Open access peer-reviewed chapter

Collagen-Based Biomaterial as Drug Delivery Module

Written By

Amit Kumar Verma

Submitted: 22 January 2022 Reviewed: 04 February 2022 Published: 04 July 2022

DOI: 10.5772/intechopen.103063

From the Edited Volume

Collagen Biomaterials

Edited by Nirmal Mazumder and Sanjiban Chakrabarty

Chapter metrics overview

345 Chapter Downloads

View Full Metrics

Abstract

In the field of medicine, controlled drug delivery has become a major challenge due to inefficiency of drug at critical parameters such as permeability, solubility, half-life, targeting ability, bio- & hemocompatibility, immunogenicity, off-target toxicity and biodegradability. Since several decades the role of drug delivery module has been a crucial parameter of research and clinical observations to improve the effectiveness of drugs. Biomaterials- natural or artificial are mainly used for medical application such as in therapeutics or in diagnostics. Among all the biomaterials, collagen based-hydrogels/ films/ composite materials have attracted the research and innovations and are the excellent objects for drug delivery, tissue engineering, wound dressings and gene therapeutics etc. due to high encapsulating capacity, mechanically strong swollen structural network and efficient mass transfer properties. Substantial developments have been performed using collagen-based drug delivery systems (DDS) to deliver biomolecules with better efficacy. In spite of significant progress, several issues at clinical trials particularly targeting of intracellular molecules such as genes is still a challenge for researchers. Experimental results, theoretical models, molecular simulations will boost the fabrication/designing of collage-based DDS, which further will enhance the understanding of controlled delivery/mechanism of therapeutics at specific targets for various disease treatments.

Keywords

  • collagen
  • biomaterial
  • drug delivery systems (DDS)
  • drug
  • hydrogels
  • films
  • composite material

1. Introduction

Collagen is unique and major structural protein of extracellular matrix (ECM) and plays crucial role to the structural integrity of tissues/organs and cellular growth in vertebrates and other organisms, constitute around 30% of the total protein content of mammal’s body, involved in mechanical protection of tissues and organs such as skin, tendons, ligaments, bones, cartilage, blood vessels, cornea and nails etc. (Figure 1) [1, 2, 3, 4, 5, 6]. More than 50% in the skin and more than 90% of extracellular proteins in the tendon and bone is made up of collagen [7, 8].

Figure 1.

Collagen’s occurrence in different body tissues.

The unique feature of collagen molecule is triple helix structure made by three identical or non-identical polypeptide chains. Each polypeptide chain comprises around 1000 amino acids and the chains are supercoiled in left handed manner around the axis with staggering of residues between adjacent chains, give rise to triple helix right handed structure [9]. Each chain is having the repeated sequence of (Gly-X-Y)n, whereas X and Y are mostly proline and hydroxyproline residues [1, 10, 11]. At present, 30 different types of collagens have been characterized and reported in literature [12]. Among all the different variants of collagen, type I, II, III, V and XI represent more than 90% of human fibrillar collagen and is majorly distributed in dermis, hair, bone, cartilage, ligament, tendon and placenta [13]. Other types such as type IV and VIII form the network frame of basement membranes [12, 14]. Type I collagen has very important role in medicine as well as in development of medical devices, artificial implants, drug carriers for controlled release and scaffolds for tissue regeneration [5, 15, 16]. Being the highly versatile, structurally unique and biocompatible protein substance, the collagen can be developed into different types of drug/active substance carrier module such as hydrogels, microparticles and films etc. [3, 7].

Advertisement

2. Collagen as a biomaterial

According to Hench and Erthridge, 1982 [17], “a biomaterial is used to make devices to replace a part or a function of the body in a safe, reliable, economic, and physiologically acceptable manner.” Raghavendra et al. [18] has mentioned the other definitions of biomaterial are “materials of synthetic as well as of natural origin in contact with tissue, blood, and biological fluids, intended for use for prosthetic, diagnostic, therapeutic, and storage applications without adversely affecting the living organism and its components” [19] and “any substance (other than drugs) or combination of substances, synthetic or natural in origin, which can be used for any period of time, as a whole or as a part of a system which treats, augments, or replaces any tissue, organ, or function of the body” [20]. Application of biomaterials in physiological systems is possible due to competent and stable features of biomaterials [21] which can be achieved with proper combination of mechanical, physical, chemical and biological attributes [22]. Modern biomaterials are designed and developed singly or in combinations of polymers, metals, composite materials and ceramics etc. [18].

In the field of medicine, collagen is one of the most studied biomaterial or biopolymer and according to Cheng et al. [23], around 260,000 literature articles (at present, the number is many more than reported) reported it as pivotal component in tissue regeneration and so called as ‘the steel of the biological material”. Collagen is the main biopolymer of ECM of vertebrates and invertebrates and has the capability to interact with large number of biomolecules leading to various biological reactions/changes under normal or pathological processes in the body, inspiring the scientists to develop the various formulations based on collagen [24, 25, 26]. The attributes for the testimony of collagen’s usage in wide scenario of medicine are its cosmopolitanism, high biocompatibility, hemocompatibility, biomimetic and biodegradability and to make composite biopolymer with biomaterials like chitosan (CHS), alginate (ALG), cellulose (CL), hyaluronic acid (HA), glycosaminoglycans (GAGs) as well as synthetic materials like carboxy methyl cellulose (CMC), poly vinyl alcohol (PVA), poly ε-caprolactone (PCL), poly ethyl methacrylate (PEMA) etc. in different formulations [27, 28, 29].

Now it is well established that collagen has good elasticity, physical, mechanical, enzymatic and thermal stability inside the body environment, but after extraction and utilization these properties are compromised at large scale [30, 31, 32], so the additional need of chemicals, chemical and physical processes are required to develop the stable biomaterial. Though extracted collagen is a promising drug delivery material in the field of ophthalmology but can be easily degradable in vitro due to disruption of natural intermolecular crosslinks of lysine and hydroxylysine residues during isolation and purification process [33]. The weaknesses of extracted collagen such as mechanical and thermal strength, enzymatic degradation can be reduced with the help of various methods of chemical, physical and enzymatic crosslinking, covalent conjugating, grafting polymerization or blending. The blended collagen-based biomaterials like hydrogels, films, microspheres and nanoparticles (NPs) have low immunogenicity, good absorption, hemostatic property and synergism with other bioactive compounds or loaded drugs and remain unaltered after several processes. The requirement of mechanical, pH, enzymatic, thermal stability is not provided alone by collagen for a controlled DDS but can be achieved with combination of CHS or ALG for release of analgesics, chemotherapeutic molecules and natural bioactive agent like curcumin or aloe vera. The blended biopolymer will have the combined desired properties of the separate material. Hydrogel of blended collagen-CHS have antibacterial, antifungal, anti-carcinogenic and immunogenic attributes [34, 35, 36, 37]. Blended collagen-CL based hydrogel has properties like biomimetic and hemostatic from collagen while mechanical strength and antibacterial characteristic from CL. In the field of ophthalmology blended collagen-based hydrogels with CHS or ALG are applied for corneal disease treatment exhibiting good mechanical and thermal attributes along with transparency. The films, microspheres, membranes and scaffolds of blended collagen-based materials are used in wound dressing due to moisture retaining, low adhesion, absorption of blood and tissue exudates, anti-infective and permeability properties [32, 38, 39, 40, 41]. The sources and possible applications of most utilized material of blended biopolymers along with important features like stability, toxicity and biomedical are presented in Table 1.

BiopolymersSourceStabilityToxicityBiomedical propertyApplicationsReferences
CollagenVertebrate protein (skin, bone, tendon, dentin, cornea, cartilage, vessels, intestine, uterus, dermis, placentaPoor mechanical, thermal, enzymatic, tensile rigidityInsignificantBiocompatible, biomimetic fibril forming, self-assembling, biodegradableFood, cosmetics, ophthalmology, inserts, shields, particles, gels, aqueous
injectables, drug delivery, Grafting, tissue engineering
[38, 40, 42, 43, 44]
AlginateBrown algae (Phaeophyceae family), Laminaria hyperborea, Laminaria digitata, Laminaria japonica, Ascophyllum nodosum & Macrocystis pyriferaPoor mechanical and chemicalLowBiocompatibiliy, mild gelationWound healing, delivery of bioactive agents like small drugs, proteins, tissue engineering, cell transplantation[45, 46, 47, 48]
ChitosanFrom chitin of insects, crustaceans, cephalopods
cephalopods
Mechanically strongInsignificantNontoxic, non-immunogenic, non-immunogenic, non-carcinogenic, biocompatible, bioabsorbable, antimicrobial, antifungal, anticoagulant, anti-tumor, hemostaticBiosensors, drug delivery, wound dressing[35, 49, 50, 51]
CelluloseWood, cotton, sugarbeet potato, tubers, onion, hemp, flax, wheat straw, mulberry
bark, algae, bacteria
Mechanically highInsignificantBiocompatible, biodegradable, biological affinity, antibacterialWound dressing, shields, dental implants, bone tissue grafting, artificial blood vessel[37, 52, 53, 54]

Table 1.

Natural and abundant biopolymers: Sources, properties, and potential applications.

Advertisement

3. Crosslinking for collagen-based biomaterial

Collagen is the most abundant vertebrate protein and mostly used biopolymer and with variety of physiological features like biocompatible, low immunogenic, self assembling fibril formation etc. The collagen is mechanically strong and durable in vivo but after the isolation and purification processes is vulnerable to degradation in vitro due to dissociation of natural crosslinks and assembly structure by neutral salt, acid alkali or proteases and quality of extracted collagen is inferior to native state (Figure 2) [42, 55]. The researchers are attempting to make the suitable collagen-based biomaterial with properties of increased mechanical strength, reduced enzymatic degradation, stability, solubility and low toxicity by introducing exogenous crosslinking [42, 56]. The introduced intermolecular crosslinking prevents the unknotting of collagen fibrils produced by heat and advancing the thermal stability along with increased tensile strength, stiffness, compressive modulus and decreased extensibility [57, 58, 59, 60]. The exogenous natural or chemical crosslinks could significantly reduce the enzymatic degradation of collagen by blocking the cleavage site [61].

Figure 2.

Native crosslinking in the collagen molecules.

Though collagen-based crosslinked biopolymers are producing significant results in the field of biomedicine and biotechnology, still no standard method is applicable to the formulation of improved non-toxic and biocompatible hydrogels, films and matrices etc. Different crosslinking strategies such as chemical, physical or enzymatic are applied to achieve the collagen-based materials with desired properties for drug delivery and other applications [62].

3.1 Chemical crosslinking

The most used crosslinking method is with chemical agents due to ease of application, less time consuming and cost effective. The most commonly used chemical reagents are formaldehyde (FA) and glutaraldehyde (GTA). FA reacts with the ε-amino group of lysine and hydroxylysine residue of collagen to form imine as an intermediate followed by crosslink with tyrosine or with amide group of asparagine or glutamine residue. FA-crosslinked products generated brittleness, significant toxicity and unfavorable reactions, hence not preferred in biomedicine [7]. Another agent GTA is widely utilized for crosslinking of collagen, based on high reactivity and low cost. The low concentration of FA and GTA produced brittle and low uniformity of composites while high concentration led to major cytotoxic effects. Several methods have been applied to remove the unreacted GTA, due to its cytotoxicity the use of GTA at present scenario is still debatable [28, 42]. Hexa-methylene-diisocyanate (HDC) was used as an alternative to GTA, but in contrast to GTA, HDC showed less severe primary and secondary cytotoxicity during cell proliferation as compared to non-crosslinked material [63]. Charulatha & Rajaram [64] evaluated the biocompatibility of collagen membranes cross-linked with 3,3′-dithio bis-propionimidate (DTBP) and dimethyl suberimidate (DMS). Both DTBP and DMS showed lower toxicity than GTA and as the better substitute for crosslinking. The polyepoxy compounds such as ethylene glycol diglycidyl ether, glycerol polyglycidyl ether and methyl glycidyl ether were used as crosslinker [65]. The epoxy group reacts with amino group of lysine residue for crosslinking similar to GTA. The polyepoxy crosslinked material showed acceptable cytotoxicity [66].

Due to non-toxicity and water solubility, the carbodiimides and acyl azides showed significant crosslinking with collagen. Carbodiimides like 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide (EDC) and N-hydroxy succinimide (NHS) are better candidate than aldehydes, HDC and polyepoxy compounds due to formation of amide bonds between –COOH and –NH2 group of collagen without becoming the part of actual linkage. Van Wachem et al. [67] compared the four crosslinking methods i.e. GTA, HDC, acyl azide and EDC for the assessment of biocompatibility and tissue regeneration ability and EDC crosslinked material showed best results among the four tested methods. Pieper et al. [68] showed that EDC crosslinked collagen expressed no cytotoxicity, slow enzymatic degradation and decreased calcification.

Non-toxic, biodegradable and biocompatible natural compounds as promising cross-linkers like alginic acid (anionic block copolymer) from brown algae, iridoid compounds from the fruits of Gardenia jasminoides and Genipa americana [69], oxidized alginate [70], dialdehyde starch [71], D, L-glyceraldehyde [72], natural polyphenols and dialdehyde- carboxymethyl cellulose [73] have been broadly examined. In recent years usage of polyphenols such as caffeic acid (CA) and tannic acid (TA) [74], proanthocyanidin [75], procyanidin [76], epigallocatechin gallate (EGCG) and epicatechin gallates (ECG) [77] and other tannins [78] have been increased due to antioxidative, anti-inflammatory, antimicrobial, cardioprotective, antithrombotic, pharmacological and therapeutic possibilities. According to Jackson et al. [77] TA, EGCG and ECG showed stabilization of collagen implant for long period at very low concentration and protection against collagenase more effectively than GTA and carbodiimides. In contrast to GTA, natural crosslinkers have disadvantages like long term storage and degree of crosslinking.

3.2 Physcial crosslinking

Physical techniques like ionizing radiation (X-ray and γ-ray), UV light and dehydrothermal treatment and dye-mediated photo-oxidation are used as crosslinkers for collagen based formulations. The physical crosslinking relies on the factors like amount of radiation, temperature, hydration conditions, electron beam intensity and UV denaturation [43]. Photosensitizer riboflavin in combination with UV-irradiation produced the similar results of crosslinking by GTA without cytotoxicity to reduce the harmful effects of physical and chemical crosslinking [45]. UV- light mediated crosslinking generated the denaturation and conformational modifications of collagen molecules which opposed the stabilization of UV-induced crosslinked product [79]. Dehydrothermal treatment resulted into the complex of collagen with anatomically accepted structures without contraction, curling or deformity for longer duration without the involvement of chemical crosslinking [80]. In contrast to chemical crosslinking the heating disrupted the triple helical conformation of collagen and increased the degradation by enzymes [81]. Overall the physical crosslinking methods are simple and safe for the production of splendid biocompatible biomaterials in comparison to exogenous cytotoxic chemical crosslinkers.

3.3 Enzymatic crosslinking

Enzymatic approaches have gained interest due to brilliant specificity and accurate reaction kinetics and to surmount the difficulties with chemical methods. Enzymatic crosslinkers can be categorized into oxidoreductases, transferases and hydrolases based on the catalytic reaction [82]. The oxidative enzymes tyrosinase and laccase [83] along with acyltransferase-transglutaminase have the capacity to modify the protein substrate to enhance the quality of crosslinked biomaterials [84]. Transglutaminases are calcium dependent and catalyze the reactions in broad range of pH and temperatures. Microbial origin biodegradable transglutaminases can catalyze the crosslinking in the concentration dependent manner [85, 86]. Exogenous lysyl oxidase catalyzes the lysine residue into highly reactive aldehydes leading to the formation of crosslinks in the ECM proteins. The pre-treatment of lysyl oxidase promoted the maturation of native and engineered collagen tissues both in vitro and in vivo with reference to increased tensile modulus and pyridinoline crosslinking [87].

Comparing all the crosslinking strategies the chemical method is the most preferred due to generation of consistent and high degree of crosslinking [88]. The physical method is used as a appurtenant crosslinking approach. Enzymatic approach alleviates the shortcomings produced by chemical and physical crosslinking methods, but it is time consuming and expensive [89]. In recent years, the use of natural and eco-friendly biocompatible crosslinkers has increased and become the very promising agents. The natural substance- genipin obtained from irridoid glucoside (geniposide) is one of the important crosslinker with great potential in the field of biomedicine. Genipin is very expensive in contrast to other natural crosslinkers, while TA is cheap and easily accessible compound for crosslinking [89].

Advertisement

4. Collagen-based formulations for drug delivery

Traditional drugs have been the main concern to effectively treat the several diseases. The introduction of classical drug in therapeutics at high concentrations generally develops the substantial and sometimes severe consequences. The development of effective DDS for the efficacious augmentation of particular drug at desired target and at optimal concentrations for necessary duration has been the critical concern of clinical investigations and research since several years. To achieve the targeted or controlled local drug delivery, both synthetic and natural drug delivery materials are playing the important role.

At present, DDS have been developed based on polymers, nanomaterials and lipids etc. for the attachment or encapsulation of drugs to target the delivery or controlled release for long duration [90, 91]. Collagen based biomaterials or composite materials have become the important DDS due to specific pore size for active drug or principal load, effective fibrillar network, enzymatic degradation, long term stability in vivo, biocompatibility, low antigenicity, highly reduced toxicity and safety features [92]. The chemical modifications of –OH, –NH2 and –COOH groups on collagen molecule making it more relevant and promising candidate in the domain of biomedicine. Collagen based biomaterials can be formulated into different forms according to desired medical application of drug delivery as mentioned in Figure 3. Various types of collagen based bioformulations such as hydrogels, films, sponges, scaffolds, matrices, aqueous injections, microspheres, micro-particles, micro-beads, NPs, nano-composites, nanofibres, shields, inserts, tubes, coatings, monolithic devices, implants and dressings etc. are applied for various drug delivery applications, tissue growth and regeneration. In Table 2, the type of formulations, synthetic or natural polymeric support material, active compound or drug and specific biomedical application has been summarized.

Figure 3.

Different types of collagen based biomaterials used for drug delivery.

Delivery formCollagen/ collagen compositeDrug/active substanceMedicinal applicationReferences
Gel/ hydrogelCollagenKeterolacInflammation[93]
CollagenTADrug release and kinetics[94]
CollagenNGF-βSustained delivery and corneal regeneration[95]
CollagenApis mellifera royal jellyDelivery vehicle for wound healing[96]
CollagenCurcuminControlled anti- proteolytic and pro-angiogenic efficacy[97]
GelatinEssential oil of Eupatorium adenophorumAntibacterial wound dressing[98]
Collagen/ ALGBSAOcular drug delivery[46]
Collagen/ ALGCell proliferation[48]
Collagen/ HEMAGA, naproxenDrug release, antimicrobial potential[29]
Collagen/ Fe3O4 NPsFluoresceinDelivery, release and cell viability[99]
Collagen/ CHSQHREDGSMyocardial infarction[100]
Gelatin/ CHS/ ALG5-FUAnti cancer drug delivery[49]
Collagen/ ALGMethylene blue imiquimodCombinatorial photothermal and immune tumor therapy[101]
Collagen/ CHSTumor necrosis factor-α (TNF- α)Drug delivery and tissue filler[50]
Collagen/ graphene oxide (GO)FGF-2Controlled release[102]
Collagen/ TA/ Poly-ethylenimine (PEI)Doxorubicin (DOX)Cancer therapy and antibacterial activity[103]
Collagen/ CHSNeuropathic diabetic foot ulcer (DBU) treatment[104]
Collagen/ 1-ethyl-3-methylimidazolium
acetate/ microbial transglutaminase
Controlled biodegradation[86]
Collagen/ carrageenanAllopurinolDrug delivery and bioavailability[105]
Collagen/ PLGA/ poly-lactic acid (PLA)/ poly-ε-caprolactone (PCL)SpironolactoneDrug delivery[106]
Collagen/ CHScell-penetrating peptide (CPP) (Oligoarginine, R8)Wound healing and antimicrobial activity[107]
Collagen/ CHSThymosin β4(Tβ4)Epicardial cell migration and angiogenesis[108]
Hydrogel as aqueous injectionCollagenCisplatin, vinblastine, 5-FU, 111In or 90Y labeled monoclonal antibodiesTumor treatment[109, 110, 111]
CollagenTransforming growth factor (TGF)-β, FGF, Insulin, growth hormone (GH)Wound healing and repair[112, 113, 114]
Collagen/ CMCInterleukin (IL)-10Retinal ischemia/ reperfusion therapy[52]
Collagen/ CHSNanobodies: 2D5 and KPUTumor treatment in cancer therapy[115]
Collagen/ ALGDoxycyclineVision-threatning diseases[116]
FilmsCollagenMedroxyprogesterone acetateDrug release[117]
CollagenTetracycline, antibiotics[118, 119]
CollagenPlatelet derived growth factor (PDGF)Sustained release[120]
CollagenProteins and polysaccharides[121]
CollagenAloe veraEnhanced biological avtivity[32]
CollagenSilver sulfadiazineAntibacterial activity[122]
CollagenL-cysteine hydrochlorideDrug delivery[123]
CollagenAnti-aging activity[124]
CollagenThymolAntibacterial activity[125]
Collagen/ elastinCell adhesion and viability[126]
Collagen/ PVARecombinant human growth hormone (rhGH)Drug delivery[127]
Collagen/ calcium phosphateCell adhesion, proliferation, differentiation, mineralization[128]
Collagen/ polyurethane (PUR)GFCell adhesion, proliferation and growth[129]
Collagen/ carboxymethyl guar gumCeftazidimeDrug release, antibacterial activity[130]
Collagen/ CHSGentamicin sulfateAntibiotic release[51]
Collagen/ CHSDOXCancer treatment[131]
Collagen/ CHSLidocaine, tetracaine, benzocaineWound healing[132]
Collagen/ CHS/ HAGentamicin sulfateAntibiotic release[133]
Collagen/ GOOvalbumin (OVA)Sustained release[134]
Collagen/ CHSAllantoin and lidocaineWound healing[135]
Collagen / CHS/ GO/ EDCBasic fibroblast growth factor (bFGF)Wound dressing[136]
MembraneCollagen/ PLGAVancomycin, gentamicin and lidocaineAntibiotic activity[137]
Collagen/ CHSNifedipine and propranolol hydrochlorideCardiac disease[138]
Collagen/ CHSNifedipineTransdermal delivery[139]
Collagen/ PVACiprofloxacin hydrochlorideAntibacterial activity and treatment of ulcerative keratitis[27]
ScaffoldCollagen as PURACOL®Human antimicrobial peptide (AMP) cathelicidin LL37Antimicrobial activity and cytotoxicity[140]
Collagenpolyethylenimine (PEI)-plasmid DNA (pDNA) encoding PDGF-B complexesBone regeneration and gene delivery[141]
Collagen/ CHSIbuprofenThermoresponsive scaffold[142]
Collagen/ CHSbFGFWound healing and skin tissue
engineering
[143]
CollagenN,N,N-trimethyl chitosan chloride (TMC)/ plasmid DNA encoding VEGF.Angiogenesis and gene delivery vector[144]
CollagenBMPPeriodontal healing[145]
Gelapin-Simvastatin ScaffoldsSimvastatinBone defect healing[146]
Collagen/ CHS/ ALGCurcuminDiabetic wound healing[147]
Collagen/ CHSNorfloxacinSkin regeneration[148]
Collagen/ elastinPenta-galloyl glucose (PGG)Diabetes related complications[149]
Collagen / HAPPTHrP 107–111, pentapetideThermal drug release[150]
Collagen/ CHS/ chondroitin (CHD)PLGA microspheresTissue engineering[151]
Collagen/ PEI polyplexescollagen-mimetic peptide (CMP)Release kinetics and improved gene activity[152]
Collagen/ CSPlatelet-rich plasma (PRP)Growth factor release and wound healing[153]
Collagen / HAPBMP, alendronateBone regeneration[154]
Collagen/ CHSCurcumin nanoparticlesWound healing and gene expression[155]
SpongeCollagenGrowth factors (GFs), FGF, BMPsWound healing and tissue regeneration[156, 157, 158, 159]
CollagenGentamicin, cefotaxim, fusidic acid, clindamycinWound healing[160]
CollagenAll trans-retinoic acidCervical dysplasia[161]
CollagenNiflumic acidDrug delivery and release[162]
CollagenGentamicinWound infection[163]
CollagenBMP-2Drug delivery comparison[164]
Collagen/ PLGACell attachment and cytotoxicity[165]
Collagen/ CHSDexamethasoneOral muscositis[166]
Collagen/ PLGAGentamicinTissue regeneration[167]
Collagen/ PCL/ HAMethylene blue and curcuminSustained drug release kinetics[168]
MatrixCollagenAmpicillinDrug release[169]
CollagenCisplatinLocal cancer therapy[170]
Collagen/ PEI/ DNA complexTissue repair[171]
Collgen as INFUSE® bone graft and MASTERGRAFT®Recombinant bone morphogenetic protein-2 (rhBMP-2)Bone fracture and spinal fusion[172]
Collagen/ HATissue development[173]
Collagen/ chondroitin-6-sulphate (CS)rhBMP-4Drug delivery[174]
Collagen/ heparinCofibrillogenesis[175]
Collagen/ tri-calcium phosphate (CP)Chondroinductive growth factorOsteochondral tissue repair[176]
3-D microsphereCollagenGlial derived neurotrophic factor (GDNF)Controlled proliferation[177]
CollagenBMP-2Local delivery[178]
Collagen/ CHS/ nano-HAPDrug delivery[179]
Collagen/ BCBSAPotential DDS[54]
Collagen/ ALGGDNFNeurodegenerative diseases[180]
Collagen/ BCBMP-2Adhesion, proliferation and osteogenic differentiation[53]
Collagen/ ALGBMP-4Proliferation and differentiation[181]
MicroparticlesCollagenRetinol, tretinoin, tetracain, lidocainDrug delivery[182]
CollagenCyclosporineAllograft implantation[183, 184]
CollagenGlucocorticosteroidsDrug delivery[185]
CollagenLysozymeProtein delivery[186]
Collagen/ hydroxyl ethylcelluloseRetinolDrug delivery[187]
Collagen/ poly oxyethylated sorbitan esterEthacridine lactateDrug delivery[188]
Collagen/ CHS/ CPGlycolic acidTissue regeneration[189]
Micro-beadsCollagen/ fibrinInsulin like growth factor (IGF)-1Regeneration of urethral sphincter muscle[190]
NanoparticleCollagenEstrogen (17-beta-estradiol-hemihydrate)Delivery in hormone replacement therapy[191]
Gelatin/ ALG/ Fe3O4Doxorubicin hydrochlorideCancer chemotherapy[192]
Collagen/ CHSDoxorubicin hydrochlorideAdvanced cancer chemotherapy[193]
Collagen peptide/ calcium/ ALGCalcium supplementation[194]
Collagen / Ferritin/ TaO NPsTGF-β1Sustained release, imaging and regeneration of oral tissue[195]
Collagen/ ALG/ AgWound healing[47]
AtelocollagensiRNA duplexDelivery for gene silencing[196]
NanocompositeCollagen/ bacterial cellulose (BC)/ apatiteOsteogenic growth peptide (OGP)Bone regeneration[197]
Collagen/ HAP/ ALGBone morphogenetic proteins (BMP)Bone filler[198]
NanofibresCollagen/ PCLCell proliferation and migration[199]
InsertsCollagenGentamicin[200]
CollagenErythromycin, erythromycin-estolate, penicillin-procaineInfection, glaucoma[201]
CollagenPilocarpine[202]
ShieldsCollagenTobramycinInfection, mycosis[203]
CollagenVancomycinInfection, mycosis[204]
CollagenGentamicinInfection, mycosis[205]
CollagenNetilmycinInfection, mycosis[206]
CollagenAmphotericin BGlaucoma, inflammation[207]
CollagenPolymyxin B sulphateGlaucoma, inflammation[208]
Collagen5-fluorouracil (5-FU)Glaucoma, inflammation[209]
CollagenSteroidsGlaucoma, inflammation[209, 210]
Monolithic devicesCollagenMinocycline, lysozymePeriodontitis[211]
CollagenInterleukin-2 (IL-2)Drug delivery[212]
CollagenInterferonDrug delivery[213]
TubesCollagen/ silk fibroinVascular tissue engineering[214]
Coating for compositeCollagen/ poly-L-lactic acid (PLLA)Cell compatibility[215]
Collagen as enteric coatingGastro-resistant tabletsDelayed release[216]
ImplantCollagenGentamicinPrevention of surgical site infection[217]
CollagenPS1 as antineoplastic glycanAntitumor activity[218]
Collagen/ PCLCorneal tissue regeneration[219]
DressingCollagen/ ALG/ HAAmpicillinWound healing and antimicrobial activity[220]
Collagen/ CHS/ glucanA. veraAntibacterial activity, wound healing of chronic wounds and ulcers[221]

Table 2.

Application of collagen and collagen based material in drug delivery systems (DDS).

4.1 Gel/hydrogel

Hydrogels are three dimensional (3-D) crosslinked arrangements of similar or different types of polymeric molecules with the property of absorbing and retaining the optimum quantity of water or biological fluid without degrading or losing the network structure. The material can be categorized as hydrogel, if the water content in the material is at least 10% of the total weight or volume of hydrogel [38]. The water molecules in the hydrogel provide the freedom of flexibility to design the natural tissue like environment. Hydrogels can be synthetic or natural with different chemical constituents and having different mechanical, physical and chemical attributes according to biomedical application. Hydrogels can be hydrophilic or hydrophobic in nature. Hydrophilic hydrogels possess hydroxyl (–OH), amine (–NH2), carboxyl (–COOH), amide (–CONH–CONH2) and sulphonic (–SO3H) group for the swelling and absorbing property. The hydrophobic polymers show low swelling feature despite having improved mechanical, physical or chemical strength.

Drug transport within hydrogel can be regulated by altering the network/mesh size or the interactions with drugs using chemical methods [172, 222]. If the loaded drug is smaller than the crosslinked network of hydrogel, it can simply diffuse through the hydrogel while the larger drug molecules are entrapped within the hydrogel network and can be released after the degradation of the mesh. The biopolymer and its crosslinked network can be degenerated via slow hydrolysis of peptide or ester linkage or cleaving the thiol-related bonds, or through the enzymatic activity [223]. By the incorporation of non-covalent or covalent drug-matrix interactions, drug release from the hydrogel can be tuned [224, 225]. The characters mainly mesh size, crosslinking chemistry and drug interactions facilitate the better handling of drug transport through hydrogel. ECM-based hydrogels are the preferred choice for local drug delivery due to mechanical and biochemical support through cell-matrix interactions and diffusion and infiltration of small drug molecules in between crosslinked polymeric network [172].

Collagen alone is used for the delivery of several drugs and active principles such as keterolac, nerve growth factor (NGF)-β, TA, curcumin and royal jelly etc. for various biomedical purposes like anti-inflammation, corneal regeneration, drug release and kinetics, angiogenesis and wound healing, respectively. According to Ramírez et al. [96] type I collagen hydrogel extracellular vehicles (EVs) with Apis mellifera royal jelly displayed effective wound healing to stimulate mesenchymal stem cell (MSC) migration and inhibition of biofilm formation by Staphylococcus aureus along with stable release kinetics up to 7 days. Curcumin crosslinked collagen aerogel system expressed the enhanced physical and mechanical features and controlled anti-proteolytic and pro-angiogenic potential, made them appropriate 3D scaffolds for medicine purposes. Here, curcumin as a nutraceutical was used as a crosslinker for further usage [97]. In a study by Chuysinuan et al. [98], gelatin-based hydrogel was prepared by mixing gelatin with GTA crosslinker and essential oil of Eupatorium adenophorum and applied on patients with open wound to assess the wound healing and anti-bacterial properties by analyzing release profiles.

Collagen-based composite materials with synthetic (CMC, PVA, PCL, PEMA, HEMA, polyurethane (PUR) etc.) or natural (CL, bacterial cellulose (BC), CHS, ALG, HA, GAGs etc.) polymers have major roles in biomedicine. Liu et al. [46] developed a composite of collagen-ALG with suitable mechanical strength and optical clarity to support human corneal epithelial cell growth using bovine serum albumin (BSA) as a model drug. The hydrogel system could be applied as therapeutic lens in patients with corneal illnesses. Collagen-based hydrogel preparation by mixing of acrylamide and 2-hydroxy ethyl methacrylate (HEMA) was used as DDS for linear release profile of gallic acid (GA) and naproxen up to 36 hours for wound healing. Addition of metal NPs such as Ag and Cu in this collagen-HEMA hydrogel films showed antimicrobial potential against Escherichia coli, Bacillus subtilis and S. aureus [29]. Bettnini et al. [99] prepared the porous collagen-based hydrogel scaffold with iron oxide (Fe3O4) NPs for the release of fluorescein and biocompatibility, cell viability of 3 T3 fibroblasts cells and proposed the safety and applications in tissue engineering and drug delivery. Reis et al. [100] developed the collagen-CHS thermoresponsive hydrogel conjugated with angiopoietin-1 derived peptide glutamine-histidine-arginine-glutamic acid-aspartic acid-glycine-serine peptide (QHREDGS) for the survival and maturation of cardiomyocytes. Hydrogels with high peptide load showed better morphology, viability, metabolic activity, success rate of beating as compared to hydrogels with low peptide concentration and control groups.

Graphene oxide (GO) sheets were inserted into the collagen-based hydrogels for the controlled release of fibroblast growth factor (FGF)-2 to induce pluripotent stem cell culture. Low permeability of GO sheets allowed the release of FGF-2 in controlled and regulated fashion while the FGF-2 interacted with collagen through electrostatic forces and partial hydrogen bonding. The release profile of FGF-2 was attained up to 400 hours using three different concentrations of GO and showed the fabricated hydrogel for better release of growth factors (GFs) for biomedical application [102]. Choi et al. [103] developed the collagen-TA-poly ethylenimine (PEI) hydrogel with layer-by-layer self-assembled films to overcome the problem of poor mechanical strength and fast release of inserted drug. Doxorubicin (DOX) was used as model cancer therapy drug. The multifunctional hydrogels showed sustained and controlled release up to 6–7 days without any cytotoxic effects along with antibacterial property against Gram positive and negative bacteria and higher strength to compression load.

Injectable hydrogels are the promising materials for cancer treatment and controlled delivery. With the minimal invasive processes injectable hydrogels can be located and remained at required position and also mitigate the irregular shape defects after the implantation. Aqueous injections of hydrogels could be used in biomedicine field such as drug release, wound healing, repair, tumor treatment, tissue regeneration, ocular/retinal disorders and cancer therapy etc. Fan et al. [115] designed the hydrogel prepared from tilapia skin collagen and CHS for the delivery of model nanobodies- 2D5 and KPU. The hydrogel was biodegradable and expedited the release of nanobodies and could pave the way for tumor treatment. Carboxymethyl cellulose (CMC)-collagen based aqueous injectable hydrogel showed promising antioxidative and drug carrier benefits to treat the retinal ischaemia or reperfusion injury in rat models and could be applied for drug based treatment of retinal illnesses in humans. Animals were treated with interleukin (IL)-10 loaded hydrogels and expressed better therapeutic results of restoration of retinal structures and reduced retinal apoptosis, significantly decreased retinal oxidative stress in comparison to control group [52].

4.2 Films/membranes

The films or membranes are very thin and flexible layer of biopolymers with or without plasticizer having optical and mechanical anisotropy with very high tensile strength making them suitable for various medical applications of sustained drug release, cell adhesion, proliferation, differentiation, cancer treatment, wound healing and tissue regeneration. The thin films are the prominent material to target sensitive locations not possible with other formulations like liquid or tablets [226]. Thin films exhibited the improvised onset of drug activity, decreased dose quantity or frequency and augmented drug efficacy, reduced side effects by drug and extensive metabolism [227, 228].

Gil et al. [122] developed the innovative chromium free-collagen film for slow drug release carrier for skin burn related complications like ulcers and infected wounds. The biocompatible films were tested for drug silver sulfadiazine and its antibacterial potential against Pseudomonas aeruginosa, E. coli, Micrococcus luteus, S. aureus, Proteus vulgaris and Klebsiella pneumoniae. The findings proposed the effective strategy for Chromium (Cr) removal from leather waste and generation of environment friendly material could be transformed into collagen films, promising candidate for drug carriers. The use of biocompatible, recyclable, biodegradable natural materials has become tremendously increased in recent decades. Langasco et al. [123] developed the natural collagen films from marine sponges for topical drug delivery application. L-cysteine loaded films were analyzed for different drug concentrations and drying parameters. The films showed the healing potential of cysteine, acted as biocompatible carrier to absorb excess of wound exudate along with drug release. The films could be the promising material and might behave as bioactive, biomimetic drug carrier for effective wound healing. Jana et al. [130] synthesized the fish scale collagen-carboxymethyl guar gum film loaded with broad spectrum antibiotic ceftazidime. Around 90–95% of ceftazidime was released after 96 hours at physiological pH. In vitro study on NIH 3 T3 fibroblast cell line showed the biocompatibility of crosslinked film and antibacterial results exhibited the inhibition of S. aureus and P. aeruginosa.

Collagen-CHS based films/membranes are important biomaterials and used for antibiotic release, wound healing, cancer treatment, transdermal delivery, cardiac illness, tissue engineering. Martino et al. [132] formulated the collagen-CHS film for the delivery of mixture of local anesthetics compounds- lidocaine, tetracaine and benzocaine. The films were developed by rapid, cost effective and highly reproducible casting approach. The films showed good mechanical strength and flexibility with high water permeability. The anesthetics were uniformly distributed in the film and controlled released from 6 to 24 hours. The film exhibited in vitro non-cytotoxicity, cell proliferative and biocompatibility properties against human dermal fibroblast cells making it better candidate for drug release and proliferation. Liu et al. [136] fabricated the collagen-CHS-GO composite film using EDC as crosslinker loaded with basic FGF for effective wound healing through controlled release. The film had improved thermal endurance and higher degree of crosslinking for advanced mechanical strength due to GO. This novel DDS prevented the initial sudden release and loss of bioactive potential of basic FGF in vivo and in vitro. In cultured L929 fibroblasts, the film showed good biocompatibility in terms of cell adhesion and proliferation. These films were implanted on rats showed the wound remodeling to repair full thickness skin wound. So these films were promising substitute as wound dressing material for drug delivery with wound healing. Daja et al. [27] developed the collagen/PVA anionic membrane for drug carrier of ciprofloxacin hydrochloride along with antibacterial efficacy and its application in the treatment of ulcerative keratitis. The membrane provided the sustained DDS and inhibited the growth of S. aureus and E. coli during 48 hours. The membrane had proper mechanical strength, water amount, hydrophilicity, permeability and pH without any stress to cornea during interaction. The collagen fibrils in membrane decreased stromal damage and improved the epithelium regeneration. The formulated membranes were cost-effective and secured biomaterial for the treatment of corneal ulcers in patients.

4.3 Scaffolds/sponges/matrices

Scaffolds are collagen sponges or matrices with three dimensional network structures. Scaffolds can be obtained with various synthesis approaches of freeze drying, electrospinning and 3D printing etc. The freeze drying is the most effective method preserving the structure and native or inherent properties of collagen along with loaded drug/active principle in the scaffold. Collagen-based scaffolds or matrices are the important and favorable materials for bone, skin and tissue regeneration, angiogenesis, gene delivery, wound healing and repair, sustained drug release and improved gene expression. Now a days several commercially available collagen-based sponges in the market are Collarx®, Collatamp® G, Collatamp®EG Sulmycin® Implant, Garamycin® Schwamm, Duracol®, Duracoll®, Gentacol®, Gentacoll®, Garacol®, Garacoll® and Cronocol® - Gentamicin surgical implants.

Elangovan et al. [141] developed the non-viral gene delivery system for bone regeneration with the help of collagen scaffold to deliver the PEI-plasmid DNA encoding platelet derived growth factor (PDGF)-B complexes. The complexes expressed low cytotoxicity and markedly higher proliferation of human bone marrow stromal cells in contrast to scaffold without DNA and PDGF-B. In rats model the complexes exhibited higher bone volume followed the 4 weeks of grafting in comparison to empty scaffolds. The results advocated the use of non-viral scaffolds for bone regeneration along with gene delivery vehicle in clinical applications. Collagen-based biopolymers are one of the most important biomaterials to formulate the matrices in the field of tissue engineering due to significant non-toxicity, biocompatibility and resorptive potential. López-Noriega et al. [150] designed the collagen-hydroxyapatite (HAP) scaffold with covalently attached thermoresponsive liposomes. The encapsulated drug with pro-osteogenic and anti-osteoclastic properties was PTHrP107–111, a pentapeptide. The regulated release of pentapeptide was correlated with enhanced expression of osteopontin and osteocalcin genes in cultured pre-osteoblastic MC3T3-E1 cells. This scaffold medicated drug release and cell regeneration has vast potential for various types of tissue regeneration.

Collagen-based 3D biomaterials are broadly used in the field of biomedicine for their properties of biocompatibility, inherent bioactivity to induce cell proliferation, hemostatic and low antigenicity. A porous and highly structured biomaterial such as sponges or matrices promotes the flexibility, permeability and biomimicry [229, 230]. Crosslinking or amalgamation of natural or synthetic biopolymers improvises the shortcomings of collagen polymer alone in terms of physico-chemical and biological parameters. Alagha et al. [166] prepared the porous muco-adhesive collagen-CHS bio-sponge as DDS for dexamethasone to treat the oral mucositis. The sponge was characterized by X-ray, FTIR, SEM, DSC and swelling behavior. The collagen-CHS sponge showed regulated drug release up to 10 hours as compared to collagen sponge for 5 hours. David et al. [168] fabricated the collagen-PCL-HA macroporous sponge to deliver the model drug methylene blue and curcumin. Several parameters such as absorption, water uptake, drug loading and delivery along with mechanical and structural features were examined for the developed sponge. In comparison to control group, the sponge showed sustained release kinetics for drugs and making the sponge as future material for applications of wound dressing and lab models.

Collagen matrices are able to deliver the gene or plasmid DNA in cultured cells and alter the gene expression in tissue engineering. Orsi et al. [171] formulated the bio-activated collagen-PEI-DNA complex to control the gene expression and attract the specific cell type. The transfected NIH3T3 cells with matrix-PEI-DNA complex secreted the plasmid encoded protein to promote the tissue repair and regeneration. The developed matrix could be the new approach for tissue repair.

4.4 3-D microspheres/ microparticles/micro-beads

Microspheres, microparticles or micro-beads are spherical particles with large surface to volume ratio for improved drug delivery, growth factors and broad surface area for cellular interactions to other biomolecules [28]. The size of collagen-based microparticles ranging from 3 to 40 μm. Berndt et al. [231] developed the collagen microspheres encapsulated astrocytes crosslinked with poly (ethylene glycol) tetrasuccinimidyl glutarate (4S-StarPEG) as growth enhancing and carrier for injured spinal cord. Astrocytes were transfected with plasmids encoding nerve growth factor (NGF)-ires-enhanced green fluorescent protein (EGFP) genes and then added to the culture of rat dorsal root ganglion and significantly improved growth was observed. The report showed the potential of microspheres as carrier of astrocytes for neural tissue regeneration. Zhang et al. [53] formulated the 3-D microsphere of collagen-BC-bone morphogenic protein (BMP)-2 for bone tissue augmentation. The 3D microporous microspheres effectively enhanced the adhesion, proliferation and osteogenic differentiation of mice MC3T3-E1 cells and expressed adequate biocompatibility.

Marine based collagen from jellyfish species Catostylus tagi was used to develop the microparticles formulation for sustained delivery of lysozyme and α-lactalbumin. The collagen microparticles were crosslinked with EDC and investigation of lysozyme activity was retained throughout the crosslinking and encapsulation process. These microparticles from marine collagen could be the promising material for controlled release of therapeutic proteins [186]. Yang and Fang [232] developed the microporous nano-HAP/collagen/phosphatidylserine scaffolds embedding collagen microparticles for the sustained release of steroidal saponins for bone tissue engineering in cultured MC-3 T3-E1 cells. The scaffolds provided scope for spatial and temporally controlled drug delivery and deposition at wounded site and reduction in adverse side effects. Vardar et al. [190] formulated the novel injectable collagen-fibrin microfluidic system loaded with recombinant insulin like growth factor-1 (α2PI1–8-MMp-IGF-1) to treat the urinary incontinence. The natural crosslinker genipin was used for collagen modification. The microbeads showed slow release of GF and positive cell behavior for the induction of in vivo smooth muscle regeneration for effective management of urinary incontinence.

4.5 Nanoparticles/ nanocomposites/nanofibres

Nanomaterials within the size of 1–100 nm are one of the best materials with admirable biochemical and pharmacological attributes [233]. Biological protein-based NPs are applied in different applications due to eco-friendly nature and biocompatibility and replacing the synthetic materials. Collagen is the preferred NP substance and by direct or indirect crosslinking to collagen NPs provide better substitute for protein based drug delivery vehicle [234]. The crosslinking of collagen with NPs is the new strategy to enhance the mechanical and physical strength of collagen tissue for various applications. Metal oxide NPs such as iron oxide (Fe3O4), zinc oxide (ZnO), alumina oxide (Al2O3), tantalum oxide (TaO), Al2O3-ZrO2 and Fe3O4-ZnO improve the mechanical features of collagen-based biomaterials [99, 195, 235]. The metallic NPs provided broad spectrum antimicrobial, antioxidative and anti-inflammatory attributes [236, 237] to collagen based material and serve as an alternative to toxic chemical crosslinkers and impede the collagen degradation by physical crosslinking. These features advertise the use of NPs based collagen biopolymers in medical field such as targeted controlled drug delivery, cell targeting and tissue engineering. Choi et al. [195] prepared the collagen hydrogel containing the ferritin NPs, TaO NPs along with transforming growth factor (TGF)-β1 for the controlled release and imaging medium for regeneration of oral tissues.

NPs based biopolymers for cancer therapeutics are effectively utilized in recent years due to targeted and controlled release kinetics. Anandhakumar et al. [193] fabricated the collagen peptide-CHS NPs for encapsulation of standard cancer drug DOX in cancer therapy. The NPs showed high encapsulation capacity of DOX and pH regulated release. NPs with DOX expressed significant anti-proliferative activity against HeLa cells in contrast to normal cells. The NPs showed excellent biocompatibility with high power as smart DDS for cancer therapeutics. Zhang et al. [47] designed the collagen-ALG biocomposite doped with silver NPs (AgNPs) with antibacterial potential and applicable as wound dressing. The biocomposite exhibited insignificant in vitro toxicity at lower concentrations of AgNPs and also inhibited the growth of S. aureus and E. coli. Saska et al. [197] fabricated the nanocomposite of BC-collagen-apatite and osteogenic growth peptide (OGP) for bone tissue regeneration. The OGP containing nanocomposite triggered the early development of osteoblastic phenotype and elevated cellular growth without any cytotoxic, genotoxic or mutagenic adverse effects. The nanocomposite could be the promising future biomaterial for bone tissue engineering.

4.6 Inserts/shields/monolithic devices or pellets

The approach of applying ocular collagen inserts to administer the drug for prolonged period was started in early 1970s. The inserts were films or as molded rods or wafers of collagen incorporated with drug such as pilocarpine, penicillin-procaine, erythromycin, erythromycin esolate and gentamicin etc. in the form of eyedrops, ointments and subconjunctival injections to treat the cornea related disorders [200, 201, 202]. In the late 1980s researches on inserts were overtaken by shields which became commercially available in the market in reproducible manner [7].

Collagen shields were formulated as corneal bandages/dressings to facilitate the wound healing, allow sufficient oxygen transmission, lubricate the eye surface to minimize stress and to regenerate the corneal epithelial linings after corneal injury or damage, transplantation, radial keratomy, glaucoma, keratitis or cornea related disorders [238, 239]. These shields could be used as carrier to deliver the ophthalmic medication such as water soluble antibiotics- gentamicin, vancomycin, tobramycin, netilmicin, polymyxin B sulphate, trimethoprim, amphotericin B, pilocarpine and flurbiprofene sodium etc. [7]. The drug delivery aspect of shields is limited by transparency, reduced visual acuity, slight irritation, complex administration procedure and prolonged durability. In current scenario, the commercial formulations like Biocora®, ProshieldO®, MediLenso®, Irvine® and Chiron® etc. are showing better future for delivery of corticosteroids and subconjuctival antibiotics etc. [240].

Collagen minipellets are cylindrical injectable controlled release drug delivery vehicle. In 1992, Takeuchi [211] prepared the little rods of 1 mm in diameter and length of 15 mm of injectable collagen minipellet for the local delivery of minocycline and lysozyme to treat periodontitis. Fujioka et al. [241] used the injectable collagen minipellet to deliver interleukin (IL)-2 molecule. Maeda et al. [242] used the collagen minipellet as a carrier to deliver the recombinant human bone morphogenic protein (rhBMP)-2 to induce the bone formation in mice models. Lofthouse et al. [243] developed the degradable collagen minipellet infused with avidin and IL-1β as vaccine carrier for clostridial antigen into sheep and mice. Higaki et al. [244] fabricated the biodegradable collagen minipellet to deliver the tetanus and diphtheria toxoid as single dose vaccine delivery system in mice.

Advertisement

5. Conclusion

Protein-based biomaterials have excellent biocompatibility with minimal cytotoxicity and biodegradability and theirs physical, chemical and biological parameters can be altered according to biomedical application. Collagen, a major protein in animal body is the attractive biopolymers for the delivery of therapeutic drugs, growth factors, hormones, proteins/enzymes, gene and imaging probes in the field of drug delivery systems, wound healing, bone grafts, implants, tissue regeneration, ocular diseases, cosmetic surgery, reconstructive surgery and cardiac treatments.

The researchers are consistently designing the protein-based hybrid materials with desired physical, mechanical, chemical and biological properties. Collagen-based hybrid biomaterials can be formed with natural polymers like CHS, CL, ALG, gelatin, HA, CHD, HAP or the chemically modified form of these natural polymers or with synthetic polymer such as CMC, PVA, PCL, PEMA, PLA and PLGA etc. through various physical, chemical and enzymatic crosslinking approaches. Collagen-based biomaterials can be fabricated into variety of physical forms hydrogel, films or membranes, scaffolds or sponges, matrices, 3-D microspheres, microparticles, nanoparticles, nanocomposites, inserts, shields and pellets for drug based delivery of synthetic and natural active biocomponents in various fields of medical science. Collagen-based biomaterials have attracted the researchers to develop efficient and controlled therapeutic vehicles for clinical applications ensuring the patient compliance. The more efforts are needed to translate the clinical results into production scale with collaboration of researchers, material scientists, clinical doctors and industry.

References

  1. 1. Ahmed M, Verma AK, Patel R. Collagen extraction and recent biological activities of collagen peptides derived from sea-food waste: A review. Sustainable Chemistry and Pharmacy. 2020;18:100315. DOI: 10.1016/j.scp.2020.100315
  2. 2. Zhang Y, Sun T, Jiang C. Biomacromolecules as carriers in drug delivery and tissue engineering. Acta Pharmaceutica Sinica B. 2018;8(1):34-50. DOI: 10.1016/j.apsb.2017.11.005
  3. 3. Jao D, Xue Y, Medina J, Hu X. Protein-based drug-delivery materials. Materials (Basel). 2017;10(5):517. DOI: 10.3390/ma100505517
  4. 4. Zhang X, Zhang J, Shi. Mesoporousbioglass/silk fibroin scaffolds as a drug delivery system: Fabrication, drug loading and release in vitro and repair calvarial defects in vivo. Journal of Wuhan University of Technology-Materials Science Edition. 2014;29:401-406. DOI: 10.1007%2Fs11595-014-0929-0
  5. 5. Albu MG, Titorencu I, Ghica MV. Collagen-based drug delivery systems for tissue engineering. In: Pignatello R, editor. Biomaterials Applications for Nanomedicine. London: InTech; 2011. pp. 333-358. Available from: http://www.intechopen.com/books/biomaterialsapplications-for-anomedicine/collagen-based-drug-delivery-systems-for-tissue-engineering
  6. 6. Fujioka K, Maeda M, Hojo T, Sano A. Protein release from collagen matrices. Advanced Drug Delivery Reviews. 1998;31:247-266
  7. 7. Friess W. Collagen – biomaterial for drug delivery. European Journal of Pharmaceutics and Biopharmaceutics. 1998;45:113-136
  8. 8. Piez KA. Collagen. In: Encyclopaedia of Polymer Science and Engineering. New York: Wiley; 1985. pp. 699-727
  9. 9. Bhagwat PK, Dandge PB. Collagen and collagenolytic proteases: A review. Biocatalysis and Agriculatral Biotechnology. 2018;15:43-55. DOI: 10.1016/j.bcab.2018.05.005
  10. 10. Yang YJ, Jung D, Yang B, Hwang BH, Cha HJ. Aquatic proteins with repetitive motifs provide insights to bioengineering of novel biomaterials. Biotechnology Journal. 2014;9(12):1493-1502. DOI: 10.1002/biot.201400070
  11. 11. Addad S, Exposito J-Y, Faye C, Ricard-Blum S, Lethias C. Isolation, characterization and biological evaluation of jellyfish collagen for use in biomedical applications. Marine Drugs. 2011;9(6):967-983. DOI: 10.3390/md9060967
  12. 12. Ricard-Blum S. The collagen family. In: Hynes R, Yamada K, editors. Additional Perspectives on Extracellular Matrix Biology. Cold Spring Harbor Perspectives in Biology. Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press; 2011. pp. 1-20. DOI: 10.1101/cshperspect.a004978
  13. 13. Varanko A, Saha S, Chilkoti A. Recent trends in protein and peptide-based biomaterials for advanced drug delivery. Advanced Drug Delivery Reviews. 2020;156:133-187. DOI: 10.1016/j.addr.2020.08.008
  14. 14. Gelse K, Pöschl E, Aigner T. Collagens - Structure, function, and biosynthesis. Advanced Drug Delivery Reviews. 2003;55:1531-1546. DOI: 10.1016/j.addr.2003.08.002
  15. 15. Felician FF, Xia C, Qi W, Xu H. Collagen from marine biological sources and medical applications. Chemistry & Biodiversity. 2018;15(5):e1700557. DOI: 10.1002/cbdv.201700557
  16. 16. Wang D-A, Williams CG, Yang F, Elisseeff H. Enhancing the tissue-biomaterial interface: Tissue-initiated integration of biomaterials. Advanced Functional Materials. 2004;14(12):1152-1159
  17. 17. Hench LL, Ethridge EC. Biomaterials: An Interfacial Approach. New York: Academic Press; 1982. p. 385
  18. 18. Raghavendra GM, Varaprasad K, Jayaramudu T. Biomaterials: Design, development and biomedical applications. In: Thomas S, Grohens Y, Ninan N, editors. Nanotechnology Applications for Tissue Engineering. Amsterdam: Elsevier Inc; 2015. DOI: 10.1016/B978-0-323-32889-0.00002-9
  19. 19. Bronzino JD. The Biomedical Engineering Handbook. 2nd ed. Vol. 2. New York: CRC Press; 1999. p. 2896
  20. 20. Helmus MN, Gibbons DF, Cebon D. Biocompatibility: Meeting a key functional requirement of next-generation medical devices. Toxicologic Pathology. 2008;36(1):70-80
  21. 21. Yoruç BH, Şener BC. Biomaterials. In: Kara S, editor. A Roadmap of Biomedical Engineers and Milestones. Rijeka: InTech; 2012. Available from: http://www.intechopen.com/books/a-roadmap-of-biomedical-engineers-and-milestones/biomaterials
  22. 22. Williams DF. Tissue biomaterial interactions. Journal of Materials Science. 1987;22(10):3421-3445. DOI: 10.1007/BF01161439
  23. 23. Cheng W, Yan-hua R, Fang-gang N, Guo-anZ. The content and ratio of type I and III collagen in skin differ with age and injury. African Journal of Biotechnology. 2011;10:2524-2529
  24. 24. Ramshaw JA, Peng YY, Glattauer V, Werkmeister J. Collagens as biomaterials. Journal of Materials Science: Materials in Medicine. 2009;20:3-8. DOI: 10.1007%2Fs10856-008-3415-4
  25. 25. Lin K, Zhang D, Macedo MH, Cui W, Sarmento B, Shen G. Advanced collagen-based biomaterials for regenerative biomedicine. Advanced Functional Materials. 2019;29:1804943. DOI: 10.1002/adfm.201804943
  26. 26. Bretaud S, Guillon E, Karppinen S-M, Pihlajaniemi T, Ruggiero F. Collagen XV, a multifaceted multiplexin present across tissues and species. Matrix Biology Plus. 2020;6-7:100023. DOI: 10.1016/j.mbplus.2020.100023
  27. 27. Daza JHU, Righetto GM, Chaud MV, Martins VCA, Camargo ILBC, Plepis AMG. PVA/anionic collagen membranes as drug carriers of ciprofloxacin hydrochloride with sustained antibacterial activity and potential use in the treatment of ulcerative keratitis. Journal of Biomaterials Applications. 2020;35(3):301-312. DOI: 10.1177/0885328220931733
  28. 28. Geanaliu-Nicolae R-E, Andronescu E. Blended natural support materials-collagen based hydrogels used in biomedicine. Materials. 2020;13(24):5641. DOI: 10.3390/ma13245641
  29. 29. Sahiner M, Alpaslan D, Bitlisli BO. Collagen-based hydrogel films as drug-delivery devices with antimicrobial properties. Polymer Bulletin. 2014;71:3017-3033. DOI: 10.1007/s00289-014-1235-x
  30. 30. Parenteau-Bareil R, Gauvin R, Berthod F. Collagen-based biomaterials for tissue engineering applications. Materials. 2010;3:1863-1887
  31. 31. Antoine EE, Vlachos PP, Rylander MN. Review of collagen I hydrogels for bioengineered tissue microenvironments: Characterization of mechanics, structure, and transport. Tissue Engineering Part B Reviews. 2014;20(20):683-696. DOI: 10.1089/ten.TEB.2014.0086
  32. 32. Andonegi M, Heras KL, Santos-Vizcaino E, Igartua M, Hernandez RM, Caba KDL, et al. Structure-properties relationship of chitosan/collagen films with potential for biomedical applications. Carbohydrate Polymers. 2020;237:116159. DOI: 10.1016/j.carbpol.2020.116159
  33. 33. Rafat M, Li F, Fagerholm P, Lagali NS, Watsky MA, Munger R, et al. Peg-stabilized carbodiimide crosslinked collagen–chitosan hydrogels for corneal tissue engineering. Biomaterials. 2008;29:3960-3972. DOI: 10.1016/j.biomaterials.2008.06.017
  34. 34. Broguiere N, Formica FA, Barreto G, Zenobi-Wong M. Sortase a as a cross-linking enzyme in tissue engineering. Acta Biomaterialia. 2018;77:182-190. DOI: 10.1016/j.actbio.2018.07.020
  35. 35. Hamedi H, Moradi S, Hudson SM, Tonelli AE. Chitosan based hydrogels and their applications for drug delivery in wound dressings: A review. Carbohydrate Polymers. 2018;1(199):445-460. DOI: 10.1016/j.carbpol.2018.06.114
  36. 36. Wu X, Liu A, Wang W, Ye R. Improved mechanical properties and thermal-stability of collagen fiber based film by crosslinking with casein, keratin or SPI: Effect of crosslinking process and concentrations of proteins. International Journal of Biological Macromolecules. 2018;109:1319-1328. DOI: 10.1016/j.ijbiomac.2017.11.144
  37. 37. Picheth GF, Pirich CL, Sierakowski MR, Woehl MA, Sakakibara CN, De Souza CF, et al. Bacterial cellulose in biomedical applications: A review. International Journal of Biological Macromolecules. 2017;104:97-106. DOI: 10.1016/j.ijbiomac.2017.05.171
  38. 38. Bahram M, Mohseni N, Moghtader M. An introduction to hydrogels and some recent applications. In: Majee SB, editor. Emerging Concepts in Analysis and Applications of Hydrogels. Rijeka: IntechOpen; 2016. DOI: 10.5772/64301. Available from: https://www.intechopen.com/chapters/51535
  39. 39. Voicu G, Geanaliu-Nicolae RE, Pîrvan A-A, Andronescu E, Florin I. Synthesis, characterization and bioevaluation of drug-collagen hybrid materials for biomedical applications. International Journal of Pharmaceutics. 2016;510:474-484. DOI: 10.1016/j.ijpharm.2015.11.054
  40. 40. Ahmadi F, Oveisi Z, Samani SM, Amoozgar Z. Chitosan based hydrogels: Characteristics and pharmaceutical applications. Research in Pharmaceutical Sciences. 2015;10(1):1-16
  41. 41. Voicu G, Geanaliu-Nicolae RE, Ghitulica CD, Ficai A, Grumezescu AM, Bleotu C. Synthesis, characterization and bioevaluation of irinotecan-collagen hybrid materials for biomedical applications as drug delivery systems in tumoral treatments. Central European Journal of Chemistry. 2013;11:2134-2143. DOI: 10.2478/s11532-013-0341-1
  42. 42. Gu L, Shan T, Ma Y-X, Tay FR, Niu L-N. Novel biomedical applications of crosslinked collagen. Trends in Biotechnology. 2019;37:464-491. DOI: 10.1016/j.tibtech.2018.10.007
  43. 43. Bazrafshan Z, Stylios GK. Spinnability of collagen as a biomimetic material: A review. International Journal of Biological Macromolecules. 2019;15(129):693-705. DOI: 10.1016/j.ijbiomac.2019.02.024
  44. 44. Ardelean IL, Gudovan D, Ficai D, Ficai A, Andronescu E, Albu-Kaya MG, et al. Collagen/hydroxyapatite bone grafts manufactured by homogeneous/heterogeneous 3D printing. Materials Letters. 2018;231:179-182. DOI: 10.1016/j.matlet.2018.08.042
  45. 45. Lee KY, Mooney DJ. Alginate: Properties and biomedical applications. Progress in Polymer Science. 2012;37(1):106-126. DOI: 10.1016/j.progpolymsci.2011.06.003
  46. 46. Liu W, Griffith M, Li F. Alginate microsphere-collagen composite hydrogel for ocular drug delivery and implantation. Journal of Materials Science: Materials in Medicine. 2008;19:3365-3371. DOI: 10.1007/s10856-008-3486-2
  47. 47. Zhang H, Peng M, Cheng T, Zhao P, Qiu L, Zhou J, et al. Silver nanoparticles-doped collagen–alginate antimicrobial biocomposite as potential wound dressing. Journal of Materials Science. 2018;53:14944-14952
  48. 48. Sang L, Luo D, Xu S, Wang X, Li X. Fabrication and evaluation of biomimetic scaffolds by using collagen–alginate fibrillar gels for potential tissue engineering applications. Materials Science and Engineering: C. 2011;31(2):262-271
  49. 49. Chen X, Fan M, Tan H, Ren B, Yuan G, Jia Y, et al. Magnetic and self-healing chitosan-alginate hydrogel encapsulated gelatin microspheres via covalent cross-linking for drug delivery. Materials Science & Engineering: C. 2019;101:619-629. DOI: 10.1016/j.msec.2019.04.012
  50. 50. Wang W, Itoh S, Aizawa T, Okawa A, Sakai K, Ohkuma T, et al. Development of an injectable chitosan/marine collagen composite gel. Biomedical Materials. 2010;5:065009. DOI: 10.1088/1748-6041/5/6/065009
  51. 51. Sionkowska A, Kaczmarek B, Gadzała-Kopciuch R. Gentamicin release from chitosan and collagen composites. Journal of Drug Delivery Science and Technology. 2016;35:353-359. DOI: 10.1016/j.jddst.2016.09.001
  52. 52. Mu H, Wang Y, Wei H, Lu H, Feng Z, Yu H, et al. Collagen peptide modified carboxymethyl cellulose as both antioxidant drug and carrier for drug delivery against retinal ischaemia/reperfusion injury. Journal of Cellular & Molecular Medicine. 2018;22:5008-5019. DOI: 10.1111/jcmm.13768
  53. 53. Zhang W, Wang X-C, Li X-Y, Zhang L-L, Jiang F. A 3D porous microsphere with multistage structure and component based on bacterial cellulose and collagen for bone tissue engineering. Carbohydrate Polymers. 2020;236:116043. DOI: 10.1016/j.carbpol.2020.116043
  54. 54. Zhang W, Wang X-C, Wang J-J, Zhang L-L. Drugs adsorption and release behavior of collagen/bacterial cellulose porous microspheres. International Journal of Biological Macromolecules. 2019;140:196-205. DOI: 10.1016/j.ijbiomac.2019.08.139
  55. 55. Gorgieva S, Kokol V. Collagen- vs. gelatine-based biomaterials and their biocompatibility: Review and perspectives. In: Pignatello R, editor. Biomaterials Applications for Nanomedicine. Rijeka: IntechOpen; 2011. DOI: 10.5772/24118. Available from: https://www.intechopen.com/chapters/23617
  56. 56. Tefft S, Bentz H, Estridge TD. Collagen and heparin matrices for growth factor delivery. Journal of Controlled Release. 1997;48(1):29-33
  57. 57. Jaikumar D, Baskaran B, Vaidyanathan VG. Effect of chromium(III) gallate complex on stabilization of collagen. International Journal of Biological Macromolecules. 2017;96:429-435. DOI: 10.1016/j.ijbiomac.2016.12.049
  58. 58. Chen Y-C, Chen M, Gaffney EA, Borwn CP. Effect of crosslinking in cartilage-like collagen microstructures. Journal of Mechanical Behavior of Biomedical Materials. 2017;66:138-143. DOI: 10.1016/j.jmbbm.2016.10.006
  59. 59. Kishore V, Iyer R, Frandsen A, Nguyen T-U. In vitro characterization of electrochemically compacted collagen matrices for corneal applications. Biomedical Materials. 2016;11:055008. DOI: 10.1088/1748-6041/11/5/055008
  60. 60. Dias J, Diakonis VF, Lorenzo M, Gonzalez F, Porras K, Douglas S, et al. Corneal stromal elasticity and viscoelasticity assessed by atomic force microscopy after different cross linking protocols. Experimental Eye Research. 2015;138:1-5. DOI: 10.1016/j.exer.2015.06.015
  61. 61. Aldahlawi NH, Hayes S, O’Brart DPS, Akhbanbetova A, Littlechild SL, Meek KM. Enzymatic resistance of corneas crosslinked using riboflavin in conjunction with low energy, high energy, and pulsed UVA irradiation modes. Investigative Ophthalmology & Visual Science. 2016;57:1547-1552. DOI: 10.1167/iovs.15-18769
  62. 62. Martinez AW, Caves JM, Ravi S, Li W, Chaikof EL. Effects of crosslinking on the mechanical properties, drug release and cytocompatibility of protein polymers. Acta Biomaterialia. 2014;10:26-33. DOI: 10.1016/j.actbio.2013.08.029
  63. 63. van Luyn MJA, van Wachem PB, Damink LO, Dijkstra PJ, Feijen J, Nieuwenhuis P. Secondary cytotoxicity of crosslinked dermal sheep collagens during repeated exposure to human fibroblasts. Biomaterials. 1992;13:1017-1024
  64. 64. Charulatha V, Rajaram A. Dimethyl 3,3′-dithiobispropionimidate: A novel crosslinking reagent for collagen. Journal of Biomedical Materials Research. 2001;54:122-128
  65. 65. Tang Z, Yue Y. Crosslinkage of collagen by polyglycidyl ethers. American Society for Artificial Internal Organs Journal. 1995;41:72-78
  66. 66. Nishi C, Nakajima N, Ikada Y. In vitro evaluation of cytotoxicity of diepoxy compounds used for biomaterial modification. Journal of Biomedical Materials Research. 1995;29:829-834
  67. 67. van Wachem PB, van Luyn MJA, Nieuwenhuis P, Koerten HK, Damink LO, Hoopen HT, et al. In vivo degradation of processed dermal sheep collagen evaluation with transmission electron microscopy. Biomaterials. 1991;12:215-223
  68. 68. Pieper JS, Oosterhof A, Dijkstra PJ, Veerkamp JH, van Kuppevelt TH. Preparation and characterization of porous crosslinked collagenous matrixes containing bioavailable chondroitin sulfate. Biomaterials. 1999;20:847-858
  69. 69. Nagaoka H, Nagaoka H, Walter R, Boushell LW, Miguez PA, Burton A, et al. Characterization of genipin-modified dentin collagen. BioMed Research International. 2014:702821. DOI: 10.1155/2014/702821
  70. 70. Balakrishnan B, Jayakrishnan A. Self-cross-linking biopolymers as injectable in situ forming biodegradable scaffolds. Biomaterials. 2005;26(18):3941-3951
  71. 71. Mu C, Liu F, Cheng Q, Li H, Wu B, Zhang G, et al. Collagen cryogel cross-linked by dialdehyde starch. Macromolecular Materials and Engineering. 2010;295(2):100-107
  72. 72. Sisson K, Zhang C, Farach-Carson MC, Chase DB, Rabolt JF. Evaluation of cross-linking methods for electrospun gelatin on cell growth and viability. Biomacromolecules. 2009;10(7):1675-1680
  73. 73. Tan H, Wu B, Li C, Mu C, Li H, Lin W. Collagen cryogel cross-linked by naturally derived dialdehyde carboxymethyl cellulose. Carbohydrate Polymers. 2015;129:17-24. DOI: 10.1016/j.carbpol.2015.04.029
  74. 74. Zhang X, Do MD, Casey P, Sulistio A, Qiao GG, Lundin L, et al. Chemical crosslinking of gelatin with natural phenolic compounds as studied by high-resolution NMR spectroscopy. Biomacromolecules. 2010;11(4):1125-1132
  75. 75. Kim S, Nimni ME, Yang Z, Han B. Chitosan/gelatin-based films crosslinked by proanthocyanidin. Journal of Biomedical Materials Research Part B: Applied Biomaterials. 2005;75(2):442-450
  76. 76. He L, Mu C, Ski J, Zhang Q, Shi B, Lin Q. Modification of collagen with a natural cross-linker, procyanidin. International Journal of Biological Macromolecules. 2011;48(2):354-359
  77. 77. Jackson JK, Zhao J, Wong W, Burt HB. The inhibition of collagenase induced degradation of collagen by the galloyl-containing polyphenols tannic acid, epigallocatechin gallate and epicatechin gallate. Journal of Materials Science: Materials for Medicine. 2010;2:1435-1443
  78. 78. Pena C, Caba K, Eceiza A, Ruseckaite R, Mondragon I. Enhancing water repellence and mechanical properties of gelatin films by tannin addition. Bio/Technology. 2010;101:6836-6842
  79. 79. Davidenko N, Bax DV, Schuster CF, Farndale RW, Hamaia SW, Best SM, et al. Optimisation of UV irradiation as a binding site conserving method for crosslinking collagen-based scaffolds. Journal of Materials Science: Materials in Medicine. 2016;27:14. DOI: 10.1007/s10856-015-5627-8
  80. 80. Rowland CR, Colucci LA, Guilak F. Fabrication of anatomically-shaped cartilage constructs using decellularized cartilage-derived matrix scaffolds. Biomaterials. 2016;91:57-72. DOI: 10.1016/j.biomaterials.2016.03.012
  81. 81. Krishnakumar GS, Gostynska N, Dapporto M, Campodoni E, Montesi M, Panseri S, et al. Evaluation of different cross-linking agents on hybrid biomimetic collagen-hydroxyapatite composites for regenerative medicine. International Journal of Biological Macromolecules. 2018;106:739-748. DOI: 10.1016/j.ijbiomac.2017.08.076
  82. 82. Heck T, Faccio G, Richter M, Linda T-ML. Enzyme-catalyzed protein crosslinking. Applied Microbiology & Biotechnology. 2013;97:461-475. DOI: 10.1007/s00253-012-4569-z
  83. 83. Jus S, Stachel I, Schloegl W, Pretzler M, Friess W, Meyer M, et al. Cross-linking of collagen with laccases and tyrosinases. Materials Science and Engineering C. 2011;31(5):1068-1077
  84. 84. Wu X, Liu Y, Liu A, Wang W. Improved thermal-stability and mechanical properties of type I collagen by crosslinking with casein, keratin and soy protein isolate using transglutaminase. International Journal of Biological Macromolecules. 2017;98:292-301. DOI: 10.1016/j.ijbiomac.2017.01.127
  85. 85. Zhao L, Li X, Zhao J, Ma S, Fan D, Zhu C, et al. A novel smart injectable hydrogel prepared by microbial transglutaminase and human-like collagen: Its characterization and biocompatibility. Materials Science & Engineering. C, Materials for Biological Applications. 2016;68:317-326. DOI: 10.1016/j.msec.2016.05.108
  86. 86. Li X, Fan D. Smart collagen hydrogels based on 1-ethyl-3-methylimidazolium acetate and microbial transglutaminase for potential applications in tissue engineering and cancer therapy. ACS Biomaterials Science & Engineering. 2019;5(7):3523-3536. DOI: 10.1021/acsbiomaterials.9b00393
  87. 87. Makris EA, Responte DJ, Paschos NK, Hu JC, Athanasiou KA. Developing functional musculoskeletal tissues through hypoxia and lysyl oxidase-induced collagen cross-linking. Proceedings of National Academy of Sciences, USA. 2014;111(45):E4832-E4841. DOI: 10.1073/pnas.1414271111
  88. 88. Choi Y, Kim HJ, Min KS. Effects of proanthocyanidin, a crosslinking agent, on physical and biological properties of collagen hydrogel scaffold. Restorative Dentistry Endodontics. 2016;41(4):296-303. DOI: 10.5395/rde.2016.41.4.296
  89. 89. Oryan A, Kamali A, Moshiri A, Baharvand H, Daemi H. Chemical crosslinking of biopolymeric scaffolds: Current knowledge and future directions of crosslinked engineered bone scaffolds. International Journal of Biological Macromolecules. 2018;107:678-688. DOI: 10.1016/j.ijbiomac.2017.08.184
  90. 90. Kanamala M, Wilson WR, Yang M, Palmer BD, Wu Z. Mechanisms and biomaterials in pH-responsive tumour targeted drug delivery: A review. Biomaterials. 2016;85:152-167. DOI: 10.1016/j.biomaterials.2016.01.061
  91. 91. Liu D, Yang F, Xiong F, Gu N. The smart drug delivery system and its clinical potential. Theranostics. 2016;6:1306-1323. DOI: 10.7150/thno.14858
  92. 92. Andersen JT, Dalhus B, Viuff D, Ravn BT, Gunnarsen KS, Plumridge A, et al. Extending serum half-life of albumin by engineering neonatal c receptor (FcRn) binding. Journal of Biological Chemistry. 2014;289:13492-13502. DOI: 10.1074/jbc.M114.549832
  93. 93. Fu C-CR, Shek E, Fleitman JS, de Leung MC. Collagen Containing Ophthalmic Formulation, Eur. Patent 90119626.1. Munich: European Patent Office; 1990
  94. 94. Ghica MV, Albu MG, Popa L, Leca M, Brăzdaru L, Cotruţ C, et al. Drug delivery systems based on collagen-tannic acid matrices. Revue Roumaine de Chimie. 2009;54(11):1103-1110
  95. 95. Xeroudaki M, Thangavelu M, Lennikov A, Ratnayake A, Bisevac J, Petrovski G, et al. A porous collagen-based hydrogel and implantation method for corneal stromal regeneration and sustained local drug delivery. Scientific Reports. 2020;10:16936. DOI: 10.1038/s41598-020-73730-9
  96. 96. Ramírez OJ, Alvarez S, Contreras-Kallens P, Barrera NP, Aguayo S, Schuh CMAP. Type I collagen hydrogels as a delivery matrix for royal jelly derived extracellular vesicles. Drug Delivery. 2020;27(1):1308-1318. DOI: 10.1080/10717544.2020.1818880
  97. 97. Dharunya G, Duraipandy N, Lakra R, Korapatti SK, Jayavel R, Kira MS. Curcumin cross-linked collagen aero-gels with controlled anti-proteolytic and pro-angiogenic efficacy. Biomedical Materials. 2016;11:045011. DOI: 10.1088/1748-6041/11/4/045011
  98. 98. Chuysinuan P, Chimnoi N, Reuk-Ngam N, Khlaychan P, Makarasen A, Wetprasit N, et al. Development of gelatin hydrogel pads incorporated with Eupatorium adenophorum essential oil as antibacterial wound dressing. Polymer Bulletin. 2018;76:701-724. DOI: 10.1007/s00289-018-2395-x
  99. 99. Bettini S, Bonfrate V, Syrgiannis Z, Sannino A, Salvatore L, Madaghiele M, et al. Biocompatible collagen paramagnetic scaffold for controlled drug release. Biomacromolecules. 2015;16:2599-2608. DOI: 10.1021/acs.biomac.5b00829
  100. 100. Reis LA, Chiu LL, Liang Y, Hyunh K, Momen A, Radisic M. A peptide-modified chitosan–collagen hydrogel for cardiac cell culture and delivery. Acta Biomaterialia. 2012;8:1022-1036
  101. 101. Mei E, Li S, Song J, Xing R, Li Z, Yan X. Self-assembling collagen/alginate hybrid hydrogels for combinatorial photothermal and immuno tumor therapy. Colloids and Surfaces A: Physicochemical and Engineering Aspects. 2019;577:570-575. DOI: 10.1016/j.colsurfa.2019.06.023
  102. 102. Choi M, Chung JH, Cho Y, Hong BY, Hong J. Nano-film modification of collagen hydrogels for controlled growth factor release. Chemical Engineering Science. 2015;137:626-630. DOI: 10.1016/j.ces.2015.07.011
  103. 103. Choi D, Heo J, Park JH, Jo Y, Jeong H, Chang M, et al. Nano-film coatings onto collagen hydrogels with desired drug release. Journal of Industrial and Engineering Chemistry. 2016;36925:326-333. DOI: 10.1016/j.jiec.2016.02.023
  104. 104. Djavid GE, Tabaie SM, Tajali SB, Totounchi M, Farhoud A, Fateh M, et al. Application of a collagen matrix dressing on a neuropathic diabetic foot ulcer: A randomised control trial. Journal of Wound Care. 2020;2(29,Sup3):S13-S18. DOI: 10.12968/jowc.2020.29.Sup3.S13
  105. 105. Nguyen CH, Vu MQ, Phan TT, Vu TQ, Vo QA, Bach GL, et al. Novel pH-sensitive hydrogel beads based on carrageenan and fish scale collagen for allopurinol drug delivery. Journal of Polymers and the Environment. 2020;28:1795-1810. DOI: 10.1007/s10924-020-01727-6
  106. 106. Wang X, Ronsin O, Gravez B, Farman N, Baumberger T, Jaisser F, et al. Nanostructured dense collagen-polyester composite hydrogels as amphiphilic platforms for drug delivery. Advanced Science. 2021;18(8):2004213. DOI: 10.1002/advs.202004213
  107. 107. Li M, Han M, Sun Y, Hua Y, Chen G, Zhang L. Oligoarginine mediated collagen/chitosan gel composite for cutaneous wound healing. International Journal of Biological Macromolecules. 2019;122:1120-1127. DOI: 10.1016/j.ijbiomac.2018.09.061
  108. 108. Chiu LL, Radisic M. Controlled release of thymosin β4 using collagen–chitosan composite hydrogels promotes epicardial cell migration and angiogenesis. Journal of Controlled Release. 2011;155:376-385. DOI: 10.1016/j.jconrel.2011.05.026
  109. 109. Davidson BS, Izzo F, Cromeens DM, Stephens LC, Siddik ZH, Curley SA. Collagen matrix cisplatin prevents local tumor growth after margin-positive resection. Journal of Surgical Research. 1995;58:618-624
  110. 110. Ning S, Trisler K, Brown DM, Yu NY, Kanekal S, Lundsten MJ, et al. Intratumoral radioimmunotherapy of a human colon cancer xenograft using a sustained-release gel. Radiotherapy and Oncology. 1996;39:179-189
  111. 111. Kanekal S, Sahai A, Jones RE, Brown D. Enhanced retention of 195mPt-cisplatin in murine tumors with a novel injectable sustained release drug delivery system. Pharmaceutical Research. 1995;12:S-228
  112. 112. Slavin J, Nash JR, Kingsnorth AN. Effect of transforming growth factor beta and basic fibroblast growth factor on steroid impaired healing intestinal wound. British Journal of Surgery. 1992;79:69-72
  113. 113. Weiner AL, Carpenter-Green SS, Soehngen EC, Lenk RP, Popescu MC. Liposome–collagen gel matrix: A novel sustained drug delivery system. Journal of Pharmaceutical Sciences. 1985;74:922-925
  114. 114. Pajean M, Herbage D. Effect of collagen on liposome permeability. International Journal of Pharmaceutics. 1993;91:209-216
  115. 115. Fan X, Liang Y, Cui Y, Li F, Sun Y, Yang J, et al. Development of tilapia collagen and chitosan composite hydrogels for nanobody delivery. Colloids Surface B Biointerfaces. 2020;195:111261. DOI: 10.1016/j.colsurfb.2020.111261
  116. 116. Wong FSY, Tsang KK, Chu AMW, Chan BP, Yao KM, Lo ACY. Injectable cell-encapsulating composite alginate-collagen platform with inducible termination switch for safer ocular drug delivery. Biomaterials. 2019;201:53-67. DOI: 10.1016/j.biomaterials.2019.01.032
  117. 117. Bradley WG, Wilkes GL. Some mechanical property considerations of reconstituted collagen for drug release supports. Biomaterials, Medical Devices, and Artificial Organs. 1977;5:159-175
  118. 118. McNamara TF, Ramamurthy NS, Golub LM. Non-Antibacterial Tetracycline Compositions Possessing Anti-Collagenolytic Properties and Methods of Preparing and Using Same. US Patent 4935412. Alexandria: US Patent and trademark office; 1990
  119. 119. Minabe M, Takeuchi K, Tamura K, Hori T, Umemoto T. Subgingival administration of tetracycline on a collagen film. Journal of Periodontology. 1989;60:552-556
  120. 120. Song S-Z, Morawiecki A, Pierce GF, Pitt CG. Collagen Film for Sustained Delivery of Proteins, Eur. Patent 92305467.3. Munich: European Patent Office; 1992
  121. 121. Friess W, Lee G, Groves MJ. Insoluble collagen matrices for prolonged delivery of proteins. Pharmaceutical Development & Technology. 1996;1:185-193
  122. 122. Gil CBS, Gil VSB, Carvalho SM, Silva GR, Magalhães JT, Rodrigo L, et al. Recycled collagen films as biomaterials for controlled drug delivery. New Journal of Chemistry. 2016;40:8502-8510. DOI: 10.1039/C6NJ00674D
  123. 123. Langasco R, Cadeddu B, Formato M, Lepedda AJ, Cossu M, Giunchedi P, et al. Natural collagenic skeleton of marine sponges in pharmaceutics: Innovative biomaterial for topical drug delivery. Materials Science and Engineering: C. 2017;70:710-720. DOI: 10.1016/j.msec.2016.09.041
  124. 124. Lee YI, Lee SG, Kim E, Jung I, Suk J, Kim J, et al. Anti-aging effect of an oral disintegrating collagen film: A prospective, single-arm study. International Journal of Dermatology. 2022;61(1):54-61. DOI: 10.1111/ijd.15675
  125. 125. Walczak M, Michalska-Sionkowska M, Kaczmarek B, Sionkowska A. Surface and antibacterial properties of thin films based on collagen and thymol. Materials Today Communications. 2020;22:100949. DOI: 10.1016/j.mtcomm.2020.100949
  126. 126. Skopinska-Wisniewska J, Sionkowska A, Kaminska A, Kaznica A, Jachimiak R, Drewa T. Surface characterization of collagen/elastin based biomaterials for tissue regeneration. Applied Surface Science. 2009;255(9):8286-8292
  127. 127. Cascone MC, Sim B, Downes S. Blends of synthetic and natural polymers as drug delivery systems for growth hormone. Biomaterials. 1995;16:569-574
  128. 128. Hong YJ, Chun J-S, LeeW-K. Association of collagen with calcium phosphate promoted osteogenic responses of osteoblast-like MG63 cells. Colloids and Surfaces B: Biointerfaces. 2011;83(2):245-253
  129. 129. Park JC, Hwang YS, Lee JE, Park KD, Matsumura K, Hyon SH, et al. Type I atelocollagen grafting onto ozone-treated polyurethane films: Cell attachment, proliferation, and collagen synthesis. Journal of Biomedical Materials Research. 2000;52:669-677. DOI: 10.1002/1097-4636(20001215)52:4<669::aid-jbm11>3.0.co;2-u
  130. 130. Jana P, Mitra T, Selvaraj TKR, Gnanamani A, Kundu PP. Preparation of guar gum scaffold film grafted with ethylenediamine and fish scale collagen, crosslinked with ceftazidime for wound healing application. Carbohydrate Polymers. 2016;153(20):573-581. DOI: 10.1016/j.carbpol.2016.07.053
  131. 131. Chen M, Huang Y-Q, Cao H, Liu Y, Guo H, Chen LS, et al. Collagen/chitosan film containing biotinylated glycol chitosan nanoparticles for localized drug delivery. Colloids and Surfaces B: Biointerfaces. 2015;128:339-346. DOI: 10.1016/j.colsurfb.2015.02.024
  132. 132. Martino AD, Drannikov A, Surgutskaia NS, Ozaltin K, Postnikov PS, Marina TE, et al. Chitosan-collagen based film for controlled delivery of a combination of short life anesthetics. International Journal of Biological Macromolecules. 2019;140:1183-1193. DOI: 10.1016/j.ijbiomac.2019.08.228
  133. 133. Michalska-Sionkowska M, Kaczmarek B, Walczak M, Sionkowska A. Antimicrobial activity of new materials based on the blends of collagen/chitosan/hyaluronic acid with gentamicin sulfate addition. Materials Science and Engineering: C. 2018;86:103-108. DOI: 10.1016/j.msec.2018.01.005
  134. 134. Tanum J, Heo J, Hong J. Spontaneous biomacromolecule absorption and long-term release by graphene oxide. ACS Omega. 2018;3(5):5903-5909. DOI: 10.1021/acsomega.8b00537
  135. 135. Yaşayan G, Karaca G, Akgüner ZP, Öztürk AB. Chitosan/collagen composite films as wound dressings encapsulating allantoin and lidocaine hydrochloride. International Journal of Polymeric Materials and Polymeric Biomaterials. 2020;70(9):623-635. DOI: 10.1080/00914037.2020.1740993
  136. 136. Liu T, Dan W, Dan N, Liu X, Liu X, Peng X. A novel grapheme oxide-modified collagen-chitosan bio-film for controlled growth factor release in wound healing applications. Materials Science and Engineering: C Materials for Biological Applications. 2017;77:202-211. DOI: 10.1016/j.msec.2017.03.256
  137. 137. Chen DW, Hsu Y-H, Liao J-Y, Liu S-J, Chen J-K, Ueng SW-N. Sustainable release of vancomycin, gentamicin and lidocaine from novel electrospun sandwich-structured PLGA/collagen nanofibrous membranes. International Journal of Pharmaceutics. 2012;430:335-341. DOI: 10.1016/j.ijpharm.2012.04.010
  138. 138. Thacharodi D. Collagen-chitosan composite membranes controlled transdermal delivery of nifedipine and propranolol hydrochloride. International Journal of Pharmaceutics. 1996;134:239-241. DOI: 10.1016/0378-5173(96)04453-5
  139. 139. Thacharodi D, Rao KP. Rate-controlling biopolymer membranes as transdermal delivery systems for nifedipine: Development and in vitro evaluations. Biomaterials. 1996;13:1307-1311
  140. 140. Lozeau LD, Grosha J, Kole D, Prifti F, Dominko T, Camesano TA, et al. Collagen tethering of synthetic human antimicrobial peptides cathelicidin LL37 and its effects on antimicrobial activity and cytotoxicity. Acta Biomaterialia. 2017;52:9-20. DOI: 10.1016/j.actbio.2016.12.047
  141. 141. Elangovan S, D’Mello SR, Hong L, Ross RD, Allamargot C, Dawson DV, et al. The enhancement of bone regeneration by gene activated matrix encoding for platelet derived growth factor. Biomaterials. 2014;35:737-747. DOI: 10.1016/j.biomaterials.2013.10.021
  142. 142. Barroso T, Viveiros R, Casimiro T, Aguiar-Ricardo A. Development of dual-responsive chitosan–collagen scaffolds for pulsatile release of bioactive molecules. Journal of Supercritical Fluids. 2014;94:102-112. DOI: 10.1016/j.supflu.2014.07.005
  143. 143. Intaraprasit S, Faikrua SA, Viyoch J. Efficacy evaluation of the fibroblast-seeded collagen/ chitosan scaffold on application in skin tissue engineering. Science. Asia. 2012;38:268-277. DOI: 10.2306/scienceasia1513-1874.2012.38.268
  144. 144. Mao Z, Shi H, Guo R, Ma L, Gao C, Han C, et al. Enhanced angiogenesis of porous collagen scaffolds by incorporation of TMC/DNA complexes encoding vascular endothelial growth factor. Acta Biomaterialia. 2009;5:2983-2994. DOI: 10.1016/j.actbio.2009.04.004
  145. 145. Kato A, Miyaji H, Ishizuka R, Tokunaga K, Inoue K, Kosen Y, et al. Combination of root surface modification with BMP-2 and collagen hydrogel scaffold implantation for periodontal healing in beagle dogs. Open Dentistry Journal. 2015;30(9):52-59. DOI: 10.2174/1874210601509010052
  146. 146. Moshiri A, Shahrezaee M, Shekarchi AK. Three-dimensional porous gelapin–simvastatin scaffolds promoted bone defect healing in rabbits. Calcified Tissue International. 2015;96:552-564. DOI: 10.1007/s00223-015-9981-9
  147. 147. Karri VVSR, Kuppusamy G, Talluri SV, Mannemala SS, Kollipara R, Wadhwani AD, et al. Curcumin loaded chitosan nanoparticles impregnated into collagen-alginate scaffolds for diabetic wound healing. International Journal of Biological Macromolecules. 2016;93:1519-1529. DOI: 10.1016/j.ijbiomac.2016.05.038
  148. 148. Mahmoud AA, Salama AH. Norfloxacin-loaded collagen/chitosan scaffolds for skin reconstruction: Preparation, evaluation and in-vivo wound healing assessment. European Journal of Pharmaceutical Sciences. 2016;83:155-165. DOI: 10.1016/j.ejps.2015.12.026
  149. 149. Chow JP, Simionescu DT, Warner H, Wang B, Patnaik SS, Liao J, et al. Mitigation ofdiabetes-related complications in implanted collagen and elastin scaffolds using matrix-binding polyphenol. Biomaterials. 2013;34:685-695. DOI: 10.1016/j.biomaterials.2012.09.081
  150. 150. López-Noriega A, Ruiz-Hernández E, Quinlan E, Storm G, Hennink WE, O'Brien FJ. Thermally triggered release of apro-osteogenic peptide from a functionalized collagen-based scaffold using thermo- sensitive liposomes. Journal of Controlled Release. 2014;187:158-166. DOI: 10.1016/j.jconrel.2014.05.043
  151. 151. Cao H, Chen M-M, Liu Y-Y, Huang Y-Q, Wang J-H, Chen J-D, et al. Fish collagen-based scaffold containing PLGA microspheres for controlled growth factor delivery in skin tissue engineering. Colloids and Surfaces B: Biointerfaces. 2015;136:1098-1106. DOI: 10.1016/j.colsurfb.2015.10.022
  152. 152. Urello MA, Kiick KL, Sullivan MO. ECM turnoverstimulated gene delivery through collagen-mimetic peptide-plasmid integration in collagen. Acta Biomaterialia. 2017;62:167-178. DOI: 10.1016/j.actbio.2017.08.038
  153. 153. do Amaral RJFC, Zayed NMA, Pascu EI, Cavanagh B, Hobbs C, Santarella F, et al. Functionalising collagen-based scaffolds with platelet-rich plasma for enhanced skin wound healing potential. Frontiers in Bioengineering and Biotechnology. 2019;3(7):371. DOI: 10.3389/fbioe.2019.00371
  154. 154. Lee D, Wufuer M, Kim I, Choi TH, Kim BJ, Jung HG, et al. Sequential dual-drug delivery of BMP-2 and alendronate from hydroxyapatite-collagen scaffolds for enhanced bone regeneration. Scientific Reports. 2021;12(11):746. DOI: 10.1038/s41598-020-80608-3
  155. 155. Rezaii M, Oryan S, Javeri A. Curcumin nanoparticles incorporated collagen-chitosan scffold promotes cutaneous wound healing through regulation of TGF-β1/Smad7 gene expression. Materials Science & Engineering: C. 2019;98:347-357. DOI: 10.1016/j.msec.2018.12.143
  156. 156. Marks MG, Doillon C, Silver FH. Effects of fibroblasts and basic fibroblast growth factor on facilitation of dermal wound healing by type I collagen matrices. Journal of Biomedical Materials Research. 1991;25:683-696
  157. 157. Kenley RA, Yim K, Abrams J, Ron E, Turek T, Marden LJ, et al. Biotechnology and bone graft substitutes. Pharmaceutical Research. 1993;10:1393-1401
  158. 158. Fujimura K, Bessho K, Kusumoto K, Ogawa Y, Iizuka T. Experimental studies on bone inducing activity of composites of a telopeptide type I collagen as a carrier for ectopic induction by rhBMP-2. Biochemical and Biophysical Research Communications. 1995;208:316-322
  159. 159. Horisaka Y, Okamoto Y, Matsumoto N, Yoshimura Y, Hirano A, Nishida M, et al. Histological changes of implanted collagen material during bone induction. Journal of Biomedical Materials Research. 1994;28:97-103
  160. 160. Wachol-Drewek Z, Pfeiffer M, Scholl E. Comparative investigation of drug delivery of collagen implants saturated in antibiotic solutions and a sponge containing gentamicin. Biomaterials. 1996;17:1733-1738
  161. 161. Peng YM, Alberts DS, Graham V, Surwit EA, Weiner S, Meyskens FL. Cervical tissue uptake of all-trans-retinoic acid delivered via a collagen sponge–cervical cap delivery device in patients with cervical dyplasia. Investigational New Drugs. 1986;4:245-249
  162. 162. Barbaresso RC, Rau I, Zgârian RG, Meghea A, Ghica MV. Niflumic acid-collagen delivery systems used as anti-inflammatory drugs and analgesics in dentistry. Comptes Rendus Chimie. 2014;17:12-17. DOI: 10.1016/j.crci.2013.07.007
  163. 163. Chia CLK, Shelat VG, Low W, George S, Rao J. The use of Collatamp® G, local gentamicin-collagen sponge, in reducing wound infection. International Surgery. 2014;99:565-570. DOI: 10.9738/INTSURG-D-13-00171.1
  164. 164. Bhakta G, Lim ZXH, Rai B, Lin T, Hui JH, Preswich GD, et al. The influence of collagen and hyaluronan matrices on the delivery and bioactivity of bone morphogenetic protein-2 and ectopic bone formation. Acta Biomaterialia. 2013;9(11):9098-9106. DOI: 10.1016/j.actbio.2013.07.008
  165. 165. Wen F, Chang S, Toh YC, Teoh SH, Yu H. Development of poly (lactic-coglycolicacid)-collagen scaffolds for tissue engineering. Materials Science and Engineering: C. 2007;27(2):285-292
  166. 166. Alagha A, Nourallah A, Hariri S. Characterization of dexamethasone loaded collagen-chitosan sponge and in vitro release study. Journal of Drug Delivery Science and Technology. 2020;55:101449. DOI: 10.1016/j.jddst.2019.101449
  167. 167. Schlapp M, Friess W. Collagen/PLGA microparticle composites for local controlled delivery of gentamicin. Journal of Pharmaceutical Sciences. 2003;92:2145-2151. DOI: 10.1002/jps.10460
  168. 168. David G, Bargan AI, Drobota M, Bele A, Rosca I. Comparative investigation of collagen-based hybrid 3D structures for potential biomedical applications. Materials. 2021;14(12):3313. DOI: 10.3390/ma14123313
  169. 169. Helary C, Abed A, Mosser G, Louedec L, Letourneur D, Coradin T, et al. Evaluation of dense collagen matrices as medicated wound dressing for the treatment of cutaneous chronic wounds. Biomaterials Science. 2015;3:373-382. DOI: 10.1039/C4BM00370E
  170. 170. De-Souza R, Zahedi P, Allen CJ, Piquette-Miller M. Polymeric drug delivery systems for localized cancer chemotherapy. Drug Delivery. 2010;2010(17):365-375. DOI: 10.3109003762854
  171. 171. Orsi S, Capua De A, Guarnieri D, Marasco D, Netti PA. Cell recruitment and transfection in gene activated collagen matrix. Biomaterials. 2010;31:570-576. DOI: 10.1016/j.biomaterials.2009.09.054
  172. 172. Li J, Mooney DJ. Designing hydrogels for controlled drug delivery. Nature Review Materials. 2016;1:16071. DOI: 10.1038/natrevmats.2016.71
  173. 173. Davidenko N, Campbell JJ, Thian ES, Watson CJ, Cameron RE. Collagen–hyaluronic acid scaffolds for adipose tissue engineering. Acta Biomaterialia. 2010;6(10):3957-3968
  174. 174. Stadlinger B, Pilling E, Huhle M, Mai R, Bierbaum S, Scharnweber D, et al. Evaluation of osseointegration of dental implants coated with collagen, chondroitin sulphate and BMP-4: An animal study. International Journal of Oral and Maxillofacial Surgery. 2008;37(1):54-59
  175. 175. Stamov D, Grimmer M, Salchert K, Pompe T, Werner C. Heparin intercalation into reconstituted collagen I fibrils: Impact on growth kinetics and morphology. Biomaterials. 2008;29(1):1-14
  176. 176. Gotterbarm T, Richter W, Jung M, Vilei SB, Mainil-Varlet P, Yamashita T, et al. An in vivo study of a growth-factor enhanced, cell free, two-layered collagen–tricalcium phosphate in deep osteochondral defects. Biomaterials. 2006;27(18):3387-3395
  177. 177. Wong HL, Wang MX, Cheung PT, Yao KM, Chan BP. A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity. Biomaterials. 2007;28:5369-5380. DOI: 10.1016/j.biomaterials.2007.08.016
  178. 178. Solorio L, Zwolinkski C, Lund AW, Farrell MJ, Stegemann JP. Gelatin microspheres crosslinked with genipin for local delivery of growth factors. Journal of Tissue Engineering and Regenerative Medicine. 2010;4:514-523. DOI: 10.1002/term.267
  179. 179. Huang Z, Feng Q, Yu B, Li S. Biomimetic properties of an injectable chitosan/nano-hydroxyapatite/collagen composite. Materials Science & Engineering: C. 2011;31:683-687. DOI: 10.1016/j.msec.2010.12.014
  180. 180. Lee M, Lo ACY, Cheung P, Wong D, Chan BP. Drug carrier systems based on collagen–alginate composite structures for improving the performance of GDNF-secreting HEK293 cells. Biomaterials. 2009;30:1214-1221. DOI: 10.1016/j.biomaterials.2008.11.017
  181. 181. Mansouri V, Salehi M, Omrani MD, Niknam Z, Ardeshirylajimi A. Collagen-alginate microspheres as a 3D culture system for mouse embryonic stem cells differentiation to primordial germ cells. Biologicals. 2017;48:114-120. DOI: 10.1016/j.biologicals.2017.04.003
  182. 182. Rӧssler B. Entwicklung und Untersuchung von Kollagen mikropartikelnalsWirkstoffträger in Hydrogelen am Beispiel all-trans Retinol [Thesis]. Frankfurt: University of Frankfurt; 1993
  183. 183. Gebhardt BM, Kaufman HE. Collagen as a delivery system for hydrophobic drugs: Studies with cyclosporine. Journal of Ocular Pharmacology and Therapeutics. 1995;11:319-327
  184. 184. Gebhardt BM, Varnell ED, Kaufman HE. Cyclosporine in collagen particles: Corneal penetration and suppression of allograft rejection. Journal of Ocular Pharmacology and Therapeutics. 1995;11:509-517
  185. 185. Berthold A, Cremer K, Kreuter J. Collagen microparticles: Carriers for glucocorticosteroids. European Journal of Pharmaceutics and Biopharmaceutics. 1998;45:23-29
  186. 186. Calejo MT, Almeida AJ, Fernandes AI. Exploring a new jellyfish collagen in the production of microparticles for protein delivery. Journal of Microencapsulation. 2012;29:520-531. DOI: 10.3109/02652048.2012.665089
  187. 187. Rӧssler B, Kreuter J, Ross G. Effect of collagen microparticles on the stability of retinol and its absorption into hairless mouse skin in vitro. Die Pharmazie. 1994;49:175-179
  188. 188. Kreuter J, Scherer D, Müller W, Roreger M. VerfahrenzurHerstellung von Kollagenpartikeln und ihreVerwendungalsWirkstofftra, German Patent 4038887. Munich: German Patent and Trademark Office; 1990
  189. 189. Zugravu MV, Smith RA, Reves BT, Jennings JA, Cooper JO, Haggard WO, et al. Physical properties and in vitro evaluation of collagen–chitosan–calcium phosphate microparticle-based scaffolds for bone tissue regeneration. Journal of Biomaterials Applications. 2013;28(4):566-579. DOI: 10.1177/0885328212465662
  190. 190. Vardar E, Larsson HM, Allazetta S, Engelhardt EM, Pinnagoda K, Vythilingam G, et al. Microfluidic production of bioactive fibrin micro-beads embedded in crosslinked collagen used as an injectable bulking agent for urinary incontinence treatment. Acta Biomaterialia. 2018;67:156-166. DOI: 10.1016/j.actbio.2017.11.034
  191. 191. Nicklas M, Schatton W, Heinemann S, Hanke T, Kreuter J. Preparation and characterization of marine sponge collagen nanoparticles and employment for the transdermal delivery of 17b-estradiol-hemihydrate. Drug Development & Industrial Pharmacy. 2009;35:1035-1042. DOI: 10.1080/03639040902755213
  192. 192. Jahanban-Esfahlan R, Derakhshankhah H, Haghshenas B, Massoumi B, Abbasian M, Jaymand M. A bio-inspired magnetic natural hydrogel containing gelatin and alginate as a drug delivery system for cancer chemotherapy. International Journal of Biological Macromolecules. 2020;156:438-445. DOI: 10.1016/j.ijbiomac.2020.04.074
  193. 193. Anandhakumar S, Krishnamoorthy G, Ramkumar K, Raichur A. Preparation of collagen peptide functionalized chitosan nanoparticles by ionic gelation method: An effective carrier system for encapsulation and release of doxorubicin for cancer drug delivery. Materials Science & Engineering: C. 2017;70:378-385. DOI: 10.1016/j.msec.2016.09.003
  194. 194. Guo H, Hong Z, Yi R. Core-shell collagen peptide chelated calcium/calcium alginate nanoparticles from fish scales for calcium supplementation. Journal of Food Science. 2015;80:N1595-N1601. DOI: 10.1111/1750-3841.12912
  195. 195. Choi J, Park H, Kim T, Jeong Y, Oh MH, Hyeon T, et al. Engineered collagen hydrogels for the sustained release of biomolecules and imaging agents: Promoting the growth of human gingival cells. International Journal of Nanomedicine. 2014;9(1):5189-5201. DOI: 10.2147/IJN.S71304
  196. 196. Minakuchi Y, Takeshita F, Kosaka N, Sasaki H, Yamamoto Y, Kouno M, et al. Ochiya T. Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo. Nucleic Acids Research. 2004;32(13):e109. DOI: 10.1093/nar/gnh093
  197. 197. Saska S, Teixeira LN, De Castro-Raucci LMS, Scarel-Caminaga RM, Franchi LP, Dos Santos RA, et al. Nanocellulose-collagen-apatite composite associated with osteogenic growth peptide for bone regeneration. International Journal of Biological Macromolecules. 2017;103:467-476. DOI: 10.1016/j.ijbiomac.2017.05.086
  198. 198. Sotome S, Uemura T, Kikuchi M, Chen J, Itoh S, Tanaka J, et al. Synthesis and in vivo evaluation of a novel hydroxyapatite/collagen–alginate as a bone filler and a drug delivery carrier of bone morphogenetic protein. Materials Science & Engineering: C. 2004;24:341-347. DOI: 10.1016/j.msec.2003.12.003
  199. 199. Schnell E, Klinkhammer K, Balzer S, Brook G, Klee D, Dalton P, et al. Guidance of glial cell migration and axonal growth on electrospun nanofibers of poly-ε-caprolactone and a collagen/poly-ε-caprolactone blend. Biomaterials. 2007;28(19):3012-3025
  200. 200. Bloomfield SE, Miyata T, Dunn MW, Bueser N, Stenzel KH, Rubin AL. Soluble gentamicin ophthalmic inserts as a drug delivery system. Ophthalmology. 1978;96:885-887
  201. 201. Punch PI, Costa ND, Edwards ME, Wilcox GE. The release of insoluble antibiotics from collagen ocular inserts in vitro and their insertion into the conjunctival sac of cattle. Journal of Veterinary Pharmacology and Therapeutics. 1987;10:37-42
  202. 202. Rubin AL, Stenzel KH, Miyata T, White MJ, Dunn MW. Collagen as a vehicle for drug delivery. Journal of Clinical Pharmacology. 1973;17:309-312
  203. 203. Poland DE, Kaufman HE. Clinical uses of collagen shields. Journal of Cataract Refractive Surgery. 1988;14:489-491
  204. 204. Phinney RB, Schwartz SD, Lee DA, Mondino BJ. Collagen shield delivery of gentamicin and vancomycin. Archives of Ophthalmology. 1988;106:1599-1604
  205. 205. Milani JK, Verbukh I, Pleyer U, Sumner H, Adamu SA, Halabi HP, et al. Collagen shields impregnated with gentamicin-dexamethasone as a potential drug delivery device. American Journal of Ophthalmology. 1993;116:622-627
  206. 206. Dorigo MT, De Natale R, Miglioli PA. Collagen shields delivery of netilmicin: A study of ocular pharmacokinetics. Chemotherapy. 1995;41:1-4
  207. 207. Mendicute J, Ondarra A, Eder F, Ostolaza JI, Salaberria M, Lamsfus JM. The use of collagen shields impregnated with amphotericin B to treat aspergillus keratomycosis. CLAO Journal. 1995;21:252-255
  208. 208. Palmer RM, McDonald MB. A corneal lens/shield system to promote postoperative corneal epithelial healing. Journal of Cataract Refractive Surgery. 1995;21:125-126
  209. 209. Finkelstein I, Trope GE, Meno IA, Rootsman D, Basu PK. Potential value of collagen shields as a subconjunctival depot release system. Current Eye Research. 1990;90:653-659
  210. 210. Hwang DG, Stern WH, Hwang PH, MacGowan-Smith LA. Collagen shield enhancement of topical dexamethasone penetration. Archives of Ophthalmology. 1989;107:1375-1380
  211. 211. Takeuchi K. Application of local drug delivery systems to periodontal therapy. Nihon-shishubyo-gakkai-kaishi. 1992;34:741-758
  212. 212. Matsuoka J, Sakagami K, Shiozaki S, Uchida S, Fujiwara T, Gohchi A, et al. Development of an interleukin-2 slow delivery system. Transactions of the American Society for Artificial Internal Organs. 1988;34:729-731
  213. 213. Fujioka Y, Takada Y. Collagen-based drug delivery system –development of minipellet system. Pharmaceutical Technology, Japan. 1991;7:402-409
  214. 214. Zhou J, Cao C, Ma X, Lin J. Electrospinning of silk fibroin and collagen for vascular tissue engineering. International Journal of Biological Macromolecules. 2010;47(4):514-519
  215. 215. Chen Y, Mak AFT, Wang M, Li J, Wong MS. PLLA scaffolds with biomimetic apatite coating and biomimetic apatite/collagen composite coating to enhance osteoblast-like cells attachment and activity. Surface and Coatings Technology. 2006;201(3–4):575-580
  216. 216. Nicklas M, Schatton W, Heinemann S, Hanke T, Kreuter J. Enteric coating derived from marine sponge collagen. Drug Development & Industrial Pharmacy. 2009;35:1384-1388. DOI: 10.3109/03639040902939239
  217. 217. de Bruin AF, Gosselink MP, van der Harst E, Rutten HJ. Local application of gentamicin collagen implants in the prophylaxis of surgical site infections following gastrointestinal surgery: A review of clinical experience. Techniques in Coloproctology. 2010;14:301-310. DOI: 10.1007/s10151-010-0593-0
  218. 218. Friess W, Zhou W, Groves MJ. In vivo activity of collagen matrices containing PS1, an anti-neoplastic glycan, against murine sarcoma cells. Journal of Pharmacy and Pharmacology. 2011;2(3):121-124. DOI: 10.1111/j.2042-7158.1996.tb00573.x
  219. 219. Sun X, Yang X, Song W, Ren L. Construction and evaluation of collagen-based corneal grafts using polycaprolactone to improve tension stress. ACS Omega. 2020;145(1):674-682. DOI: 10.1021/acsomega.9b03297
  220. 220. Lin Z, Wu T, Wang W, Li B, Wang M, Chen L, et al. Biofunctions of antimicrobial peptide-conjugated alginate/hyaluronic acid/collagen wound dressings promote wound healing of a mixed-bacteria-infected wound. International Journal of Biological Macromolecules. 2019;140:330-342. DOI: 10.1016/j.ijbiomac.2019.08.087
  221. 221. Abdel-Mohsen AM, Frankova J, Abdel-Rahman RM, Salem AA, Sahffiee NM, Kubena I, et al. Chitosan-glucan complex hollow fibers reinforced collagen wound dressing embedded with aloe vera. II. Multifunctional properties to promote cutaneous wound healing. International Journal of Pharmaceutics. 2020;582:119349. DOI: 10.1016/j.ijpharm.2020.119349
  222. 222. Dimatteo R, Darling NJ, Segura T. In situ forming injectable hydrogels for drug delivery and wound repair. Advanced Drug Delivery Reviews. 2018;127:167-184. DOI: 10.1016/j.addr.2018.03.007
  223. 223. Zustiak SP, Leach JB. Hydrolytically degradable poly(ethylene glycol) hydrogel scaffolds with tunable degradation and mechanical properties. Biomacromolecules. 2010;11:1348-1357. DOI: 10.1021/bm100137q
  224. 224. Narayanaswamy R, Torchilin VP. Hydrogels and their applications in targeted drug delivery. Molecules. 2019;24:E603. DOI: 10.3390/molecules24030603
  225. 225. Ruskowitz RE, DeForest AC. Photoresponsive biomaterials for targeted drug delivery and 4D cell culture. Nature Reviews Materials. 2018;3:1-17. DOI: 10.1038/natrevmats2017.87
  226. 226. Karki S, Kim H, Na S-J, Shin D, Jo K, Lee J. Thin films as an emerging platform for drug delivery. Asian Journal of Pharmaceutical Sciences. 2016;11:559-574. DOI: 10.1016/j.ajps.2016.05.004
  227. 227. Borges AF, Silva C, Coelho JF, Simões S. Oral films: Current status and future perspectives: I - Galenical development and quality attributes. Journal of Controlled Release. 2015;206:1-19. DOI: 10.1016/j.jconrel.2015.03.006
  228. 228. Castro PM, Fonte P, Sousa F, Madureira RA, Sarmento B, Pintado ME. Oral films as breakthrough tools for oral delivery of proteins/peptides. Journal of Controlled Release. 2015;211:63-73. DOI: 10.1016/j.jconrel.2015.05.258
  229. 229. Zhang Q, Lu H, Kawazoe N, Chen G. Pore size effect of collagen scaffolds on cartilage regeneration. Acta Biomaterilia. 2014;10:2005-2013. DOI: 10.1016/j.actbio.2013.12.042
  230. 230. Higuchi A, Ling Q-D, Hsu S-T, Umezawa A. Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chemical Reviews. 2012;112:4507-4540. DOI: 10.1021/cr3000169
  231. 231. Berndt M, Li Y, Seyedhassantehrani N, Yao L. Fabrication and characterization of microspheres encapsulating astrocytes for neural regeneration. ACS Biomaterials Science & Engineering. 2017;3:1313-1321. DOI: 10.1021/acsbiomaterials.6b00229
  232. 232. Yang C, Fang C. Microporous nano-hydroxyapatite/collagen/phosphatidylserine scaffolds embedding collagen microparticles for controlled drug delivery in bone tissue engineering. Materials Research. 2015;18(5):1077-1081. DOI: 10.1590/1516-1439.042115
  233. 233. Padovani GC, Feitosa VP, Sauro S, Tay FR, Durán G, Paula AJ, et al. Advances in dental materials through nanotechnology: Facts, perspectives and toxicological aspects. Trends in Biotechnology. 2015;33:621-636. DOI: 10.1016/j.tibtech.2015.09.005
  234. 234. DeFrates K, Markiewicz T, Gallo P, Rack A, Weyhmiller A, Jarmusik B, et al. Protein polymer-based nanoparticles: Fabrication and medical applications. International Journal of Molecular Sciences. 2018;19:E1717. DOI: 10.3390/ijms19061717
  235. 235. Mandal A, Farhan MK, Sastry TP. Effect of reinforced Al2O3 nanoparticles on collagen nanobiocomposite from chrome-containing leather waste for biomedical applications. Clean Technologies & Environmental Policy. 2016;18:765-773. DOI: 10.1007/s10098-015-1045-3
  236. 236. Srivatsan KV, Duraipandy N, Behum S, Lakra R, Ramamutrthy U, Saikorrapati P, et al. Effect of curcumin caged silver nanoparticle on collagen stabilization for biomedical applica-tions. International Journal of Biological Macromolecules. 2015;75:306-315. DOI: 10.1016/j.ijbiomac.2015.01.050
  237. 237. Saleh T, Ahmed E, Yu L, Hussein K, Park K-M, Lee Y-S, et al. Silver nanoparticles improve structural stability and biocompatibility of decellularized porcine liver. Artificial Cells, Nanomedicine & Biotechnology. 2018;46(2):273-284. DOI: 10.1080/21691401.2018.1457037
  238. 238. Robin JB, Keys CL, Kaminski LA, Viana MA. The effect of collagen shields on rabbit corneal reepithelialization after chemical debridement. Investigative Ophthalmology & Visual Science. 1990;31:1294-1300
  239. 239. Shaker GJ, Ueda S, LoCascio JA, Aquavella JV. Effect of collagen shield on cat corneal epithelial wound healing. Investigative Ophthalmology & Visual Science. 1989;30:1565-1568
  240. 240. Sahithi B, Ansari SK, Hameeda SK, Sahithya G, Durga PM, Yogitha L. A review on collagen based drug delivery systems. Indian Journal of Research in Pharmacy and Biotechnology. 2013;1(3):461-468
  241. 241. Fujioka K, Takada Y, Sato S, Miyata T. Novel delivery system for proteins using collagen as a carrier material: The minipellet. Journal of Controlled Release. 1995;33:307-315
  242. 242. Maeda H, Sano A, Fujioka K. Profile of rhBMP-2 release from collagen minipellet and induction of ectopic bone formation. Drug Development and Industrial Pharmacy. 2004;30(5):473-480. DOI: 10.1081/DDC-120037475
  243. 243. Lofthouse S, Nagahara S, Sedgmen B, Barcham G, Brandon M, Sano A. The application of biodegradable collagen minipellets as vaccine delivery vehicles in mice and sheep. Vaccine. 2001;19(30):4318-4327. DOI: 10.1016/s0264-410x(01)00153-0
  244. 244. Higaki M, Azechi Y, Takase T, Igarashi R, Nagahara S, Sano A, et al. Collagen minipellet as a controlled release delivery system for tetanus and diphtheria toxoid. Vaccine. 2001;19(23-24):3091-3096. DOI: 10.1016/s0264-410x(01)00039-1

Written By

Amit Kumar Verma

Submitted: 22 January 2022 Reviewed: 04 February 2022 Published: 04 July 2022