You are on page 1of 11

Special Issue

Revolutionary advances in 2-methacryloyloxyethyl phosphorylcholine


polymers as biomaterials

Kazuhiko Ishihara
Department of Materials Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan

Received 6 June 2018; accepted 24 January 2019


Published online 13 February 2019 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/jbm.a.36635

Abstract: In the last 40 years, many strategies to fabricate bio- due to the extreme hydrophilicity and electrically neutral
compatible and antithrombogenic polymers have been pro- nature of the polymers, as well as to the ability of phosphoryl-
posed, especially in Japan. The development of one such choline to induce bulk-like behavior in surrounding waters.
polymers composed of 2-methacryloyloxyethyl phosphoryl- Accordingly, these polymers have been used worldwide to
choline unit, is described in this review, with specific exam- modify the surfaces and improve the overall biocompatibility
ples of use in biomedical devices. These polymers are of such medical devices as long-term implantable artificial
typically incorporated into other materials to effectively pre- organs. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A:
vent unfavorable biological responses and reactions. For 107A: 933–943, 2019.
example, the polymers suppress protein adsorption and cell
adhesion to materials in contact with plasma or whole blood, Key Words: antithrombogenicity, phosphorylcholine group,
even in the absence of anticoagulant. These properties are surface modification, protein adsorption, cell adhesion

How to cite this article: Ishihara K. 2019. Revolutionary advances in 2-methacryloyloxyethyl phosphorylcholine polymers as bio-
materials. J Biomed Mater Res Part A 2019:107A:933–943.

DEVELOPMENTS OF ANTITHROMBOGENIC POLYMERS IN Society for Biomaterials, and produced a variety of antithrom-
JAPAN IN THE LAST 40 YEARS bogenic polymers (Fig. 1), including hydrophobic–hydrophilic
Many medical devices, including implantable cardiovascular segmented block-type polymers synthesized by Okano,10,11
artificial organs, come into contact with blood, and thrombus polyion complexes derived by Kataoka from polystyrene,12
formation at such devices is a critical issue. Accordingly, the and amorphous–crystalline surfaces fabricated by Yui.13 These
development of antithrombogenic materials has been one of polymers suppress platelet adhesion and activation on struc-
the most active areas in biomedical, medical, and pharma- turally heterogeneous, neutral, or totally hydrophilic sur-
ceutical research, starting in the 1960s. By the 1970s, sev- faces, as measured in vitro by flowing whole blood and
eral concepts had been proposed,1–9 including grafting of platelets through column packed with the polymer-coated
water-soluble polymers to surfaces, deposition of microdo- microspheres.7 Subsequently, polyion complexes and
mains using block-type segmented polymers, controlling sur- amorphous–crystalline surfaces were applied to temporary-
face charge using polyelectrolytes, and hybridization with use medical devices such as cell separation filters and cannu-
biologically active molecules. For instance, heparinization of las, respectively. Poly(2-hydroxyethyl methacrylate [HEMA]-
surfaces was found to reliably delay blood coagulation, and block-styrene) was also applied to a small-diameter vascular
thus enabled deployment of temporary-use medical devices prosthesis,10,14 which, however, was not usable as implant
such as catheters and oxygenators.9 despite good patency in animal models over 3 years.
Of note, members of the Japanese Society for Biomaterials Drawing inspiration from an article which has been pub-
were pivotal to the development and testing of antithrombo- lished in Journal of Biomedical Materials Research in 1976,15
genic materials, especially polymers. One project, “Design of in which Noishiki reported that poly(ethylene terephthalate)
multiple-phase biomedical materials,” was active from 1982 (Dacron) vascular prostheses coated with phospholipids exhibit
to 1984 with support from the Ministry of Education, Japan. long-term patency in animals, Nakabayashi of Tokyo Medical
This project brought together many members of the Japanese and Dental University synthesized 2-methacryloyloxyethyl

Correspondence to: K. Ishihara; e-mail: ishihara@mpc.t.u-tokyo.ac.jp


Contract grant sponsor: Japan Science and Technology Agency

© 2019 WILEY PERIODICALS, INC. 933


FIGURE 1. Molecular design concepts for antithrombogenic polymers proposed during last 40 years.

phosphorylcholine (MPC) and its polymers, which were PREPARATION OF MPC POLYMERS
described in 1978.16 At that time, yields of the MPC were An elegant synthetic procedure of MPC with well-defined
poor from early synthetic attempts, which prevented suffi- reaction conditions was first developed in 1987, and then
cient testing of the antithrombogenicity. After that, an effi- reported in 1990 (Fig. 2).21 This method was licensed to the
cient synthetic route has since been developed, and different Japanese chemical company, NOF Corp. Ltd., with support
types of MPC polymers have been obtained and evaluated from the Japan Science and Technology Agency (JST). An
actively for antithrombogenicity and biocompatibility.17–20 In industrial-scale facility was set up in 1994 and completed in
this review, the properties of MPC polymer-coated surfaces 1999. During this period, small modifications were made to
were summarized, as well as the biological reactions that the synthetic procedure to increase the yield to 80% and
occur at such surfaces. Some examples of MPC polymers use achieve purity >98.0%, thus enabling MPC use in bio-related
in clinical devices are also described. industry and medical industry. This plant continues to

FIGURE 2. Development process of MPC and its polymers.

934 ISHIHARA REVOLUTIONARY ADVANCES IN 2-METHACRYLOYLOXYETHYL PHOSPHORYLCHOLINE POLYMERS


SPECIAL ISSUE

FIGURE 3. Control of molecular structure of the MPC polymers.

operate today, producing almost 10 tons of MPC each year.22 Administration as Lipidure-CM5206 (NOF Corp., Japan). The
A worldwide distribution network for MPC as a reagent molecular weight is important factor to determine the solubil-
means that researchers can obtain it easily. This opened up ity of the PMB30 in water. That is, the low molecular weight
the field of MPC polymer science for application in the bio- PMB30 named PUREBRIGHT (NOF), which has below 5 × 104
medical engineering and clinical medicine fields after high as molecular weight, can dissolve in aqueous medium and
purity MPC was successfully obtained. form polymer aggregate through hydrophobic interaction.33,34
Copolymerization of MPC with other vinyl compounds So, it can be applied to the solubilizer for the poorly water-
was also systematically investigated by conventional23 or soluble bioactive compounds by the polymer aggregate.
advanced living radical polymerization24–27 into various archi-
tectures, such as block-type and graft-type copolymers.24–29
Recently, surface-initiated living radical polymerization of SURFACE PROPERTIES OF MPC POLYMERS
MPC was also demonstrated to form polymer brushes with As MPC polymers are soluble in alcohol, substrates such as
unique properties arising from the high density of polymer general metals, ceramics, and plastics can be coated by sim-
graft chains.30–32 Typically, MPC copolymerizes efficiently ple solvent evaporation. In the case of the water-insoluble
with styrene and with various alkyl methacrylates such as n- PMB30, the hydrophilic phosphorylcholine groups are large
butyl methacrylate (BMA), n-hexyl methacrylate, n-dodecyl enough so that the substrate is covered entirely, with static
methacrylate (DMA), or n-stearyl methacrylate, resulting in water contact angle 30  at equilibrium. However, a large
polymers with statistically random sequence. Naturally, the hysteresis in the contact angle is observed when the surface
subunit composition of the resulting polymer can be altered is immersed in and withdrawn from water,35 suggesting high
by simply changing the ratio of monomers added. In addition, mobility of the polymer chain at the air/water interface, as
the solubility of these polymers depends on the proportion of well as a change in the orientation of phosphorylcholine
MPC units incorporated, the hydrophobicity of the alkyl meth- groups. Consequently, equilibration is required to establish
acrylate, and the molecular weight of the polymer (Fig. 3). It surface hydrophilicity, although this is quickly achieved since
is noted that MPC polymers are soluble in water and alcohol, PMB30 is deposited at a thickness of only about 100 nm.
but not in 60–92 vol % aqueous ethanol. Poly(MPC-co-BMA) Importantly, the zeta potential of PMB30 is nearly zero, since
(PMB) is the most valuable MPC polymer, of which three internal salt bridges are formed between phosphate anions
types with different MPC unit compositions and molecular and trimethylammonium cations in phosphorylcholine.
weights are commercially available today.22,23 One is the Accordingly, many substrates have nearly zero surface zeta
water-soluble PMB80, which contains 80 mol % MPC units potential after coating with PMB30,17,22,36 even metal and
and is used as a moisture trap in contact lenses and as a com- glass, which typically have large negative zeta potential
ponent in cosmetics. The other is the water-insoluble PMB30, before coating. The lack of electrical charges may also reduce
which is used as coating in medical devices. Water insolubility interactions with biomolecules.37 Coating with MPC poly-
and chemical stability in physiological conditions (pH 7.4, mers also dramatically enhances lubricity in aqueous
ionic strength 150 mM) and during sterilization are achieved medium that depends on hydrophilicity.38–40 The increase in
by controlling the polymer molecular weight (>5 × 105) and lubricity is due to the low viscosity of the polymer, which, in
the MPC unit content (about 30 mol %). This polymer is reg- turn, arises from its structural properties, and from weak
istered in the Device Master File of the US Food and Drug interactions with surrounding waters.

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A | MAY 2019 VOL 107A, ISSUE 5 935
FIGURE 4. Hydration state of the MPC polymer at aqueous interface.

Interfacial phenomena at MPC polymers are mostly attrib- aqueous medium containing ions and eventually covered
utable to waters surrounding phosphorylcholine groups with proteins and cells. Protein adsorption to surfaces has
(Fig. 4).41 As a rule, water molecules prefer to interact with been extensively studied, including in systems consisting of
each other by strong hydrogen bonding, and thereby form a buffer supplemented with a single protein, and was found
clusters. Accordingly, ions and other hydrogen bonding part- to strongly influence the subsequent adhesion of cells.48 The
ners disrupt such clusters by directly interacting with water amount and orientation of adsorbed proteins are essential to
molecules. In contrast, hydrophobic groups tend to reinforce cell adhesion, as is the protein that accumulates at the top of
such clusters, since interactions between hydrophobic groups what could be multiple distinct protein layers, since this pro-
and water molecules consist mostly of van der Waals forces, tein will be in direct contact with environment.
which are weaker than hydrogen bonds. Using Raman spec- In cooperation with Anderson and Ziats at Case Western
troscopy, it has been revealed that the poly(MPC) aqueous Reserve University, eight human plasma proteins were found
solution and PMB30-coated surface show less destroying the to adsorb to glass and poly(BMA), as described in Journal of
hydrogen bonding between water molecules in the bulk water Biomedical Materials Research in 1991.49 These proteins
phase.42,43 It is due to weak interaction of PMB30 with water were identified by sandwich-type immunoassay, which
molecules even the phosphorylcholine group takes hydro- detects only the proteins at the top layer, and include the
philic nature. In MPC polymers, the three hydrophobic methyl major plasma proteins albumin, γ-globulin, and fibrinogen.
groups bonded to nitrogen in phosphorylcholine are surface The minor plasma proteins high-molecular-weight kininogen,
exposed because the phosphate anion and trimethylammo- complement protein C5, fibronectin, and coagulation factors
nium cation are closely linked by electrostatics. Accordingly, XII (Hageman factor XII) and VIII (von Willebrand factor)
the methyl groups induce hydrophobic hydration, in which were also detected, but at lower concentrations than the
water molecules aggregate into bulk-like structures.44,45 In major proteins. Of note, these proteins are essential to blood
other words, MPC polymers are covered with a layer of bulk- coagulation and cell adhesion. Importantly, coating with
like water.46,47 Thus, protein molecules do not accept signifi- PMB significantly reduced the adsorption of these proteins,
cant molecular interaction such as electrostatic interaction regardless of plasma concentration and contact time. These
and hydrophobic interaction on the surface. Also, water move- results were confirmed by immunoassays with fibrinogen
ment at the interface is quick as that same as that in aqueous antibodies conjugated to gold colloids, which accumulated
medium, so that, protein molecules attached on the surface strongly on glass and poly(BMA), but accumulated to a far
can detach from the surface quickly and easily. As the results, lower extent on PMB-coated devices.
amount of protein adsorbed tightly on the surface is In general, MPC polymers effectively inhibit the adhesion
extremely small on the PMB30 surface. of many cell types, including platelets and white blood cells,
by altering the composition, chemical structure, and architec-
ture of surfaces. Platelet adhesion, morphology, and activa-
INTERACTION WITH BLOOD COMPONENTS tion have been directly observed by using platelet-rich-
When hydrophilic polymer-modified substrates come into plasma and whole blood.23,24,50–58 Although MPC unit con-
contact with blood and body fluids, surfaces are wetted with tent was found to be critical, 30 mol % MPC unit was

936 ISHIHARA REVOLUTIONARY ADVANCES IN 2-METHACRYLOYLOXYETHYL PHOSPHORYLCHOLINE POLYMERS


SPECIAL ISSUE

FIGURE 5. Effects of MPC polymer modification on the surface antithrombogenicity and biocompatibility. Protein adsorption was evaluated atomic
force microscope after contact with protein solution for 90 min. Cell adhesion was evaluated by culturing Cos 7 kidney cells for 24 h. Thrombus forma-
tion was examined using small diameter polyester vascular grafts coated with SPU and SPU/MPC polymer blend. The vascular grafts were implanted
into rabbit artery for 90 min in the SPU case and 1 month in the SPU/MPC polymer case. Tissue response was observed using SPU and SPU/MPC
polymer membranes after implantation subcutaneously for 2 weeks. Tissues surrounding the specimen are stained by hematoxylin–eosin method.

sufficient to significantly suppress platelet adhesion to and tetrahydrofuran are useful in this case. These include
activation on microspheres under flow, and also even after poly(MPC-co-DMA) (PMD) and poly(MPC-co-2-ethylhexyl
1–3 h of static contact. Short-term contact with human methacrylate), which are soluble in tetrahydrofuran at
whole blood also indicated effective suppression of blood 0.5 wt %. These polymers are also more adhesive and less
coagulation on PMB30, even in the absence of anticoagu- easily stripped off due to relatively lower glass-transition
lants.51 Cells flowed on PMB also do not adhere, but instead temperatures.
survive for 3 days as floating aggregates (Fig. 5). Similar Since some conventional polymers are already used in
behaviors were observed with macrophages,53 HeLa cells, medical devices, blending with MPC polymers during device
human umbilical vein endothelial cells, and embryonic stem fabrication is a straightforward alternative to coating after
cells.58 Collectively, these observations indicate that MPC the fact. Physical incorporation of MPC polymers into the
polymer is an excellent inhibitor of cell adhesion and activa- base polymer may enhance the long-term stability of the sur-
tion as a result of suppressed protein adsorption. face. Accordingly, blending of MPC polymers was attempted to
polyethylene [PE]),68–70 polysulfone (PSf),75–77 segmented
polyurethane (SPU),28,29,53,78–82 and polydimethylsiloxane.82–85
SURFACE MODIFICATION OF BIOMATERIALS WITH THE MPC Notably, no significant phase separation was observed,
POLYMERS although microdomains may have formed. A blend of PSf and
MPC polymers are themselves mechanically unsuitable as MPC polymer was molded into a hollow fiber membrane for
medical devices. Therefore, these polymers have been used use in blood purification devices.76 This hollow fiber was fabri-
in the last 25 years only to modify the surfaces of conven- cated on a double-nozzle system with water as inner liquid
tional materials used in medical devices. These are summa- phase. Consequently, hydrophilic nature of the MPC polymers
rized in Table I. The representative MPC polymer, PMB is induced migration and accumulation at the water contact
applied widely. Surface modification using the PMB is phase, resulting in a dense inner surface with concentrated
achieved conveniently by solvent evaporation, and the thick- MPC unit. This layer is encased mainly by PSf, to which the
ness of the coat is controlled via the concentration of the hydrophobic elements of MPC polymer are also anchored.
polymer, for example, a coat of 20–100 nm is deposited at Thus, the final device has asymmetric structure, and has excel-
polymer concentrations of 0.1–1.0 wt %. However, this pro- lent mechanical properties and blood compatibility, in compar-
cess is unsuitable for coating polyolefins, which are not suffi- ison to conventional PSf hollow fibers, which are fabricated
ciently wetted by ethanol, the solvent typically used to with poly(vinylpyrrolidone) (PVPy) as pore forming reagent.
dissolve MPC polymers. Thus, MPC polymers variants with However, PVPy was, in the end, incorporated evenly across
enhanced solubility in less polar solvents such as the device, and its hydration degraded the mechanical

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A | MAY 2019 VOL 107A, ISSUE 5 937
TABLE I. Conventional Medical Substrates Modified with the MPC Polymers

Materials Method Functions References


PMMA, acrylic polymer Coating, reaction Reduced protein adsorption and bacteria 59–63
adhesion
PET Coating, grafting Reduced protein adsorption 35,64
Polyurethane Coating Antithrombogenicity, improved lubricity 39,65
PVC Coating Antibiofouling 66
PP Coating Reduced protein adsorption 23,67
PE Blending Cell adhesion resistance 68–70
UHMWPE Grafting Improved lubrication 71,72
PTFE Grafting Antithrombogenicity 73,74
PSf Blending, coating Antithrombogenicity 75–77
SPU Coating, blending Antithrombogenicity 28,29,53,78–82
PDMS Grafting, coating, blending Reduced protein adsorption 83–86
Cellulose Grafting, reacting, coating Reduced protein adsorption, 87,88
antithrombogenicity
PLA, PLGA Blending, coating Reduced protein adsorption 89,90
PEEK Grafting Improved lubrication, antithrombogenicity 91–97
Co-Cr alloy Reacting Improved lubrication 40,98,99
Ti alloy Coating, reacting Reduced protein adsorption, antibiofouling 100–105
SUS Coating Suppression of bacteria adhesion and biofilm 106
formation
HAp Coating Antibiofouling 107
DLC Coating antithrombogenicity 108
Abbreviations of materials are DLC; diamond-like carbon, HAp; hydroxyapatite, PET; poly(ethylene terephthalate), PDMS; polydimethylsiloxane,
PE; polyethylene, PEEK: poly(ether ether ketone), PLA; poly(lactic acid), PLGA; poly(lactic acid-co-glycolic acid), PP; polypropylene, PSf; polysul-
fone, PTFE; polytetrafluoroethylene, PVA; poly(vinyl chloride), SPU; segmented polyurethane, SUS; stainless steel, UHMWPE; ultra-high-
molecular weight polyethylene.

properties of the hollow fiber. In addition, conventional PSf These findings have made clear conclusions that the MPC poly-
hollow fibers became occluded within 1 h of exposure to mers can prevent unfavorable biological responses and biofoul-
whole blood circulating ex vivo through a hemodialyzer. On ing, and improve lubrication at the surface.
the other hand, PSf/MPC polymer hybrid hollow fibers did not
induce blood coagulation, even in the absence of anticoagulant. APPLICATION OF MPC POLYMERS IN MEDICAL DEVICES
The water-soluble and amphiphilic PMB80 is an especially Cardiovascular devices
convenient modifier of PSf hollow fibers during follow fiber As shown in Table II, many medical devices using the MPC
fabrication process,77 since it is soluble in aqueous media, and polymer have been approved and clinically used. These have
is adsorbed and fixed spontaneously as described to blood- been summarized well in the book chapters.109–111 Coronary
facing surfaces. This device also does not induce coagulation in guide wires developed in the United Kingdom were the first
blood circulating ex vivo for more than 1 h without anticoagu- medical devices to be coated with MPC polymers, specifically
lant treatment. PMD.112 The coat significantly suppresses thrombogenesis,
Some MPC polymers have also been blended with SPU, which occurs at 48% of uncoated guide wires. MPC polymer
such as Tecoflex.28,29,78–81 The MPC polymers are dissolved coating has also been investigated in other cardiovascular
to a mixture of chloroform and ethanol, because the SPU is devices, including left ventricular assist devices (LVADs),
soluble in the former solvent while the MPC polymer is solu- extracorporeal circuits, vascular grafts, and coronary stents.
ble in the latter solvent. Upon casting on glass, the SPU is Of these, BiodivYsio coronary stents stably coated with
deposited more quickly and forming a thin membrane at the crosslinked MPC polymers have been approved in the United
air contacting surface by faster evaporation of chloroform States and the European Union, specifically those designed
than that of ethanol. Subsequently, the MPC polymer accu- for small vessels around 2.0 mm in diameter. The coat con-
mulates on glass facing surface as ethanol evaporates. sists of poly(MPC-co-DMA-co-2-hydroxypropyl methacrylate
Importantly, addition of only 5 wt % MPC polymer against (HPMA)-co-3-(trimethoxysilyl)propyl methacrylate [TSMA])
SPU is sufficient to inhibit coagulation at the resulting sur- self-crosslinked by reaction of HPMA unit with TSMA
face, as evaluated in vitro using platelet-rich plasma and unit.113,114 The performance of the MPC polymer coating on
whole blood. To evaluate in vivo performance,80 tubings with the stent application has been summarized in a literature.115
diameter 2 mm, which are prepared by polyester microfi- To avoid eventual occlusion by abnormal growth of the
bers, were coated with the SPU/MPC polymer blend, and blood vessel, stents are coated with 1 μm thick MPC poly-
implanted as vascular prostheses in a rabbit artery (Fig. 5). mers infused with drugs, which are then slowly released.
These prostheses remained functional and patent 1 month after For example, an MPC polymer-coated stent that delivers an
implantation. In contrast, polyester tubings coated with pure anti-inflammatory steroid suppresses inflammation and
SPU completely occluded within 90 min after implantation. neointimal hyperplasia in animal models.116 Based on such

938 ISHIHARA REVOLUTIONARY ADVANCES IN 2-METHACRYLOYLOXYETHYL PHOSPHORYLCHOLINE POLYMERS


SPECIAL ISSUE

TABLE II. Medical Devices used the MPC Polymers

Medical Device Product name Manufacturer Clinical Introduction Year


Guide wire Hunter Biocompatible 1997 (FDA approval: K970938)
Stent BiodivYsio Biocompatible 2000
TriMaxx Abbott Laboratories 2007
Drug eluting stent Dexamet Biocompatible 2001
Endeavor Medtronic 2008
Artificial lung (oxygenator) Physio Sorin Biomedica 2006 (FDA approval: K061031)
Oxia ICN (Legacoat) JMS 2015
Microcatheter Londis Clinical Supply/Termo MHLW approval
Catheter Eliminate Clinical Supply/Termo MHLW approval
Artificial heart (LVAD) Evaheart Sun Medical 2011
Contact lens Proclear Cooper Vision 1998
aquair evolution Cooper Vision 2009
Tympanostomy tube – Gyrus, Grace Medical 2000 (FDA approval: K062672)
Artificial hip joint Aquala KYOCERA 2011
MHLW; Ministry of Health, Labour and Welfare, Japan.

properties, the Dexamet was commercialized in 2001 as a contact lens approved by the US Food and Drug Administra-
stent coated with crosslinked MPC polymer and preloaded tion to reduce dry-eye syndrome. The proteins and lipids in
with the anti-inflammatory drug dexamethasone. Subse- tears pass through the lenses without fouling them, provid-
quently, MPC polymers were used by Abbott Vascular ing nutrients and protection against bacterial infection. Being
Devices in the TriMaxx stent, and are also being developed strongly hydrophilic, MPC polymer also retains water, allow-
and clinically evaluated by Medtronic as coating for the ing the tear film to wet the lens evenly and thereby prevent
Endeavor stent. formation of islands that may become fouled. Ultimately, this
Extracorporeal treatments of blood require systems reduces on-eye dehydration, and is particularly helpful for
equipped with specific devices such as membrane oxygena- dry-eye sufferers.
tors and tubings. Accordingly, blood-facing surfaces have
been modified with PMD, an MPC polymer, to improve blood NOTES AND FUTURE PERSPECTIVE
compatibility. Italian company Sorin Biomedica has adopted
MPC polymers have been clearly demonstrated over the
this approach for a range of extracorporeal devices to
years as extremely wettable, and remain hydrated even
enhance platelet preservation, as well as to suppress platelet
when exposed to the atmosphere. The substrates modified
activity and complement activation.117 These devices also
with MPC polymer have high lubricity, and have low friction
suppress intraoperative thrombin formation and the associ-
coefficients similar to that of natural cartilage.71,72,120,121
ated consumption of platelets, fibrinogen, and antithrombin.
Hence, developing new joints using poly(MPC) at interfaces
Moreover, EVEARHEART, an implantable LVAD, was
is an intriguing possibility.122
approved in 2011 in Japan after 6 years in clinical trial with
Extremely low-friction poly(MPC)-modified surfaces are
supporting by the JST.118 The device is a titanium alloy
obtained using established methods of surface-initiated graft
coated with PMB30.100,119 Notably, PMB30 withstands blood
polymerization. However, a new and simple technique of pho-
flow even after 7 months postimplantation, despite being
toinduced MPC polymerization was reported.68,71,72 In this
not chemical fixation on the surface. This resistance is attrib-
approach, the water-insoluble photoinitiator benzophenone is
uted to its large molecular weight of the PMB30 (>5 × 105).
deposited onto the substrate from acetone, and is then
EVEARHEART has been implanted in more than 160 patients
exposed to ultraviolet light in an aqueous solution of MPC to
in Japan, and is in clinical trials in the United States.
trigger polymerization. An artificial hip joint cup fabricated
from crosslinked ultra-high-molecular weight PE was modi-
Ophthalmic devices fied in this manner.110 The resulting surface was found to
Ocular devices are another major commercial application of have high lubricity, also known as fluidic friction, and signifi-
MPC polymer.109 For example, soft contact lenses made from cantly reduced wear. Based on these results and support by
poly(HEMA) hydrogels, which have been available since the the JST, conventional Aquala artificial hip joint systems from
1980s, may sometimes prevent oxygen diffusion and provide KYOCERA Co. Ltd., Japan are now grafted with poly(MPC).
persistent mechanical irritation following adsorption of pro- More than 52,000 of such systems have been implanted in
teins and lipids from tears. Thus, MPC polymer was investi- the patients from November 2011 to January 2019.
gated as surface modifier to suppress protein adsorption The history of MPC polymer-based biomaterials now
and enhance wettability. Subsequently, Biocompatible spans about 40 years, like that of the Japanese Society for Bio-
Co. Ltd. has produced soft contact lenses (Proclear, Omafil- materials. In 1987, when Nakabayashi introduced to me an
con A) that contain 20% MPC unit, 80% HEMA unit, and initial history of the MPC polymer research, it was a great and
small amount of crosslinker, and which are now commer- bright trigger for me to start the syntheses and characteriza-
cially available from CooperVision Co.. Proclear is the only tions including biological evaluations of the MPC polymers. In

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A | MAY 2019 VOL 107A, ISSUE 5 939
particularly, in the last 30 years, the MPC polymers have been 13. Yui N, Sanui K, Ogata N, Kataoka K, Okano T, Sakurai Y. Effect of
microstructure of poly(propylene-oxide)-segmented polyamides
demonstrated over all biocompatibility, that is, preventing sig-
on platelet adhesion. J Biomed Mater Res 1986;20:929–943.
nificant cell adhesion and tissue reactions by reducing protein 14. Nojiri C, Nakahama S, Senshu K, Okano T, Kawagoishi N, Kido T,
adsorption. Indeed, some MPC polymer-coated implantable Sakai K, Koyanagi H, Akutsu T. A new amphiphilic block co-
medical devices have been approved and are in use. In addi- polymer with improved elastomeric properties for application in
various medical devices. ASAIO J 1993;39:M322–M326.
tion, MPC and MPC polymers have been commercialized 15. Noishiki Y. Biochemical response to dacron vascular prosthesis.
worldwide as reagents and as base of new polymer designs J Biomed Mater Res 1976;10:759–767.
with even better performance or novel features. Recently, 16. Kadoma Y, Nakabayashi N, Masuhara E, Yamauchi J.
MPC polymer-like zwitterionic polymers, such as those based Synthesis and hemolysis test of the polymer containing phos-
phorylcholine groups. Kobunshi Ronbunshu 1978;35:423–427.
on sulfobetaine and carboxybetaine, have been synthesized (in Japanese).
and evaluated as the potential for biomaterials.67,123–127 It is 17. Ishihara K. Bioinspired phospholipid polymer biomaterials for
expected that continued research into polymer biomaterials making high performance artificial organs. Sci Technol Adv Mater
2000;1:131–138.
will advance minimally invasive medical care and regenera-
18. Lewis AL. Phosphorylcholine-based polymers and their use in the
tive medicine in support of an aging society. prevention of biofouling. Colloid Surf B: Biointerfaces 2000;18:
261–275.
19. Iwasaki Y, Ishihara K. Cell membrane-inspired phospholipid poly-
ACKNOWLEDGMENTS mers for developing medical devices with excellent biointerfaces.
Researches into MPC polymers as biomaterials, from the fun- Sci Technol Adv Mater 2012;13:064101.
20. Iwasaki Y, Ishihara K. Phosphorylcholine-containing polymers for
damental stage to the development of medical devices, have
biomedical applications. Anal Bioanal Chem 2005;381:534–546.
been strongly supported by the Japanese Society for Bioma- 21. Ishihara K, Ueda T, Nakabayashi N. Preparation of phospholipid
terials (JSB), and as well as by many members of the Society polymers and their properties as polymer hydrogel membrane.
for Biomaterials (SFB). Also, as industrialization and some Polym J 1990;22:355–360.
22. Ishihara K, Fukazawa K. 2-Methacryloyloxyethyl phosphorylcho-
clinical trials have been supported by Japan Science and line polymers phosphorus-based polymers: From synthesis to
Technology Agency (JST). The author expresses sincere applications. In: Monge S, David G, editors. Bio- and Bioinspired
appreciation for these supports. Nanomaterials. Weinheim: Wiley-VCH; 2014. p 369–389.
23. Ueda T, Oshida H, Kurita K, Ishihara K, Nakabayashi N. Prepara-
tion of 2-methacryloyloxyethyl phosphorylcholine copolymers
REFERENCES with alkyl methacrylates and their blood compatibility. Polym J
1. Salyer IO, Blardinelli AJ, Ball GL 3rd, Weesner WE, Gott VL, 1992;24:1259–1269.
Rámos MD, Furuse A. New blood-compatible polymers for artifi- 24. Ishihara K, Tsuji T, Kurosaki T, Nakabayashi N. Hemocompatibility
cial heart applications. J Biomed Mater Res 1971;5:105–127. on graft copolymers composed of poly(2-methacryloyloxyethyl
2. Nyilas E, Ward RS Jr. Development of blood-compatible elasto- phosphorylcholine) side chain and poly(n-butyl methacrylate)
mers. V. Surface structure and blood compatibility of avcothane backbone. J Biomed Mater Res 1994;28:225–232.
elastomers. J Biomed Mater Res 1977;11:69–68. 25. Ma Y, Tang Y, Billingham NC, Armes SP, Lewis AL, Lloyd AW,
3. Coleman DL, Gregonis DE, Andrade JD. Blood-materials interac- Salvage JP. Well-defined biocompatible block copolymers via atom
tions: The minimum interfacial free energy and the optimum transfer radical polymerization of 2-methacryloyloxyethyl phosphor-
polar/apolar ratio hypotheses. J Biomed Mater Res 1982;16: ylcholine in protic media. Macromolecules 2003;36:3475–3484.
381–398. 26. Yusa S, Fukuda K, Yamamoto T, Ishihara K, Morishima Y. Synthe-
4. Mori Y, Nagaoka S, Takiuchi H, Kikuchi T, Noguchi N, Tanzawa H, sis of well-defined amphiphilic block copolymers having phospho-
Noishiki Y. A new antithrombogenic material with long polyethyle- lipid polymer sequences as a novel biocompatible polymer
neoxide chains. Trans Am Soc Artif Intern Organs 1982;28: micelle reagent. Biomacromolecules 2005;6:663–670.
459–463. 27. Bhuchar N, Deng Z, Ishihara K, Narain R. Detailed study of the
5. Haycox CL, Ratner BD. In vitro platelet interactions in whole reversible addition-fragmentation chain transfer polymerization
human blood exposed to biomaterial surfaces: Insights on blood and co-polymerization of 2-methacryloyloxyethyl phosphorylcho-
compatibility. J Biomed Mater Res 1993;27:1181–1193. line. Polym Chem 2011;2:632–639.
6. de Mel A, Cousins BG, Seifalian AM. Surface modification of bio- 28. Asanuma Y, Inoue Y, Yusa S, Ishihara K. Hybridization of poly
materials: A quest for blood compatibility. Int J Biomater 2012; (2-methacryloyloxyethyl phosphorylcholine-block-2-ethylhexyl
2012:707863. methacrylate) with segmented polyurethane for reducing throm-
7. Kishida A, Ueno Y, Fukudome N, Yashima E, Maruyama I, bogenicity. Colloids Surf B Biointerfaces 2013;108:239–245.
Akashi M. Immobilization of human thrombomodulin onto 29. Liu Y, Inoue Y, Sakata S, Kakinoki S, Yamaoka T, Ishihara K.
poly(ether urethane urea) for developing antithrombogenic blood- Effects of molecular architecture of phospholipid polymers on sur-
contacting materials. Biomaterials 1994;15:848–852. face modification of segmented polyurethanes. J Biomater Sci
8. Miyama H, Harumiya N, Mori Y, Tanzawa H. A new antithrombo- Polym Ed 2014;25:474–486.
genic heparinized polymer. J Biomed Mater Res 1977;11:251–265. 30. Iwata R, Suk-In P, Hoven VP, Takahara A, Akiyoshi K, Iwasaki Y.
9. Kim SW, Jacobs H. Design of nonthrombogenic polymer surfaces Control of nanobiointerfaces generated from well-defined biomi-
for blood-contacting medical devices. Blood Purif 1996;14:357–372. metic polymer brushes for protein and cell manipulations. Bioma-
10. Okano T, Nishiyama S, Shinohara I, Akaike T, Sakurai Y, cromolecules 2004;5:2308–2314.
Kataoka K, Tsuruta T. Effect of hydrophilic and hydrophobic micro- 31. Feng W, Zhu S, Ishihara K, Brash JL. Adsorption of fibrinogen and
domains on mode of interaction between block polymer and blood lysozyme on silicon grafted with poly(2-methacryloyloxyethyl
platelets. J Biomed Mater Res 1981;15:393–402. phosphorylcholine) via surface-initiated atom transfer radical poly-
11. Okano T, Aoyagi T, Kataoka K, Abe K, Sakurai Y, Shimada M, merization. Langmuir 2005;21:5980–5987.
Shinohara I. Hydrophilic-hydrophobic microdomain surfaces hav- 32. Sakata S, Inoue Y, Ishihara K. Molecular interaction forces gener-
ing an ability to suppress platelet aggregation and their in vitro ated during protein adsorption to well-defined polymer brush sur-
antithrombogenicity. J Biomed Mater Res 1986;20:919–927. faces. Langmuir 2015;31:3108–3114.
12. Kataoka K, Okano T, Sakurai Y, Nishimura T, Maeda M, Inoue S, 33. Ishihara K, Iwasaki Y, Nakabayashi N. Polymeric lipid nano-
Tsuruta T. Effect of microphase separated structure of sphere consisting of water soluble poly(2-methacryloyloxyethyl
polystyrene/polyamine graft copolymer on adhering rat platelets phosphorylcholine-co-n-butyl methacrylate). Polym J 1999;31:
in vitro. Biomaterials 1982;3:237–240. 1231–1236.

940 ISHIHARA REVOLUTIONARY ADVANCES IN 2-METHACRYLOYLOXYETHYL PHOSPHORYLCHOLINE POLYMERS


SPECIAL ISSUE

34. Ishihara K, Mu M, Konno T. Water-soluble and amphiphilic phos- 55. Sawada S, Iwasaki Y, Nakabayashi N, Ishihara K. Stress response
pholipid copolymers having 2-methacryloyloxuethyl phosphoryl- of adherent cells on a polymer blend surface composed of a seg-
choline units for the solubilization of bioactive compounds. mented polyurethane and MPC copolymers. J Biomed Mater Res
J Biomater Sci Polym Ed 2018;29:844–862. A 2006;79:476–484.
35. Ueda T, Ishihara K, Nakabayashi N. Adsorption-desorption of pro- 56. Yumoto H, Hirota K, Hirao K, Miyazaki T, Yamamoto N,
teins on phospholipid polymer surfaces evaluated by dynamic Miyamoto K, Murakami K, Fujiwara N, Matsuo T, Miyake Y. Anti-
contact angle measurement. J Biomed Mater Res 1995;29: inflammatory and protective effects of 2-methacryloyloxyethyl
381–387. phosphorylcholine polymer on oral epithelial cells. J Biomed
36. Kikuchi M, Terayama Y, Ishikawa T, Hoshino T, Kobayashi M, Mater Res A 2015;103:555–563.
Ogawa H, Masunaga H, Koike J-I, Horigome M, Ishihara K, 57. Liu G, Iwata K, Ogasawara T, Watanabe J, Fukazawa K, Ishihara K,
Takahara A. Chain dimension of polyampholytes in solution and Asawa Y, Fujihara Y, Chung UL, Moro T, Takatori Y, Takato T,
immobilized brush states. Polym J 2012;44:121–130. Nakamura K, Kawaguchi H, Hoshi K. Selection of highly osteo-
37. Ishihara K, Kitagawa T, Inoue Y. Initial cell adhesion on well- genic and chondrogenic cells from bone marrow stromal cells in
defined surface by polymer brush layers with varying chemical biocompatible polymer-coated plates. J Biomed Mater Res A
structures. ACS Appl Mater Interface 2012;1:I103–I109. 2010;92:1273–1282.
38. Foy JR, Williams PF 3rd, Powell GL, Ishihara K, Nakabayashi N, 58. Koike M, Kurosawa H, Amano Y. A round-bottom 96-well polysty-
LaBerge M. Effect of phospholipidic boundary lubrication in rigid rene plate coated with 2-methacryloyloxyethyl phosphorylcholine
and compliant hemiarthroplasty models. Proc Inst Mech Eng H as an effective tool for embryoid body formation. Cytotechnology
1999;213:5–18. 2005;47:3–10.
39. Ho SP, Nakabayashi N, Iwasaki Y, Boland T, LaBerge M. Frictional 59. Shigeta M, Tanaka T, Koike N, Yamakawa N, Usui M. Suppression
properties of poly(MPC-co-BMA) phospholipid polymer for cathe- of fibroblast and bacterial adhesion by MPC coating on acrylic
ter applications. Biomaterials 2003;24:5121–5129. intraocular lenses. J Cataract Refract Surg 2006;32:859–866.
40. Kyomoto M, Moro T, Saiga K, Miyaji F, Kawaguchi H, Takatori Y, 60. Takahashi N, Iwasa F, Inoue Y, Morisaki H, Ishihara K, Baba K.
Nakamura K, Ishihara K. Lubricity and stability of poly Evaluation of the durability and antiadhesive action of
(2-methacryloyloxyethyl phosphorylcholine) polymer layer on Co- 2-methacryloyloxyethyl phosphorylcholine grafting on an acrylic
Cr-Mo surface for hemi-arthroplasty to prevent degeneration of resin denture base material. J Prosthet Dent 2014;112:194–203.
articular cartilage. Biomaterials 2010;31:658–668. 61. Fukunishi M, Inoue Y, Morisaki H, Kuwata H, Ishihara K, Baba K. A
41. Ishihara K, Mu M, Konno T, Inoue Y, Fukazawa K. The unique polymethyl methacrylate-based acrylic dental resin surface bound
hydration state of poly(2-methacryloyloxyethyl phosphorylcho- with a photoreactive polymer inhibits accumulation of bacterial
line). J Biomater Sci Polym Ed 2017;28:884–899. plaque. Int J Prosthodont 2017;30:533–540.
42. Kitano H, Imai M, Mori T, Gemmei-Ide M, Yokoyama Y, Ishihara K. 62. Ikeya K, Iwasa F, Inoue Y, Fukunishi M, Takahashi N, Ishihara K,
Structure of water in the vicinity of phospholipid analogue copoly- Baba K. Inhibition of denture plaque deposition on complete den-
mers as studied by vibrational spectroscopy. Langmuir 2003;19: tures by 2-methacryloyloxyethyl phosphorylcholine polymer coat-
10260–10266. ing: A clinical study. J Prosthet Dent 2018;119:67–74.
43. Kitano H, Sudo K, Ichikawa K, Ide M, Ishihara K. Raman spectro- 63. Huang X, Luo C, Lin L, Zhang L, Li H, Yao K, Xu Z. UV-assisted
scopic study on the structure of water in aqueous polyelectrolyte treatment on hydrophobic acrylic IOLs anterior surface with
solutions. J Phys Chem B 2000;104:11425–11429. methacryloyloxyethyl phosphorylcholine: Reducing inflammation
44. Némethy G, Scheraga HA. Structure of water and hydrophobic and maintaining low posterior capsular opacification properties.
bonding in proteins. I. A model for the thermodynamic properties Korean J Couns Psychother 2017;75:1289–1298.
of liquid water. J Chem Phys 1962;36:3382–3400. 64. Zheng Z, Ren L, Zhai Z, Wang Y, Hang F. Surface modification on
45. Némethy G, Scheraga HA. Structure of water and hydrophobic polyethylene terephthalate films with 2-methacryloyloxyethyl phos-
bonding in proteins. II. Model for the thermodynamic properties of phorylcholine. Korean J Couns Psychother 2013;33:3041–3046.
aqueous solution of hydrocarbons. J Chem Phys 1962;36: 65. Lee I, Kobayashi K, Sun HY, Takatani S, Zhong LG. Biomembrane
3401–3417. mimetic polymer poly(2-methacryloyloxyethyl phosphorylcholine-
46. Ishihara K, Nomura H, Mihara T, Kurita K, Iwasaki Y, co-n-butyl methacrylate) at the interface of polyurethane surfaces.
Nakabayashi N. Why do phospholipid polymers reduce protein J Biomed Mater Res A 2007;82:316–322.
adsorption? J Biomed Mater Res 1998;39:323–330. 66. Campbell EJ, O’Byrne V, Stratford PW, Quirk I, Vick TA, Wiles MC,
47. Morita S, Tanaka M. Effect of sodium chloride on hydration struc- Yianni YP. Biocompatible surfaces using methacryloylphosphoryl-
tures of PMEA and P(MPC-r-BMA). Langmuir 2014;30: choline laurylmethacrylate copolymer. ASAIO J 1994;40:
10698–10703. M853–M857.
48. Horbett T, Brash JL, Norde W, editors. Proteins at Interfaces III State 67. Chen SH, Chang Y, Ishihara K. Reduced blood cell adhesion on
of the Art. Washington, DC: American Chemical Society; 2012. polypropylene substrates through a simple surface zwitterioniza-
49. Ishihara K, Ziats NP, Tierney BP, Nakabayashi N, Anderson JM. tion. Langmuir 2017;33:611–621.
Protein adsorption from human plasma is reduced on phospho- 68. Ishihara K, Iwasaki Y, Ebihara S, Shindo Y, Nakabayashi N. Photo-
lipid polymers. J Biomed Mater Res 1991;25:1397–1407. induced graft polymerization of 2-methacryloyloxyethyl phosphor-
50. Ishihara K, Aragaki R, Ueda T, Watenabe A, Nakabayashi N. ylcholine on polyethylene membrane surface for obtaining blood
Reduced thrombogenicity of polymers having phospholipid polar cell adhesion resistance. Colloids Surf B Biointerfaces 2000;18:
groups. J Biomed Mater Res 1990;24:1069–1077. 325–335.
51. Ishihara K, Oshida H, Endo Y, Ueda T, Watanabe A, 69. Iwasaki Y, Sawada S, Nakabayashi N, Khang G, Lee HB,
Nakabayashi N. Hemocompatibility of human whole blood on Ishihara K. The effect of the chemical structure of the phospholipid
polymers with a phospholipid polar group and its mechanism. polymer on fibronectin adsorption and fibroblast adhesion on the
J Biomed Mater Res 1992;25:1397–1407. gradient phospholipid surface. Biomaterials 1999;20:2185–2191.
52. Ishihara K, Ishikawa E, Iwasaki Y, Nakabayashi N. Inhibition of 70. Ishihara K, Nishiuchi D, Watanabe J, Iwasaki Y. Polyethylene/-
fibroblast cell adhesion on substrate by coating with phospholipid polymer alloy as an alternative to poly(vinyl chlo-
2-methacryloyloxyethyl phosphorylcholine polymers. J Biomater ride)-based materials. Biomaterials 2004;25:1115–1122.
Sci Polym Ed 1999;10:1047–1061. 71. Moro T, Takatori Y, Ishihara K, Konno T, Takigawa Y,
53. DeFife KM, Yun JK, Azeez A, Stack S, Ishihara K, Nakabayashi N, Matsushita T, Chung UI, Nakamura K, Kawaguchi H. Surface graft-
Colton E, Anderson JM. Adhesion and cytokine production by mono- ing of artificial joints with a biocompatible polymer for preventing
cytes on poly(2-methacryloyloxyethyl phosphorylcholine-co-alkyl periprosthetic osteolysis. Nat Mater 2004;3:829–836.
methacrylate)-coated polymers. J Biomed Mater Res 1995;29:431–439. 72. Kyomoto M, Moro T, Yamane S, Hashimoto M, Takatori Y,
54. Sawada S, Sakaki S, Iwasaki Y, Nakabayashi N, Ishihara K. Sup- Ishihara K. Effect of UV-irradiation intensity on graft polymerization
pression of the inflammatory response from adherent cells on of 2-methacryloyloxyethyl phosphorylcholine on orthopedic bear-
phospholipid polymers. J Biomed Mater Res A 2003;64:411–416. ing substrate. J Biomed Mater Res A 2014;102:3012–3023.

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A | MAY 2019 VOL 107A, ISSUE 5 941
73. Chen C, Lumsden AB, Ofenloch JC, Noe B, Campbell EJ, 90. Iwasaki Y, Sawada S, Ishihara K, Khang G, Lee HB. Reduction of
Stratford PW, Yianni YP, Taylor AS, Hanson SR. Phosphorylcho- surface-induced inflammatory reaction on PLGA/MPC polymer
line coating of ePTFE grafts reduces neointimal hyperplasia in blend. Biomaterials 2002;23:3897–3903.
canine model. Ann Vasc Surg 1997;11:74–79. 91. Kyomoto M, Ishihara K. Self-initiated surface graft polymerization of
74. Chen C, Ofenloch JC, Yianni YP, Hanson SR, Lumsden AB. Phos- 2-methacryloyloxyethyl phosphorylcholine on poly(ether ether ketone)
phorylcholine coating of ePTFE reduces platelet deposition and by photoirradiation. ACS Appl Mater Interfaces 2009;1:537–542.
neointimal hyperplasia in arteriovenous grafts. J Surg Res 1998; 92. Kyomoto M, Moro T, Takatori Y, Kawaguchi H, Nakamura K,
77:119–125. Ishihara K. Self-initiated surface grafting with poly
75. Ishihara K, Fukumoto K, Iwasaki Y, Nakabayashi N. Modification of (2-methacryloyloxyethyl phosphorylcholine) on poly(ether-ether-
polysulfone with phospholipid polymer for improvement of the ketone). Biomaterials 2010;31:1017–1024.
blood compatibility. Part 1. Surface characterization. Biomaterials 93. Kyomoto M, Moro T, Yamane S, Hashimoto M, Takatori Y,
1999;20:1545–1551. Ishihara K. Poly(ether-ether-ketone) orthopedic bearing surface mod-
76. Hasegawa T, Iwasaki Y, Ishihara K. Preparation of blood- ified by self-initiated surface grafting of poly(2-methacryloyloxyethyl
compatible hollow fibers from a polymer alloy composed of poly- phosphorylcholine). Biomaterials 2013;34:7829–7839.
sulfone and 2-methacryloyloxyethyl phosphorylcholine polymer. 94. Tateishi T, Kyomoto M, Kakinoki S, Yamaoka T, Ishihara K.
J Biomed Mater Res 2002;63:333–341. Reduced platelets and bacteria adhesion on poly(ether ether
77. Iwasaki Y, Nakabayashi N, Ishihara K. In vitro and ex vivo blood ketone) by photoinduced and self-initiated graft polymerization of
compatibility study of 2-methacryloyloxyethyl phosphorylcholine 2-methacryloyloxyethyl phosphorylcholine. J Biomed Mater Res A
(MPC) copolymer-coated hemodialysis hollow fibers. J Artif 2014;102:1342–1349.
Organs 2003;6:260–266. 95. Kawasaki Y, Iwasaki Y. Surface modification of poly(ether ether
78. Ishihara K, Tanaka S, Furukawa N, Kurita K, Nakabayashi N. ketone) with methacryloyl-functionalized phospholipid polymers
Improved blood compatibility of segmented polyurethanes by via self-initiation graft polymerization. J Biomater Sci Polym Ed
polymeric additives having phospholipid polar groups. 2014;25:895–906.
I. Molecular design of polymeric additives and their functions. 96. Shiojima T, Inoue Y, Kyomoto M, Ishihara K. High-efficiency prep-
J Biomed Mater Res 1996;32:391–399. aration of poly(2-methacryloyloxyethyl phosphorylcholine) graft-
79. Ishihara K, Shibata N, Tanaka S, Iwasaki Y, Kurosaki T, ing layer on poly(ether ether ketone) by photoinduced and self-
Nakabayashi N. Improved blood compatibility of segmented initiated graft polymerization in an aqueous solution in the pres-
polyurethane by polymeric additives having phospholipid polar ence of inorganic salt additives. Acta Biomater 2016;40:38–45.
group. II. Dispersion state of the polymeric additive and protein 97. Yamane S, Kyomoto M, Moro T, Hashimoto M, Takatori Y,
adsorption on the surface. J Biomed Mater Res 1996;32:401–408. Tanaka S, Ishihara K. Wear resistance of poly
80. Yoneyama T, Ishihara K, Nakabayashi N, Ito M, Mishima Y. Short- (2-methacryloyloxyethyl phosphorylcholine)-grafted carbon fiber
term in vivo evaluation of small-diameter vascular prosthesis com- reinforced poly(ether ether ketone) liners against metal and
posed of segmented poly(etherurethane)/2-methacryloyloxyethyl ceramic femoral heads. J Biomed Mater Res B Appl Biomater
phosphorylcholine polymer blend. J Biomed Mater Res 1998;43: 2018;106:1028–1037.
15–20. 98. Kyomoto M, Iwasaki Y, Moro T, Konno T, Miyaji F, Kawaguchi H,
81. Ogawa R, Iwasaki Y, Ishihara K. Thermal property and processabil- Takatori Y, Nakamura K, Ishihara K. High lubricious surface of
ity of elastomeric polymer alloy composed of segmented polyure- cobalt-chromium-molybdenum alloy prepared by grafting poly
thane and phospholipid polymer. J Biomed Mater Res 2002;62: (2-methacryloyloxyethyl phosphorylcholine). Biomaterials 2007;28:
214–221. 3121–3130.
82. Iwasaki Y, Aiba Y, Morimoto N, Nakabayashi N, Ishihara K. Semi- 99. Kyomoto M, Moro T, Iwasaki Y, Miyaji F, Kawaguchi H, Takatori Y,
interpenetrating polymer networks composed of biocompatible Nakamura K, Ishihara K. Superlubricious surface mimicking articu-
phospholipid polymer and segmented polyurethane. J Biomed lar cartilage by grafting poly(2-methacryloyloxyethyl phosphoryl-
Mater Res 2000;52:701–708. choline) on orthopaedic metal bearings. J Biomed Mater Res A
83. Seo JH, Matsuno R, Konno T, Takai M, Ishihara K. Surface tether- 2009;91:730–741.
ing of phosphorylcholine groups onto poly(dimethylsiloxane) 100. Snyder TA, Tsukui H, Kihara S, Akimoto T, Litwak KN,
through swelling—Deswelling methods with phospholipids moiety Kameneva MV, Yamazaki K, Wagner WR. Preclinical biocompati-
containing ABA-type block copolymers. Biomaterials 2008;29: bility assessment of the EVAHEART ventricular assist device: Coat-
1367–1376. ing comparison and platelet activation. J Biomed Mater Res A
84. Fukazawa K, Ishihara K. Simple surface treatment using amphi- 2007;81:85–92.
philic phospholipid polymers to obtain wetting and lubricity on 101. Ye SH, Johnson CA Jr, Woolley JR, Oh HI, Gamble LJ, Ishihara K,
polydimethylsiloxane-based substrates. Colloids Surf B Biointer- Wagner WR. Surface modification of a titanium alloy with a phos-
faces 2012;97:70–76. pholipid polymer prepared by a plasma-induced grafting tech-
85. Huang XD, Li HY, Lin L, Yao K. Reduced silicone oil adherence to nique to improve surface thromboresistance. Colloids Surf B
silicone intraocular lens by surface modification with Biointerfaces 2009;74:96–102.
2-methacryloyloxyethyl phosphoryl-choline. Curr Eye Res 2013;38: 102. Ye SH, Johnson CA Jr, Woolley JR, Snyder TA, Gamble LJ,
91–96. Wagner WR. Covalent surface modification of a titanium alloy with
86. Hsiue GH, Lee SD, Chang PC, Kao CY. Surface characterization a phosphorylcholine-containing copolymer for reduced thrombo-
and biological properties study of silicone rubber membrane genicity in cardiovascular devices. J Biomed Mater Res A 2009;91:
grafted with phospholipid as biomaterial via plasma induced graft 18–28.
copolymerization. J Biomed Mater Res 1998;42:134–147. 103. Yao Y, Fukazawa K, Huang N, Ishihara K. Effects of
87. Ishihara K, Nakabayashi N, Fukumoto K, Aoki J. Improvement of 3,4-dihydrophenyl groups in water-soluble phospholipid polymer
blood compatibility on cellulose dialysis membrane. I. Grafting of on stable surface modification of titanium alloy. Colloids Surf B
2-methacryloyloxyethyl phosphorylcholine on to a cellulose mem- Biointerfaces 2011;88(1):215–220.
brane surface. Biomaterials 1992;13:145–149. 104. Pan CJ, Hou YH, Liu HQ, Ding HY, Dong YX. Improved anticoagu-
88. Ishihara K, Miyazaki H, Kurosaki T, Nakabayashi N. Improvement lation of titanium by sequential immobilization of oligo(ethylene
of blood compatibility on cellulose dialysis membrane. III. Synthe- glycol) and 2-methacryloyloxyethyl phosphorylcholine. Colloids
sis and performance of water-soluble cellulose grafted with phos- Surf B Biointerfaces 2013;112:508–512.
pholipid polymer as coating material on cellulose dialysis 105. Fukuhara Y, Kyuzo M, Tsutsumi Y, Nagai A, Chen P, Hanawa T.
membrane. J Biomed Mater Res 1995;29:181–188. Phospholipid polymer electrodeposited on titanium inhibits plate-
89. Bao LL, Huang HQ, Zhao J, Nakashima K, Gong YK. Preparation let adhesion. J Biomed Mater Res B Appl Biomater 2016;104:
and characterization of zwitterionic phospholipid polymer-coated 554–560.
poly(lactic acid) nanoparticles. J Biomater Sci Polym Ed 2014;25: 106. Fujii K, Matsumoto HN, Koyama Y, Iwasaki Y, Ishihara K,
1703–1716. Takakuda K. Prevention of biofilm formation with a coating of

942 ISHIHARA REVOLUTIONARY ADVANCES IN 2-METHACRYLOYLOXYETHYL PHOSPHORYLCHOLINE POLYMERS


SPECIAL ISSUE

2-methacryloyloxyethyl phosphorylcholine polymer. J Vet Med 117. Myers GJ, Johnstone DR, Swyer WJ, McTeer S, Maxwell SL,
Sci 2008;70:167–173. Squires C, Ditmore SN, Power CV, Mitchell LB, Ditmore JE,
107. Kang S, Lee M, Kang M, Noh M, Jeon J, Lee Y, Seo JH. Develop- Aniuk LD, Hirsch GM, Buth KJ. Evaluation of Mimesys phosphoryl-
ment of anti-biofouling interface on hydroxyapatite surface by coat- choline (PC)-coated oxygenators during cardiopulmonary bypass
ing zwitterionic MPC polymer containing calcium-binding moieties in adults. J Extra Corpor Technol 2003;35:6–12.
to prevent oral bacterial adhesion. Acta Biomater 2016;40:70–77. 118. Yamane T, Nishida M, Kawamura H, Miyakoshi T, Yamazaki K.
108. Bito K, Hasebe T, Maegawa S, Maeda T, Matsumoto T, Suzuki T, Flow visualization for the implantable ventricular assist device
Hotta A. In vitro basic fibroblast growth factor (bFGF) delivery EVAHEART®. J Artif Organs 2013;16:42–48.
using an antithrombogenic 2-methacryloyloxyethyl phosphoryl- 119. Kihara S, Yamazaki K, Litwak KN, Litwak P, Kameneva MV,
choline (MPC) polymer coated with a micropatterned diamond-like Ushiyama H, Tokuno T, Borzelleca DC, Umezu M, Tomioka J,
carbon (DLC) film. J Biomed Mater Res A 2017;105:3384–3391. Tagusari O, Akimoto T, Koyanagi H, Kurosawa H, Kormos RL,
109. Goda T, Shimizu T, Ishihara K. Bioinspired biomaterials for soft Griffith BP. In vivo evaluation of a MPC polymer coated continu-
contact lenses. In: Chirila TV, editor. Biomaterials and Regenera- ous flow left ventricular assist system. Artif Organs 2003;27:
tive Medicine in Ophthalmology. Cambridge: Woodhead Publish- 188–192.
ing; 2010. p 263–279. 120. Kobayashi M, Takahara A. Tribological properties of hydrophilic
110. Kyomoto M, Moro T, Ishihara K. Phospholipid polymer grafted polymer brushes under wet conditions. Chem Rec 2010;10:
highly cross-linked UHMWPE. In: Kurtz SM, editor. UHMWPE Bio- 208–216.
materials Handbook. Cambridge: Elsevier; 2015. p 352–368. 121. Chen M, Briscoe WH, Armes SP, Cohen H, Klein J. Lubrication at
111. Lewis AL, Lloyd AW. Biomedical applications of biomimetic poly- physiological pressures by polyzwitterionic brushes. Science 2009;
mers: The phosphorylcholine-containing polymers. In: Santin M, 323:1698–1701.
Phillips G, editors. Biomimetic, Bioresponsive, and Bioactive Mate- 122. Ishihara K. Highly lubricious polymer interfaces for advanced arti-
rials: An Introduction to Integrating Materials with Tissues. ficial hip joint through biomimetic design. Polym J 2015;47:
Hoboken, NJ: Wiley; 2012. p 95–140. 585–597.
112. Gobeil F, Juneau C, Plante S. Thrombus formation on guide wires 123. Mi L, Jiang S. Integrated antimicrobial and nonfouling
during routine PTCA procedures: A scanning electron microscopic zwitterionic polymers. Angew Chem Int Ed Engl 2014;53:
evaluation. Can J Cardiol 2002;18(3):263–269. 1746–1754.
113. Lewis AL, Tolhurst LA, Stratford PW. Analysis of a 124. Schlenoff JB. Zwitteration: Coating surfaces with zwitterionic func-
phosphorylcholine-based polymer coating on a coronary stent tionality to reduce nonspecific adsorption. Langmuir 2014;30:
pre- and post-implantation. Biomaterials 2002;23(7):1697–1706. 9625–9636.
114. Lewis AL, Furze JD, Small S, Robertson JD, Higgins BJ, Taylor S, 125. Cao B, Tang Q, Cheng G. Recent advances of zwitterionic carboxy-
Ricci DR. Long-term stability of a coronary stent coating post- betaine materials and their derivatives. J Biomater Sci Polym Ed
implantation. J Biomed Mater Res 2002;63:699–705. 2014;25:1502–1513.
115. Lewis AL, Stratford PW. Phosphorylcholine-coated stents. J Long 126. Bernards M, He Y. Polyampholyte polymers as a versatile zwitter-
Term Eff Med Implants 2002;12:231–250. ionic biomaterial platform. J Biomater Sci Polym Ed 2014;25:
116. Lewis AL, Vick TA, Collias AC, Hughes LG, Palmer RR, 1479–1488.
Leppard SW, Furze JD, Taylor AS, Stratford PW. Phosphorylcho- 127. Leng C, Sun S, Zhang K, Jiang S, Chen Z. Molecular level studies
line-based polymer coatings for stent drug delivery. J Mater Sci on interfacial hydration of zwitterionic and other antifouling poly-
Mater Med 2001;12:865–870. mers in situ. Acta Biomater 2016;40:6–15.

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A | MAY 2019 VOL 107A, ISSUE 5 943

You might also like