of the injectable silk fibroin scaffold particles, as shown in Table 1, were assessed. In some embodiments, injectable silk fibroin scaffold particles smaller than several microns can also be produced. In some embodiments, as the silk fibroin scaffold particles are degrading, they do not produce an adverse inflammatory response due to their size. As shown in Table 1, the silk fibroin scaffold particles can be dry or hydrated. One of the advantages of using dry silk fibroin scaffold particles is that they can be used off the shelf. Another benefit of the dry particles is their ability to infuse into the scaffold particles while eliminating the need to pre-hydrate.

The different injection locations, as shown in Table 1, are meant to represent examples of clinical injection sites for a variety of applications, but they should not be construed as limiting. For example, subcutaneous injections can be representative of the soft tissue defect fillers; subcutaneous, submuscular and intramuscular injections can be representative of breast reconstructions.

In some embodiments, the injectable silk fibroin scaffolds (e.g., unseeded or cell-seeded) can also increase vascularity in a tissue to be treated by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or higher, as compared to a non-treated tissue or a tissue treated with non-silk injectable compositions.

In some embodiments, the injectable silk fibroin scaffolds seeded with cells, e.g., ASCs or lipoaspirate, can also increase its integration with the host tissue by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or higher, as compared to a tissue treated with unseeded injectable silk fibroin scaffolds.

In some embodiments, the injectable silk fibroin scaffolds seeded with cells, e.g., ASCs or lipoaspirate, can increase adipose tissue regeneration by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or higher, as compared to a tissue treated with unseeded injectable silk fibroin scaffolds.

Example 2. In Vivo Studies of Injectable Silk Fibroin Porous Scaffolds Exemplary Materials and Methods

Preparation of injectable silk fibroin porous scaffolds: In particular embodiments, silk fibroin porous scaffolds were formed from solvent based silk solutions (17% w/v). Porogens can be used to create pores with the scaffolds. In certain embodiments, NaCl porogens with a size ranging from about 300-500 microns were used. In certain embodiments, NaCl porogens with a size ranging from about 850-1000 microns were used. NaCl porogens were packed into a Teflon coated container, and silk solution was poured atop the salt. The container was covered and allowed to sit at room temperature for 1-3 days. The container was then left uncovered for 1 day. The container was placed into distilled water to leach out the salt. In some embodiments, the silk resultant scaffolds were placed in a methanol bath for 1 day to further induce beta-sheet formation. Injectable silk fibroin porous scaffolds can then be produced using the methods described in Example 1, e.g., by reducing the silk fibroin porous scaffold into smaller pieces. See, for example, FIG. 2.

Preparation of processed lipoaspirate: Lipoaspirate from elective plastic surgery was obtained on the same day of scaffold injection. Lipoaspirate was transported aseptically at room temperature just after surgery. Approximately 30 ml of lipoaspirate was added to a 50 ml conical tube and centrifuged at room temperature at 1000 rpm for 10 minutes. The blood and free lipids were removed. The remaining tissue was placed in sterile Petri dishes. The injectable silk fibroin porous scaffolds were placed into the processed lipoaspirate for 1 hour prior to injection.

Injection methods: One ml of chopped silk fibroin porous scaffolds and lipoaspirate was drawn up into a 1 ml syringe. The final volume ratios of lipoaspirate to silk scaffolds could range from 19:3 (low dose) to 19:6 (high dose). In some embodiments, the injectable scaffold and lipoaspirate mixture was injected subcutaneously through a 24 gauge needle, in a hydrated state with lipoaspirate, on the back of an athymic mouse. See, e.g., FIG. 1.

Histology: Explant sample and constructs were processed according standard histology protocols. Formalin fixed samples were put through a series of dehydration solvents and finally paraffin using an automated tissue processor. Samples were embedded in paraffin, cut in 10 micron sections, and let to adhere on glass slides. The sections were rehydrated and stained with hematoxylin and eosin, and imaged.

Results

At 6 weeks post-injection, the mice were sacrificed and the injected materials were harvested. The explanted masses were measured for their weight and mass. No differences in relative weight change or mass change were found with silk fibroin particles produced from solid-state porous silk fibroin of varying porosities (e.g., 300-500 micron pores vs. 850-1000 micron pores) or relative ratios of silk fibroin scaffold to lipoaspirate (e.g., 3:19 vs 6:19) at such time point.

As shown in FIG. 3, the injected lipoaspirate (line arrows) was detected in all groups. Within the injected lipoaspirate, injectable silk fibroin scaffold pieces were detected (dotted arrows). The injected silk fibroin materials did not elicit a pro-inflammatory response; however macrophages were detected at the periphery of the injected mass.

Claims

1.-110. (canceled)

111. A method for repairing or augmenting a tissue in a subject, the method comprising:

injecting a composition in the tissue to be repaired or augmented, the composition comprising a plurality of silk fibroin particles and an injection carrier,
wherein at least a portion of the silk fibroin particles retain at least 50% of their original volume within the tissue for at least 6 weeks, and
wherein the silk fibroin particles have (i) a porosity of at least 70%, (ii) have a size from 1 μm to 2,000 μm, and (iii) a pore size from 10 nm to 1000 μm.

112. The method of claim 111, wherein the silk fibroin particles are fabricated by forming a solid-state silk fibroin from a silk solution consisting of 4% (w/v) to 20% (w/v) silk fibroin and water, and by reducing the solid-state silk fibroin into particles.

113. The method of claim 111, wherein the composition further comprises a hydrogel distributed within the silk fibroin particles.

114. The method of claim 111, wherein reducing the solid-state silk fibroin particles is performed by a mechanical means selected from the group consisting of micronizing, milling, pulverizing, crushing, grinding, cutting, and any combinations thereof.

115. The method of claim 111, wherein the solid-state silk fibroin formed by freeze-drying the silk solution.

116. The method of claim 111, wherein at least a portion of the silk fibroin particles retain at least 70% of their original volume within the tissue for at least 6 weeks.

117. The method of claim 111, wherein at least a portion of the silk fibroin particles retain at least 50% of their original volume within the tissue for at least 3 months.

118. The method of claim 111, wherein at least a portion of the silk fibroin particles are adapted to degrade no more than 50% of their original volume in at least 3 months.

119. The method of claim 111, wherein the silk fibroin is mixed with a biocompatible polymer to form mixed polymer particles, wherein the biocompatible polymer comprises polycaprolactone.

120. The method of claim 111, wherein the composition comprises at least one active agent.

121. The method of claim 120, wherein the active agent is an anesthetic.

122. The method of claim 111, wherein the composition comprises hyaluronic acid.

123. The method of claim 111, wherein the composition further comprises a dermal filler.

124. The method of claim 123, wherein the dermal filler is selected from the group consisting of poly(methyl methacrylate) microspheres, hydroxylapatite, poly(L-lactic acid), hyaluronic acid, collagen, and any combinations thereof.

125. The method of claim 111, wherein the injection is performed subcutaneously, submuscularly, or intramuscularly.

126. The method of claim 111, wherein the tissue is a soft tissue.

127. The method of claim 126, wherein the soft tissue is selected from the group consisting of a tendon, a ligament, skin, a breast tissue, a fibrous tissue, a connective tissue, a muscle, and any combinations thereof.

128. The method of claim 111, wherein the silk fibroin particles have a size from 10 μm to 1500 μm.

129. The method of claim 111, wherein the pores have a size from 1 μm to 1000 μm.

130. The method of claim 111, wherein the injection carrier comprises a saline solution.

Patent History
Publication number: 20210244850
Type: Application
Filed: Nov 6, 2020
Publication Date: Aug 12, 2021
Inventors: Evangelia Bellas (Philadelphia, PA), Kacey Marra (Canonsburg, PA), J. Peter Rubin (Pittsburgh, PA), David L. Kaplan (Concord, MA), James J. Yoo (Winston-Salem, NC)
Application Number: 17/091,835
Classifications
International Classification: A61L 27/22 (20060101); A61L 27/54 (20060101); A61L 27/36 (20060101); A61L 27/38 (20060101); A61L 27/48 (20060101); A61L 27/56 (20060101); A61L 27/58 (20060101);