01.12.2012 Views

Final Program Syllabus - MCJ Consulting

Final Program Syllabus - MCJ Consulting

Final Program Syllabus - MCJ Consulting

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

January 27, 2012<br />

Dear Colleagues,<br />

On behalf of the Santa Monica Sports Medicine Foundation and Kerlan Jobe Orthopedic<br />

Foundation, we are pleased to welcome you to the 12 th International Sports Medicine Fellows<br />

Conference: Comprehensive Approaches to Articular Cartilage Repair and Hip Arthroscopy, to<br />

Carlsbad, California.<br />

This two day meeting includes a variety of lectures on topics related to Articular Cartilage<br />

Repair and Hip Arthroscopy. Hands-on labs will serve to enhance your educational experience.<br />

We hope you take the time to enjoy the beautiful California coast, with its natural beauty and<br />

variety of activities.<br />

The 12 th Annual International Sports Medicine Fellows Conference will be successful as we<br />

welcome fellows from around the country, hungry for information related to the practice of<br />

sports medicine.<br />

Best Regards,<br />

Bert R. Mandelbaum, MD<br />

Course Chairman<br />

Medical Director, Santa Monica Orthopaedic and Sports Medicine Group<br />

Ralph A. Gambardella, MD<br />

Course Chairman<br />

Medical Director, Kerlan Jobe Orthopaedic Clinic<br />

ISMF <strong>Program</strong> Office: <strong>MCJ</strong> <strong>Consulting</strong> ● 2678 Bishop Drive ● Suite 250 ● San Ramon, California USA ● Tel +1 (925) 807 1190


12 th International Sports Medicine Fellows Conference:<br />

Comprehensive Approaches to Articular Cartilage Repair and Hip Arthroscopy<br />

January 27-29, 2012 • Carlsbad, California<br />

The 12 th International Sports Medicine Fellows Conference gratefully<br />

acknowledges the generous support of:<br />

Platinum Sponsors:<br />

Gold Sponsors:<br />

Silver Sponsors:


12 th International Sports Medicine Fellows Conference:<br />

Comprehensive Approaches to Articular Cartilage Repair and Hip Arthroscopy<br />

January 27-29, 2012 • Carlsbad, California<br />

Wayne K. Augé II, MD<br />

Fall River, MA, USA<br />

Robert H. Brophy, MD<br />

Chesterfield, MO, USA<br />

William Bugbee, MD<br />

La Jolla, CA, USA<br />

J.W. Thomas Byrd, MD<br />

Nashville, TN, USA<br />

Susan Chubinskaya, PhD<br />

Chicago, IL, USA<br />

Brian J. Cole, MD<br />

Chicago, IL, USA<br />

Jack Farr, MD<br />

Greenwood, Indiana, USA<br />

Wayne K. Gersoff, MD<br />

Denver, CO, USA<br />

Alberto Gobbi, MD<br />

Milan, ITALY<br />

Course Chairs:<br />

Bert R. Mandelbaum, MD, Course Co-Chair<br />

Santa Monica, California, USA<br />

Ralph A. Gambardella, MD, Course Co-Chair<br />

Los Angeles, California, USA<br />

Michael B. Gerhardt, MD, Hip Course Chair<br />

Manhattan Beach, California, USA<br />

Course Faculty<br />

William Hutchinson, MD<br />

Santa Monica, CA, USA<br />

Warren Kramer, MD<br />

Newport Beach, CA, USA<br />

David R. McAllister, MD<br />

Los Angeles, CA, USA<br />

Kai Mithoefer<br />

Boston, MA, USA<br />

Marc Safran, MD<br />

Redwood City, CA, USA<br />

Robert L. Sah, MD, ScD<br />

La Jolla, CA, USA<br />

Jason M. Scopp, MD<br />

Salisbury, Maryland, USA<br />

Jason Snibbe, MD<br />

Beverley Hill, CA, USA<br />

Jason Theodosakis, MD<br />

Tucson, AZ, USA<br />

C. Thomas Vangsness, MD<br />

Los Angeles, CA, USA


Driving Directions from Hilton Garden Inn to DJO Inc.<br />

Start: Hilton Garden Inn Carlsbad Beach<br />

6450 Carlsbad Blvd<br />

Carlsbad, CA 92011<br />

(760) 476-0800<br />

End: DJO Incorporated<br />

1430 Decision Street<br />

Vista, CA 92081<br />

Directions Distance Time<br />

Start: Drive (West) toward Carlsbad Blvd. 0.2 0:01<br />

1: Turn RIGHT to merge onto Palomar Airport Rd [CR-S12] 6.3 0:07<br />

2: Turn LEFT (North) onto Business Park Dr. 0.6 0:01<br />

3: Take 2 nd RIGHT (North-East) onto Scott St 0.1 0:01<br />

4: Take 1 st RIGHT (South) onto Decision St < 0.1 0:01<br />

End: Arrive (DJO Inc) 1340 Decision Street is on the RIGHT < 0.1 < 1min<br />

Total Route (approximate-driving times vary) 7.1 mi 11 mins<br />

If you have problems getting into the lab call:<br />

Cheresse Edwards, CERF Coordinator<br />

760-597-3942 office<br />

760-535-4704 cell - Best number to reach her


12 th International Sports Medicine Fellows Conference<br />

January 27-29, 2012● Carlsbad, California<br />

Session I<br />

7:15 AM - 7:30 AM Basics of Articular Cartilage/Repair Robert Sah, MD, ScD USA<br />

7:30 AM - 7:45 AM Articular Cartilage Imaging: New Horizons Robert Brophy, MD USA<br />

7:45 AM - 8:00 AM Microfracture Kai Mithoefer, MD USA<br />

8:00 AM - 8:15 AM Medium and Small Defects OC AutoGrafts and New Scaffolds Ralph Gambardella, MD USA<br />

8:15 AM - 8:30 AM CAIS and Juvenile Allogenic Cells Clinical Trials Bert Mandelbaum, MD USA<br />

8:30 AM - 8:45 AM Osteochondral Allograft: Basic Science William Bugbee, MD USA<br />

8:45 AM - 9:00 AM Osteochondral Allograft: Clinical Experience David McAllister, MD USA<br />

9:00 AM - 9:15 AM Discussion


Basics of Articular Cartilage & Synovial Joints<br />

11 th Annual Articular Cartilage Repair Course<br />

for Sports Medicine Fellows<br />

January 15, 2011<br />

Robert L. Sah, MD, ScD<br />

Professor of Bioengineering &<br />

Adjunct Professor of Orthopaedic Surgery<br />

University of California, San Diego<br />

La Jolla, California 92093-0412<br />

Professor,<br />

Howard Hughes Medical Institute<br />

rsah@ucsd.edu


Articular Cartilage Repair Course for Sports Medicine Fellows Robert L. Sah, MD, ScD<br />

Basics of Articular Cartilage & Synovial Joints January 16, 2011<br />

OUTLINE<br />

1) Principles of load-bearing, low-friction, and wear-resistant biomechanical functions of<br />

articular cartilage in healthy synovial joints.<br />

� Articular cartilage bears load that is >NxBW, over contact areas of ~(cm) 2 , with<br />

corresponding stress levels of ~MPa.<br />

� Normally, articular cartilage (synovial joints) has an apparent friction coefficient, μ, of<br />

~0.001-0.05 that varies markedly with the method of measurement.<br />

� Normally, articular cartilage (synovial joints) has a wear coefficient, κ, of ~0.001 (μm) 2 /N.<br />

2) The basis of the biomechanical functions of cartilage and synovial joints.<br />

� The load-bearing properties of articular cartilage exhibit visco(poro)-elasticity.<br />

o Under compressive loading, normal cartilage becomes pressurized with little<br />

compressive strain, and then slowly depressurizes as fluid exudes out and<br />

compressive strain increases.<br />

o The content and organization of its matrix constituents, especially proteoglycan and<br />

collagens, contribute to high compressive modulus and low hydraulic permeability.<br />

o In osteoarthritis, the balance between proteoglycan swelling and collagen restraint is<br />

altered due to collagen network dysfunction, resulting in cartilage swelling.<br />

� The low-friction properties of articular cartilage are due to pressure-dependent and pressureindependent<br />

phenomena (intrinsic friction coefficient with normal synovial fluid is 0.025).<br />

o In the boundary mode of lubrication, the low-friction properties of cartilage are<br />

dependent on the lubricating effects of synovial fluid (Schmidt+, Osteoarthritis<br />

Cartilage, 2007) and lubricant molecules, proteoglycan-4 (aka lubricin, superficial<br />

zone protein, PRG4) and hyaluronan (Schmidt+, Arthritis Rheum, 2007).<br />

o The “intrinsic” (boundary-mode) steady-state friction coefficient of articular cartilage,<br />

lubricated with normal synovial fluid, is ~0.025 (Schmidt+, Osteoarthritis Cartilage,<br />

2007).<br />

o The boundary lubricants in synovial fluid normally serve to lower cartilage-cartilage<br />

friction, and, consequently, shear strain of cartilage during loading.<br />

o After injury, synovial fluid is deficient in lubricating properties (Elsaid+, Arthritis<br />

Rheum, 2005), in association with decreased hyaluronan (Ballard+, JBJS, 2012)<br />

and/or PRG4 (Jay+, Arthritis Rheum, 2007).<br />

� The wear-resistance of cartilage depends on synovial fluid components, probably also<br />

boundary lubricant molecules.<br />

p. 2


Articular Cartilage Repair Course for Sports Medicine Fellows Robert L. Sah, MD, ScD<br />

Basics of Articular Cartilage & Synovial Joints January 16, 2011<br />

3) The biological and biophysical basis of articular cartilage maintenance and repair.<br />

� Heterogeneity in the Cells and Matrix of Articular Cartilage and Synovial Tissues.<br />

o The chondrocytes within articular cartilage are heterogeneous.<br />

1. Cells in the superficial zones synthesize and secrete large amounts of<br />

proteoglycan-4 lubricant (Flannery CR+, BBRC, 1999), whereas cells in the<br />

middle and deep zones synthesize and secrete large amounts of aggrecan.<br />

Don’t let the articular cartilage surface dry out (killing the surface chondrocytes) during<br />

surgery.<br />

2. Progenitor cells within cartilage and surrounding tissues may be sources for<br />

repair.<br />

o The matrix of articular cartilage is heterogeneous and anisotropic.<br />

1. The tangentially-oriented collagen network of the superficial zone provides<br />

high tensile stiffness (Temple MM+, Osteoarthritis Cartilage, 2007) and<br />

resistance to wear.<br />

2. The aggrecan content of the middle and deep zones provides high<br />

compressive stiffness and low hydraulic permeability (Chen AC+, J<br />

Biomechanics, 2001).<br />

3. The depth-variation in aggrecan content, in association with collagen network<br />

content and function (Han EH+, Biophys J, 2011), underlies the depth-varying<br />

compressive modulus of articular cartilage (Schinagl RM+, J Orthop Res,<br />

1997)<br />

� Mechanobiological Homeostasis of Load-Bearing.<br />

o The dynamics of load-bearing proteoglycan and collagen molecules in cartilage<br />

extracellular matrix depends (Hascall and Sandy) on the balance between<br />

1. synthesis and secretion by chondrocytes<br />

2. degradation by enzymes and loss driven by fluid flow.<br />

o Articular cartilage responds to dynamic compression (Sah RL+, J Orthop Res, 1989)<br />

and shear (Jin M+, Arch Biochem Biophys, 2001) via chondrocyte altering synthesis<br />

of matrix constituents, important for load-bearing.<br />

o Excessive compression can induce lethal strains to the chondrocytes.<br />

Be gentle with articular cartilage. Do not poke it or whack it too hard.<br />

� Mechanobiological Homeostasis of Articulation.<br />

o The dynamics of lubricant molecules in synovial fluid depends (Blewis ME+., Eur<br />

Cell Mater, 2007) on the balance between<br />

1. the cells lining the synovial joint, the chondrocytes and synoviocytes,<br />

secreting proteoglycan-4 and hyaluronan lubricant into synovial fluid<br />

2. degradation/loss of lubricant molecules from synovial fluid.<br />

p. 3


Articular Cartilage Repair Course for Sports Medicine Fellows Robert L. Sah, MD, ScD<br />

Basics of Articular Cartilage & Synovial Joints January 16, 2011<br />

o Articular cartilage responds to dynamic shear via chondrocytes in superficial zone<br />

altering synthesis of PRG4 lubricant into the synovial fluid (Nugent GE+, Arthritis<br />

Rheum, 2006).<br />

o PRG4 lubricant molecules are absorbed to the articular surface through interactions<br />

allowing it to be depleted and then repleted (Nugent GE+, J Orthop Res, 2007).<br />

o Continuous passive motion (of knee joints ex vivo) enhances secretion of PRG4<br />

(Nugent-Derfus GE+, Osteoarthritis Cartilage, 2007).<br />

p. 4


ARTHRITIS & RHEUMATISM<br />

Vol. 58, No. 7, July 2008, pp 2065–2074<br />

DOI 10.1002/art.23548<br />

© 2008, American College of Rheumatology<br />

Biomechanics of Cartilage Articulation<br />

Effects of Lubrication and Degeneration on Shear Deformation<br />

Benjamin L. Wong, 1 Won C. Bae, 1 June Chun, 1 Kenneth R. Gratz, 1<br />

Martin Lotz, 2 and Robert L. Sah 1<br />

Objective. To characterize cartilage shear strain<br />

during articulation, and the effects of lubrication and<br />

degeneration.<br />

Methods. Human osteochondral cores from lateral<br />

femoral condyles, characterized as normal or<br />

mildly degenerated based on surface structure, were<br />

selected. Under video microscopy, pairs of osteochondral<br />

blocks from each core were apposed, compressed<br />

15%, and subjected to relative lateral motion with<br />

synovial fluid (SF) or phosphate buffered saline (PBS)<br />

as lubricant. When cartilage surfaces began to slide<br />

steadily, shear strain (E xz) and modulus (G) overall in<br />

the full tissue thickness and also as a function of depth<br />

from the surface were determined.<br />

Results. In normal tissue with SF as lubricant, E xz<br />

was highest (0.056) near the articular surface and<br />

diminished monotonically with depth, with an overall<br />

average E xz of 0.028. In degenerated cartilage with SF as<br />

lubricant, E xz near the surface (0.28) was 5-fold that of<br />

normal cartilage and localized there, with an overall E xz<br />

of 0.041. With PBS as lubricant, E xz values near the<br />

articular surface were �50% higher than those observed<br />

with SF, and overall E xz was 0.045 and 0.062 in normal<br />

and degenerated tissue, respectively. Near the articular<br />

Supported by the National Science Foundation and the NIH.<br />

Mr. Wong’s work was supported in part by the San Diego Fellowship.<br />

Dr. Sah’s work was supported by the Howard Hughes Medical Institute<br />

through the Professors <strong>Program</strong> Grant to the University of California,<br />

San Diego.<br />

1 Benjamin L. Wong, MS, Won C. Bae, PhD, June Chun, BS,<br />

Kenneth R. Gratz, MS, Robert L. Sah, MD, ScD: University of<br />

California, San Diego, La Jolla, California; 2 Martin Lotz, MD: Scripps<br />

Research Institute, La Jolla, California.<br />

Mr. Wong and Dr. Bae contributed equally to this work.<br />

Address correspondence and reprint requests to Robert L.<br />

Sah, MD, ScD, Department of Bioengineering, Mail Code 0412,<br />

University of California, San Diego, 9500 Gilman Drive, La Jolla, CA<br />

92093-0412. E-mail: rsah@ucsd.edu.<br />

Submitted for publication September 20, 2007; accepted in<br />

revised form March 18, 2008.<br />

2065<br />

surface, G was lower with degeneration (0.06 MPa,<br />

versus 0.18 MPa in normal cartilage). In both normal<br />

and degenerated cartilage, G increased with tissue<br />

depth to 3–4 MPa, with an overall G of 0.26–0.32 MPa.<br />

Conclusion. During articulation, peak cartilage<br />

shear is highest near the articular surface and decreases<br />

markedly with depth. With degeneration and diminished<br />

lubrication, the markedly increased cartilage<br />

shear near the articular surface may contribute to<br />

progressive cartilage deterioration and osteoarthritis.<br />

The tissue-scale mechanics of articular cartilage<br />

during the compressive and shear loading of joints with<br />

normal movement have not been fully characterized.<br />

After repeated knee bending (1) and running (2), articular<br />

cartilage is compressed by �5–20% of its overall<br />

thickness. Within the cartilage tissue of dynamically<br />

compressed osteochondral blocks (3), the magnitude of<br />

compressive strain is highest near the articular surface<br />

and minimal in deeper regions, a pattern consistent with<br />

the depth-varying compressive modulus of cartilage (4).<br />

Despite extensive knowledge of cartilage compressive<br />

behavior, the overall and depth-varying shear deformation<br />

of cartilage during joint movement remains to be<br />

elucidated. Theoretical models of articulating joints can<br />

be used to predict local cartilage deformation (5,6), but<br />

are highly dependent on assumptions about boundary<br />

conditions at articulating surfaces (e.g., frictionless or<br />

adherent) and depth-varying mechanical properties (5).<br />

Detailed experimental characterization of apposing cartilage<br />

samples sliding relative to one another would<br />

further the understanding of cartilage mechanics during<br />

joint movement by elucidating the actual boundary<br />

condition at the articulating cartilage surface as well as<br />

the deformation and properties of cartilage in shear.<br />

Lubrication of articulating cartilage by synovial<br />

fluid (SF) facilitates low friction in the boundary mode


2066 WONG ET AL<br />

and may therefore affect the shear deformation of<br />

cartilage. When articulating cartilage is tested in a<br />

configuration that reveals boundary lubrication effects,<br />

SF and lubricant molecules in SF are shown to reduce<br />

articular surface interaction, as indicated by decreased<br />

friction (7,8). The replacement of SF lubricant with<br />

phosphate buffered saline (PBS) results in an elevation<br />

of boundary-mode friction (7). The dependence of local<br />

and overall shear deformation on SF may be important<br />

for maintenance of cartilage health, since acute injury or<br />

inflammatory arthritis results in reduced lubricating<br />

function of SF, and this may be involved in postinjury<br />

cartilage degeneration (9).<br />

Cartilage degeneration may also affect the way in<br />

which the tissue deforms during joint movement. As<br />

cartilage undergoes degeneration, for example with aging<br />

(10), articular surfaces become fibrillated and roughened<br />

(11). Concomitantly, cartilage mechanical properties<br />

deteriorate under compressive, tensile, and shear<br />

loading (12), i.e., cartilage deformation and strain magnitude<br />

increase in response to a particular amplitude of<br />

applied load. Such mechanical deformation of cartilage<br />

affects chondrocyte metabolism, with moderate levels of<br />

dynamic compressive and shear strain stimulating matrix<br />

synthesis, but excessive levels inhibiting matrix synthesis<br />

(13). Thus, determination of the deformation and strain<br />

of articulating cartilage, with both normal and degenerated<br />

surfaces, would provide insight into the local mechanical<br />

cues that regulate chondrocyte behavior and<br />

the consequences of degenerative changes on such cues.<br />

One experimental approach used to elucidate the<br />

local strain deformation and strain of cartilage is to track<br />

displacement of fiducial markers using a video microscopy<br />

system. Previous studies have focused on cartilage<br />

deformation and strain when samples were subjected to<br />

compression in the absence of applied shear (4,14,15).<br />

The depth-dependent compressive behavior of cartilage<br />

found in these microscale studies was consistent with<br />

findings in macroscopic tissue explants investigated by<br />

magnetic resonance imaging (3), and provided a resolution<br />

sufficient to resolve small magnitudes of strain<br />

(�1%). Thus, microscale analysis may also provide<br />

insight into cartilage shear deformation, both locally and<br />

overall. For studying cartilage shear deformation during<br />

joint movement (Figure 1A), a pair of osteochondral<br />

blocks (Figure 1B) can be compressed in apposition<br />

(Figure 1C) and subjected to lateral shearing motion<br />

(Figure 1D). Such a configuration can be controlled to<br />

mimic the biomechanical behavior of articulating cartilage<br />

at one scale (full-thickness tissue) and allow elucidation<br />

of cartilage deformation at a finer scale (regions<br />

Figure 1. A and B, Schematic representation of knee joint movements<br />

at multiple scales (A), and deformation of cartilage under no load,<br />

compression (Comp.), and compression plus shear loading (B). C,<br />

Material points in compressed cartilage before and during shear<br />

loading used to determine u�(x,z) (depth-dependent displacement<br />

[Disp.] vectors), which were then used to assess intratissue deformation.<br />

D, Schematic representation of experimental setup and loading<br />

protocol for microscale shear testing.<br />

of tissue measurements). Combined measurements of<br />

strain and stress allow determination of local and overall<br />

biomechanical properties such as shear modulus.


LUBRICATION AND DEGENERATION EFFECTS ON CARTILAGE SHEAR DURING ARTICULATION 2067<br />

Table 1. Age of the donors, and thickness and Mankin-Shapiro<br />

scores of the cartilage samples*<br />

Normal<br />

(n � 3)<br />

The hypothesis of the present study was that<br />

during joint articulation, the shear deformation of cartilage<br />

is affected by both lubrication and degeneration.<br />

To test this, we implemented a cartilage-on-cartilage<br />

sliding test to microscopically observe and analyze cartilage<br />

deformation during compression and shear. With<br />

this system, we assessed the effects of lubrication (by SF<br />

versus PBS) and degeneration (normal versus mildly<br />

degenerated) on overall and depth-varying shear strain,<br />

and determined the overall and depth-varying shear<br />

modulus of normal and mildly degenerated cartilage.<br />

MATERIALS AND METHODS<br />

Mildly degenerated<br />

(n � 3)<br />

Age, years 48 � 1.7 78 � 9.2<br />

Thickness, mm 1.86 � 0.12 2.08 � 0.15<br />

Mankin-Shapiro score 1.89 � 0.29 9.11 � 1.24<br />

* Values are the mean � SEM.<br />

Sample isolation. Osteochondral cores (each with a<br />

diameter of 10 mm) were isolated from the anterior lateral<br />

femoral condyle of a single knee block from each of 6 fresh<br />

human cadaveric tissue donors (Table 1). Cores were chosen<br />

for this study based on the gross appearance of the articular<br />

surface, i.e., normal (n � 3) or mildly degenerated (n � 3). The<br />

surface was considered normal if the modified Outerbridge<br />

grade (16) was 1, and mildly degenerated if the grade was 3.<br />

Normal specimens were from donors ages 41–60 years at the<br />

time of death, and degenerated specimens were from donors<br />

age �60 years (Figures 2A and B). The cores were immersed<br />

in PBS containing proteinase inhibitors (PIs) and stored at<br />

�80°C until use.<br />

Experimental design. On the day of testing, each core<br />

was thawed and prepared. The core was scored vertically in the<br />

cartilage using a razor blade and cut through the bone using a<br />

low-speed saw with a 0.3-mm–thick diamond-edge blade<br />

(Isomet; Buehler, Lake Bluff, IL) to yield an osteochondral<br />

fragment for histopathologic analysis and 2 approximately<br />

rectangular blocks for biomechanical testing. Each of the 2<br />

blocks had a cartilage surface area of �3 � 8mm 2 and a total<br />

thickness of �7 mm.<br />

Histopathologic analysis. Histopathologic analysis was<br />

performed to assess and confirm the state of degeneration.<br />

Samples were fixed in 4% paraformaldehyde in PBS (pH 7.0)<br />

for 24 hours, decalcified with 18% disodium EDTA in PBS for<br />

7 days, and sectioned to 7 �m using a cryostat. Some sections<br />

were stained with Alcian blue to localize sulfated glycosaminoglycans<br />

(GAGs), as previously described (17), and other<br />

sections were stained with hematoxylin and eosin to highlight<br />

cellular detail (18). Staining of samples resulted in a gradation<br />

of intensity (Figures 2C and D), reflecting the variation in<br />

GAG concentration with depth; thus, the staining method was<br />

sufficient to delineate GAG variation, such as loss due to<br />

pathology, and allow qualitative scoring. Transmitted light<br />

micrographs of the stained sections were obtained, and histologic<br />

features (19) analyzed using the Mankin-Shapiro semiquantitative<br />

scale (20). Briefly, the histopathologic characteristics<br />

assessed included structural integrity (surface irregularity<br />

and vertical and horizontal clefts), cellularity (cloning and<br />

hypocellularity), and GAG loss. A relatively high score represented<br />

more degenerated cartilage. Grades from 3 independent<br />

observers were averaged for each sample. Interobserver<br />

errors (SD) for normal and degenerated samples were reasonably<br />

small (average 1.0 and 2.1, respectively).<br />

Biomechanical testing. Samples were first tested by<br />

microscale shear testing as described below, with PBS plus PI<br />

as the lubricant. Then, samples were allowed to reswell for �4<br />

hours at 4°C.<br />

Next, samples were tested again by microscale shear<br />

Figure 2. A and B, Photographs of cross-sections of samples of<br />

normal and degenerated cartilage. C and D, Representative micrographs<br />

of Alcian blue–stained sections, showing structure detail of<br />

normal (C) and degenerated (D) cartilage. Color figure can be viewed<br />

in the online issue, which is available at http://www.arthritisrheum.org.


2068 WONG ET AL<br />

testing, this time with SF (pooled from adult bovine knees and<br />

stored at �80°C) plus PI as the lubricant. The SF had been<br />

characterized previously for boundary lubrication properties<br />

(7) and for levels of lubricant molecules (�1 mg/ml of hyaluronan<br />

and 0.45 mg/ml of proteoglycan 4 [21]). The same<br />

regions of interest as used in studies with PBS were imaged.<br />

Then, samples were allowed to reswell.<br />

<strong>Final</strong>ly, samples were tested in a macroscale shear<br />

system to assess overall mechanical properties of cartilage in<br />

shear, as described below.<br />

Microscale shear testing. Each sample, consisting of<br />

paired osteochondral blocks, was bathed for �14–18 hours in<br />

test lubricant containing PI and propidium iodide (20 �g/ml) at<br />

4°C to fluorescently highlight cell nuclei prior to microscale<br />

shear testing. Each pair of osteochondral blocks (Figures 1B<br />

and D) was then placed with cartilage in apposition in a custom<br />

biaxial loading chamber mounted onto an epifluorescence<br />

microscope for digital video imaging (4). The chamber secured<br />

one block at the bone and allowed in-plane movement of the<br />

apposing mobile block along the x-directed 8-mm lengths of<br />

the samples (Figure 1D), with orthogonally positioned plungers<br />

interfaced with either a micrometer (for axial displacement)<br />

(model 262RL; Starrett, Athol, MA) or motioncontroller<br />

(for lateral displacement) (model MFN25PP;<br />

Newport, Irvine, CA). Fluorescence images (G-2A filter; Nikon,<br />

Melville, NY) with a field of view of �3 � 2mm 2 were<br />

obtained at 5 frames/second, showing a full-thickness region of<br />

cartilage in the secured block and a partial-thickness region of<br />

cartilage in the apposing block.<br />

Cartilage deformation in the secured block was assessed<br />

during axial and shear loading. First, the block was<br />

imaged in the reference state (uncompressed). Then, axial<br />

displacement (�40 �m/second) was applied by the micrometer<br />

to induce 15% compression (1 �� z, where � z is the stretch<br />

ratio [22]) of the cartilage tissue (Figure 1C), during which<br />

time sequential images (�1.5% compression increment/frame)<br />

were acquired. Samples were then allowed to stress-relax for 1<br />

hour, which was calculated to be sufficient to reach an approximate<br />

equilibrium stress based on consideration of the characteristic<br />

time constant. Experimentally, this duration of stressrelaxation<br />

was validated to be sufficient with load decreasing to<br />

50% of the peak by 130 seconds, and load at 1 hour being only<br />

3 � 1% (mean � SEM; n � 3) higher than the load at 16<br />

hours. Subsequently, lateral motion was applied to the mobile<br />

osteochondral block (Figure 1D). Two sets of lateral displacements<br />

(�x), each consisting of �1 mm and then �1 mm<br />

(returning to initial position), were applied at 100 �m/second<br />

to the bone portion of the mobile block. The first set, followed<br />

by a pause of 12 seconds, was for preconditioning (7), while the<br />

second set was recorded for analysis. The sliding velocity was<br />

chosen based on the range of velocities (0–0.1 m/second)<br />

occurring during the loading (stance) phase of gait (23,24).<br />

Before and during the application of lateral displacements<br />

(Figure 1D), sequential images, with �10 �m of lateral<br />

movement of the mobile block between frames, were obtained.<br />

Macroscale shear testing. To assess the overall compressive<br />

and shear biomechanical properties of the cartilage of the<br />

secured block, following microscale shear tests, each secured<br />

block was subjected to a macroscale test using a biaxial<br />

mechanical tester (Mach-1 V500CS; BioSyntech, Montreal,<br />

Quebec, Canada). Test conditions were chosen to match the<br />

mechanical deformation observed in the microscale shear test,<br />

but with instrumentation allowing measurement of axial and<br />

shear loads. Each sample block was affixed, compressed 15%<br />

at a rate of 0.03%/second using a rigid stainless steel platen<br />

(�2 �m pore size, providing a no-slip boundary condition),<br />

and allowed to stress-relax for 1 hour to reach equilibrium.<br />

With increasing lateral displacement, continuous and monotonically<br />

increasing shear load waveforms were observed,<br />

confirming no-slip conditions; a plateau in shear load, which<br />

would indicate slip, was not observed. Equilibrium force was<br />

recorded and normalized to the 3 � 8mm 2 area to yield an<br />

estimate of the equilibrium compressive stress. Next, 2 sets of<br />

lateral displacements of the amplitude occurring at the surface<br />

at the onset of sliding in microscale shear tests were applied at<br />

a rate of 100 �m/second, held for 10 seconds while shear load<br />

was being recorded, and released. A pause of �12 seconds<br />

followed the first set of lateral displacements, during which the<br />

cartilage sample was continuously hydrated in PBS plus PI. As<br />

in the microscale tests, the first set was for preconditioning,<br />

while the second set was used for analysis. The lateral displacements<br />

were then repeated in the reverse direction following a<br />

5-minute pause, and shear loads occurring in the last second of<br />

the 10-second load-capture period in both directions were<br />

averaged. This average shear load was normalized to the 3 �<br />

8mm 2 area to yield an estimate of the shear stress (at which<br />

sliding occurred in the microscale test).<br />

Data collection and calculations. Digital micrographs<br />

were analyzed to determine the depth-varying and overall<br />

strains in cartilage. Images were analyzed with MatLab 7.0<br />

(MathWorks, Natick, MA) using software routines similar to<br />

those described previously (14,25,26). Briefly, an evenly distributed<br />

set of cell nuclei (�250 cells/mm 2 ), which served as<br />

fiducial markers, was selected and tracked by maximizing<br />

cross-correlation of regions surrounding each marker with the<br />

preceding, and then initial, frames. Generally, chondrocyte<br />

nuclei in normal tissue were available, and were chosen in<br />

regions up to the true articular surface (defined by a discontinuity<br />

in displacement during sliding), while for degenerated<br />

tissue, nuclei were visible up to �50–70 �m (�3%) below the<br />

true surface, due to fibrillation. Local affine mappings of<br />

nuclei were used to calculate the displacement (u�) of uniformly<br />

spaced (10-pixel) mesh points in the region of interest (�1<br />

mm � full thickness) during deformation (Figure 1D). Displacement<br />

gradients (�u�/�z, �u�/�x) were then determined by<br />

finite difference approximations, and in turn, used to determine<br />

Lagrangian compressive strain (E zz) after axial compression<br />

and shear strain (E xz) after lateral shearing, with these<br />

Lagrangian strains being appropriate for finite strains as well<br />

as small strains (27).<br />

This method was validated, and interobserver variability<br />

was assessed. Using mathematically transformed images,<br />

the calculated strain values deviated by less than 0.005 � 0.005<br />

(mean � SEM) from the theoretical values, with the error<br />

being proportionately smaller in regions exhibiting lower amplitudes<br />

of strain. Interobserver variability, assessed as the<br />

average of the SD in calculated strain between 3 observers, was<br />

�0.01 for a subset of data from all sample types (n � 4). Here


LUBRICATION AND DEGENERATION EFFECTS ON CARTILAGE SHEAR DURING ARTICULATION 2069<br />

also, the variability was proportionately lower in regions of<br />

relatively small shear magnitudes.<br />

The calculated shear strain was interpolated, averaged<br />

depthwise, and plotted as a function of tissue depth, normalized<br />

to the cartilage thickness in the compressed state. Shear<br />

strain was interpolated linearly at every 5% of the normalized<br />

tissue thickness near the articular surface (i.e., 0–0.3) and<br />

every 10% for remaining regions of the tissue depth (i.e.,<br />

0.3–1) after applied compression and prior to lateral motion.<br />

To consolidate data, strain values were averaged at the same<br />

normalized depth (0 [surface] and 1 [tidemark]) to yield a<br />

depth profile. Shear strain results were determined when<br />

surfaces were sliding, at which time shear strain was at a peak<br />

and no further changes in deformation occurred with additional<br />

lateral displacement (i.e., sliding). The overall Lagrangian<br />

shear strain was defined as half the lateral surface displacement<br />

normalized to the compressed cartilage thickness. Since<br />

the shear strain peaked near the articular surface, shear strain<br />

occurring at the top 5% was also compared.<br />

Overall cartilage properties were determined from<br />

forces recorded during compression and shear. Under compression<br />

alone, the compressive modulus of cartilage was<br />

defined as the increment in stress divided by the increment in<br />

applied compressive strain. The overall shear modulus (G) for<br />

each sample was determined as the increment in shear stress<br />

(shear load divided by surface area) divided by the increment<br />

in shear strain.<br />

The depth-varying shear moduli were estimated from<br />

the overall shear modulus and local shear strain in each<br />

sample. Since shear modulus is inversely related to shear<br />

strain, and with shear stress assumed to be constant at all<br />

depths, the local shear modulus was estimated as the overall<br />

shear modulus multiplied by the overall shear strain and<br />

divided by the local shear strain. Because these estimates<br />

depend on shear strain in the denominator, shear modulus was<br />

calculated at every 10% of the tissue depth near the articular<br />

surface (up to 20% of the depth) and every 20% of the tissue<br />

depth in the subsequent regions, to reduce noise.<br />

Statistical analysis. Data are reported as the mean �<br />

SEM, unless noted otherwise. Repeated-measures analysis of<br />

variance was used to determine the effects of normalized tissue<br />

depth (0–1 [surface–bone]), lubricant (PBS or SF), and degeneration<br />

(normal or degenerated) on tissue shear strain, and to<br />

determine the effects of tissue depth and degeneration on<br />

shear modulus.<br />

RESULTS<br />

Histopathologic findings. The overall histopathology<br />

scores were consistent with the gross (Figures<br />

2A and B) and histologic (Figures 2C and D) appearance<br />

of normal and degenerated samples. Degenerated<br />

samples had significantly (P � 0.01) higher histopathology<br />

scores than normal samples (Table 1). In degenerated<br />

samples, structural (surface irregularity, vertical<br />

clefts to transitional zone, and transverse clefts) and<br />

cellular (cloning) features exhibited mild degeneration<br />

and were scored �1-point higher than the corresponding<br />

features in normal samples. In contrast, cellularity<br />

and GAG staining scores were similarly low (i.e., normal)<br />

in normal and degenerated samples. These results<br />

confirmed that gross visualization resulted in an appropriate<br />

selection of normal and mildly degenerated samples,<br />

for articulation testing.<br />

Compressive deformation and properties. The<br />

axial compression of cartilage in the secured block<br />

resulted in compressive strain that varied significantly<br />

with depth from the articular surface (P � 0.001) but not<br />

between normal and degenerated samples (P � 0.6). E zz<br />

was relatively high near the articular surface (mean �<br />

SEM 0.38 � 0.05) and relatively low in the deepest<br />

regions (0.06 � 0.01). Transverse-oriented (E xx) and<br />

shear (E xz) strain resulting from the applied compression<br />

was low, averaging �0.03 in all samples. The<br />

equilibrium compressive modulus (E) in the fullthickness<br />

cartilage was not significantly different between<br />

sample groups (P � 0.5) (mean � SEM 0.79 �<br />

0.13 MPa in normal cartilage and 0.61 � 0.23 MPa in<br />

degenerated cartilage).<br />

Shear deformation. Qualitatively, cartilage shear<br />

loading resulted in a sequence of 4 events: 1) At the<br />

onset of applied lateral displacement, cartilage surfaces<br />

initially adhered and began to move laterally in unison.<br />

2) With increasing lateral displacement, lateral deformation<br />

and hence E xz increased and occurred throughout<br />

the tissue depth. 3) Next, lateral deformation and E xz of<br />

cartilage peaked, just as the surfaces detached and slid<br />

relative to each other. 4) With additional lateral displacement,<br />

the cartilage deformation and E xz were<br />

maintained at a steady-state peak.<br />

At the steady state, tissue displacement and E xz<br />

(Figure 3) were nonuniform and depth-varying, with<br />

shear magnitudes that were high near the articular<br />

surface and low (almost indistinguishable) near the<br />

bone. The mean � SEM maximum lateral displacement<br />

of apposing surfaces before slipping when lubricated<br />

with SF was 115 � 14 �m and 82 � 16 �m in normal and<br />

degenerated tissue, respectively. When lubricated with<br />

PBS, maximum lateral displacement in normal and<br />

degenerated tissue was 169 � 12 �m and 125 � 18 �m,<br />

respectively, before slipping. Differences in shear deformation<br />

between normal and degenerated samples were<br />

also evident (Figure 3). Shear deformation in degenerated<br />

samples (Figures 3C and D) was concentrated near<br />

the surface, while in normal samples, deformation occurred<br />

further into the tissue depth (Figures 3A and B).<br />

Quantitatively, the E xz of articulating cartilage


2070 WONG ET AL<br />

Figure 3. Micrographs obtained during shear loading of apposing<br />

normal (A and B) and degenerated (C and D) cartilage samples<br />

lubricated with phosphate buffered saline (PBS) (A and C) or synovial<br />

fluid (SF) (B and D), after maximum shear strain was achieved. The<br />

cell nuclei tracking method was used to create maps of shear strain<br />

(magnified above each panel; boxes with dotted lines indicate magnified<br />

areas). Dashed lines surround the analyzed regions on the<br />

undeformed images. Schematic at the bottom depicts the sample and<br />

testing configuration, with the dotted-line box representing the area<br />

from which the images shown were obtained and analyzed. Bars � 100<br />

�m.<br />

varied with depth from the articular surface, lubricant,<br />

and surface degeneration. In the depth profile of E xz<br />

(Figures 4A and B), the highest values were obtained<br />

near the articular surface, and E xz decreased monotonically<br />

with depth (P � 0.001). The variation of E xz with<br />

depth depended on surface degeneration (P � 0.001 for<br />

the interaction), with E xz decreasing more with depth in<br />

the degenerated sample. A similar trend was noted with<br />

testing using PBS, where E xz decreased at greater rates<br />

with depth than when tested with SF (P � 0.001 for the<br />

interaction) in both normal and degenerated samples.<br />

The combined effect of depth and either degeneration<br />

or lubricant on shear strain was evidenced as varying E xz<br />

values near the surface, but similarly low values of E xz<br />

(�0.01) in the deep layer of cartilage.<br />

Peak E xz at the surface (z � 0) (Figure 5A) and<br />

overall E xz (Figure 5B) were each significantly affected<br />

by both the lubricant used and degeneration status. The<br />

peak surface E xz (Figure 5A) was �3–5 times higher in<br />

Figure 4. Biomechanical measures of adult human articular cartilage<br />

in shear. A and B, Local shear strain (E xz) versus normalized tissue<br />

depth in normal and degenerated cartilage, as determined by microscale<br />

shear testing. Samples were tested with phosphate buffered<br />

saline (PBS) (A) or synovial fluid (SF) (B) as lubricant. C, Local shear<br />

modulus (G) versus normalized tissue depth in normal and degenerated<br />

samples, as determined by macroscale testing. Values are the<br />

mean and SEM.


LUBRICATION AND DEGENERATION EFFECTS ON CARTILAGE SHEAR DURING ARTICULATION 2071<br />

Figure 5. Effect of sample group (normal or degenerated cartilage)<br />

and of lubricant (phosphate buffered saline [PBS] or synovial fluid<br />

[SF]) on A, peak surface shear strain and B, overall shear strain. Values<br />

are the mean and SEM.<br />

samples with surface degeneration than in normal tissue<br />

(P � 0.05) with both surface lubricants. With PBS, the<br />

mean of the peak surface (Figure 5A) E xz increased<br />

from 0.11 (normal samples) to 0.41 (degenerated samples).<br />

With SF, E xz increased from 0.056 (normal samples)<br />

to 0.28 (degenerated samples). In addition, peak<br />

surface E xz values obtained when tested with PBS were<br />

�1.5–2 times the values obtained when tested with SF<br />

(P � 0.05). Overall E xz (Figure 5B) was not detectably<br />

different between normal and degenerated samples (P �<br />

0.10) when tested using the same lubricant. However,<br />

overall E xz did vary with lubricant. The overall E xz of<br />

normal tissue was significantly lower with SF than with<br />

PBS (0.028 versus 0.045), and the overall E xz of degenerated<br />

tissue was 0.041 with SF versus 0.062 with PBS (P<br />

� 0.05 for both).<br />

Shear properties. To replicate sample shear deformation<br />

in microscale shear tests, lateral surface dis-<br />

placements in macroscale shear tests ranging from 70 to<br />

125 �m (measured from microscale shear tests) were<br />

applied to normal samples, and 120–160 �m was applied<br />

to degenerated samples. Overall G was not significantly<br />

different between normal and mildly degenerated tissue<br />

(mean � SEM 0.32 � 0.09 MPa in normal and 0.26 �<br />

0.07 MPa in degenerated cartilage; P � 0.7). G was<br />

lowest near the articular surface and increased significantly<br />

with tissue depth (P � 0.01) (0.2–-0.6 times and<br />

10–15 times the overall G near the surface and in the<br />

deepest region, respectively) (Figure 4C). At all depths<br />

below the surface, G was not significantly different<br />

between normal and degenerated samples (P � 0.5).<br />

Near the articular surface, there was a trend toward<br />

lower G values with mild surface degeneration (P �<br />

0.10) (surface G � 0.18 � 0.06 MPa and 0.06 � 0.02<br />

MPa in normal and degenerated samples, respectively).<br />

DISCUSSION<br />

This study elucidated the shear deformation and<br />

strain of cartilage during contact and sliding, as well as<br />

the effects of lubrication and mild degeneration, using a<br />

microscale cartilage-on-cartilage testing system. The<br />

present results indicate that cartilage–cartilage articulation<br />

results in 4 sequential events: adherence, adherence<br />

and deformation, detachment, and sliding. Peak E xz was<br />

highest near the surface and modulated by both the<br />

lubricant used and the condition of the sample itself<br />

(Figures 4A and B). Relative to the use of normal SF as<br />

lubricant, E xz increased with the use of PBS by �100%<br />

near the surface (Figure 5A) and �55% overall (Figure<br />

5B). In addition, when degenerated samples were tested,<br />

their E xz values near the surface were �3–5 times higher<br />

than those of normal samples (Figure 5A).<br />

Osteochondral samples used in this study were<br />

prepared from a site that is affected by age-associated<br />

degeneration and osteoarthritis. The samples were from<br />

the lateral femoral condyle, a load-bearing region where<br />

�22% of arthroscopically diagnosed chondral defects<br />

occur (28,29). The biomechanical factors associated with<br />

early degeneration in such load-bearing regions may<br />

contribute to the progressive degeneration of articular<br />

cartilage. Although the samples used had been previously<br />

frozen, a single freeze–thaw cycle does not result in<br />

marked changes in compressive properties of normal<br />

(30) or degenerated (31) articular cartilage or its structure<br />

(32), and is thus unlikely to have marked effects on<br />

shear properties. Although the distal femoral condyle<br />

normally apposes the tibial plateau, cartilage from the<br />

femoral condyle was apposed against itself to create a


2072 WONG ET AL<br />

simplified symmetric loading situation. Cartilage from<br />

other sites could be examined in future studies.<br />

The testing protocol used in this study provides a<br />

mechanical environment mimicking the compression<br />

and sliding of articular cartilage during normal joint<br />

loading. A knee undergoes a wide range of dynamic<br />

compression (up to 5–20%) (1,2) and sliding (up to �50<br />

mm) during normal activities (estimated from refs. 23<br />

and 24). The present analysis addresses certain parts of<br />

the gait cycle (e.g., contralateral toe-off and heel rise<br />

[23]) in which compressive loading is high and sliding<br />

velocity is low, during which time interaction of apposing<br />

tissue surfaces is likely to be initiated. Since samples<br />

were allowed to stress-relax for 1 hour, the pattern of<br />

cartilage deformation and strain is likely to be representative<br />

of that occurring after prolonged cyclic loading<br />

and sliding, rather than that which occurs at the onset of<br />

cyclic loading. Cartilage gradually depressurizes and<br />

reaches an averaged steady-state compression under<br />

prolonged cyclic loading. During this time, interstitial<br />

fluid pressure diminishes (33), and boundary lubrication<br />

becomes increasingly important (7). The ratedependence<br />

of shear deformation was not assessed;<br />

however, shear deformation may not change markedly<br />

with rate of lateral displacement since kinetic friction<br />

remains fairly constant with sliding velocity (7). Additional<br />

studies might elucidate the time course of cartilage<br />

shear after the onset of loading, as well as effects of<br />

sliding velocity on shear deformation.<br />

Although edge effects may occur in this shear<br />

testing protocol, such effects are likely minimal in the<br />

central area of tissue that was analyzed. Boundaries of<br />

the analyzed region were �2 mm away from both the<br />

leading and the trailing edges of the samples. Recent<br />

studies have shown that intratissue deformation (Gratz<br />

KR, et al: unpublished observations) and stiffness (34)<br />

are affected only �1–2 mm from vertical boundaries of<br />

cartilage. Thus, the results are likely to be representative<br />

of the major areas of cartilage contact.<br />

The reduction in E xz with SF as lubricant is<br />

consistent with predictions derived from a variety of past<br />

studies on cartilage friction. The friction coefficient<br />

decreases with the use of SF as lubricant compared with<br />

saline, both in cartilage-on-cartilage in the boundary<br />

mode (7,35) and in whole joint (36–38) friction tests.<br />

Assuming cartilage material properties (i.e., compressive<br />

and shear modulus) are maintained with the use of<br />

SF and PBS, the friction-reducing property of SF would<br />

be predicted to result in a reduction of tissue E xz.<br />

The overall E xz values obtained in this study are<br />

consistent with values estimated during physiologic joint<br />

articulation based on previously reported material properties<br />

of cartilage, as follows. To provide a first-order<br />

estimate of tissue properties, cartilage can be modeled<br />

as a linear, homogeneous, isotropic, and elastic tissue.<br />

With such assumptions and under small strain conditions,<br />

the overall stiffness of cartilage in shear (G) can be<br />

related to the shear stress (� xz) and the resulting overall<br />

E xz (27) as follows: G � � xz/2E xz (equation 1). In<br />

addition, � xz acting on cartilage can be related to the<br />

friction coefficient (�) and normal contact stress (� n)as<br />

follows: � xz � �� n (equation 2). Substituting equation 2<br />

into equation 1 and rearranging terms, cartilage E xz can<br />

be expressed as E xz � �� n/G (equation 3).<br />

Using reported values of � (�0.01) (39) and � n<br />

(�2 Mpa) (40) and the overall G values of �0.2–0.4<br />

MPa found in this study, the estimated overall E xz is<br />

0.02–0.05. The estimate of E xz is consistent with the<br />

range of overall E xz determined in this study (Figure<br />

5B). This suggests that an overall E xz ranging from 0.01<br />

to 0.1 may be suitable for biomimetic in vitro simulation<br />

of physiologic shear loading, for example during mechanical<br />

evaluation and mechanical stimulation for tissue<br />

engineering of cartilage.<br />

The increased E xz with degeneration, particularly<br />

near the surface with SF as lubricant (Figure 5A),<br />

appears to be due to both increased friction between<br />

sliding surfaces and a reduction in tissue shear modulus.<br />

Roughened surfaces have local asperities that can cause<br />

increased interaction and adherence between tissue surfaces,<br />

enhancing friction during sliding. Since boundary<br />

lubricants modulate friction, higher E xz in degenerated<br />

cartilage when tested in PBS (Figure 5A) indicates that<br />

friction between degenerated surfaces is higher than that<br />

between normal surfaces. Additionally, with degeneration,<br />

cartilage mechanical properties, such as G, become<br />

impaired (12). Diminished G would result in increased<br />

cartilage E xz at the same magnitude of � xz (equation 3),<br />

even if friction coefficient were similar. Since surface E xz<br />

increased with degeneration with the use of SF as<br />

lubricant, and G near the surface tended to diminish<br />

with degeneration, the elevated E xz at the surface in<br />

mildly degenerated tissue is likely a result of both<br />

increased � and reduced G (equation 3 and Figure 5A).<br />

Depth variations in E xz and G in the cartilage<br />

samples used in the present study provide additional<br />

information on the depth-varying biomechanical properties<br />

of articular cartilage. Compressive strain is depthvarying<br />

in both statically (4) and dynamically (3) compressed<br />

cartilage, decreasing monotonically with depth<br />

from the articular surface. E xz observed in this study was<br />

similarly depth-varying, being highest near the surface


LUBRICATION AND DEGENERATION EFFECTS ON CARTILAGE SHEAR DURING ARTICULATION 2073<br />

and lowest near the tidemark (Figures 4A and B).<br />

Compressive modulus, deduced from the overall compressive<br />

stress and local strain, was reflective of the<br />

depth-varying strain magnitudes, with cartilage increasing<br />

in stiffness with increasing depth (4). Similarly, G<br />

was lowest near the articular surface and increased<br />

monotonically to a maximum (20%) in the deepest<br />

region (Figure 4C). While G near the surface only<br />

tended to decrease with degeneration, the lack of statistical<br />

significance was likely due to the calculation from 2<br />

measurements, shear stress and strain (each having<br />

variability), as well as donor variability.<br />

The overall unconfined compressive modulus at<br />

equilibrium found in this study (E � 0.79 MPa) is<br />

consistent with previously reported values in human<br />

cartilage (0.58 MPa) (41). The overall shear properties<br />

of human cartilage have yet to be reported; however,<br />

overall G can be estimated from Poisson’s ratio (�) and<br />

the aggregate modulus (H A) demonstrated in indentation<br />

tests, using the relationship<br />

G � H A�1 � ���1 � 2��<br />

2�1 � ���1 � ��<br />

(27,42). The overall G found in this study (0.32 MPa) is<br />

consistent with that estimated from indentation tests<br />

(0.2–0.4 MPa) (43). Thus, for cartilage in compression,<br />

the deformation and mechanical properties we observed<br />

are consistent with findings of previous studies, and for<br />

cartilage in shear, the newly described depth-varying<br />

strain and shear properties are within values predicted<br />

for full-thickness human cartilage.<br />

With cartilage degeneration, tissue structure and<br />

low cellularity may affect the analysis and interpretation<br />

of deformation and strain. Degenerated cartilage contains<br />

clefts and areas of erosion (20), both of which<br />

result in material discontinuities within cartilage. Thus,<br />

continuum assumptions are not strictly valid. Also, compatibility<br />

conditions (27) are not maintained when fibrillated<br />

tissues overlap during deformation. Nevertheless,<br />

the present samples were only mildly degenerated and<br />

relatively intact except at the surface, and, whereas there<br />

were few visible cells at the surface of the degenerated<br />

samples, cells near the surface (3–5% depth) could be<br />

tracked. From a linear extrapolation, this may have<br />

resulted in a slight (�15%) underestimation of the<br />

absolute magnitude of superficial shear strain.<br />

The present results suggest that changes in E xz<br />

magnitudes and shear behavior may contribute to cartilage<br />

degeneration and pathogenesis. While samples<br />

were tested with a common overall compressive strain in<br />

this study, marked differences in E xz between groups<br />

occurred even with this conservative testing protocol.<br />

Under similar loads instead of overall strain, softer<br />

tissues (i.e., degenerated cartilage) would exhibit larger<br />

compressive and shear strain, excessive levels of which<br />

can contribute to cartilage degeneration. Excessive magnitudes<br />

of compressive strain result in mechanical injury<br />

to cells (44,45) and matrix (45,46), leading to reduced<br />

cartilage remodeling, maintenance, and repair. Likewise,<br />

high E xz that results from degenerated matrix,<br />

along with deficient lubrication, may lead to additional<br />

degeneration. Cell death and matrix damage may spread<br />

with continued exposure to high E xz, diminishing local<br />

mechanical properties of cartilage in both compression<br />

and shear. Cartilage E xz may then further increase and<br />

further the changes in tissue structure, resulting in a<br />

self-propagating cycle of degeneration. Such mechanical<br />

effects may also induce changes in surrounding joint<br />

tissues and the biochemical environment, which could<br />

supplement cartilage deterioration.<br />

ACKNOWLEDGMENTS<br />

We thank the many residents and staff at Dr. Lotz’s<br />

Laboratory at the Scripps Research Institute for harvesting<br />

and providing the human tissue used in this study, and Barbara<br />

Schumacher for guidance on histopathologic processing.<br />

AUTHOR CONTRIBUTIONS<br />

Dr. Sah had full access to all of the data in the study and takes<br />

responsibility for the integrity of the data and the accuracy of the data<br />

analysis.<br />

Study design. Wong, Bae, Sah.<br />

Acquisition of data. Wong, Chun, Lotz.<br />

Analysis and interpretation of data. Wong, Bae, Gratz, Sah.<br />

Manuscript preparation. Wong, Bae, Sah.<br />

Statistical analysis. Wong, Bae, Sah.<br />

REFERENCES<br />

1. Eckstein F, Lemberger B, Stammberger T, Englmeier KH, Reiser<br />

M. Patellar cartilage deformation in vivo after static versus dynamic<br />

loading. J Biomech 2000;33:819–25.<br />

2. Kersting UG, Stubendorff JJ, Schmidt MC, Bruggemann GP.<br />

Changes in knee cartilage volume and serum COMP concentration<br />

after running exercise. Osteoarthritis Cartilage 2005;13:<br />

925–34.<br />

3. Neu CP, Hull ML, Walton JH. Heterogeneous three-dimensional<br />

strain fields during unconfined cyclic compression in bovine<br />

articular cartilage explants. J Orthop Res 2005;23:1390–8.<br />

4. Schinagl RM, Gurskis D, Chen AC, Sah RL. Depth-dependent<br />

confined compression modulus of full-thickness bovine articular<br />

cartilage. J Orthop Res 1997;15:499–506.<br />

5. Ateshian GA, Lai WM, Zhu WB, Mow VC. An asymptotic<br />

solution for the contact of two biphasic cartilage layers. J Biomech<br />

1994;27:1347–60.<br />

6. Swanson SAV. Friction, wear, and lubrication. In: Freeman MAR,


2074 WONG ET AL<br />

editor. Adult articular cartilage. 2nd ed. Tunbridge Wells (UK):<br />

Pitman Medical; 1979. p. 415–60.<br />

7. Schmidt TA, Sah RL. Effect of synovial fluid on boundary<br />

lubrication of articular cartilage. Osteoarthritis Cartilage 2007;15:<br />

35–47.<br />

8. Schmidt TA, Gastelum NS, Nguyen QT, Schumacher BL, Sah RL.<br />

Boundary lubrication of articular cartilage: role of synovial fluid<br />

constituents. Arthritis Rheum 2007;56:882–91.<br />

9. Elsaid KA, Jay GD, Warman ML, Rhee DK, Chichester CO.<br />

Association of articular cartilage degradation and loss of boundary-lubricating<br />

ability of synovial fluid following injury and inflammatory<br />

arthritis. Arthritis Rheum 2005;52:1746–55.<br />

10. Clark JM, Simonian PT. Scanning electron microscopy of “fibrillated”<br />

and “malacic” human articular cartilage: technical considerations.<br />

Microsc Res Tech 1997;37:299–313.<br />

11. Meachim G, Emery IH. Quantitative aspects of patello-femoral<br />

cartilage fibrillation in Liverpool necropsies. Ann Rheum Dis<br />

1974;33:39–47.<br />

12. Setton LA, Elliott DM, Mow VC. Altered mechanics of cartilage<br />

with osteoarthritis: human osteoarthritis and an experimental<br />

model of joint degeneration. Osteoarthritis Cartilage 1999;7:2–14.<br />

13. Sah RL, Grodzinsky AJ, Plaas AHK, Sandy JD. Effects of static<br />

and dynamic compression on matrix metabolism in cartilage<br />

explants. In: Kuettner KE, Schleyerbach R, Peyron JG, Hascall<br />

VC, editors. Articular cartilage and osteoarthritis. New York:<br />

Raven Press; 1992. p. 373–92.<br />

14. Wang CC, Deng JM, Ateshian GA, Hung CT. An automated<br />

approach for direct measurement of two-dimensional strain distributions<br />

within articular cartilage under unconfined compression.<br />

J Biomech Eng 2002;124:557–67.<br />

15. Chahine NO, Wang CC, Hung CT, Ateshian GA. Anisotropic<br />

strain-dependent material properties of bovine articular cartilage<br />

in the transitional range from tension to compression. J Biomech<br />

2004;37:1251–61.<br />

16. Yamada K, Healey R, Amiel D, Lotz M, Coutts R. Subchondral<br />

bone of the human knee joint in aging and osteoarthritis. Osteoarthritis<br />

Cartilage 2002;10:360–9.<br />

17. Scott JE, Dorling J. Differential staining of acid glycosaminoglycans<br />

(mucopolysaccharides) by alcian blue in salt solutions. Histochemie<br />

1965;5:221–33.<br />

18. Ross MH, Kaye GI, Pawlina W. Histology: a text and atlas. 4th ed.<br />

Philadelphia: Lippincott Williams & Wilkins; 2003.<br />

19. Bae WC, Temple MM, Amiel D, Coutts RD, Niederauer GG, Sah<br />

RL. Indentation testing of human cartilage: sensitivity to articular<br />

surface degeneration. Arthritis Rheum 2003;48:3382-94.<br />

20. Shapiro F, Glimcher MJ. Induction of osteoarthrosis in the rabbit<br />

knee joint: histologic changes following menisectomy and meniscal<br />

lesions. Clin Orthop Rel Res 1980;147:287–95.<br />

21. Mazzucco D, Scott R, Spector M. Composition of joint fluid in<br />

patients undergoing total knee replacement and revision arthroplasty:<br />

correlation with flow properties. Biomaterials 2004;25:<br />

4433–45.<br />

22. Fung YC. Biomechanics: mechanical properties of living tissues.<br />

2nd ed. New York: Springer-Verlag; 1993.<br />

23. Whittle M. Gait analysis: an introduction. 3rd ed. Oxford (UK):<br />

Butterworth-Heinemann; 2002.<br />

24. Shelburne KB, Torry MR, Pandy MG. Muscle, ligament, and<br />

joint-contact forces at the knee during walking. Med Sci Sports<br />

Exerc 2005;37:1948–56.<br />

25. Bae WC, Lewis CW, Levenston ME, Sah RL. Indentation testing<br />

of human articular cartilage: effects of probe tip geometry and<br />

indentation depth on intra-tissue strain. J Biomech 2006;39:<br />

1039–47.<br />

26. Gratz KR, Sah RL. Experimental measurement and quantification<br />

of frictional contact between biological surfaces experiencing large<br />

deformation and slip. J Biomech 2008;41:1333–40.<br />

27. Fung YC. A first course in continuum mechanics. 2nd ed. Englewood<br />

Cliffs (NJ): Prentice-Hall; 1977.<br />

28. Hjelle K, Solheim E, Strand T, Muri R, Brittberg M. Articular<br />

cartilage defects in 1,000 knee arthroscopies. Arthroscopy 2002;<br />

18:730–4.<br />

29. Curl WW, Krome J, Gordon ES, Rushing J, Smith BP, Poehling<br />

GG. Cartilage injuries: a review of 31,516 knee arthroscopies.<br />

Arthroscopy 1997;13:456–60.<br />

30. Swann AC, Seedhom BB. The stiffness of normal articular cartilage<br />

and the predominant acting stress levels: implications for the<br />

aetiology of osteoarthrosis. Br J Rheumatol 1993;32:16–25.<br />

31. Kempson GE, Spivey CJ, Swanson SA, Freeman MA. Patterns of<br />

cartilage stiffness on normal and degenerate human femoral<br />

heads. J Biomech 1971;4:597–609.<br />

32. Kiefer GN, Sundby K, McAllister D, Shrive NG, Frank CB, Lam<br />

T, et al. The effect of cryopreservation on the biomechanical<br />

behavior of bovine articular cartilage. J Orthop Res 1989;7:<br />

494–501.<br />

33. Armstrong CG, Lai WM, Mow VC. An analysis of the unconfined<br />

compression of articular cartilage. J Biomech Eng 1984;106:<br />

165–73.<br />

34. Bae WC, Law AW, Amiel D, Sah RL. Sensitivity of indentation<br />

testing to step-off edges and interface integrity in cartilage repair.<br />

Ann Biomed Eng 2004;32:360–9.<br />

35. Forster H, Fisher J. The influence of loading time and lubricant on<br />

the friction of articular cartilage. Proc Inst Mech Eng [H] 1996;<br />

210:109–19.<br />

36. Wright V, Dowson D. Lubrication and cartilage. J Anat 1976;121:<br />

107–18.<br />

37. Linn FC. Lubrication of animal joints. II. The mechanism. J Biomech<br />

1968;1:193–205.<br />

38. Mabuchi K, Tsukamoto Y, Obara T, Yamaguchi T. The effect of<br />

additive hyaluronic acid on animal joints with experimentally<br />

reduced lubricating ability. J Biomed Mater Res 1994;28:865–70.<br />

39. Tanaka E, Kawai N, Tanaka M, Todoh M, van Eijden T, Hanaoka<br />

K, et al. The frictional coefficient of the temporomandibular joint<br />

and its dependency on the magnitude and duration of joint<br />

loading. J Dent Res 2004;83:404–7.<br />

40. Raimondi MT, Pietrabissa R. Contact pressures at grafted cartilage<br />

lesions in the knee. Knee Surg Sports Traumatol Arthrosc<br />

2005;13:444–50.<br />

41. Jurvelin JS, Buschmann MD, Hunziker EB. Mechanical anisotropy<br />

of the human knee articular cartilage in compression. Proc<br />

Inst Mech Eng [H] 2003;217:215–9.<br />

42. Mow VC, Hayes WC, editors. Basic orthopaedic biomechanics.<br />

2nd ed. New York: Raven Press; 1997.<br />

43. Athanasiou KA, Rosenwasser MP, Buckwlter JA, Malinin TI,<br />

Mow VC. Interspecies comparisons of in situ intrinsic mechanical<br />

properties of distal femoral cartilage. J Orthop Res 1991;9:330–40.<br />

44. Loening A, Levenston M, James I, Nuttal M, Hung H, Gowen M,<br />

et al. Injurious mechanical compression of bovine articular cartilage<br />

induces chondrocyte apoptosis. Arch Biochem Biophys 2000;<br />

381:205–12.<br />

45. Chen CT, Bhargava M, Lin PM, Torzilli PA. Time, stress, and<br />

location dependent chondrocyte death and collagen damage in<br />

cyclically loaded articular cartilage. J Orthop Res 2003;21:888–98.<br />

46. Thibault M, Poole AR, Buschmann MD. Cyclic compression of<br />

cartilage/bone explants in vitro leads to physical weakening, mechanical<br />

breakdown of collagen and release of matrix fragments.<br />

J Orthop Res 2002;20:1265–73.


Effect of a Focal Articular Defect on Cartilage Deformation during<br />

Patello-Femoral Articulation<br />

Benjamin L. Wong, Robert L. Sah<br />

Department of Bioengineering, University of California—San Diego, 9500 Gilman Drive, La Jolla, California<br />

Received 3 August 2009; accepted 22 April 2009<br />

Published online 2 July 2010 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jor.21187<br />

ABSTRACT: The objective of this study was to determine cartilage strains near, and in apposition to, a focal defect during patello-femoral<br />

articulation. Bovine osteochondral blocks from the trochlea (TRO) and patella (PAT) were apposed, compressed 12%, and subjected to<br />

sliding under video microscopy. Samples, lubricated with synovial fluid, were tested intact and then with a full-thickness defect in PAT<br />

cartilage. Shear (Exz), axial (Ezz), and lateral (Exx) strains were determined locally for TRO and PAT cartilage. For articulation with a<br />

focal defect, the strain amplitudes of PAT cartilage near the surface were ∼2–8× lower in Exz and ∼1.4× higher in −Ezz than intact PAT<br />

cartilage. At 20% depth, Exz and Exx for PAT cartilage with a focal defect were ∼2× and ∼10–25× higher than intact PAT, respectively.<br />

For TRO articulating against a focal defect, Exz and −Ezz near the surface and at 20% depth were ∼2–4× lower than that for articulation<br />

against intact cartilage. The results elucidate dramatic region-specific changes in strain due to lateral motion. In these regions, such<br />

altered cartilage mechanics during knee movement may cause focal defects to extend by induction of damaging levels of strain to bordering<br />

regions of cartilage. © 2010 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J. Orthop. Res. 28: 1554–1561, 2010<br />

Keywords: focal defect; articulation; biomechanics; deformation; cartilage strains<br />

Focal articular defects are prevalent in symptomatic<br />

knees and are associated with progressive cartilage<br />

degeneration. In patients evaluated arthroscopically,<br />

focal defects were found in 20–60% of all symptomatic<br />

knees, occurring most frequently in the medial condyle<br />

and patella, both of which are load-bearing regions (Fig.<br />

1). 1–4 Such, focal defects typically ranged from 0.5 to<br />

4cm 2 in area with an average area of 2.1 cm 2 , and<br />

extended beyond half the cartilage thickness in depth<br />

in 50% of all visualized articular defects. 1 With continued<br />

joint loading and time, untreated defects enlarge, 5<br />

are associated with cartilage volume loss, 6 and exhibit<br />

histopathological signs of cartilage degeneration adjacent<br />

to the focal defect. 7,8 While such findings suggest<br />

that the presence of a focal defect predisposes joints<br />

to secondary osteoarthritis, the mechanism by which it<br />

causes cartilage to degenerate within the proximity of a<br />

focal defect remains to be elucidated.<br />

The effect of a focal defect on cartilage deformation<br />

has been examined during axially directed loading.<br />

Under axial compression alone, contact stress and<br />

stress gradients are elevated in areas of cartilage near<br />

the edges of a focal defect. 9,10 As a result, macroscopic<br />

tissue deformation 11 and local strains 12 are<br />

increased markedly in these regions. Elevated strains<br />

may reach levels that induce cell death 13 and matrix<br />

damage, 14 and thus, the elevated strains resulting from<br />

a focal defect may become injurious to cartilage and<br />

induce degeneration. Such tissue deformation may be<br />

altered by or accentuated by the addition of lateral<br />

motion superimposed on compression, as both occur<br />

Additional Supporting Information may be found in the online<br />

version of this article.<br />

Correspondence to: Robert L. Sah (T: 858-534-0821;<br />

F: 858-822-1614.; E-mail: rsah@ucsd.edu)<br />

© 2010 Orthopaedic Research Society. Published by Wiley Periodicals, Inc.<br />

1554 JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010<br />

in load-bearing regions of cartilage during joint movement.<br />

During knee movement, articular surfaces contact,<br />

compress, and articulate against each other. Within the<br />

patellofemoral groove of the knee, patellar cartilage<br />

contacts and slides against trochlear cartilage during<br />

normal joint movement and loading (Fig. 2A). Under<br />

applied compressive loads of ∼1.5× the body weight,<br />

patellofemoral cartilage compresses ∼10% of its overall<br />

thickness in vitro following 14 min of static loading, 15<br />

and following knee bending, patellar cartilage alone<br />

compresses about ∼5–10% overall in vivo. 16 Collectively,<br />

such results provide physiologic mechanical parameters<br />

for the in vitro testing of patello-femoral articulation.<br />

Recently, local and overall deformation of cartilage<br />

during compression and cartilage articulation 17 were<br />

determined using video microscopy 18 and image correlation<br />

to track the displacement of fiducial markers. 19,20<br />

Such a test configuration can be applied to study contact<br />

of patellar and trochlear cartilage surfaces, 21,22 to<br />

examine the effects of a focal articular defect on the<br />

deformation of cartilage near, and in apposition to, the<br />

proximal defect edge that experiences oncoming forward<br />

motion of the apposing surface during compression and<br />

lateral displacement (Fig. 2). Effects of a focal defect on<br />

cartilage deformation may be distinctly and markedly<br />

different during compression than during shear.<br />

Therefore, the hypothesis of this study was that during<br />

patellofemoral cartilage articulation, the cartilage<br />

deformation of the patella and trochlea are markedly<br />

altered with the presence of a focal articular defect.<br />

The specific objective of this study was to determine the<br />

effects of a focal articular defect, created in the patellar<br />

tissue, on the local and overall strains of the patellar<br />

and trochlear cartilage during compression and sliding<br />

motion.


Figure 1. Anatomical distribution of focal articular defects averaged<br />

from four previous arthroscopy studies on the prevalence of<br />

cartilage defects in symptomatic knees. 1–4<br />

Figure 2. Schematic of (A) knee joint movements at multiple<br />

scales, (B) experimental setup and loading protocol for micro-shear<br />

testing, (C) and locations of sub-regions (EDGE, MID, FAR) used<br />

for statistical analysis of strains in patella and trochlear cartilage.<br />

EFFECT OF A FOCAL DEFECT ON CARTILAGE DEFORMATION 1555<br />

MATERIALS AND METHODS<br />

Sample Isolation and Preparation<br />

Osteochondral cores with macroscopically normal cartilage<br />

were harvested from the trochlea (TRO) and patella (PAT)<br />

of four adult bovine animals (1–2 years). Using a low speed<br />

drill press with custom stainless steel coring bits, a 10 mm<br />

diameter osteochondral core was isolated from both the TRO<br />

and PAT of each joint in a manner similar to that described<br />

previously. 23 The TRO and PAT cores were then trimmed to<br />

each yield one approximately rectangular block for biomechanical<br />

testing. 17 Each rectangular block had a cartilage surface<br />

area of ∼3 × 10 mm 2 and a total thickness of ∼1 cm (Fig. 2A).<br />

Each sample consisted of one TRO and one PAT block from the<br />

same knee, and was stored in phosphate buffered saline (PBS)<br />

containing proteinase inhibitors (PI) until testing.<br />

Prior to mechanical testing, samples were stained for<br />

∼2–4 h at 4 ◦ C in PBS + PI and propidium iodide (20 �g/mL) to<br />

fluorescently highlight cell nuclei. Blocks were then bathed in<br />

normal bovine synovial fluid (SF) containing PI and propidium<br />

iodide (20 �g/mL) at 4 ◦ C for 12–16 h to lubricate surfaces. The<br />

SF was pooled from adult bovine knees, previously characterized<br />

for boundary lubrication properties and lubricant levels<br />

and stored at −80 ◦ C, and characterized previously for boundary<br />

lubrication properties and lubricant molecules levels. 23<br />

Experimental Design<br />

To characterize the effect of a focal defect on cartilage deformation<br />

during articulation, samples were mechanically tested,<br />

first intact and then with a focal defect. In between tests, samples<br />

were rinsed, allowed to reswell, and incubated for ∼2–4 h<br />

in PBS + PI and then in SF + PI for an additional 12–16 h at<br />

4 ◦ C. Following the mechanical testing of intact samples and<br />

reincubation, a full thickness (∼2 mm in depth), 3 mm wide<br />

focal articular defect was created in the center of the PAT cartilage<br />

(Fig. 2A) as described previously. 12 The width of the focal<br />

defect was chosen so to be wide relative to the articulation distance<br />

(described below) in order to focus on the initial stages<br />

of articulation.<br />

Micro-Scale Shear Testing<br />

Samples were shear tested under video microscopy essentially<br />

as described previously. 17 Briefly, each TRO and PAT pair<br />

was secured in a custom bi-axial loading chamber mounted<br />

onto an epi-fluorescence microscope for digital video imaging<br />

(Fig. 2B). 18 The chamber secured the PAT block at the bone<br />

and allowed in-plane movement of the apposing mobile TRO<br />

block with orthogonally positioned plungers. 17 Subsequently,<br />

an axial displacement was applied (∼40 �m/s) to induce 12%<br />

compression (1 − �z, where �z is the stretch ratio 24 ) of the overall<br />

cartilage thickness (Fig. 2B). Samples were then allowed<br />

to stress relax for 1 h, determined to be sufficient to reach<br />

an approximate equilibrium stress for the current sample<br />

geometries. 17 Cartilage deformation was then captured during<br />

lateral motion separately in the TRO and PAT cartilage<br />

following axial compression. Three sets of applied lateral displacements<br />

(�x), each consisting of +1 mm and then −1mm<br />

(returning to initial position) were applied at 100 �m/s to the<br />

bone portion of the TRO block (Fig. 2B). The first set, was for<br />

preconditioning, 23 while the second and third set were recorded<br />

for the PAT and TRO blocks for analysis, respectively. Deformation<br />

during patello-femoral cartilage articulation was captured<br />

with sequential fluorescence images taken at ∼25 �m increments<br />

of lateral displacement.<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010


1556 WONG AND SAH<br />

Data Collection and Calculations<br />

Acquired images were analyzed as described previously 17,20<br />

to determine the depth-varying and overall deformation<br />

and strain in cartilage. Evenly distributed cell nuclei<br />

(∼250 cells/mm 2 ) were tracked during lateral motion to determine<br />

the displacements of uniformly spaced (10 pixel) mesh<br />

points in local regions. Subsequently, Lagrangian shear (Exz),<br />

axial (Ezz), and lateral (Exx) strains were determined relative<br />

to the unloaded state, 24 when articular surfaces were sliding<br />

(�x = 0.8 mm) and strains had become steady. Local strains<br />

were determined by averaging both depth-wise and at various<br />

lateral distances from the defect edge. First, sample thickness<br />

was normalized and divided into 8 intervals, with 4 intervals<br />

being 0.083 times the normalized thickness near the articular<br />

surface (i.e., 0–0.333) and 0.167 times for the remaining tissue<br />

depth (i.e., 0.333–1). To reduce noise and consolidate data, PAT<br />

cartilage strains near the proximal defect edge, which experiences<br />

forward lateral motion, were averaged depth-wise for<br />

lateral regions (∼0.2 mm × full cartilage thickness) at (EDGE),<br />

∼0.4 mm (MID) and ∼0.8 mm (FAR) away from the defect edge<br />

to yield a depth-profile at varying lateral distances (Fig. 2C).<br />

For TRO cartilage in direct apposition to the focal defect prior<br />

to lateral motion, strains following lateral articulation were<br />

averaged depth-wise in lateral regions (∼0.15 mm × full cartilage<br />

thickness) at (EDGE), ∼0.3 mm (MID), and ∼0.5 mm<br />

(FAR) away from the defect edge. For intact tissue, strains<br />

in corresponding lateral regions were determined similarly.<br />

Overall strain values were determined as the mean of all local<br />

values.<br />

Statistical Analysis<br />

Data are reported as mean ± standard error of the mean<br />

(SEM). Repeated measures ANOVA was used to determine the<br />

effects of a focal defect (intact vs. defect), tissue depth, and lateral<br />

location from the defect edge (EDGE, MID, FAR) on local<br />

and overall strains. Differences between defect and intact samples<br />

at the various lateral locations were assessed by planned<br />

pair-wise comparisons.<br />

Figure 3. Representative micrographs of patellar cartilage as (A,D,G) intact or (B,E,H) with a focal defect with superimposed colormaps<br />

of (A,B) shear (Exz), (D,E) axial (−Ezz), and (G,H) lateral (Exx) strain maps when articulating against trochlear samples after articular<br />

surfaces have slid. Strain map boundaries encompass the corresponding deformed states. Local (C) shear (Exz), (F) axial (−Ezz), and (I)<br />

lateral (Exx) strain averaged depth-wise versus normalized tissue depth for patella cartilage as intact or with a focal defect. For samples<br />

with a focal defect, strains were also determined as a function of lateral distance from the defect edge (EDGE, MID, FAR).<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010


RESULTS<br />

Qualitatively, the deformations of intact PAT and TRO<br />

cartilage were similar, being depth-varying during axial<br />

compression as well as during lateral motion. Cartilage<br />

thickness was similar between intact samples, being<br />

1.97 ± 0.13 mm and 1.93 ± 0.14 mm for PAT and TRO<br />

tissue, respectively. Under compression, axial deformation<br />

of intact PAT and TRO cartilage were similarly<br />

depth-varying, being highest near the surface and lowest<br />

near the tidemark. Also during compression, shear<br />

and lateral deformation were low (


1558 WONG AND SAH<br />

Figure 5. Representative micrographs of trochlear cartilage with superimposed colormaps of (A,B) shear (Exz), (D,E) axial (−Ezz), and<br />

(G,H) lateral (Exx) strain when in apposition with patellar cartilage as (A,D,G) intact or (B,E,H) with a focal defect during lateral motion.<br />

Dashed lines (– –) encompass the analyzed regions prior to lateral motion, while boundaries of strain maps encompass the corresponding<br />

deformed states. Local (C) shear (Exz), (F) axial (−Ezz), and (I) lateral (Exx) strain averaged depth-wise versus normalized tissue depth<br />

for trochlear cartilage, intact or with a focal defect. For samples apposing a focal defect, strains were also determined as a function of<br />

lateral distance from the defect edge (EDGE, MID, FAR).<br />

a defect decreased with increasing lateral distance from<br />

the defect edge (p < 0.01). Thus, when TRO cartilage filling<br />

the focal defect slides over the proximal defect edge,<br />

PAT cartilage near a focal defect compresses more than<br />

intact cartilage near the surface and overall.<br />

Lateral Strain (Exx)<br />

With articulation, the resultant Exx for intact cartilage<br />

did not vary significantly with tissue depth (p = 0.2) and<br />

lateral location (p = 1.0) (Fig. 3G), while for cartilage<br />

with a defect, Exx varied with tissue depth (p < 0.001)<br />

and lateral region (p < 0.01) (Fig. 3H). For all lateral<br />

regions, Exx remained negligible (≤0.01) for intact cartilage<br />

throughout tissue depth and peaked at ∼20% depth<br />

for defect samples (Fig. 3I). Near the articular surface,<br />

Exx for cartilage with a defect were statistically indifferent<br />

(p = 0.4) from that for intact cartilage and did not<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010<br />

significantly (p = 0.6) vary with lateral distance (Fig.<br />

4G). At 20% tissue depth (Fig. 4H) and overall (Fig.<br />

4I), cartilage Exx peaked at a value of ∼0.08 and ∼0.03,<br />

respectively, in the MID region for cartilage with a defect<br />

(p < 0.01) and were significantly higher than Exx for<br />

intact cartilage (p < 0.01). Thus, PAT cartilage expands<br />

laterally at 20% tissue depth as it becomes further compressed<br />

when TRO cartilage filling the focal defect slides<br />

over the defect edge.<br />

Trochlear Cartilage Deformation<br />

During patello-femoral articulation, the depth-variation<br />

in strains of TRO cartilage in articulation with an intact<br />

and defect-containing PAT surface were similar to that<br />

of PAT cartilage (Fig. 5), and the effect of a focal defect<br />

on TRO cartilage strains were also marked (Fig. 6).<br />

For TRO cartilage in articulation with an intact PAT


Figure 6. Effect of a focal defect (intact (I) vs. defect (D)) on (A–C)<br />

shear, (D–F) axial, and (G–I) lateral strain (A,D,G) near the articular<br />

surface, (B,E,H) at 20% tissue depth, and (C,F,I) overall of<br />

trochlear cartilage during articulation.<br />

surface, Exz and −Ezz were highest near the surface and<br />

decreased with increasing depth (p < 0.001), while Exx<br />

remained negligible throughout tissue depth (p = 0.6).<br />

All strains did not vary with lateral region for TRO<br />

articulation with intact PAT cartilage (p = 0.15–0.6). For<br />

TRO cartilage in articulation with a focal defect, Exz<br />

and −Ezz were ∼2–3× (p < 0.05, Fig. 6A) and ∼2–4×<br />

(p < 0.01, Fig. 6D), respectively, less near the surface<br />

than when in articulation with an intact surface. Furthermore,<br />

TRO cartilage compressed laterally in the<br />

EDGE region near the surface, and Exx near the surface<br />

markedly increased with lateral distance (p < 0.05),<br />

transitioning into lateral tension in the MID and FAR<br />

regions (Fig. 6G). The effects of tissue depth, lateral<br />

region, and a focal defect on TRO strains during lateral<br />

articulation are described in further detail in the<br />

supplementary data.<br />

DISCUSSION<br />

This study elucidated the deformation of cartilage<br />

within the proximity of, and directly in apposition to,<br />

EFFECT OF A FOCAL DEFECT ON CARTILAGE DEFORMATION 1559<br />

a focal articular defect during patello-femoral cartilage<br />

articulation. As the TRO surface displaced laterally, the<br />

tissue partially filling the focal defect pushed and then<br />

plowed over the proximal defect edge. As a result, PAT<br />

cartilage proximal to the defect sheared markedly less<br />

(∼70–700%) near the surface and more (∼50–60%) at<br />

20% tissue depth than intact cartilage during lateral<br />

motion (Fig. 7). PAT cartilage near the focal defect also<br />

compressed more (30–40%) than intact cartilage, and<br />

expanded laterally ∼10–25× more at 20% tissue depth<br />

than intact cartilage as TRO cartilage translated laterally<br />

over the proximal defect edge. For regions directly in<br />

apposition to the focal defect, TRO cartilage sheared and<br />

compressed less (∼50–70%) than when it slid over intact<br />

cartilage. As the TRO surface plowed over the proximal<br />

defect edge, TRO cartilage near the surface also compressed<br />

laterally at the EDGE region, and expanded<br />

laterally at MID and FAR regions. Collectively, the current<br />

results indicate that with articulation, the tissue<br />

deformation of both the cartilage adjacent to, and in<br />

apposition to, a focal defect are altered drastically by<br />

the presence of a focal defect, extending the findings of<br />

strain analysis of cartilage when compressed. 12<br />

To replicate the physiologic articulation and lubrication<br />

of cartilage surfaces, osteochondral samples were<br />

isolated from the trochlear and patella and tested in<br />

apposition with normal bovine synovial fluid. While<br />

efforts were not taken to ensure trochlear and patellar<br />

samples were in exact apposition and registration<br />

in the native anatomical joint, patellar cartilage translates<br />

a distance of ∼2.7 cm over the trochlear surface<br />

(estimated from Refs. 25,26 ). Since samples were isolated<br />

from matching anatomical aspects (i.e., lateral or<br />

medial), samples likely apposed and slid against one<br />

another during anatomic articulation. Since the medial<br />

femoral condyle is the site of greatest incidence of focal<br />

lesions, 1–4 condylar cartilage are of interest. However, it<br />

has a greater curvature in the saggital plane and articulates<br />

against both tibial and meniscal cartilage during<br />

knee movement. Thus, effects of a focal defect on condylar<br />

cartilage are likely to be somewhat more complex and<br />

remain to be investigated. A relative sliding distance of<br />

1 mm was sufficient to result in deformations reaching<br />

a steady-state for both intact and defect samples (data<br />

not shown). Thus, deformations described in the present<br />

study would likely be representative of greater, more<br />

physiologic, sliding distances.<br />

The defect geometry for the present samples was<br />

a full thickness slot with sharp edges in a rectangular<br />

osteochondral block and is a simple model of focal<br />

articular defects. Such a configuration would be most<br />

representative of a partially apposed and loaded defect,<br />

since completely covered defects would have a significant<br />

hoop stress around the defect rim that would help<br />

resistance deformation during articulation. Additional<br />

analyses to address complex three-dimensional issues<br />

would be useful in future studies. Furthermore, defect<br />

edges that are rounded, smoothed, or blunted may also<br />

occur, which may minimize tissue strains near focal<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010


1560 WONG AND SAH<br />

Figure 7. (A) Schematic depicting the effect of a focal defect on tissue deformation of trochlear and patellar cartilage when unloaded,<br />

compressed, and compressed and sheared in apposition. (B) Table of relative changes in shear (Exz), axial (−Ezz), and lateral (Exx)<br />

strains compared to normal intact cartilage (i.e., without a focal defect) for both trochlear and patellar cartilage when they are unloaded,<br />

compressed, and compressed and sheared in apposition.<br />

defects. While the present study provides analysis for<br />

a particular set of defect geometries, others remain to<br />

be investigated.<br />

While patellar cartilage was analyzed up to the articular<br />

surface, the region very close to the trochlear<br />

surface (∼10% of tissue depth), which partially filled the<br />

defect, was not analyzed because during lateral motion,<br />

tissue compression was so extensive in that region that<br />

cell nuclei coalesced and could not be discriminated and<br />

tracked. However, regions directly in apposition of the<br />

defect edge (EDGE) prior to lateral motion were tracked<br />

appropriately near the surface. Thus, strains in other<br />

lateral regions (MID, FAR) of the trochlear cartilage may<br />

be somewhat (∼10–15%) underestimated and may be<br />

more similar to the values of the EDGE lateral region.<br />

The dramatic elevation in cartilage strains near a<br />

focal defect during lateral articulation may be related<br />

and possibly contribute to, the progressive tissue degeneration<br />

associated with focal defects. With joint loading<br />

and time, increased fibrillation and decreased matrix<br />

staining were noted in regions adjacent to a focal defect<br />

in animal models. 7,8 Also, untreated defects in human<br />

knee joints have been shown to enlarge 5 and be associated<br />

with cartilage volume loss. 6 Markedly elevated<br />

strains likely make cartilage susceptible to damage in<br />

these regions, via cell death 13 and matrix damage, 14<br />

which can be induced by high magnitudes of compression.<br />

With repeated loading during joint movement, the<br />

increased Exz and Exx at 20% tissue depth, as well as<br />

the elevated −Ezz near the surface, may all contribute<br />

to inducing the indicators of tissue degeneration characterized<br />

previously near the focal defect edge. Thus, focal<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010<br />

defects may enlarge due to articulation-induced strain,<br />

and damage the bordering regions of cartilage.<br />

For cartilage in direct apposition to focal defects, the<br />

reduction in localized strains during lateral articulation<br />

may be related to the low incidences of two focal defects<br />

being in direct apposition (i.e., “kissing lesions”). Focal<br />

defects found on both apposing cartilage surfaces were<br />

found in only ∼2% of the symptomatic knees arthroscopically<br />

diagnosed with focal defects. 3 The low frequency<br />

of kissing lesions may be attributed the reduction in<br />

Exz and −Ezz of cartilage when in articulation with<br />

a focal defect. Since strain magnitudes are lowered, a<br />

chondral defect may be unlikely to develop from a preexisting<br />

focal defect on the opposing surface. Instead,<br />

kissing lesions may form with the defects being initiated<br />

concurrently at the time of injury or with one lesion<br />

enlarging big enough to become full-thickness in depth<br />

and cause the apposing chondral surface to be in articulation<br />

against the subchondral bone, inducing abrasive<br />

wear.<br />

The present study suggests that focal articular<br />

defects drastically alter cartilage deformation of both<br />

apposing cartilage surfaces not only during axial loading,<br />

but also distinctly during lateral articulation. Focal<br />

defects, of which a majority (∼61%) has been associated<br />

with acute injury or trauma, 1 markedly alter the<br />

mechanical environment of cartilage, and the resulting<br />

abnormal strains may be injurious to cells and<br />

matrix. Mechanically induced cell death and tissue loss<br />

reduce cell population and likely compromise the overall<br />

biosynthetic response of the tissue. 27 The metabolic<br />

activities of the remaining cells that may continue


to experience injurious levels of strain, are markedly<br />

altered. 13,14 As a result, tissue repair and remodeling<br />

responses are likely compromised, facilitating changes<br />

in cartilage structure by degeneration and wear. Thus,<br />

rehabilitation protocols, such as a period of non-weight<br />

bearing following acute injury, may be beneficial for<br />

allowing repair as well as limiting damage to the<br />

remaining native cartilage following surgical procedures<br />

such as debridement. Thus with normal loading,<br />

the changes in cartilage deformation associated with<br />

focal effects during both axial loading and lateral articulation<br />

may contribute to the enlargement of focal defects<br />

and predispose joints to secondary osteoarthritis.<br />

ACKNOWLEDGMENTS<br />

This work was supported by NIH and the Howard Hughes<br />

Medical Institute through the Professors <strong>Program</strong> to UCSD (to<br />

R.L.S.). We thank Chris Kim and Alexander Cigan for assistance<br />

with the sample preparations.<br />

REFERENCES<br />

1. Hjelle K, Solheim E, Strand T, et al. 2002. Articular cartilage<br />

defects in 1,000 knee arthroscopies. Arthroscopy 18:730–734.<br />

2. Curl WW, Krome J, Gordon ES, et al. 1997. Cartilage<br />

injuries: A review of 31,516 knee arthroscopies. Arthroscopy<br />

13:456–460.<br />

3. Aroen A, Loken S, Heir S, et al. 2004. Articular cartilage<br />

lesions in 993 consecutive knee arthroscopies. Am J Sports<br />

Med 32:211–215.<br />

4. Widuchowski W, Widuchowski J, Trzaska T. 2007. Articular<br />

cartilage defects: Study of 25,124 knee arthroscopies. Knee<br />

14:177–182.<br />

5. Wang Y, Ding C, Wluka AE, et al. 2006. Factors affecting<br />

progression of knee cartilage defects in normal subjects over<br />

2 years. Rheumatology (Oxford) 45:79–84.<br />

6. Cicuttini F, Ding C, Wluka A, et al. 2005. Association of cartilage<br />

defects with loss of knee cartilage in healthy, middle-age<br />

adults: A prospective study. Arthritis Rheum 52:2033–2039.<br />

7. Lefkoe TP, Trafton PG, Ehrlich MG, et al. 1993. An<br />

experimental model of femoral condylar defect leading to<br />

osteoarthrosis. J Orthop Trauma 7:458–467.<br />

8. Jackson DW, Lalor PA, Aberman HM, et al. 2001. Spontaneous<br />

repair of full-thickness defects of articular cartilage<br />

in a goat model. A preliminary study. J Bone Joint Surg Am<br />

83-A:53–64.<br />

9. Brown TD, Pope DF, Hale JE, et al. 1991. Effects of osteochondral<br />

defect size on cartilage contact stress. J Orthop Res<br />

9:559–567.<br />

10. Guettler JH, Demetropoulos CK, Yang KH, et al. 2004. Osteochondral<br />

defects in the human knee: Influence of defect size<br />

on cartilage rim stress and load redistribution to surrounding<br />

cartilage. Am J Sports Med 32:1451–1458.<br />

EFFECT OF A FOCAL DEFECT ON CARTILAGE DEFORMATION 1561<br />

11. Braman JP, Bruckner JD, Clark JM, et al. 2005. Articular<br />

cartilage adjacent to experimental defects is subject to<br />

atypical strains. Clin Orthop Relat Res 430:202–207.<br />

12. Gratz KR, Wong BL, Bae WC, et al. 2009. The effects of focal<br />

articular defects on cartilage contact mechanics. J Orthop<br />

Res 27:584–592.<br />

13. Kurz B, Jin M, Patwari P, et al. 2001. Biosynthetic response<br />

and mechanical properties of articular cartilage after injurious<br />

compression. J Orthop Res 19:1140–1146.<br />

14. Quinn TM, Allen RG, Schalet BJ, et al. 2001. Matrix and cell<br />

injury due to sub-impact loading of adult bovine articular<br />

cartilage explants: Effects of strain rate and peak stress. J<br />

Orthop Res 19:242–249.<br />

15. Herberhold C, Faber S, Stammberger T, et al. 1999. In<br />

situ measurement of articular cartilage deformation in<br />

intact femoropatellar joints under static loading. J Biomech<br />

32:1287–1295.<br />

16. Eckstein F, Lemberger B, Stammberger T, et al. 2000. Patellar<br />

cartilage deformation in vivo after static versus dynamic<br />

loading. J Biomech 33:819–825.<br />

17. Wong BL, Bae WC, Chun J, et al. 2008. Biomechanics of cartilage<br />

articulation: Effects of lubrication and degeneration<br />

on shear deformation. Arthritis Rheum 58:2065–2074.<br />

18. Schinagl RM, Gurskis D, Chen AC, et al. 1997. Depthdependent<br />

confined compression modulus of full-thickness<br />

bovine articular cartilage. J Orthop Res 15:499–506.<br />

19. Wang CC, Deng JM, Ateshian GA, et al. 2002. An automated<br />

approach for direct measurement of two-dimensional<br />

strain distributions within articular cartilage under unconfined<br />

compression. J Biomech Eng 124:557–567.<br />

20. Gratz KR, Sah RL. 2008. Experimental measurement and<br />

quantification of frictional contact between biological surfaces<br />

experiencing large deformation and slip. J Biomech<br />

41:1333–1340.<br />

21. Erne OK, Reid JB, Ehmke LW, et al. 2005. Depth-dependent<br />

strain of patellofemoral articular cartilage in unconfined<br />

compression. J Biomech 38:667–672.<br />

22. Guterl CC, Gardner TR, Rajan V, et al. 2009. Twodimensional<br />

strain fields on the cross-section of the human<br />

patellofemoral joint under physiological loading. J Biomech<br />

42:1275–1281.<br />

23. Schmidt TA, Sah RL. 2007. Effect of synovial fluid on boundary<br />

lubrication of articular cartilage. Osteoarthr Cartilage<br />

15:35–47.<br />

24. Fung YC. 1977. A first course in continuum mechanics.<br />

Englewood Cliffs: Prentice-Hall.<br />

25. Whittle M. 2002. Gait analysis: An introduction. Oxford;<br />

Boston: Butterworth-Heinemann.<br />

26. Shelburne KB, Torry MR, Pandy MG. 2005. Muscle, ligament,<br />

and joint-contact forces at the knee during walking.<br />

Med Sci Sports Exerc 37:1948–1956.<br />

27. Hunziker EB, Quinn TM. 2003. Surgical removal of articular<br />

cartilage leads to loss of chondrocytes from cartilage<br />

bordering the wound edge. J Bone Joint Surg Am 85-A(Suppl<br />

2):85–92.<br />

JOURNAL OF ORTHOPAEDIC RESEARCH DECEMBER 2010


The Journal of Bone and Joint Surgery Page Proof 1<br />

Author(s): Paragraph text formatting will be adjusted prior to publication.<br />

NIH: Yes<br />

Article type: Scientific Articles<br />

Cover: Trauma, Basic Science<br />

Disclosure: One or more of the authors received payments<br />

or services, either directly or indirectly (i.e., via his or her<br />

institution), from a third party in support of an aspect of<br />

this work. In addition, one or more of the authors, or his or<br />

her institution, has had a financial relationship, in the<br />

thirty-six months prior to submission of this work, with an<br />

entity in the biomedical arena that could be perceived to<br />

influence or have the potential to influence what is written<br />

in this work. Also, one or more of the authors has had<br />

another relationship, or has engaged in another activity, that<br />

could be perceived to influence or have the potential to<br />

influence what is written in this work. The complete<br />

Disclosures of Potential Conflicts of Interest submitted<br />

by authors are always provided with the online version of<br />

the article.<br />

doi:10.2106/JBJS.K.00046<br />

Effect of Tibial Plateau Fracture on Lubrication<br />

Function and Composition of Synovial Fluid<br />

Brooke L. Ballard, MD*, Jennifer M. Antonacci, PhD*, Michele M. Temple-Wong, PhD, Alexander Y.<br />

Hui, BS, Barbara L. Schumacher, BS, William D. Bugbee, MD, Alexandra K. Schwartz, MD, Paul J.<br />

Girard, MD, and Robert L. Sah, MD, ScD<br />

Investigation performed at the Department of Orthopaedic Surgery, University of California, San Diego, San<br />

Diego, and Department of Bioengineering, University of California, San Diego, La Jolla, California


Effect of Tibial Plateau Fracture on Page 3 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 2 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

regression analysis indicated that kinetic friction coefficient increased as hyaluronan<br />

1<br />

Abstract<br />

1<br />

concentration decreased.<br />

2<br />

Background: Intra-articular fractures may hasten post-traumatic arthritis in patients who are<br />

2<br />

Conclusions: Joints afflicted with a tibial plateau fracture have synovial fluid with decreased<br />

3<br />

typically too active and young for joint replacement. Current orthopaedic treatment principles,<br />

3<br />

lubrication properties in association with a decreased concentration of hyaluronan.<br />

4<br />

including recreating anatomic alignment and establishing articular congruity, have not eliminated<br />

4<br />

Clinical Relevance: Tibial plateau fractures result in a post-traumatic deficiency in synovial fluid<br />

5<br />

post-traumatic arthritis. Additional biomechanical and biological factors may contribute to the<br />

5<br />

lubrication function.<br />

6<br />

development of arthritis. The objective of the present study was to evaluate human synovial fluid<br />

6<br />

7<br />

following tibial plateau fractures for friction-lowering function and the concentrations of putative<br />

7<br />

lubricant molecules.<br />

8<br />

Methods: Synovial fluid specimens were obtained from the knees of eight patients (25-57 years<br />

9<br />

old) with a tibial plateau fracture, with five specimens from the injured knee as injury<br />

10<br />

synovial fluid and six specimens from contralateral knees as control synovial fluid. Each<br />

11<br />

specimen was centrifuged to obtain a fluid sample, separated from a cell pellet, for further<br />

12<br />

analysis. For each fluid sample, the start-up (static) and steady-state (kinetic) friction coefficients<br />

13<br />

in the boundary mode of lubrication were determined from a cartilage-on-cartilage<br />

14<br />

biomechanical test of friction. Also, concentrations of the putative lubricants, hyaluronan and<br />

15<br />

proteoglycan-4, as well as total protein, were determined for fluid samples.<br />

16<br />

Results: The injury group of experimental samples was from patients 45±13 years old<br />

17<br />

(mean±SD), after a post-injury period of 11±9 days. Start-up and kinetic friction coefficient<br />

18<br />

demonstrated similar trends and dependencies. The kinetic friction coefficients for human<br />

19<br />

plateau fracture synovial fluid were ~100% higher than that of control human synovial fluid.<br />

20<br />

Hyaluronan concentrations were 9-fold lower for plateau fracture synovial fluid compared with<br />

21<br />

the control synovial fluid, whereas proteoglycan-4 concentrations were >2-fold higher in plateau<br />

22<br />

fracture synovial fluid compared with the control synovial fluid. Univariate and multivariate<br />

23


Effect of Tibial Plateau Fracture on Page 5 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 4 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

cellular components from damaged tissues and bone marrow infiltrate the joint space 13,19-22 .<br />

1<br />

Introduction<br />

1<br />

Such alteration of synovial fluid may disrupt its lubrication functions.<br />

2<br />

Traumatic intra-articular fractures, such as those of the tibial plateau, are at risk of joint<br />

2<br />

Alteration in the lubricating function and lubricant composition of synovial fluid appears to<br />

3<br />

degeneration and post-traumatic arthritis even when treated according to traditional orthopaedic<br />

3<br />

be involved in cartilage deterioration after anterior cruciate ligament (ACL) injury, as well as<br />

4<br />

principles to restore articular congruency and anatomic alignment 1-7 . The consequences of poor<br />

4<br />

other injuries of the knee joint. After ACL injury of human knees, the level of proteoglycan-4 in<br />

5<br />

results can be life-changing–painful weight-bearing, restricted activity and lost time in the work<br />

5<br />

synovial fluid was reduced, while levels of degradative enzymes, cartilage matrix degradation<br />

6<br />

force 1,8-10 . The costs of post-traumatic arthritis are a component of the estimated >$100 billion<br />

6<br />

products, and inflammatory markers were increased for up to 6-18 months after injury 15,23 .<br />

7<br />

annual burden of osteoarthritis on the U.S. economy, with 9.8% of knee osteoarthritis estimated<br />

7<br />

Deficient lubrication after an injury has similarly been detected in the synovial fluid from acutely<br />

8<br />

to be of post-trauma etiology; furthermore, patients with post-traumatic arthritis and its chronic<br />

8<br />

injured equine joints, in association with a decreased concentration of hyaluronan 24 . In guinea<br />

9<br />

consequences tend to be younger with an additional impact on employment 9 . Compared to<br />

9<br />

pig 25 and rat 26,27 models of knee joint injury, synovial fluid lubrication function and lubricant<br />

10<br />

patients without post-traumatic arthritis, patients with post-traumatic arthritis have worse clinical<br />

10<br />

levels are also diminished. These studies suggest that human knee trauma, and specifically intra-<br />

11<br />

outcomes after arthrodesis or arthroplasty 11,12 . Post-traumatic arthritis and its consequences have<br />

11<br />

articular tibial plateau fracture, may lead to a deficiency in synovial fluid lubrication.<br />

12<br />

not been eliminated by employing traditional orthopedic principles, suggesting a possible role for<br />

12<br />

The hypothesis tested in the present study was that tibial plateau fractures impair the friction-<br />

13<br />

additional biological and biomechanical factors.<br />

13<br />

lowering lubrication function of human synovial fluid in association with changes in the<br />

14<br />

The pathogenesis of post-traumatic arthritis is complex and multifactorial. Recent studies<br />

14<br />

concentrations of proteoglycan-4 and hyaluronan lubricant molecules. The objectives of this<br />

15<br />

have focused on articular chondrocyte death and cartilage damage due to direct trauma, enzyme-<br />

15<br />

study were to compare the trauma synovial fluid from joints afflicted with acute tibial plateau<br />

16<br />

mediated cartilage degradation, and the role of reactive oxygen species 13-16 . Deficient lubrication<br />

16<br />

fractures with normal synovial fluid in terms of 1) friction-lowering boundary lubrication<br />

17<br />

may also contribute to cartilage deterioration after trauma. Lubrication typically allows articular<br />

17<br />

function and 2) biochemical composition, including hyaluronan and proteoglycan-4<br />

18<br />

cartilage to bear load and slide with low friction and low wear 17 , and is mediated by high levels<br />

18<br />

concentrations. In addition, the possible biomechanical basis for impaired lubricant function was<br />

19<br />

of proteoglycan-4 and hyaluronan in synovial fluid 18 . Synovial fluid lubricant molecules are<br />

19<br />

assessed by correlating friction coefficient and lubricant concentration.<br />

20<br />

secreted by chondrocytes and synoviocytes lining the joint, and are concentrated through<br />

20<br />

21<br />

selective retention by synovium. However, after an intra-articular fracture, soft tissues that<br />

21<br />

Materials and Methods<br />

22<br />

normally produce and retain synovial fluid lubricants are damaged, and in addition, blood and<br />

22


Effect of Tibial Plateau Fracture on Page 7 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 6 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

but Ctrl-hSF were not of sufficient volume. Thus, three other Ctrl-hSF samples were used from<br />

1<br />

Materials. Materials for lubrication testing were obtained as previously described 28 . The<br />

1<br />

other patients. Radiographs of the contralateral knee were evaluated by radiologists and<br />

2<br />

antibody to proteoglycan-4 was that to Lubricin from AbCam (Cambridge, MA); Streptomyces<br />

2<br />

orthopaedists and confirmed to not have an acute injury. To obtain fluid, following sterilization<br />

3<br />

hyaluronidase was from Seikagaku (Tokyo, Japan); SeaKem gold agaraose (Lonza) was from<br />

3<br />

of skin overlying each joint, a standard 18 gauge hollow bore spinal needle from Becton-<br />

4<br />

Fisher Scientific (Pittsburg, PA) and SDS-horizontal agarose gel electrophoresis and Western<br />

4<br />

Dickinson attached to a 60cc syringe barrel from Becton-Dickinson (Laguna Hills, CA) was<br />

5<br />

blot materials were from Life Technologies (Carlsbad, CA). EDTA-treated adult human blood<br />

5<br />

introduced into the joint space and SF was aspirated. The site of needle introduction into the<br />

6<br />

was purchased from Golden West Biologicals (Temecula, CA). EDTA-treated bovine blood was<br />

6<br />

knee joint was at the level of the joint line, 1 cm medial or lateral to the patellar ligament on the<br />

7<br />

from Animal Technologies (Tyler, TX).<br />

7<br />

anterior aspect of the knee 30 .<br />

8<br />

Patient Samples and Fracture Classification. Samples of human synovial fluid (hSF) were<br />

8<br />

Experimental Design. Gross Analysis of hSF Samples. Samples were initially studied by<br />

9<br />

aspirated from consented patients at a level one trauma center under the auspices of an IRB-<br />

9<br />

gross examination for color and clarity. Samples were noted to be straw-colored or bloody.<br />

10<br />

approved research plan. Patients 21 years and older, scheduled for surgery after sustaining a<br />

10<br />

Clarity was described by the degree of opacity as clear or opaque based on the ability to read<br />

11<br />

closed tibial plateau fracture, were asked to be included in the research study prior to the<br />

11<br />

markings through a ~1cm distance through the tube. Samples were then centrifuged (3,000g for<br />

12<br />

procedure by an orthopaedic surgeon, and consented. A particular age range was not sought.<br />

12<br />

30 minutes at 4°C) to obtain discrete fractions. The relative volume of fractions was then<br />

13<br />

Exclusion criteria consisted of open fractures, and vulnerable groups including minors, known<br />

13<br />

estimated directly from the centrifuged tubes. Fluid samples were photographed before and<br />

14<br />

cognitively impaired or institutionalized individuals, patients with known HIV or HCV or HBV<br />

14<br />

after centrifugation. Following centrifugation, the supernatant and pellet were separated and<br />

15<br />

infections, and patients unable to provide consent.<br />

15<br />

stored in aliquots at -80°C. Light microscopy analysis of selected supernatants and pellets<br />

16<br />

Radiographs, and CT scans if available based on routine clinical care, were graded by an<br />

16<br />

indicated that the processing was sufficient to separate cells such that they were present in the<br />

17<br />

attending orthopaedic surgeon using the Schatzker classification system for tibial plateau<br />

17<br />

pellet and absent from the supernatant (data not shown). Samples with adequate volume (1 ml)<br />

18<br />

fractures 29 .<br />

18<br />

after processing were selected for use in the study.<br />

19<br />

A total of 11 synovial fluid (SF) samples collected from 8 patients were used in the present<br />

19<br />

Assay Validation for Biochemical Analysis of In Vitro Mixtures of SF and Blood. Due to the<br />

20<br />

study. Attempts were made in all consented patients for aspiration from both the injured joint and<br />

20<br />

blood present in the synovial fluid aspirated from patients with a tibial plateau fracture, it<br />

21<br />

contralateral knee. Synovial fluid samples of sufficient volume (see below) were successfully<br />

21<br />

was necessary to ensure that the presence of blood components in the synovial fluid did not<br />

22<br />

withdrawn from both the injured knee (Plat-hSF) and contralateral control knee (Ctrl-hSF) of<br />

22<br />

interfere with the accuracy of the biochemical assays used. Therefore, prior to biochemical<br />

23<br />

three donors with an acute tibial plateau fracture. For two other patients, Plat-hSF was obtained,<br />

23


Effect of Tibial Plateau Fracture on Page 9 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 8 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

subsequent test lubricant supplemented with PIs with the cartilage completely immersed for 16-<br />

1<br />

analysis of the clinical human synovial fluid samples, an in vitro model of mixtures of fresh<br />

1<br />

24 hours at 4°C prior to lubrication testing at room temperature. The lubricant sample and<br />

2<br />

EDTA-treated adult human blood (Golden West Biologicals) and human synovial fluid<br />

2<br />

cartilage were then tested at room temperature by preconditioning, compressing to 18% of the<br />

3<br />

collected during routine arthroscopic surgery was developed, in order to mimic the Plat-<br />

3<br />

total cartilage thickness, and allowing 30 minutes for stress relaxation and interstitial fluid<br />

4<br />

hSF, and used to validate the assays for protein, proteoglycan-4, and hyaluronan. First, the<br />

4<br />

depressurization. Then samples were rotated at an effective velocity of 0.3 mm/second with pre-<br />

5<br />

macroscopic appearance of in vitro mixtures of human synovial fluid and blood of varying<br />

5<br />

sliding durations (Tps; the duration the sample is stationary prior to rotation) of 120, 12, and 1.2<br />

6<br />

proportions were examined to identify variations after mixing and separation by centrifugation<br />

6<br />

seconds. Friction coefficients (μ) were calculated from the expression μ = ���Reff X�Neq), where<br />

7<br />

(3,000g for 30 minutes at 4°C). Portions of in vitro human mixtures prior to centrifugation<br />

7<br />

��is torque, Neq�is the equilibrium axial load after 30 minutes of stress relaxation, and Reff is the<br />

8<br />

(Mixture), and supernatant and pellet portions of in vitro mixtures after centrifugation were then<br />

8<br />

effective radius of the cartilage, as described previously 28,31 . Briefly, a static friction coefficient<br />

9<br />

assayed for the concentrations of total protein, hyaluronan and proteoglycan-4, relative to the<br />

9<br />

(μstatic) was calculated using the peak | τ |, measured immediately after (within 10° of) the start of<br />

10<br />

initial mixture volume. To demonstrate the visual appearance of in vitro mixtures, analogous<br />

10<br />

rotation, and Neq. A kinetic friction coefficient (μkinetic) was calculated using both the | τ |<br />

11<br />

but larger volumes (~5 ml) of bovine blood and bovine synovial fluid (Animal Technologies)<br />

11<br />

averaged during the second complete revolution of the test sample and also Neq.<br />

12<br />

were generated and photographed.<br />

12<br />

Consistent with previous results 31 comparing friction coefficients with increasing Tps, all test<br />

13<br />

Biomechanical and Biochemical Analyses of Ctrl- and Plat-hSF Samples. Portions of Ctrl-<br />

13<br />

lubricants demonstrated little variation in kinetic friction coefficients with values at Tps = 1.2<br />

14<br />

and Plat-hSF samples were analyzed by biomechanical lubrication tests for friction lowering<br />

14<br />

seconds remaining within 11% of values at Tps = 120 seconds; therefore, for brevity and clarity<br />

15<br />

properties, in addition to analysis by biochemical assays for the concentrations of total protein,<br />

15<br />

μkinetic data are presented as the average at all pre-sliding durations. As expected, the mean static<br />

16<br />

hyaluronan, and proteoglycan-4.<br />

16<br />

and kinetic friction coefficients measured for PBS were greater than 0.20 for all Tps, consistent<br />

17<br />

Analytical Methods. Lubrication Test. Portions of individual Ctrl- and Plat-hSF samples<br />

17<br />

with previous results 31 , so PBS data were not analyzed further.<br />

18<br />

were analyzed for static and kinetic friction coefficients in the boundary lubrication mode on<br />

18<br />

Biochemical Analyses. Portions of synovial fluid samples were analyzed biochemically for<br />

19<br />

articulating cartilage surfaces (one friction test per synovial fluid specimen) as described<br />

19<br />

absolute concentrations of lubricant molecules hyaluronan and proteoglycan-4, as well as for<br />

20<br />

previously 28,31 . Intact articular surfaces were in the form of osteochondral cores and annuli from<br />

20<br />

total protein. A minimum number of two replicate samples per specimen were used for<br />

21<br />

adult bovine knees, and stored in phosphate buffered saline (PBS) supplemented with protease<br />

21<br />

concentration measurements. Hyaluronan. The concentration of hyaluronan in SF samples was<br />

22<br />

inhibitors (PIs; 2 mM Na-EDTA, 1 mM PMSF, 5 mM Benz-HCL, and 10 mM NEM) at –80°C.<br />

22<br />

determined by an ELISA-like assay using hyaluronan binding protein (Corgenix). Proteoglycan-<br />

23<br />

For lubrication testing, cartilage samples were thawed at 4°C, and then bathed in ~0.5 ml of the<br />

23


Effect of Tibial Plateau Fracture on Page 11 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 10 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

RESULTS<br />

1<br />

4 (PRG4). The concentration of PRG4 in hSF samples was quantified by Western Blot using<br />

1<br />

Patient Demographics. A total of eleven synovial fluid samples from eight patients were<br />

2<br />

antibody to Lubricin after SDS-horizontal agarose gel electrophoresis on 2% 3mm thick agarose<br />

2<br />

used in the present study (Table I). For these patients, the tibial plateau fracture types ranged<br />

3<br />

gels and transfer to PVDF membrane (100mA, overnight). The immunoreactive proteins were<br />

3<br />

from Schatzker type II to type VI and from OTA class 41B2.2 to 41C3.3. The control group of<br />

4<br />

visualized by ECL-Plus detection and digital scanning with a STORM 840 Imaging System<br />

4<br />

normal samples (n=6) was from patients 42±16 years old (mean±SD), with a post-injury period<br />

5<br />

(Molecular Dynamics, Fairfield, CT). ImageQuant (Molecular Dynamics) was used to generate<br />

5<br />

before synovial fluid acquisition of 11±7 days. The injury group of experimental samples (n=5)<br />

6<br />

densitometric scans. The PRG4 in hSF was quantified using standards of PRG4 that were<br />

6<br />

was from patients 45±13 years old, with a post-injury period of 11±9 days. Of the eight patients,<br />

7<br />

purified 32 from conditioned medium of human cartilage explants and run on the same gels.<br />

7<br />

six were male, and two were female; both control and experimental synovial fluid samples were<br />

8<br />

Statistical Analysis. The data of continuous variables are presented as mean±SEM. The<br />

8<br />

successfully obtained from the two female patients.<br />

9<br />

effects of joint injury (trauma versus control) on SF properties (μkinetic and the concentration of<br />

9<br />

Gross Appearance of Control and Plateau Fracture Human Synovial Fluid Samples.<br />

10<br />

lubricants) were assessed by ANOVA. The effects of joint injury (trauma versus control, as a<br />

10<br />

The synovial fluid samples from the control and injured joints appeared markedly different.<br />

11<br />

fixed factor) and Tps (1.2, 12, and 120s, as a repeated factor) on synovial fluid static friction<br />

11<br />

Before centrifugation, the control synovial fluid showed only traces of blood (Fig. 1-A-i),<br />

12<br />

coefficient (μstatic) were assessed by two-level ANOVA. Planned comparisons for μstatic were<br />

12<br />

whereas the injury synovial fluid was grossly bloody (Fig. 1-A-ii). After centrifugation, the<br />

13<br />

performed between trauma and control groups at each Tps value. The dependencies of friction<br />

13<br />

supernatant of control synovial fluid samples was clarified and appeared clear or straw-colored<br />

14<br />

coefficients on the concentrations of putative lubricants (hyaluronan and proteoglycan-4) were<br />

14<br />

(Fig. 1-B-i). In contrast, the injury synovial fluid samples separated into a yellow supernatant<br />

15<br />

analyzed by univariate and also multivariate regression, with both the absolute and log10 value<br />

15<br />

and a dark red pellet of varying relative volumes (Fig. 1-B-ii).<br />

16<br />

of hyaluronan concentrations (since it varied by several orders of magnitude). Statistical analysis<br />

16<br />

Biochemical Assay Validation with in vitro Mixtures of SF and Blood. The in vitro<br />

17<br />

was performed using Systat 10.2 (Systat; Richmond, CA).<br />

17<br />

mixtures of bovine synovial fluid and bovine blood appeared similar to the range of control and<br />

18<br />

18<br />

injury human synovial fluid samples. Before centrifugation, mixtures with increasing proportion<br />

19<br />

Source of Funding<br />

19<br />

of blood were of an increasingly darker shade of red (Fig. 1-C). After centrifugation, samples<br />

20<br />

Supported by a grant from the Orthopaedic Trauma Association, NIH R01AR051565, an<br />

20<br />

separated into a supernatant that was increasingly yellow and a pellet that was increasing in size<br />

21<br />

NSF Graduate Research Fellowship, NIH 5T35HL007491, and an award under the Howard<br />

21<br />

as the proportion of blood increased (Fig. 1-D). The in vitro mixtures of human synovial fluid<br />

22<br />

Hughes Medical Institute Professors <strong>Program</strong>.<br />

22<br />

23


Effect of Tibial Plateau Fracture on Page 13 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 12 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

values were 49-120% higher for fracture synovial fluid than control synovial fluid, with the<br />

1<br />

and human blood appeared to separate similarly, although they were more difficult to visualize<br />

1<br />

percentage difference increasing as the pre-spin duration decreased from 120s to 1.2s. At the<br />

2<br />

due to the small volumes available.<br />

2<br />

short pre-spin duration of 1.2s, the μstatic values approached the μkinetic values (Fig. 3-A and B).<br />

3<br />

Biochemical analysis of the in vitro mixtures of human synovial fluid and blood, as well as<br />

3<br />

The average μkinetic value for fracture synovial fluid was double that of control synovial fluid<br />

4<br />

the supernatants and pellets, revealed that the hyaluronan and proteoglycan-4 constituents, when<br />

4<br />

(0.044 versus 0.022, p


Effect of Tibial Plateau Fracture on Page 15 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 14 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

fluid properties were not determined. Additionally, the number of samples was limited due to<br />

1<br />

friction coefficients (data not shown). Multivariate regression yielded similar results (elimination<br />

1<br />

the relatively small number of patients meeting the inclusion criteria and consenting to the<br />

2<br />

of variation due to proteoglycan-4, leaving only correlation with hyaluronan concentration).<br />

2<br />

study. Furthermore, the control samples were not only from contralateral knees due to the<br />

3<br />

3<br />

difficulty in obtaining such fluid.<br />

4<br />

Discussion<br />

4<br />

The present study expands upon previous research on the effects of human knee injuries on<br />

5<br />

The present study identifies marked alterations in the lubrication function and lubricant<br />

5<br />

synovial fluid lubricant properties by focusing on synovial fluid after tibial plateau fracture. In a<br />

6<br />

composition of SF from patients with intra-articular tibial plateau fractures in the initial stages of<br />

6<br />

previous study, synovial fluid aspirated from Emergency Department patients presenting with<br />

7<br />

treatment. Compared with control human synovial fluid, synovial fluid from knees with intra-<br />

7<br />

mono-articular knee effusions and no radiographic abnormalities (e.g., without intra-articular<br />

8<br />

articular fractures had markedly decreased lubrication ability (+100% increase in μkinetic, 0.022<br />

8<br />

fracture) also exhibited poor lubrication relative to saline (Δμ= –0.045) compared with human<br />

9<br />

versus 0.044) in the acute post-injury time period that was studied. Concomitantly, tibial plateau<br />

9<br />

control synovial fluid relative to saline (Δμ = –0.095) 33 . Though the lubrication test methods<br />

10<br />

fractures led to changes in the concentration of putative lubricant molecules, with a decrease in<br />

10<br />

were different, with the former study using a glass-on-latex arthrotripsometer versus articulating<br />

11<br />

hyaluronan (–87%, 2.53 mg/ml to 0.27 mg/ml) and an increase in proteoglycan-4 (+156%, 139<br />

11<br />

cartilage in the present study, the synovial fluid from intra-articular tibial plateau fractures also<br />

12<br />

μg/ml to 356 μg/ml). Regression analysis indicated that poorly lubricating synovial fluid was<br />

12<br />

resulted in a kinetic coefficient of friction approximately double that of the controls (Fig. 3).<br />

13<br />

associated with diminished hyaluronan concentration.<br />

13<br />

With attention to sample preparation and a previously validated boundary-mode friction<br />

14<br />

The experimental approach of the present study involved a number of considerations.<br />

14<br />

test protocol 28 , subsequent characterization of friction properties of synovial fluid at a<br />

15<br />

Lubrication testing of the human synovial fluid samples was performed with normal adult bovine<br />

15<br />

cartilage-cartilage interface can be obtained with low variability in measured friction<br />

16<br />

cartilage surfaces, minimizing effects of variation in cartilage surface properties. The test<br />

16<br />

coefficient with a coefficient of variation (CV) of 12 – 28%. However, the amount of<br />

17<br />

configuration facilitates boundary lubrication by apposed articular cartilage surfaces, maintaining<br />

17<br />

variability in the composition of the injury fluid may be due to a number of factors,<br />

18<br />

possible interactions between synovial fluid and the articular surfaces, and also minimizing<br />

18<br />

including donor age, severity of injury, days post injury that the sample was collected and<br />

19<br />

confounding factors such as ploughing 28 . Adult bovine cartilage appears to be lubricated<br />

19<br />

assay sensitivity. Thus, the lubrication properties of human knee synovial fluid may be disrupted<br />

20<br />

similarly by normal bovine synovial fluid (μkinetic=0.025) 31 and by control human synovial fluid<br />

20<br />

in a variety of scenarios of acute injury or inflammation.<br />

21<br />

(μkinetic=0.022, Fig. 3). Also, human synovial fluid was obtained only during surgical procedures<br />

21<br />

The present study is also consistent with and adds to information on the concentration of<br />

22<br />

under general anesthesia. These procedures occurred at various time points up to 30 days after<br />

22<br />

lubricant molecules in joint synovial fluid from humans and animals. The decreased<br />

23<br />

the injury, so the long-term and time-dependent effects of tibial plateau fracture on synovial<br />

23


Effect of Tibial Plateau Fracture on Page 17 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 16 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

and were present in the samples analyzed for both chemical constituents and lubrication<br />

1<br />

concentration of hyaluronan is consistent with effects of disease and injury of human knees 34,35 .<br />

1<br />

properties.<br />

2<br />

At the time of total knee arthroplasty, the hyaluronan concentration of synovial fluid from<br />

2<br />

Further studies on the components of the injury synovial fluid samples would be useful<br />

3<br />

osteoarthritic knees was 1.3 mg/ml 36 . Compared to these values and the control value of 2.53<br />

3<br />

to clarify the mechanism of altered lubrication. The structure of hyaluronan and<br />

4<br />

mg/ml in the present study, hyaluronan concentrations in synovial fluid from knees with tibial<br />

4<br />

proteoglycan-4 in intra-articular fracture conditions may affect their roles in the boundary<br />

5<br />

plateau fractures was markedly lower at 0.27 mg/ml. In horses, joint injury was also associated<br />

5<br />

lubrication of articular cartilage after joint injury. Although proteoglycan-4 levels were<br />

6<br />

with a decreased concentration of hyaluronan 24,37 . Studies of SF lubrication in experimental<br />

6<br />

higher in the plat-hSF, this elevated concentration may not have been sufficient to<br />

7<br />

animal injury models have focused on proteoglycan-4 25,27 but not hyaluronan. The correlation<br />

7<br />

compensate for the alterations in lubrication function after joint injury. Concurrently, the<br />

8<br />

between decreased lubrication properties and decreased HA concentration in the present study<br />

8<br />

structural quality of hyaluronan and proteoglycan-4 may have been compromised during<br />

9<br />

suggests the importance of diminished HA concentration in post-traumatic human synovial fluid.<br />

9<br />

injury, such as a reduction in the concentration of high molecular weight hyaluronan<br />

10<br />

The reported effects of injury and osteoarthritis on PRG4 concentration in humans and<br />

10<br />

present in the injury synovial fluid. Blood-derived components may also have contributed<br />

11<br />

animal models are quite varied, with some studies indicating an injury-associated decrease in<br />

11<br />

to deficient lubrication in plat-hSF.<br />

12<br />

concentration of PRG4 15,23,33 and others indicating an increase 24,38 . In osteoarthritic knees,<br />

12<br />

The consequences of increased friction coefficient, in the context of intra-articular<br />

13<br />

increasing PRG4 concentration correlated with worsening lubrication and OA severity 38 .<br />

13<br />

fracture and other joint injuries, remain to be fully elucidated. Increased friction and wear<br />

14<br />

Differences in the concentration of PRG4 in synovial fluid of human osteoarthritis (151 μg/ml)<br />

14<br />

appear to be related. It is possible that even a short period of deficient synovial fluid<br />

15<br />

and human trauma (356 μg/ml), may be due a difference in analysis methods including<br />

15<br />

lubrication, in the setting of soft-tissue or weight-bearing loads are sufficient to initiate<br />

16<br />

standards, or a difference in patient populations. Total protein levels were also markedly<br />

16<br />

damage that can have longer-term consequences. Studying early time points is the first step<br />

17<br />

different with 27 mg/ml for synovial fluid from end-stage osteoarthritic knees at the time of total<br />

17<br />

to studying and identifying a longer-term effect.<br />

18<br />

knee arthroplasty 36 , compared to 38 mg/ml in trauma knees and 22 mg/ml in control knees in the<br />

18<br />

The collective results here indicate that lubricant molecules in synovial fluid after a tibial<br />

19<br />

present study. This may be due to differences in the pathologic processes, since after trauma the<br />

19<br />

plateau fracture are acutely altered with diminished hyaluronan concentration, and elevated<br />

20<br />

joint space is compromised and SF may be diluted by extra-articular contents and infiltrated by<br />

20<br />

concentrations of proteoglycan-4 and total protein compared with controls. The correlation of<br />

21<br />

protein in blood. Although the specimens were processed to remove red blood cells, it is<br />

21<br />

impaired lubrication of trauma SF with diminished hyaluronan and elevated proteoglycan-4<br />

22<br />

likely that a considerable amount of blood components, such as serum proteins, remained<br />

22<br />

remains to be evaluated further. The proteoglycan-4 that is elevated may not be functional alone,<br />

23


Effect of Tibial Plateau Fracture on Page 19 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 18 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

References<br />

or functional in a manner dependent on high concentrations of hyaluronan. The presence of<br />

1<br />

1. Volpin G, Dowd GS, Stein H, Bentley G. Degenerative arthritis after intra-articular<br />

blood in the knee joint may lead to prolonged derangement of the articular cartilage<br />

2<br />

fractures of the knee. Long-term results. J Bone Joint Surg Br. 1990; 72:634-8.<br />

permanently 22,39,40 . The results and conclusions of the present study may also be pertinent to<br />

3<br />

2. Marsh JL, Buckwalter J, Gelberman R, Dirschl D, Olson S, Brown T, Llinias A. Articular<br />

other intra-articular fractures 20 , such as those of the distal radius, tibial plafond, calcaneus,<br />

4<br />

fractures: does an anatomic reduction really change the result? J Bone Joint Surg Am.<br />

and metacarpal. Post-traumatic changes in synovial fluid may lead to prolonged changes in<br />

5<br />

2002; 84:1259-71.<br />

synovial joint biomechanical function as well as biological function 15 . In view of these results,<br />

6<br />

3. Rasmussen PS. Tibial condylar fractures. Impairment of knee joint stability as an<br />

correction of lubrication and other pathologic changes may offer an opportunity to protect and<br />

7<br />

indication for surgical treatment. J Bone Joint Surg Am. 1973; 55:1331-50.<br />

preserve cartilage, facilitate lubrication for early range of motion activities, and ultimately<br />

8<br />

4. Jensen DB, Rude C, Duus B, Bjerg-Nielsen A. Tibial plateau fractures. A comparison of<br />

modulate the development of post-traumatic arthritis.<br />

9<br />

conservative and surgical treatment. J Bone Joint Surg Br. 1990; 72:49-52.<br />

5. Honkonen SE. Degenerative arthritis after tibial plateau fractures. J Orthop Trauma.<br />

1995; 9:273-7.<br />

6. Duwelius PJ, Connolly JF. Closed reduction of tibial plateau fractures. A comparison of<br />

functional and roentgenographic end results. Clin Orthop Relat Res. 1988; 230:116-26.<br />

7. Lucht U, Pilgaard S. Fractures of the tibial condyles. Acta Orthop Scand. 1971; 42:366-<br />

76.<br />

8. Stevens DG, Beharry R, McKee MD, Waddell JP, Schemitsch EH. The long-term<br />

functional outcome of operatively treated tibial plateau fractures. J Orthop Trauma. 2001;<br />

15:312-20.<br />

9. Buckwalter JA, Saltzman C, Brown T. The impact of osteoarthritis: implications for<br />

research. Clin Orthop Rel Res Relat Res. 2004; 427S:S6-15.<br />

10. Honkonen SE. Degenerative arthritis after tibial plateau fractures. J Orthop Trauma.<br />

1995; 9:273-7.


Effect of Tibial Plateau Fracture on Page 21 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 20 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

20. Dirschl DR, Marsh JL, Buckwalter JA, Gelberman R, Olson SA, Brown TD, Llinias A.<br />

11. Weiss NG, Parvizi J, Trousdale RT, Bryce RD, Lewallen DG. Total knee arthroplasty in<br />

Articular fractures. J Am Acad Orthop Surg. 2004; 12:416-23.<br />

patients with a prior fracture of the tibial plateau. J Bone Joint Surg Am. 2003; 85:218-<br />

21. Repo RU, Finlay JB. Survival of articular cartilage after controlled impact. J Bone Joint<br />

21.<br />

Surg Am. 1977; 59:1068-76.<br />

12. Civinini R, Carulli C, Matassi F, Villano M, Innocenti M. Total knee arthroplasty after<br />

22. Hooiveld M, Roosendaal G, Vianen M, van den Berg M, Bijlsma J, Lafeber F. Blood-<br />

complex tibial plateau fractures. Musculoskelet Surg. 2009; 93:143-7.<br />

induced joint damage: longterm effects in vitro and in vivo. J Rheumatol. 2003; 30:339-<br />

13. Furman BD, Olson SA, Guilak F. The development of posttraumatic arthritis after<br />

44.<br />

articular fracture. J Orthop Trauma. 2006; 20:719-25.<br />

23. Elsaid KA, Fleming BC, Oksendahl HL, Machan JT, Fadale PD, Hulstyn MJ, Shalvoy R,<br />

14. Ulrich-Vinther M, Maloney MD, Schwarz EM, Rosier R, O'Keefe RJ. Articular cartilage<br />

Jay GD. Decreased lubricin concentrations and markers of joint inflammation in the<br />

biology. J Am Acad Orthop Surg. 2003; 11:421-30.<br />

synovial fluid of patients with anterior cruciate ligament injury. Arthritis Rheum. 2008;<br />

15. Lohmander LS, Roos H, Dahlberg L, Hoerrner LA, Lark MW. Temporal patterns of<br />

58:1707-15.<br />

stromelysin-1, tissue inhibitor, and proteoglycan fragments in human knee joint fluid<br />

24. Antonacci JM, Schmidt TA, Serventi LA, Shu YL, Gastelum NS, Schumacher BL,<br />

after injury to the cruciate ligament or meniscus. J Orthop Res. 1994; 12:21-28.<br />

McIlwraith CW, Sah RL. Effects of joint injury on synovial fluid and boundary<br />

16. Martin JA, Buckwalter JA. Post-traumatic osteoarthritis: the role of stress induced<br />

lubrication of cartilage. Trans Orthop Res Soc. 2007; 32:156.<br />

chondrocyte damage. Biorheology. 2006; 43:517-21.<br />

25. Teeple E, Elsaid KA, Fleming BC, Jay GD, Aslani K, Crisco JJ, Mechrefe AP.<br />

17. Ateshian GA, Mow VC, Friction, lubrication, and wear of articular cartilage and<br />

Coefficients of friction, lubricin, and cartilage damage in the anterior cruciate ligament-<br />

diarthrodial joints, in Basic Orthopaedic Biomechanics and Mechano-Biology, Mow VC,<br />

deficient guinea pig knee. J Orthop Res. 2008; 26:231-7.<br />

Huiskes R, Editors. 2005, Lippincott Williams & Wilkins: Philadelphia. p. 447-494.<br />

26. Flannery CR, Zollner R, Corcoran C, Jones AR, Root A, Rivera-Bermudez MA, Blanchet<br />

18. Swanson SAV, Friction, wear, and lubrication, in Adult Articular Cartilage, Freeman<br />

T, Gleghorn JP, Bonassar LJ, Bendele AM, Morris EA, Glasson SS. Prevention of<br />

MAR, Editor. 1979, Pitman Medical: Tunbridge Wells, England. p. 415-460.<br />

cartilage degeneration in a rat model of osteoarthritis by intraarticular treatment with<br />

19. Guilak F, Fermor B, Keefe FJ, Kraus VB, Olson SA, Pisetsky DS, Setton LA, Weinberg<br />

recombinant lubricin. Arthritis Rheum. 2009; 60:840-7.<br />

JB. The role of biomechanics and inflammation in cartilage injury and repair. Clin<br />

27. Jay GD, Fleming BC, Watkins BA, McHugh KA, Anderson SC, Zhang LX, Teeple E,<br />

Orthop Relat Res. 2004; 423:17-26.<br />

Waller KA, Elsaid KA. Prevention of cartilage degeneration and restoration of


Effect of Tibial Plateau Fracture on Page 23 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 22 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

36. Mazzucco D, Scott R, Spector M. Composition of joint fluid in patients undergoing total<br />

chondroprotection by lubricin tribosupplementation in the rat following anterior cruciate<br />

knee replacement and revision arthroplasty: correlation with flow properties.<br />

ligament transection. Arthritis Rheum. 2010; 62:2382-91.<br />

Biomaterials. 2004; 25:4433-45.<br />

28. Schmidt TA, Sah RL. Effect of synovial fluid on boundary lubrication of articular<br />

37. Saari H, Konttinen YT, Tulamo RM, Antti-Poika I, Honkanen V. Concentration and<br />

cartilage. Osteoarthritis Cartilage. 2007; 15:35-47.<br />

degree of polymerization of hyaluronate in equine synovial fluid. Am J Vet Res. 1989;<br />

29. Schatzker J, McBroom R, Bruce D. The tibial plateau fracture: the Toronto experience<br />

50:2060-3.<br />

1968-1975. Clin Orthop Rel Res. 1979; 138:94-104.<br />

38. Neu CP, Reddi AH, Komvopoulos K, Schmid TM, DiCesare PE. Increased friction<br />

30. Pascual E, Doherty M. Aspiration of normal or asymptomatic pathological joints for<br />

coefficient and superficial zone protein expression in patients with advanced<br />

diagnosis and research: indications, technique and success rate. Ann Rheum Dis. 2009;<br />

osteoarthritis. Arthritis Rheum. 2010; 62:2680-7.<br />

68:3-7.<br />

39. Hooiveld M, Roosendaal G, Wenting M, van den Berg M, Bijlsma J, Lafeber F. Short-<br />

31. Schmidt TA, Gastelum NS, Nguyen QT, Schumacher BL, Sah RL. Boundary lubrication<br />

term exposure of cartilage to blood results in chondrocyte apoptosis. Am J Pathol. 2003;<br />

of articular cartilage: role of synovial fluid constituents. Arthritis Rheum. 2007; 56:882-<br />

162:943-51.<br />

91.<br />

40. Jansen NW, Roosendaal G, Wenting MJ, Bijlsma JW, Theobald M, Hazewinkel HA,<br />

32. Schumacher BL, Block JA, Schmid TM, Aydelotte MB, Kuettner KE. A novel<br />

Lafeber FP. Very rapid clearance after a joint bleed in the canine knee cannot prevent<br />

proteoglycan synthesized and secreted by chondrocytes of the superficial zone of<br />

adverse effects on cartilage and synovial tissue. Osteoarthritis Cartilage. 2009; 17:433-<br />

articular cartilage. Arch Biochem Biophys. 1994; 311:144-52.<br />

40.<br />

33. Jay GD, Elsaid KA, Zack J, Robinson K, Trespalacios F, Cha CJ, Chichester CO.<br />

Lubricating ability of aspirated synovial fluid from emergency department patients with<br />

knee joint synovitis. J Rheumatol. 2004; 31:557-564.<br />

34. Asari A, Miyauchi S, Sekiguchi T, Machida A, Kuriyama S, Miyazaki K, Namiki O.<br />

Hyaluronan, cartilage destruction and hydrarthrosis in traumatic arthritis. Osteoarthritis<br />

Cartilage. 1994; 2:79-89.<br />

35. Praest BM, Greiling H, Kock R. Assay of synovial fluid parameters: hyaluronan<br />

concentration as a potential marker for joint diseases. Clin Chim Acta. 1997; 266:117-28.


Effect of Tibial Plateau Fracture on Page 25 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Effect of Tibial Plateau Fracture on Page 24 of 25<br />

Lubrication Function and Composition of Synovial Fluid 8/22/2011<br />

Figure 4. Effect of tibial plateau fracture on (A) hyaluronan, (B,C) proteoglycan-4, and (D)<br />

TABLE LEGENDS<br />

protein. (A,C,D) Concentrations were determined for synovial fluid from control (Ctrl-hSF, n=6)<br />

Table I. Summary of donor demographics. Tibial plateau fractures are categorized according<br />

or fractured (Plat-hSF, n=5) knees. Data are Mean±SEM. * indicates p


CME Question(s) Provisionally Accepted Manuscripts Only<br />

Acknowledgement<br />

CME Questions<br />

Acknowledgments<br />

1. Proposed mechanisms of post-traumatic arthritis do NOT include<br />

a. enzyme-mediated cartilage degradation<br />

b. articular chondrocyte death and cartilage damage due to direct trauma<br />

c. reactive oxygen species<br />

d. synovial fluid electrolyte abnormalities<br />

e. impaired lubrication<br />

Vedant Kulkarni, MD, Jonah Hulst, MD, Hugo Sanchez, MD, Suzanne Steinman, MD, UCSD<br />

orthopaedics residents, graciously contributed to clinical sample acquisition for this study. This<br />

work was supported by a grant from the Orthopaedic Trauma Association (BLB), NIH<br />

R01AR051565 (RLS), an NSF Graduate Research Fellowship (JMA), NIH 5T35HL007491 (for<br />

Page 4, lines 14-17: “Recent studies have focused on articular chondrocyte death and<br />

cartilage damage due to direct trauma, enzyme-mediated cartilage degradation, and the<br />

role of reactive oxygen species 13-16 . Deficient lubrication may also contribute to cartilage<br />

deterioration after trauma.”<br />

AYH), and an award to the University of California, San Diego under the Howard Hughes<br />

Medical Institute Professors <strong>Program</strong> (for RLS).<br />

2. In the early (day 1-30) period after tibial plateau fracture, synovial fluid exhibits a<br />

a. decrease in lubrication properties<br />

b. decrease in hyaluronan concentration<br />

c. decrease in PRG4 (lubricin) concentration<br />

d. decrease in total protein concentration<br />

e. decrease in RBC count<br />

Page 13, lines 2-9: “The concentration of hyaluronan was markedly lower in fracture<br />

synovial fluid than control synovial fluid (0.27 mg/ml versus 2.53 mg/ml, –87%,<br />

p460 kDa in synovial fluid samples (Fig. 4-B), the concentration<br />

of proteoglycan-4 was markedly higher in fracture synovial fluid than control synovial<br />

fluid (356 versus 139 μg/ml, +156%, p


Figure 1<br />

Click here to download high resolution image<br />

1 1 1 III 41B2.2 7 1 of 1 ORIF 25 F pedestrian v. auto<br />

2 2 2 V 41C3.3 8 1 of 2 ex fix 55 M fall from fence<br />

3 3 3 II 41B3.1 11 2 of 2 ORIF 57 F motor vehicle accident<br />

4 4 V 41C3.3 20 2 of 2 ORIF 57 M loading dock box crush<br />

5 5 VI 41C3.3 10 2 of 2 ORIF 25 M altercation<br />

6 6 II 41B3.3 8 1 of 1 ORIF 31 M fall from telephone pole<br />

7 4 V 41C3.3 27 1 of 1 ORIF 44 M soccer<br />

8 5 II 41B3.1 3 1 of 1 ORIF 42 M ATV acccident<br />

Gender Trauma Mechanism<br />

Age<br />

Procedure<br />

Type<br />

Procedures<br />

Procedure #<br />

of Total # of<br />

Day<br />

Postinjury<br />

Schatzker OTA<br />

Classification<br />

#<br />

Trauma<br />

Sample<br />

#<br />

Control<br />

Sample<br />

Donor<br />

#<br />

Table I


Figure 3<br />

Click here to download high resolution image<br />

Figure 2<br />

Click here to download high resolution image


Figure 5<br />

Click here to download high resolution image<br />

Figure 4<br />

Click here to download high resolution image


OSTEOCHONDRAL<br />

GRAFTING<br />

WHO,WHEN,WHERE,WHY,& HOW<br />

12/17/2010<br />

Ralph A. Gambardella, MD<br />

Kerlan-Jobe Orthopaedic Clinic<br />

Los Angeles, California<br />

Treatment Options<br />

Presentation Goals<br />

• Review of Clinical History<br />

• Review of Bioengineering History<br />

• Review of Surgical Technique<br />

Surgical Options<br />

• Lavage, debridement<br />

• Drilling, abrasion, microfracture<br />

• Osteochondral autograft g<br />

• Chondrocyte Implantation<br />

• Osteochondral allograft<br />

12/17/2010<br />

1


WHO ?<br />

• NO age limit<br />

• Generally under the age of 50 years<br />

• Focal Focal, full thickness articular cartilage<br />

defects<br />

WHEN ?<br />

• Defect size variable in the literature<br />

–One cm<br />

– Five cm<br />

• Literature supports best results in young<br />

• Femoral Condyle> Trochlea> Patella<br />

WHEN ?<br />

• Most often after failed debridement and or<br />

microfracture<br />

• Appropriate as the initial surgical treatment<br />

of focal full thickness lesions<br />

WHEN ?<br />

• Osteochondritis Dissecans<br />

• More Controversial<br />

– Osteonecrosis<br />

– Osteoarthritis<br />

2


WHERE ?<br />

ANYWHERE<br />

• Knee<br />

• Ankle<br />

• Shoulder<br />

•Hip<br />

WHERE ?<br />

WHY ? Osteochondral Autografts<br />

• Non-inflammatory healing response<br />

• Fill defects with osteochondral bone graft<br />

• Favorable results at 7-10 years<br />

• Immediate access to graft<br />

• Minimally-invasive procedure<br />

12/17/2010<br />

3


WHY ?<br />

CLINICAL STUDIES<br />

Bobic (1995)<br />

• Condyle lesion with ACL injury<br />

• Arthroscopic technique with ACL<br />

reconstruction<br />

• 29 patients with >1cm lesion<br />

• 19/22 excellent results @ 2-3 year followup<br />

• Hyaline cartilage biopsy specimens<br />

Autograft Development<br />

• Open mosaicplasty<br />

– Hangody<br />

• Open/arthroscopic OATS<br />

– Bobic<br />

• Open/arthroscopic COR<br />

– Barber-Chow<br />

Hangody (1997)<br />

• Preliminary report<br />

• 44 patients<br />

• Open technique with autograft<br />

• Open technique with autograft<br />

• HSS score<br />

–Pre: 62<br />

– Post: 94<br />

4


Hangody Results<br />

• Multicenter prospective study<br />

• 417 patients<br />

• 1992 to 1996<br />

• Arthroscopic technique<br />

• Femoral condyle lesions<br />

Hangody 2004 JBJS<br />

• 831 Patients<br />

• Up to 10 year follow-up<br />

• Good to excellent results:<br />

– 92% Femoral<br />

– 87% Tibial<br />

– 79% Trochlear and Patellar<br />

– 94% Talar Dome<br />

Post-op Improvement<br />

Modified Cincinnati Knee Score<br />

1 year 3 year 5 year<br />

Abrasion 58 % 28 % 0 %<br />

Microfracture 57 % 33 % 34 %<br />

Drilling 21 % 33 % 34 %<br />

Mosaicplasty 89 % 88 % 87 %<br />

Hangody 2004 JBJS<br />

• 3% morbidity<br />

• 69 of 83 second look<br />

arthroscopy with<br />

congruent surface and<br />

viable chondrocytes<br />

with histology<br />

5


Hangody 2004 JBJS<br />

• Histologic evidence of<br />

long term graft<br />

survival<br />

• Fibrocartilage filling<br />

of donor sites<br />

Marcacci 2005 Arthoscopy<br />

• 37 patients in a prospective study<br />

• 2 year f/u<br />

• 78 % good to excellent using ICRS Score<br />

• Young patients and LFC lesions did the best<br />

Hangody 2004 JBJS<br />

• Recommendations<br />

– Defects 1-4 square cms. in size<br />

– Attention to detail in technique<br />

– Upper limit age of 50 yrs<br />

WHY ?<br />

BASIC SCIENCE<br />

STUDIES<br />

6


Bioengineering Concerns<br />

• Proud Plug<br />

– Sees increased joint load<br />

– Progressive loss of surface<br />

– Damage to opposing surface<br />

• Recessed Plug<br />

– Sees decreased joint load<br />

– Integration of soft fibrous tissue<br />

– Decreased nutrition (fluid – bone)<br />

Topographic Considerations<br />

Osteochondral Grafting<br />

Ahmad et al AOSSM March 2001<br />

• Donor Sites<br />

– Medial trochlea, lateral trochlea, intercondylar notch<br />

• Small nonloading region<br />

• Si Similar il cartilage il thickness hi k (2.1mm (2 1 ave.) )<br />

• Recipient Sites<br />

– Lateral and medial trochlea curvature best match for<br />

femoral condyles<br />

– Intercondylar notch curvature best match for central<br />

trochlea<br />

– Cartilage thickness (2.5mm ave.)<br />

Topographic Matching for<br />

Osteochondral Grafting<br />

Bartz et al AOSSM March 2001<br />

• Loadbearing condylar recipient sites<br />

– MMost tmedial di l or lateral l t lpatellar t ll groove donors d are bbest t<br />

– Most inferior groove donor site provides best match<br />

• Intercondylar notch donor sites<br />

– Accurate surface restoration for 4-6mm defects<br />

– Inadequate surface restoration for >8mm defects<br />

Cole 2002 AOSSM<br />

• Contact pressure at donor site of<br />

patellofemoral joint<br />

• Pressure is not uniform<br />

Pressure is not uniform<br />

• Pressure is higher on the lateral condyle<br />

• Harvest grafts from medial condyle<br />

7


Koh 2002 AOSSM<br />

• Graft height mismatch<br />

• Small incongruities lead to significantly<br />

elevated contact pressures<br />

– 0.5mm proud worse than 0.5mm sunk<br />

Bioengineering Concerns<br />

Evans 2004 Arthroscopy<br />

• Manual versus Power Punch for Harvest<br />

• Chondrocyte viability better with the use of<br />

a manual punch<br />

Burks 2002 AOSSM<br />

• Pressure changes from defects in femoral<br />

condyle dl<br />

• 15mm diameter leads to 150% increase in<br />

pressure transference to normal cartilage<br />

Bioengineering Concerns<br />

Lane 2004 AJSM<br />

• Goat 6 Month Study<br />

• Cleft between host and transferred region<br />

remains<br />

8


Bioengineering Concerns<br />

Huntley 2005 JBJS<br />

• Chondrocyte Death From Graft Harvesting<br />

– Fresh human tissue<br />

– Confocal microscopy<br />

• Central 99 % viable<br />

• 382 micron margin of cell death<br />

• No change in 2 hours<br />

Autograft Systems<br />

•COR<br />

• OATS<br />

• Mosaicplasty<br />

• DBCS<br />

HOW ?<br />

Cartilage Repair System<br />

NEW GENERATION IN OSTEOCHONDRAL TRANSPLANTATION<br />

Improved Accuracy<br />

Reproducible and focused graft harvest and<br />

drilling with a first-of-kind perpendicularity device<br />

Protecting Chondrocyte Viability<br />

“No impact transfer” & “Low impact delivery”<br />

Ease of Use<br />

Intuitive handling and efficiency<br />

combined in a completely disposable system<br />

9


Improved Accuracy<br />

Harvester and Drill Guide w/ Perpendicularity<br />

Rod<br />

Cutting<br />

Tooth<br />

Underscores<br />

Graft<br />

Ensures<br />

complete<br />

uniform plug<br />

Ease of Use<br />

Improved Drill Bits 5mm-20mm<br />

depth<br />

Spade Cutting Tip<br />

• Single Use guarantees<br />

sharp tip<br />

• Minimizes tip wandering<br />

and cartilage damage<br />

Fluted Channels<br />

• Reduces drilling force<br />

by removing bone<br />

• Reduces friction and heat<br />

that may cause cell damage<br />

COR Precision Targeting… Easy to Use System<br />

INTUITIVE HANDLING – Easily identified components – labeled and color coded<br />

EFFICIENCY – Complete disposable system always available, no missing parts, no<br />

sterilization<br />

Protects Chondrocyte Viability<br />

Protecting Chondrocyte Viability with “No Impact<br />

Transfer”<br />

Harvester/Delivery Guide Cutter<br />

Interface<br />

• Preloaded System<br />

• Cutter protects and stores plug outside the<br />

guide tube until ready for transfer<br />

• No contact with cartilage surface at any time<br />

Technique Keys<br />

• Perpendicular graft insertion<br />

• Joint congruity restoration<br />

8<br />

5<br />

Graft Loader<br />

• No impact on cartilage surface<br />

• Single step<br />

• Loads plug with minimal<br />

force on cancellous bone<br />

10


Indications<br />

• Contained lesion<br />

• 1cm to 3cm defects<br />

• Normal mechanical alignment<br />

• No kissing lesions<br />

THE<br />

FUTURE<br />

Contraindications<br />

• Osteoarthritis<br />

• Instability<br />

• Patellar maltracking<br />

• Mechanical malalignment<br />

Ideal Cartilage Scaffold<br />

• Synthetic and biodegradable<br />

• Designed to match the physical and mechanical<br />

properties of the recipient tissue<br />

• Integrates to reproduce the native properties of<br />

articular cartilage<br />

– Biomechanical<br />

– Histological<br />

– Biochemical<br />

• Safe, effective and durable results<br />

– IKDC, Cincinnati, KOOS or other validated measures<br />

– MRI and/or similar quantitative assessments<br />

– Minimal adverse events, reoperations and failures<br />

11


Synthetic Osteochondral<br />

Grafting<br />

OBI<br />

SMITH &<br />

NEPHEW<br />

OBI Implant Design<br />

• OBI's multilayered product<br />

addresses the three critical<br />

portions of osteochondral<br />

healing:<br />

– articular cartilage<br />

– tidemark<br />

– subchondral bone<br />

• Each layer is designed to match<br />

the physical and mechanical<br />

properties of the adjacent tissue<br />

articular cartilage<br />

tidemark<br />

subchondral bone<br />

Patent 5,607,474 plus others issued and pending<br />

TruFit Surgical Technique<br />

12


Defect<br />

Preparation of Recipient Site<br />

BGS Plug - Case Study<br />

Plug Implantation<br />

9.0 mm<br />

�� ~ 88 % of treatment site had filled with cartilage<br />

�� Excellent integration with surrounding<br />

�� Normal thickness, homogeneous neocartilage<br />

2 nd Look - 8 months<br />

2.0 mm<br />

Presented by David Caborn, M.D.<br />

Original implant margin<br />

Original implant margin<br />

BGS Plug - Case Study<br />

2nd Look Arthroscopy at 8 months<br />

57-76<br />

51-80<br />

�� Indentation Stiffness measured<br />

with ACTAEON ACTAEON Probe<br />

�� Normal stiffness over 65 65-70% 70% of<br />

original defect area<br />

�� Stiffness above 55 shown to be hyaline (Bae Bae et al, Arth & Rheum 2003 2003)<br />

�� Directly comparable to animal study at same timeframe<br />

Presented by David Caborn, M.D.<br />

13


BGS Plug - Case Study<br />

3 rd Look - 21 months<br />

Original implant margin<br />

�� Entire site had filled with cartilage with<br />

smooth integration<br />

�� Cartilage stiffness of repair and<br />

surrounding tissue within normal range of<br />

hyaline cartilage<br />

�� Area of fibrillation caused by meniscal tear<br />

Presented by David Caborn, M.D.<br />

2007 ICRS WARSAW<br />

Cartilage Repair with TruFit CB<br />

Plug<br />

Spaulding, et.al.<br />

• 8 patients<br />

• Failed debridement or microfracture<br />

• IKDC from 44.6 to 79<br />

• 8 month f/u<br />

Biopsy:<br />

anterior area of<br />

treated site<br />

near center<br />

Saf O/Fast Green shows normal<br />

hyaline like cartilage and<br />

complete subchondral bone fill<br />

BGS Plug - Case Study<br />

3 rd Look - 21 months<br />

Original implant margin<br />

Presented by David Caborn, M.D.<br />

2007 ICRS WARSAW<br />

TruFit Early Results<br />

Sciarretta, et.al.<br />

• 15 patients<br />

15 patients<br />

• 11 mm plugs<br />

• Early improvemrnt with IKDC scoring<br />

14


Point-of-Care Cell Therapy:<br />

Autologous Tissue Repair and<br />

RRegeneration ti<br />

Regenerative Medicine at Johnson & Johnson<br />

CAIS Point-of-Care Cell Therapy<br />

1: BIOPSY<br />

Harvest/Mince Small Tissue Sample<br />

Tissue Loaded<br />

Scaffold<br />

Holding Ring<br />

2: AUTOGRAFT PREPARATION<br />

Fibrin Glue<br />

(Crosseal)<br />

3: SURGICAL IMPLANTATION<br />

Minced tissue<br />

loaded scaffold<br />

+<br />

Disposable Patient Kit:<br />

• Harvesting tool<br />

• Loading Device<br />

• Proprietary Scaffold<br />

• Fixation Staples<br />

Staple<br />

J&J’s Intraoperative, Point-of-Care,<br />

Cell Therapy Paradigm (CAIS)*<br />

Harvest<br />

Same day<br />

Implantation<br />

CAIS DEVICE KIT<br />

Minces tissue<br />

Prepares Implant<br />

*(Cartilage Autograft Implantation System)<br />

CAIS Preclinical Evidence<br />

(Goats and Horses)<br />

• Surgically created, clinically relevant cartilage defects<br />

– Goat: 7 mm diameter, one staple fixation<br />

– Horse: 15 mm diameter, three staple fixation<br />

Implant<br />

Goat<br />

Horse<br />

15


Controlled Rehabilitation in Horses:<br />

Progressive Treadmill Exercise<br />

30 mph Gallop at 6 Months<br />

Histological Analysis of Repair Tissue<br />

Goats (6-mos)<br />

Empty<br />

untreated<br />

Scaffold<br />

alone<br />

CAIS<br />

Horses (12-mos)<br />

Repair Site Appearance<br />

CAIS: one stage procedure<br />

(Horse) at 12 Months<br />

0 5 10 15 mm<br />

Two stage procedure Empty<br />

• CAIS provides excellent fill, uniformity and<br />

integration of repair tissue with the host<br />

CONCLUSIONS<br />

16


Indications<br />

• Contained lesion<br />

• 1cm to 3cm defects<br />

• Normal mechanical alignment<br />

• No kissing lesions<br />

FUTURE NEEDS<br />

• More clinical studies<br />

• Long term<br />

• MRI evaluations<br />

• Bone scan evaluations<br />

• Ultrasound evaluations<br />

• Computerized mapping techniques<br />

Contraindications<br />

• Osteoarthritis<br />

• Instability<br />

• Patellar maltracking<br />

• Mechanical malalignment<br />

FUTURE NEEDS<br />

• More basic science studies<br />

–Stiffness and biomechanical<br />

studies<br />

–Edge integration studies<br />

–Plug depth and Press-fit stability<br />

–Pulsed Electromagnetic field<br />

studies<br />

17


THANK YOU<br />

18


� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety<br />

� Human Clinical Trials<br />

Efficacy<br />

� Future<br />

CAIS and Allogeneic<br />

Transplantion: Clinical<br />

trials<br />

Bert R. Mandelbaum MD DHL<br />

(hon)<br />

FIFA Medical Committee<br />

F-MARC Member<br />

Team Physician US Soccer, LA Galaxy,<br />

Chivas USA, Pepperdine University<br />

CAIS<br />

Overview<br />

Historical Overview<br />

Evolving Concepts/Technology 2009<br />

Autologous Chondrocye Implantation<br />

� 1982-Grande, Peterson Rabbit<br />

study<br />

�� 1987 1987- Clinical Application in<br />

Gothenburg Sweden<br />

� 1996- Carticel Registry<br />

� 2000 -Multiple Labs, new scaffolds<br />

� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

1st Surgery: Su ge y<br />

Harvest<br />

Day 1 Transport<br />

& Process Tissue<br />

CAIS<br />

Overview<br />

ACI Generation l Paradigm:<br />

Complex Two-Stage Procedure<br />

Day 28+:<br />

2 nd Surgery:<br />

Implantation<br />

� 2002-07<br />

Day 28<br />

Transport Day 24<br />

Cell Isolation Expansion<br />

Day 2<br />

Day 23<br />

Formulation<br />

Historical Overview<br />

Evolving Concepts/Technology 2012<br />

Autologous Chondrocye Implantation<br />

� RCT microfracture equivalence<br />

JBJS 2004 /2007<br />

Packaging<br />

ChondroCelect vs MFX<br />

� 2007-2011 Landmark StudiesKOOS<br />

Clinical benefit at 36m<br />

30<br />

� Trochlear, Patella, youth, athletes<br />

20<br />

� 10-20 year outcomes<br />

� RCT superiority over MFX<br />

10<br />

0<br />

BL 6M 12M 18M 24M 30M 36M<br />

1


Generation l<br />

Conclusions<br />

Favorable factors for ACI:<br />

1. Younger patient<br />

2. High pre-op. clinical score<br />

3. Duration


� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

1: BIOPSY<br />

Harvest/Mince Small Tissue Sample<br />

Tissue Loaded<br />

Scaffold<br />

Holding Ring<br />

2: AUTOGRAFT PREPARATION<br />

Cartilage Alone<br />

CAIS<br />

Overview<br />

CAIS<br />

Technique<br />

Fibrin Glue<br />

(Crosseal)<br />

3: SURGICAL IMPLANTATION<br />

Minced tissue<br />

loaded scaffold Staple<br />

+<br />

Disposable Patient Kit:<br />

• Harvesting tool<br />

• Loading Device<br />

• Proprietary Scaffold<br />

• Fixation Staples<br />

Tissue Specific Response<br />

SCID Mouse<br />

Bone/cartilage Paste<br />

In Vitro<br />

H/E<br />

IIn Vi Vivo<br />

Saf-O<br />

In Vivo<br />

MMAB<br />

Cartilage Alone<br />

Bone/cartilage Paste<br />

Cartilage Autograft Implantation<br />

System (CAIS) DePuy Mitek J & J<br />

Harvest<br />

Same day<br />

Implantation<br />

CAIS DEVICE KIT<br />

� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

CAIS<br />

Overview<br />

Minces tissue<br />

Prepares Implant<br />

Cole, Farr, Hosea, Richmond,<br />

Gambardella Mandelbaum<br />

Clinical Trial 2006-07<br />

30 pateints complete March<br />

31,2007<br />

CAIS Preclinical Evidence (Goats<br />

and Horses)<br />

�Surgically created, clinically relevant cartilage<br />

defects<br />

�Goat: 7 mm diameter, one staple fixation<br />

�Horse: 15 mm diameter, three staple fixation<br />

Goat<br />

Implant<br />

Horse<br />

3


Controlled Rehabilitation in<br />

Horses:<br />

Progressive Treadmill Exercise<br />

30 mph Gallop at 6 Months<br />

CAIS<br />

Histological Analysis of Repair Tissue<br />

Goats (6-mos)<br />

Horses (12-mos)<br />

Empty<br />

untreated<br />

Scaffold<br />

alone<br />

CAIS<br />

Summary of Goat and Horse<br />

Study<br />

Efficacy<br />

� Good integration with adjacent<br />

cartilage and bone<br />

� Cartilage loaded implant performed<br />

better than scaffold alone and empty<br />

ddefect f t<br />

� Long term under strenuous<br />

mechanical challenge in horses at<br />

12 months<br />

� More consistent fill<br />

� More uniform saffranin-O staining<br />

� More hyaline-like appearance<br />

� phenotypic expression of cartilage<br />

markers: Type II vs Type I collagen<br />

CAIS<br />

Morphologic Repair (Horse) 12m<br />

CAIS: one stage procedure<br />

0 5 10 15 mm<br />

Two stage procedure Empty<br />

�excellent fill, uniformity and lateral and basal<br />

integration of repair tissue<br />

CAIS<br />

Immunostaining (goats) 6 months<br />

Tissue<br />

+<br />

Scaffold<br />

Scaffold<br />

alone<br />

Summary of Goat and Horse<br />

Study<br />

Safety<br />

� No adverse events<br />

� graft preparation,<br />

� surgical implantation,<br />

� post-implantation rehab<br />

� clinical examinations<br />

� final necropsy analysis<br />

� Biocompatible<br />

Type I<br />

Type yp II<br />

Type I<br />

Type II<br />

4


� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

CAIS<br />

Overview<br />

Articular Cartilage lesions (Grade lll-IV +OCD<br />

full thickness)<br />

1-8 cm2<br />

Repair Options<br />

� Cellular/Biologic<br />

Transplantation (ACI)<br />

� Periosteum<br />

� scaffolds (HA, collagen MACI,<br />

PLA/PGA fleece) with<br />

chondrocytes<br />

X-rays<br />

Key Milestone: Clinical Trial<br />

Initiation<br />

� Internal development program at<br />

DePuy Mitek and JNJ<br />

Regenerative Therapeutics<br />

� Ongoing g g IDE Clinical study y<br />

� Randomized Controlled Multi-center<br />

� 6 sites<br />

� Ongoing EU clinical study<br />

� Randomized Controlled Multi-center<br />

� 6 countries<br />

CAIS<br />

Case History<br />

Before After<br />

Large focal defect before and after<br />

implant placement using CAIS<br />

�44 y/o male<br />

�Twisted R knee getting out of bed 1-05. He<br />

developed R knee pain, locking and mechanical<br />

symptoms. t<br />

�MRI R knee revealed OCD lesion of MFC<br />

�6-05 underwent arthroscopic debridement and<br />

chondral biopsy followed by ACI 9-05.<br />

�R knee has done well<br />

MRI<br />

5


� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

� Weeks 6-12<br />

CAIS<br />

Overview<br />

CAIS Post Operative and<br />

Rehabilitation <strong>Program</strong><br />

� Advance to FWB as tolerated<br />

�Discard crutch when pain free and gait pattern<br />

normalizes<br />

�� Discontinue brace when quad control achieved<br />

� Gain full A/PROM<br />

� Begin with open chain isometrics, progress to<br />

closed chain through pain free arc of motion;<br />

resistance less than patient’s body weight.<br />

Balance, proprioception, PRE hamstrings, calves,<br />

hips, upper quadrant.<br />

AJSM 2012<br />

Summary Cole, Farr, Mandelbaum of Goat et and al Horse<br />

Study<br />

Safety<br />

� No adverse events<br />

� graft preparation,<br />

� surgical implantation,<br />

� post-implantation rehab<br />

� clinical examinations<br />

� final necropsy analysis<br />

� Biocompatible<br />

Change from Baseline<br />

60<br />

40<br />

20<br />

0<br />

CAIS Post Operative and<br />

Rehabilitation <strong>Program</strong><br />

�Weeks 0-2<br />

�TDWB, brace locked in full extension<br />

�Begin CPM post-op<br />

�6-8 hours/day, 0-45� advance to 90� as tolerated<br />

�Weeks �Weeks 22-6 6<br />

�TDWB, brace unlocked<br />

�Continue CPM through week 3<br />

�Increase PROM as tolerated<br />

�Weeks 0-6<br />

�Isometric quad strengthening, SLR, Hip abd/add,<br />

hamstring isometrics, stationary bike for PROM<br />

CAIS Post Operative and Rehabilitation<br />

<strong>Program</strong><br />

� 12 weeks and beyond<br />

� FWB with normalized gait pattern<br />

� No brace<br />

� Full pain-free AROM<br />

� 3-6 months<br />

� Treadmill walking, g, bilateral closed chain exercises as tolerated, ,<br />

progression of stationary bike resistance, Stairmaster


� Overview<br />

� Previous experience<br />

�� Rationale for CAIS<br />

� Animal study<br />

� Human Clinical Trials<br />

Safety and Efficacy<br />

� Rehabilitation and postoperative<br />

care<br />

� Future<br />

CAIS<br />

Overview<br />

Autologous vs Allogeneic Cells<br />

• Autologous Cell-based Technologies<br />

- Advantages<br />

o No disease transmission concern<br />

o No immuno-rejection j concern<br />

o No donor tissue/cell shortage issue<br />

- Disadvantages<br />

o Repair efficacy with autologous tissue or cell grafts is<br />

effected by the age of the patient<br />

o Donor site morbidity concern<br />

o Limited availability of autologous donor tissue/cells<br />

o Two surgeries are required in the case of autologous<br />

cultured chondrocyte implantation<br />

Clinical Literature Related to<br />

Immunological Properties of Cartilage Allograft<br />

� Cartilage is immune privileged<br />

– Gibson et al. Brit J Plast Surg 1958; 11:177<br />

� Three decades of success with fresh osteochondral allograft<br />

transplantation – immune suppressive agents g not required<br />

– Gross (Clin Orthop Relat Res. 2005 435:79-87)<br />

– Bugbee (J Knee Surg. 2002 Summer;15(3):191-5)<br />

– Convery (Clin Orthop Relat Res. 1991:(273):139-45)<br />

� Osseous component occasionally could give rise to moderate<br />

immunologic reaction<br />

– Elves & Zervas Br J Exp Pathol 1974; 55:344-351<br />

� Pivotal study<br />

CAIS<br />

Future<br />

� Profiling and stratification of patients,<br />

timing and other cohort details<br />

� Arthroscopic techniques<br />

� Other technologies in conjunction<br />

…growth factors<br />

Autologous vs Allogeneic Cells<br />

• Allogeneic Cell-based Technologies<br />

- Advantages<br />

o No donor site morbidity issue<br />

o Si Single l surgery<br />

o Possible to obtain tissues/cells younger than patients’ own<br />

tissues/cells<br />

- Disadvantages<br />

o Disease transmission concern (actual risk is very low)<br />

o Immuno-rejection concern (actual risk is very low)<br />

o Donor joint supply limitation<br />

Immune-privilege of Juvenile Chondrocytes<br />

H. Nochi, P.R. Streeter, C. Milliman C, K.A. Hruska and H.D. Adkisson (2004),<br />

Transaction of the 50 th Annual Meeting of Orthopaedic Research Society<br />

San Francisco, CA. p.594<br />

Mixed lymphocyte reaction (MLR) assay was conducted to determine the proliferation of human<br />

peripheral blood lymphocytes when co-cultured with allogeneic human juvenile chondrocytes.<br />

Key y findings g from the study y are:<br />

• Juvenile chondrocytes do not elicit an allogeneic immune response.<br />

• Juvenile chondrocytes lack the expression of specific co-stimulatory molecules required for<br />

alloreactivity.<br />

• Juvenile chondrocytes express cell surface proteins that suppress lymphocytes proliferation.<br />

7


Adult vs Juvenile Cells<br />

Juvenile cartilage cell density ≈ 10 x Adult cartilage cell density<br />

Juvenile Cartilage Adult Cartilage<br />

Tissue Engineered Cartilage Grafts with Allogeneic<br />

Juvenile Chondrocytes<br />

(DeNovo ® ET)<br />

Developed by Zimmer, Inc. and ISTO Technologies, Inc.<br />

(under clinical investigation in the US; not approved for sale in US or European<br />

Union)<br />

Cell Morphology and collagen fibrillar structure<br />

Native Juvenile cartilage<br />

Juvenile Adult DeNovo Neocartilage ET<br />

Native Tissue Engineered Tissue DeNovo ET<br />

Adult vs Juvenile Cells<br />

Juvenile cells have better biosynthetic activities<br />

S-GAGG/DNA/Day<br />

3<br />

2<br />

1<br />

0<br />

0 10 20 30 40 50 60 70 80<br />

Age (years)<br />

J. Feder, H.D. Adkisson, N. Kizer, K.A. Hruska, R. Cheung, A. Grodzinsky, Y. Lu, J. Bogdanske and M. Markel (2004)<br />

In: Tissue Engineering in Musculoskeletal Clinical Practice (eds. L. Sandell & A. Grodzinsky),<br />

American Academy of Orthopaedic Surgeons, Rosemont, IL, pp.219-226<br />

DeNovo ET Implant: What is it?<br />

� Cartilage allograft grown with juvenile human chondrocytes<br />

(NO FETAL OR STILL-BORN INFANT TISSUES/CELLS ARE USED)<br />

� Secured Juvenile Donor into cartilage Minced defects with a fibrin adhesive<br />

Tissue<br />

Tissue<br />

Digestion<br />

Primary Cells<br />

Thaw and Seed<br />

Released Cells<br />

Expansion<br />

Culture<br />

Expanded Cells<br />

Implant<br />

Culture<br />

No scaffold<br />

Cryo-Preserve<br />

NC lot # 1<br />

NC lot # 2<br />

NC lot # 3<br />

NC lot # 4<br />

Donor “Cell Bank”<br />

DeNovo ET graft characteristics<br />

• intense staining for GAG indicating the abundance<br />

of proteoglycans in the extracellular matrix<br />

• Biochemical composition indicative of hyaline cartilage<br />

� Type II, VI, IX and XI collagens deposited in the ECM<br />

� No measurable Type I or X collagen in the ECM)<br />

DeNovo ET<br />

8


Allogeneic Particulated Cartilage Graft<br />

(DeNovo ® NT)<br />

Processed by ISTO Technologies, Inc.<br />

Marketed by Zimmer, Inc.<br />

(only available to surgeons in US currently in a limited release)<br />

(not approved for sale in the European Union)<br />

DeNovo NT Allograft Cartilage<br />

THANK YOU<br />

Bert R. Mandelbaum MD DHL<br />

What is it?<br />

� Cartilage tissue allograft composed of juvenile donor hyaline cartilage<br />

pieces<br />

� Mechanically minced with a high cell viability<br />

� Secured into cartilage defects with a fibrin adhesive<br />

Juvenile Donor Joint Minced<br />

DeNovo NT Implant<br />

Tissue<br />

Frisbie et. al. (2006)<br />

(a) DeNovo NT human cartilage graft<br />

(b) Implanted in chondral defects of the horse knee<br />

(c) Good qualities of both cartilage repair tissue and subchondral bone<br />

6-month horse study<br />

9


Osteochondral Allograft<br />

Transplantation:<br />

Basic Science<br />

William Bugbee, MD<br />

Att Attending di Ph Physician, i i Scripps S i Clinic Cli i<br />

Associate Professor<br />

Department of Orthopaedics<br />

University of California, San Diego<br />

Governing Principles for<br />

Osteochondral Allografting<br />

• Whole tissue or organ transplant<br />

• Viable chondrocytes<br />

• Mature hyaline cartilage matrix<br />

• Chondrocytes survive<br />

transplantation and maintain<br />

matrix<br />

• Cells within matrix are<br />

immunopriveleged<br />

How Do We Establish These<br />

“Governing Principles”?<br />

Basic Science Collaboration<br />

David Amiel, PhD<br />

Connective Tissue Biochemistry Lab<br />

Department of Orthopedics, UCSD<br />

Robert Sah, MD, ScD<br />

Cartilage Tissue Engineering Lab<br />

Department of Bioengineering, UCSD<br />

Basic Science<br />

• Historical Context<br />

• Surgical technique<br />

• Immunology<br />

• Disease transmission<br />

• Retrieval analysis<br />

• Allograft recovery and processing<br />

• Allograft storage<br />

• Fresh vs Frozen<br />

• Tissue engineering with allografts<br />

Governing Principles for<br />

Osteochondral Allografting<br />

• Osseous portion is a nonliving<br />

osteoconductive scaffold<br />

• Interface for attachment and<br />

integration<br />

• Transplant p minimal bone volume<br />

necessary for restoration or fixation<br />

• Graft incorporates by creeping<br />

substitution<br />

• Potential site for immunologic<br />

response<br />

• Behavior of osseous component is<br />

most important factor in clinical<br />

outcome<br />

Fresh Allograft<br />

Recovery and Processing<br />

• Before 2002<br />

– Institution associated tissue banks<br />

– “Exceptional release criteria”<br />

– IRB, independently developed recovery,<br />

processing and release protocols<br />

– Recovery y to implantation p less than 7 days y<br />

– Less than 100 per year<br />

• After 2002<br />

– Commercial distribution by national tissue<br />

banks<br />

– Standardized protocols, AATB and FDA<br />

oversight (CFR 1271)<br />

– Recovery to implantation 7-42 days<br />

– More than 2000 per year<br />

1/23/2012<br />

1


“Logistics” of Osteochondral<br />

Allograft Transplantation<br />

Donor<br />

Patient<br />

• Donor recovery<br />

• Evaluation and indication<br />

• Donor suitability<br />

• Consent and counseling<br />

evaluation<br />

• Communication with<br />

• Processing and testing tissue bank<br />

• Graft storage<br />

• Anatomic sizing<br />

• Donor and recipient • Insurance approval<br />

matching<br />

• Patient notification of<br />

• <strong>Final</strong> graft release<br />

potential donor<br />

• Shipping<br />

• Graft release<br />

• 28 day window to implant • Surgical scheduling<br />

Fresh Osteochondral Allografts are<br />

More Effective than Frozen<br />

Osteochondral Allografts in Repair<br />

of Cartilage Defects in the Goat<br />

*Andrea L Pallante, ‡Simon Görtz, +Won C Bae,<br />

*Albert C Chen, ‡Derek Chase, ‡Scott T Ball,<br />

+Christine B Chung, +Graeme M Bydder, ‡David Amiel, *Robert L Sah, ‡°William D<br />

Bugbee<br />

University of California-San Diego,<br />

Departments of *Bioengineering, +Radiology, ‡Orthopaedic Surgery, La Jolla, CA<br />

°Scripps Clinic, Division of Orthopaedic Surgery, La Jolla, CA<br />

Department of<br />

Bioengineering<br />

skeletally,<br />

mature goat, n = 8<br />

Non-OPERATED OPERATED<br />

O = 8 mm<br />

h = 5 mm<br />

Experimental Design<br />

MFC<br />

Treatment Storage<br />

Time [d]<br />

MFC<br />

n<br />

LT<br />

n<br />

Non-OP ── 8 8<br />

FROZEN ── 4 4<br />

FRESH 3 4 4<br />

LT<br />

harvest donor cores<br />

+<br />

mark k & ddrill ill recipient i i t sites it grafted ft d knee k<br />

LT<br />

1 cm<br />

MFC<br />

=<br />

Chondrocyte Viability<br />

• Living chondrocytes are the fundamental basis<br />

for fresh allografting<br />

• Maintenance of intact hyaline cartilage matrix<br />

(fresh vs. frozen)<br />

• Long term graft function<br />

Allograft Efficacy May Be Dependent on<br />

Viable Chondrocytes & Mechanical Stability<br />

• retrieved fresh grafts<br />

– patients required revision/TKA � bias<br />

– contain viable cells up to 29 yrs after implant 1<br />

– failures demonstrate: 2<br />

low # chondrocytes ↑ surface f roughness necrotic ti bbone<br />

articular surface<br />

articular surface<br />

cartilage<br />

tidemark<br />

• pannus-like covering, subchondral irregularities3 H & E<br />

Saf-O bone H & E<br />

1Jamali+ 2007. 2Gross 2008. 3Williams+ 2007.<br />

Goat Knee Analysis (6 months)<br />

STRUCTURE: MRI<br />

• en bloc knee samples<br />

• clinical sequences<br />

– T1w: T1-weighted w/o fat saturation<br />

– T2w FS: T2-weighted w/ fat saturation<br />

STRUCTURE: Gross Grade<br />

1 cm<br />

Non-OPERATED OPERATED<br />

LT<br />

Harvest Plugs LT<br />

MFC<br />

MFC<br />

HOST GRAFT HOST<br />

T1w T2w FS<br />

Loadd<br />

1 cm<br />

TISSUE MATRIX:<br />

Indentation Stiffness<br />

Time<br />

CHONDROCYTE<br />

VIABILITY &<br />

CELLULARITY<br />

• LIVE/DEAD: vertical & en face<br />

TISSUE MATRIX:<br />

mCT<br />

• w/ Hexabrix<br />

contrast<br />

Histology<br />

•H & E<br />

•Saf-O<br />

1/23/2012<br />

2


LIVE<br />

DEAD<br />

LIVE+DEAD<br />

Viability in Grafts @<br />

t=0 (implant)<br />

100 μm<br />

FROZEN<br />

FRESH<br />

LIVE<br />

DEAD<br />

LIVE+DEAD<br />

Reflects Cell Content @<br />

t = 6 months<br />

Non-OP<br />

250 μm<br />

FROZEN<br />

FRESH<br />

Media Optimization for the Storage<br />

of Fresh Osteochondral Allografts<br />

• Ongoing translational research project<br />

• Collaborative, interdisciplinary<br />

• NIH supported t d<br />

• Now part of standardized tissue bank recovery<br />

and processing protocols<br />

• Partially solved graft availability problem<br />

• Facilitated osteochondral transplantation surgery<br />

outside of specialized centers<br />

% Viability V<br />

100%<br />

90%<br />

80%<br />

70%<br />

60%<br />

50%<br />

40%<br />

30%<br />

20%<br />

10%<br />

0%<br />

FBS versus SFM: Viability by Layer<br />

superficial<br />

middle<br />

deep<br />

Day 1 Control Day 28 SFM Day 28 FBS<br />

MFC<br />

LT<br />

Frozen Grafts Exhibit ↓<br />

Matrix Content<br />

Non-OP FROZEN<br />

FRESH<br />

Allograft Storage Studies<br />

2002-2010<br />

• Williams SK, Amiel D, Ball ST, Allen RT, Wong VW, Chen AC, Sah RL, Bugbee WD: Prolonged<br />

Storage Effects on the Articular Cartilage of Fresh Human Osteochondral Allografts. J Bone Joint<br />

Surg Am 85-A(11):2111–20, November 2003.<br />

• Ball ST, Amiel D, Williams SK, Tontz W Jr., Chen A, Sah RL, Bugbee WD: The Effects of Storage<br />

on Fresh Human Osteochondral Allografts. Clin Orthop Relat Res (418): 246–56, January 2004.<br />

• Allen RT, Robertson CM, Pennock AT, Bugbee WD, Harwood FL, Wong VW, Chen AC, Sah RL,<br />

Amiel D: Analysis of Stored Osteochondral Allografts at the Time of Surgical Implantation. Am J<br />

Sports p Med 33(10):1479–94. ( ) October 2005.<br />

• Pennock AT, Robertson CM, Wagner F, Harwood FL, Bugbee WD, Amiel D: Does Subchondral<br />

Bone Affect the Fate of Osteochondral Allografts During Storage? Am J Sports Med 34(4):586–<br />

91, April 2006.<br />

• Robertson CM, Allen TR, Bugbee WD, Harwood FL, Healey RM, Amiel D: Upregulation of<br />

Apoptotic and Matrix-related Gene Expression During Fresh Osteochondral Allograft Storage.<br />

Clin Orthop Relat Res 442:260–66, January 2006.<br />

• Pennock AT, Wagner F, Harwood FL, Bugbee WD, Amiel D: Prolonged Storage of<br />

Osteochondral Allografts: Does the Addition of Fetal Bovine Serum Improve Chondrocyte<br />

Viability? J Knee Surg 19(4):265–72, October 2006.<br />

• Pallante AL, Bae WC, Chen AC, Görtz S, Bugbee WD, Sah RL: Chondrocyte Viability is Higher<br />

after Prolonged Storage at 37°C than at 4°C for Osteochondral Grafts. Am J Sports Med., October<br />

2009<br />

Superficial<br />

Middle<br />

Deep<br />

Day 1 Day 28 SFM Day 28 FBS<br />

1/23/2012<br />

3


CPM/mg Carrtilage<br />

80.0<br />

70.0<br />

60.0<br />

50.0<br />

40 40.00<br />

30.0<br />

20.0<br />

10.0<br />

0.0<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

0<br />

Proteoglycan Synthesis<br />

35 SO4 Uptake<br />

Day 1 Day 7 Day 14 Day 28<br />

Williams et al., JBJS Am 2003<br />

Biomechanics Results:<br />

Indentation Stiffness [IRHI]<br />

57.2 ±<br />

10.9<br />

Not significant<br />

61.0 ±<br />

10.2<br />

7 Day 28 Day<br />

Gene Expression in<br />

Stored Allografts<br />

Day 1<br />

Day 21<br />

4.5%<br />

4.0%<br />

3.5%<br />

3.0%<br />

2.5%<br />

2.0%<br />

1.5%<br />

1.0%<br />

0.5%<br />

0.0%<br />

Results:<br />

GAG Content [% hexosamine/dry wt]<br />

Not significant<br />

Not significant<br />

Not significant<br />

4.2 ± 0.7 4.1± 1.3 4.0 ± 0.5 4.2 ± 0.5<br />

1 Day 7 Day 14 Day 28 day<br />

Gene Expression in<br />

Stored Allografts<br />

Microarray Technology Microarray Technology<br />

• RNA �DNA, labeled<br />

with fluorescent dyes, and<br />

then washed over a glass<br />

slide bearing a grid<br />

spotted with DNA<br />

sequences from known<br />

genes.<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Affymetrix<br />

• By analyzing the location<br />

and intensity of the<br />

fluorescent signals, you<br />

can determine the level of<br />

activity for each gene<br />

Affymetrix<br />

Gene Array Results<br />

Up Regulated Apoptosis Genes<br />

Baseline<br />

Day 21<br />

Day 35<br />

Casp6 Casp7 CD30 CD30L Fas FasL TNFa<br />

Robertson, et al. CORR 2006<br />

1/23/2012<br />

4


Mechanism of Cell Death<br />

Apoptosis Pathway<br />

Storage Studies:<br />

Conclusions<br />

• Storage does not affect cartilage matrix or<br />

biomechanical properties.<br />

• Cell viability, y, cell density y and cell metabolism<br />

decreased with increasing storage time.<br />

• Optimum storage conditions not yet defined<br />

– DMEM with FBS<br />

• Euthermic storage vs. refrigerated<br />

– Other potential anabolic/ anti-catabolic substances<br />

• Anti TNF agents (Etanercept)<br />

• In vivo studies are needed to define the<br />

parameters of acceptable graft tissue quality.<br />

Goat Allografts Stored in Media at 4°C<br />

Role of Etanercept<br />

Baseline With Etanercept Without Etanercept<br />

4 Weeks<br />

10%<br />

Superficial<br />

50% Middle<br />

40% Deep<br />

Future Allograft Innovations:<br />

The Next 100 Years<br />

• Changes in tissue banking paradigm<br />

– universal donation, testing/ processing<br />

• Storage or cryopreservation technology<br />

• Molecular modulation of graft<br />

– bone and cartilage growth factors<br />

– Immunoreactivity<br />

• Allografts as scaffolds for cell based technologies<br />

– mature matrix<br />

• Allograft cartilage as a cell source for tissue<br />

engineered repair<br />

• Joint fabrication<br />

1/23/2012<br />

5


Thank You<br />

Summary<br />

• Fresh grafts perform better than frozen<br />

grafts<br />

• Storage necessary for processing but<br />

decreases chondrocyte viability<br />

• If you use osteochondral allografts,<br />

understand what happens before the graft<br />

is delivered to your OR and after it is<br />

implanted.<br />

1/23/2012<br />

6


Osteochondral Allograft: Clinical Experience<br />

David R. McAllister, MD<br />

Professor<br />

Chief, Sports Medicine Service<br />

Department of Orthopaedic Surgery<br />

David Geffen School of Medicine at UCLA<br />

Indications<br />

• Focal articular cartilage lesions of the femoral condyles of the knee<br />

• Large with full-thickness articular cartilage loss (with or without subchondral bone<br />

loss-i.e. OCD)<br />

• Failed previous articular cartilage treatment<br />

Rationale<br />

• Replacing damaged articular cartilage with size-matched LIVE articular cartilage<br />

• Large single plug minimizes fibrocartilage formation (i.e. “grout”)<br />

• Allograft source alleviates donor site morbidity concerns seen with autogenous<br />

cartilage transplantation<br />

• Only articular cartilage procedure which results in hyaline cartilage<br />

Results<br />

• Improved with good fit, unipolar surface, and young graft donor<br />

• Survivorship:<br />

5 years-95%<br />

10 years-71%<br />

20 years-66%<br />

Shasha, et al: Cell and Tissue Banking, 2002<br />

Results<br />

• 73% G/E results after 10 years<br />

• 76 % G/E, overall<br />

• 84% G/E for unipolar lesions<br />

• 50% G/E for bipolar lesions<br />

Chu,et al, CORR, 1999<br />

Factors Affecting Success<br />

• Size<br />

• Fit<br />

• Match<br />

• Alignment<br />

• Fresh/Frozen


Drawbacks<br />

• Can be difficult to obtain fresh non-irradiated grafts<br />

• Risk of disease transmission and infection<br />

• Logistically challenging<br />

Osteochondral Allograft Transplantation<br />

• Allografts are most effective when transplanted fresh. When kept in tissue culture<br />

medium at 4°C, articular graft chondrocytes remain alive although viability decreases<br />

with time.<br />

• Avoid use of use fresh frozen grafts<br />

ACT Instruments Overview<br />

Lesion Gauge<br />

• Sized to Cover entire Lesion<br />

• Tripod Base<br />

– 12, 4, and 8 o’clock Positions<br />

– Best Stability on a Curved Surface<br />

• Cannulated to Accept Guide Pin<br />

• Color Coded Plastic<br />

Guide Pin<br />

• 2.4mm<br />

• Drill Tip<br />

• 30cm in Length<br />

• Calibrated from 0mm to 50mm<br />

– 1.0 cm Increments<br />

– Read from the Proximal End of Lesion Gauge<br />

Guide Pin<br />

Lesion Reamer<br />

• Dual-Cutting Surface<br />

– Outer Edge<br />

• Large Channels to Expel Debris<br />

• Calibrated in 5mm Increments<br />

• Cannulated<br />

• Color Code Banding<br />

Depth Gauge<br />

• Cylinder – One per Size<br />

• Calibrated Circumferentially in 1mm Increments<br />

– Numbers Appear at 12, 4, & 8 O’Clock<br />

• Use Skin Pen to Mark Three Spots<br />

• Transfer Readings to Graft<br />

• Stainless Steel<br />

• Color Code Banding


GraftStation <br />

• Holds Allograft for Graft Preparation<br />

• Guide Arm Adjusts<br />

• Insert Lesion Gauge<br />

• Lock Arm<br />

• Insert GraftMaker <br />

• Create Graft<br />

• Pads on Base Fine-Tune Depth and Chamfer Edges to Aid Insertion<br />

GraftMaker <br />

• Two-Piece Instrument<br />

– Cutting Section<br />

– Driving Section<br />

• Creates Cartilage Graft (core)<br />

• Windows in Cutting Section<br />

– 5mm Increments<br />

– Depth Reading<br />

GraftMaker <br />

• Cutting Section Detaches<br />

– Push Graft Out w/o Touching Cartilage<br />

• Cannulated<br />

• Color Code Banding<br />

Sizing Forceps<br />

• Capture Graft Circumferentially<br />

• Cartilage Not Touched<br />

• Act as a Cutting Guide<br />

– <strong>Final</strong> Depth Cut Made Here<br />

• Ratchet Handle Clamps and Holds Position<br />

Dilator<br />

• Use to Dilate Graft Site, If Needed<br />

• Optional Use to Push Graft Out of GraftMaker, If Using This Technique<br />

• Color Code Banding<br />

Tamp<br />

• Plastic, High-Concave Cut-out<br />

– Protects Cartilage<br />

– Impacts Only on Edge<br />

• Color-Coded Plastic<br />

Retrieval Pin<br />

• Threaded Tip<br />

• 30cm Length<br />

• Attach to Drill to Advance into Graft<br />

• Pull Graft Out of Misaligned Position<br />

• Reposition Graft


Mallet<br />

• Used for Tamping and Dilating<br />

• Slotted for Cannulated Option<br />

• Special Fitting Acts as Wrench<br />

• Used to Tighten GraftStation Clamp<br />

– Wrench Also Included<br />

Two Instrument Cases<br />

• Common Instruments<br />

– Guide Pins<br />

– Retrieval Pin<br />

– Hudson Adaptor<br />

– Pusher Rod<br />

– GraftStation <br />

– Mallet<br />

– Wrench<br />

• Size-Specific Instruments<br />

– Lesion Gauge<br />

– Lesion Reamer<br />

– GraftMaker <br />

– Depth Gauge<br />

– Sizing Forceps<br />

– Dilator<br />

– Tamp<br />

• 12 Steps to OC Tx Success!<br />

Step 1: Size Defect<br />

Step 2: Place Guidepin<br />

Step 3: Ream Lesion<br />

Step 4: Size Lesion and Measure Depth<br />

Step 5: Place donor in Workstation<br />

Step 6: Cut Graft<br />

Step 7: Make Crosscut<br />

Step 8: Remove Graft<br />

Step 9: Mark depth<br />

Step 10: Cut to proper depth<br />

Step 11: Place into defect<br />

Step 12: Tamp into place


12 th International Sports Medicine Fellows Conference<br />

January 27-29, 2012● Carlsbad, California<br />

Session II<br />

9:30 AM - 9:45 AM Autologous Chondrocyte Implantation from Bench to Present Jason Scopp, MD USA<br />

9:45 AM - 10:15 AM<br />

Complex Reconstruction and Osteotomy: Pre-Operative Planning<br />

and Technique (Presentation via Live Video Stream)<br />

Brian Cole, MD USA<br />

10:15 AM - 10:30 AM Patellofemoral Joint Algorithm and Osteotomy to Arthroplasty Jack Farr, MD USA<br />

10:30 AM - 10:45 AM Meniscus Allografts and Meniscal Implants Wayne Gersoff, MD USA<br />

10:45 AM - 11:00 AM<br />

Next Generation Experience BMAC Other Scaffolds: Europe and<br />

Beyond<br />

Alberto Gobbi, MD ITALY<br />

11:00 AM - 11:15 AM Stem Cells: Fact or Fiction and Opportunity C. Thomas Vangsness, MD USA<br />

11:15 AM - 11:30 AM Post Fellowship Award Presentation Christopher Kreulen, MD USA


Autologous Chondrocyte<br />

Implantation<br />

Jason M. Scopp, M.D.<br />

Director, Joint Preservation Center<br />

Peninsula Orthopaedic Associates, PA<br />

Salisbury, Maryland<br />

Functional Unit<br />

The Macroenvironment<br />

• Limb alignment<br />

• Meniscus status<br />

• Ligamentous status<br />

• Cartilage status<br />

• Any change in one factor can affect the<br />

others via Chondral Overload Syndrome<br />

Be a cartilage doctor<br />

An intact surface is an important<br />

thing<br />

Decision Making<br />

• Small lesions ≠ large lesions<br />

• Articular cartilage defect ≠ osteoarthritis<br />

2 Stage Technique<br />

1/4/12<br />

1


Indications<br />

• Symptomatic ICRS Grade III and IV<br />

lesions<br />

• High demand patients age 15-55 with<br />

motivation and potential for compliance<br />

and realistic expectations<br />

• Size >2cm² and < 12cm² (???)<br />

• Continued pain after primary treatment<br />

Arthroscopic Evaluation<br />

• Manage concomitant<br />

pathology<br />

• Size defect<br />

• Initiate primary<br />

treatment<br />

• Biopsy<br />

• Policy<br />

Insurance Policy<br />

Autologous chondrocyte implantation for the treatment of cartilage<br />

defects is considered medically necessary for patients with cartilage<br />

defects of the femoral condyle (medial, lateral or trochlear) when the<br />

case meets the criteria set forth in the Policy Guidelines.<br />

• Patients for treatment with autologous chondrocyte implantation will<br />

present with the following:<br />

– a cartilage defect within a range of 1.5 cm to 3.0 cm in largest<br />

diameter, and<br />

– for whom more conservative treatment has failed, or is likely to fail<br />

to relieve the symptoms.<br />

– The Medical Director may consider patients with variations in<br />

lesion size or location, or those patients with more than one lesion<br />

No mention of ACI as secondary treatment option for this size<br />

The Biopsy<br />

Implant Day The Tools<br />

• Superomedial or<br />

superolateral trochlea<br />

• Lateral intercondylar<br />

notch<br />

• 200-300 mg (2 tic<br />

tacs)<br />

• Cartilage and bone<br />

• 200Kcells à� 12<br />

million<br />

1/4/12<br />

2


Retractors Position<br />

Exposure<br />

• Consider concomitant procedures<br />

• Epi soaked pledgettes<br />

• Thrombin spray<br />

• Gel foam<br />

• Sponge<br />

• Fibrin glue<br />

• Needle-tip bovie<br />

• NO tourniquet<br />

Hemostasis<br />

Defect Preparation<br />

• Debride to normal<br />

cartilage margins<br />

• 15 blade and ring<br />

curette<br />

• Maintain calcified<br />

cartilage layer<br />

• Hemostasis<br />

• Make template for<br />

periosteum<br />

Periosteum Harvest<br />

• Oversize by 2 -3mm<br />

• 3-5 fb below pes<br />

– Thin to win<br />

• Remove fascia and<br />

fat with wet sponge or<br />

fine scissors<br />

– Be careful<br />

• Femoral metaphysis<br />

– Thicker--hypertrophy<br />

– Adhesions (Seprafilm)<br />

1/4/12<br />

3


Don’t forget to mark Graft fixation<br />

Tips for uncontained<br />

• Anchors<br />

– Mitek Microfix<br />

• Drill keith needles<br />

• OATS plug<br />

• 6-O vicryl DYED<br />

• 4 corners of compass<br />

– Trochlea and patella<br />

– Avoid manhole cover<br />

• Leave open region to<br />

implant<br />

• Seal with fibrin glue<br />

• Test for watertight<br />

– epi<br />

Sandwich Technique Lesions<br />

>8mm Deep<br />

Periosteal Replacement Post-op<br />

• Nerve blocks à� Outpatient<br />

• CPM 6-8 hours a day<br />

• PWB 6-8 weeks<br />

• Proliferation Phase: 0-6wà� Soft<br />

• Transition Phase: 7w-6mà� Putty<br />

• Remondeling Phase: 6m-36mà� Stiffens<br />

1/4/12<br />

4


Results<br />

• ACI can be used for defects


Profiling the Optimal Patient<br />

• Fewer surgeries


Brian J. Cole, MD, MBA Office: 312-243-4244 1<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Notes:<br />

Cartilage Repair: Complex Reconstruction with Pre-Operative<br />

Planning and Techniques<br />

1) Presentation<br />

a) Pain<br />

b) Instability<br />

c) Deformity<br />

d) Varus/Lateral Thrust<br />

e) Prior Meniscectomy<br />

2) Treatment Goals<br />

a) Reduced pain<br />

b) Protect cartilage repair<br />

c) Eliminate instability<br />

d) Improve function<br />

3) Cartilage Resurfacing<br />

a) Patient Evaluation<br />

i) Hx<br />

ii) PE<br />

iii) Ancillary Testing<br />

(1) Rosenberg<br />

(2) Long-leg alignment<br />

(3) MRI<br />

iv) Prior arthroscopic hx/records/photos<br />

b) Decision Variables<br />

i) Defect-Specific<br />

(1) Location<br />

(2) Size/Depth<br />

(3) Geometry<br />

(4) Containment<br />

ii) Patient-Specific<br />

(1) Age<br />

(2) Demand-Match<br />

(3) Response to prior tx<br />

(4) Comorbidities<br />

(a) OCD<br />

(b) Alignment<br />

(i) T-F<br />

(ii) P-F<br />

(c) Stability<br />

(d) Meniscal Deficiency<br />

c) General Rules


Brian J. Cole, MD, MBA Office: 312-243-4244 2<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Notes:<br />

i) Avoid thinking linearly<br />

ii) Repair when possible<br />

iii) Re-establish subchondral bone<br />

iv) Do not correct alignment with grafts<br />

v) Avoid burning bridges to future treatment options<br />

d) Options<br />

i) Palliative<br />

(1) Debridement/Lavage<br />

ii) Reparative<br />

(1) Microfracture<br />

(2) Abrasion<br />

(3) Drilling<br />

iii) Restorative<br />

(1) ACI<br />

(2) OC Grafting<br />

(a) Autografts<br />

(b) Allografts


Brian J. Cole, MD, MBA Office: 312-243-4244 3<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Notes:<br />

e) General Algorithm


Brian J. Cole, MD, MBA Office: 312-243-4244 4<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

4) Osteotomy<br />

a) Osteotomy Goals (Mechanical Axis)<br />

i) Pain/Arthrosis � Overcorrect (62% across plateau)<br />

ii) Cartilage Restoration � Correct (50% across plateau minimum)<br />

iii) Instability � Sagittal Slope Correction<br />

Notes:<br />

b) Opening Wedge Advantages<br />

i) Avoid TF Joint/Peroneal Nerve<br />

ii) Maintains Proximal Tibial Shape<br />

iii) Avoids medial tibial displacement<br />

iv) Reduced Infera (less complicated arthroplasty)<br />

v) Intraoperative Correction<br />

vi) Biplane Osteotomy (Sagittal/Coronal)<br />

vii) Simplicity<br />

viii) Lower Morbidity<br />

(1) Single cut<br />

(2) No anterolateral dissection<br />

c) Disadvantages<br />

i) Bone Graft<br />

ii) Non-Union<br />

iii) Large Corrections Difficult<br />

d) Clinical Indications<br />

i) Malalignment + Arthrosis<br />

ii) Malalgnment + Instability<br />

iii) Malalignment + Arthrosis + Instability<br />

iv) Malalgnment + Cartilage Restoration (Meniscus/Articular)<br />

e) Contraindications<br />

i) Contralateral compartment arthrosis/prior lateral meniscectomy<br />

ii) Motion loss (< 0-75 o )<br />

iii) Inability to tolerate postoperative restrictions<br />

iv) Joint subluxation<br />

f) Patient Assessment<br />

i) History/PE<br />

ii) Operative Record Review<br />

iii) Instability Assessment<br />

iv) Short-Leg Cast/Brace Trial<br />

(1) Gait-Analysis<br />

g) Radiographic Assessment<br />

i) Bilateral Standing Hip to Ankle<br />

(1) Single-Leg Standing Hip to Ankle<br />

h) Alignment Assessment<br />

i) Tibiofemoral Angle (5-7 o Valgus): Single-Leg Standing Hip to Ankle


Brian J. Cole, MD, MBA Office: 312-243-4244 5<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Notes:<br />

ii) Mechanical Axis (Neutral): Deviation of weight-bearing line (preferred)<br />

iii) Sagittal Plane: Tibial Slope (1-10 o )<br />

(1) Increased slope = Increased Anterior Translation = Worsens ACL = Helps PCL<br />

(2) Decreased slope = Increases Posterior Translation = Helps ACL = Worsens ACL<br />

i) Surgical Technique<br />

i) Incision: Medial, 5 cm, start 1 cm below joint line extend to bottom of pes anserinus<br />

(similar to ACL incision for tibial tunnel centered b/w TT and posteromedial tibia)<br />

ii) Incise pes anserinus longitudinally and elevate and protect MCL<br />

iii) Elevate superficial MCL<br />

iv) Expose junction b/w fat pad and patellar tendon insertion<br />

v) Right angle under patellar tendon<br />

vi) Guide pin placement<br />

(1) Fluoroscopy<br />

(2) Perpendicular to tibia (coronal)<br />

(3) Start just below beginning of TT (4 cm below joint line)<br />

(4) Cross tibia obliquely at patellar tendon insertion<br />

(5) End at proximal fibula head (1 cm below joint line)<br />

(6) Place 2 nd pin more posterior/attach cutting guide<br />

(7) Cut below guide under fluoroscopic guidance leave lateral 1 cm<br />

(8) Finish with osteotomes<br />

(9) Gradual opening<br />

(10) Plate placement<br />

(a) 2-6.5 mm cancellous screws proximally<br />

(b) 2-4.5 mm cortical screws distally<br />

(11) Bone Graft<br />

(a) ICBG<br />

(b) Cortical ring allograft<br />

(c) DBM supplement<br />

j) Pearls and Pitfalls<br />

i) Expose patellar tendon insertion at tibial tubercle<br />

ii) Flouroscopic guidance<br />

iii) More horizontal cut for larger corrections to improve fixation stability<br />

iv) Osteotomy parallel to sagittal slope joint line<br />

v) Osteotomy below guide pin to avoid fx<br />

vi) Thin flexible osteotomes<br />

k) Rehabilitation<br />

i) Hinged knee brace<br />

ii) Immediate ROM<br />

iii) NWB/HTWB 4-6 Weeks<br />

iv) Gradual WB at 5-6 Weeks (Radiographic healing??)<br />

v) CPM with intraarticular work<br />

vi) 6-12 weeks: functional program strengthening<br />

vii) > 12 weeks: muscular endurance<br />

l) Complications<br />

i) Intraoperative<br />

(1) Fracture


Brian J. Cole, MD, MBA Office: 312-243-4244 6<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Notes:<br />

(2) Intraarticular screw<br />

(3) Medial cortex violation<br />

ii) Early Postoperative<br />

(1) Hematoma<br />

(2) Compartment syndrome<br />

iii) Late Postoperative<br />

(1) Delayed union<br />

(2) Non union<br />

(3) Hardware failure<br />

m) Special Situations<br />

i) ACL & Varus<br />

(a) ACL Alone (Shelbourne)<br />

(b) HTO Alone<br />

(i) Pain<br />

(ii) Open wedge posteriorly (decrease slope)<br />

(c) ACL & HTO<br />

(i) Pain & Instability<br />

(ii) Order<br />

1. Intra-articular preparation<br />

2. Shorter tibial tunnel<br />

3. Femoral tunnel<br />

4. Osteotomy (oblique and start low)<br />

5. Fix osteotomy<br />

6. Pass graft<br />

7. Recess femoral plug to handle graft mismatch<br />

8. Cut tibial plug after fixation<br />

(2) PCL & Varus<br />

(a) Opening wedge anterior (increased tibial slope)<br />

(3) Meniscus Transplant<br />

(a) Young: combine<br />

(b) Old: Osteotomy first, then transplant prn<br />

(4) Articular Cartilage<br />

(a) Young: combine<br />

(b) Old: Osteotomy first, then transplant prn<br />

5) Ligament Reconstruction<br />

a) Cartilage Resurfacing: ACL Always reconstruct<br />

i) Revision common<br />

ii) Avoid tunnel communication (meniscus transplantation)<br />

iii) Bone graft tunnel enlargement first and return<br />

iv) Timing<br />

(1) ACL & Meniscus transplant : Simultaneous<br />

(2) ACL & Malalignment & Cartilage<br />

(a) ACL first<br />

(b) Alighment and Cartilage Resurfacing<br />

(c) May try osteotomy first and then return prn<br />

(d) May try ligament first and then return prn


Brian J. Cole, MD, MBA Office: 312-243-4244 7<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Questions<br />

1. When performing marrow stimulation of the femoral condyle, it is important to:<br />

A. Create vertical walls at the transition zone<br />

B. Violate the calcified layer<br />

C. Initially protect the defect from weigthbearing<br />

D. Implement immediate post operative range of motion<br />

E. All of the above<br />

2. All of the following are true regarding osteochondral allograft transplantation except:<br />

A. The graft should be initially maintained at 37 o C for no more than 28 days<br />

B. A minimal amount of subchondral bone should be transplanted with the graft<br />

C. It is acceptable to utilize a contralateral femoral condyle for a donor graft<br />

D. The graft should be gently impacted<br />

E. Bipolar grafts are associated with a guarded prognosis<br />

3. Classic indications for autologous chondrocyte implantation assume all of the<br />

following except:<br />

A. The defect is associated with a minimal amount of bone loss<br />

B. The defect must be contained<br />

C. Sclerotic areas can be debrided even if bleeding occurs<br />

D. Patellar alignment can be corrected simultaneously or in a staged fashion when<br />

treating trochlear defects<br />

E. A meniscus deficient knee should undergo allograft meniscus transplantation at<br />

the same time the femoral condyle is treated.<br />

4. In a patient with ACL deficiency who is in varus, a high tibial osteotomy can help by:<br />

A. Increasing the posterior slope of the tibia in the sagital plane<br />

B. Decreasing the posterior slope of the tibia in the sagital plane<br />

C. Increasing contact pressures in the lateral compartment<br />

D. Protecting an ACL reconstruction by reducing stress on the ligament<br />

E. Both A and D<br />

Answers<br />

Question 1: E, All of the above<br />

Question 2: B, The graft should be initially maintained at 37 o C for no more than 28 days<br />

Question 3: B, The defect must be contained<br />

Question 4: E, Decreases the posterior slope of the tibia and protects the ACL reconstruction<br />

Notes:


Brian J. Cole, MD, MBA Office: 312-243-4244 8<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

CPT Codes<br />

Meniscus Transplant 29868<br />

Microfracture 29879<br />

Osteochondral Allograft 27415<br />

Osteochondral Autograft 29866<br />

HTO 27457<br />

DFO 27450<br />

Notes:


Brian J. Cole, MD, MBA Office: 312-243-4244 9<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

REFERENCES<br />

Cartilage Restoration<br />

Garretson RB, Katolik LI, Bach BR, Verma N, Cole BJ: Dynamic contact pressure in the<br />

patellofemoral joint. In Press: Am J Sports Med, 2004<br />

Cole BJ, Lee SJ: Complex knee reconstruction: Articular cartilage treatment options.<br />

Arthroscopy, (19) Supplement 1, 1-10, 2003.<br />

Freedman K, Nho S, Cole B: Marrow stimulating techniques to augment meniscus repair,<br />

Arthroscopy, 19(7):794-798, 2003.<br />

Fox JA, Kalsi RS, Cole BJ. Update on Articular Cartilage Restoration, Tech in Knee Surg<br />

2(1):2-17, 2003.<br />

Farr J, Meneghini RM, Cole BJ: Allograft interference screw fixation in meniscus transplantation.<br />

Arthroscopy, In press, March, 2004.<br />

Cole BJ, Rodeo S, Carter T: Allograft Meniscus Transplantation: Indications, Techniques,<br />

Results, J Bone Joint Surg, 84A:1236-1250, 2002.<br />

Fox J, Cole BJ. Management of Articular Cartilage Injuries. Orthopedic Knowledge Update in<br />

Sports Medicine, Volume #3, American Academy of Orthopedic Surgeons, 2003.<br />

Cole BJ, Fox JA, Lee SJ, Farr, J. Bone bridge in slot technique for meniscal transplantation. Op<br />

Tech Sports Med, 11:2, 144-155, April, 2003.<br />

Fox JA, Lee SJ, Cole BJ. Bone plug technique for meniscal transplantation. Op Tech Sports<br />

Med, 11:2, 161-169, April, 2003.<br />

Farr J, Cole BJ: Meniscus transplantation: Bone Bridge in slot technique. Op Tech Sports Med,<br />

10(3):150-156, 2002.<br />

Fox JA, Freedman KB, Lee SJ, Cole BJ. Fresh osteochondral allograft transplantation for<br />

articular cartilage defects. Op Tech Sports Med, 10(3): 168-173, 2002.<br />

Freedman JB, Coleman SH, Olenac C, Cole BJ: The biology of articular cartilage injury and the<br />

microfracture technique for the treatment of articular cartilage lesions. Seminar in Arthroplasty,<br />

13(3): 202-209, 2002.<br />

D’Amato M, Cole BJ: Autologous Chondrocyte Implantation, Orthopedic Techniques, 11:115-<br />

131, 2001.<br />

Cole BJ, Farr J: Putting it all together, Orthopedic Techniques, 11:151-154, 2001.<br />

Miller M, Cole BJ: Atlas on chondral injury, Orthopedic Techniques, 11:145-150:2001.<br />

Notes:


Brian J. Cole, MD, MBA Office: 312-243-4244 10<br />

Section Head, Rush Cartilage Restoration Center` Fax: 312-942-1517<br />

Section of Sports Medicine EM: bcole@rushortho.com<br />

Rush University Medical Center<br />

Chicago, IL<br />

Website: WWW.cartilagedoc.org<br />

Cole BJ, DiMasi M: Single-stage autologous chondrocyte implantation and lateral meniscus<br />

allograft reconstruction. Orthoped Tech Rev. 2:3, 44-59, 2000.<br />

Cole BJ (Section Ed.): Cartilage Restoration: Alternative techniques for the management of<br />

articular cartilage disease. In: The Arthritic Knee, Rosenberg, A.G., Mabrey, J.D., and Woolson,<br />

S.T. (Eds.). Rosemont, Illinois, American Academy of Orthopaedic Surgeons, 2000.<br />

Osteotomy<br />

Brown G, Amendola A: Radiographic evaluation and pre-operative planning for high tibial<br />

osteotomy. Op Tech Sports Med, 8:1, January 2000.<br />

Cole BJ, Freedman KB, Takasali S, Hingtgen B, DiMasi M, Bach Jr. BR, Hurwitz DE: Use of a<br />

lateral offset short-leg walking cast before high tibial osteotomy. Clin Ortho Rel Res, 408:209-<br />

217, 2003.<br />

Puddu G, Cipolla M, Franco V. A plate for open wedge tibial and femoral osteotomies.<br />

Presented at The Congress of the International Society of Arthroscopy, Knee Surgery and<br />

Orthopaedic Sports Medicine; 1999; Washington, DC.<br />

Hernigou P, Medevill D, Debeyre J, et al. Proximal tibial osteotomy with varus deformity: a ten- to<br />

thirteen-year follow-up study. J Bone Joint Surg Am, 69:332, 1987.<br />

Koshino T, Murase T, Saito T. Medial Opening-Wedge High Tibial Osteotomy with Use of Porous<br />

Hydroxyapatite to Treat Medial Compartment Osteoarthritis of the Knee. J Bone and Joint Surg<br />

Am, 85:78-85, January 2003.<br />

Koshino T, Morii T, Wada J, et al. High Tibial Osteotomy with Fixation by a Blade Plate for<br />

Medial Compartment Osteoarthritis of the Knee. Orthopedic Clinics of North America, 20:227-<br />

243, April 1989.<br />

Notes:


12 th Annual International Sports Medicine Fellows Conference<br />

Patellofemoral Joint Algorithm and Osteotomy to Arthroplasty<br />

January 28, 2012 10:15-10:30am<br />

Jack Farr, M.D.<br />

OrthoIndy Knee Care Institute<br />

1260 Innovation Parkway #100<br />

Greenwood, IN 46143<br />

Ph: 317-884-5200<br />

Fax: 317-884-5365<br />

E-mail: indyknee@hotmail.com<br />

Patellofemoral Compartment: AMZ and PFA<br />

Osteotomy Planning<br />

1. Step One: Define the Pathology<br />

a. Classifications to aid in Treatment<br />

i. Morphology<br />

ii. Traumatic vs. Atraumatic (Primary DJD)<br />

iii. Fulkerson Alignment Classification<br />

1. Fulkerson Alignment Classification: Alignment & TT-TG<br />

a. Correlate Patellar position with contribution of TT-TG<br />

b. Mean of Asymptomatic patients: 13 mm<br />

c. Outlier symptomatic group: >20 mm<br />

iv. Regional Chondrosis Mapping<br />

1. Normalization of PF stress without Cartilage Restoration: AMZ<br />

(Pidoriano and Fulkerson, 1997)<br />

a. Inferior Pole and Lateral Facet: 87% G/E<br />

b. Medial Facet: 55% G/E<br />

c. Proximal Pole and Diffuse: 20% GE<br />

d. Concomitant Central Trochlear Involvement: All Poor<br />

v. Soft Tissue Envelope<br />

1. Medial Patellofemoral Ligament Status<br />

2. Lateral Retinaculum Status<br />

2. Step Two: Identify Why There Is Pain<br />

a. Chondrosis ≠ pain<br />

Diagnosis of Exclusion<br />

i. Articular cartilage is aneural<br />

ii. Pain therefore originates from:<br />

1. Soft tissue (Synovium, Capsule, Tendons and Ligaments)<br />

2. Nerves (Local or Remote, e.g., saphenous, neuroma)<br />

3. Bone (Local or Remote, subchondral, referred hip)<br />

4. Need to identify those patients with pain on the basis of the<br />

mechanically identifiable factors and associated chondral defects;


exclude CRPS and dehabilitation (exceeding Dye Envelope of<br />

Function)<br />

3. Step Three: Right Surgery for the Right Patient - Patient Specific Demand Matching<br />

a. Treat both Mechanical and Chondral Pathologies<br />

4. Step Four: Applying the Specific Surgery<br />

a. Tibial Tuberosity Medialization<br />

i. TT-TG distance > 20mm<br />

ii. If suspected Femoral/Tibial rotational abnormalities (e.g., Increased<br />

femoral anteversion): CT assess hip/knee/ankle rotation (Teitge)<br />

b. Deciding on Tuberosity Transfer<br />

i. Degree of Anterization:<br />

1. Ferrandez 10 mm Clin Ortho 1989<br />

2. Ferguson 12.5 JBJS 1979<br />

3. Fulkerson 15 mm AJSM 1990<br />

ii. Medialization<br />

1. Do NOT over-medialize—Andrish 2005<br />

2. Goal to normalize in range of TT-TG 10-15 mm<br />

iii. FEA Patient-Specific Models for Tuberosity Transfer: AMZ<br />

20% mean decrease in stress<br />

c. Planning Tuberosity Transfer<br />

i. Preop Planning: Knee Specific<br />

ii. Steepest slope approx. 60 degrees<br />

iii. Applying Trigonometry to a constant elevation of 15mm:<br />

1. 60 degree slope & elevation 15 = 8.7 mm medialization<br />

2. 50 degree slope & elevation 15 = 12.5 mm medialization<br />

3. 45 degree slope & elevation 15 = 15 mm medialization<br />

4. Example of Typical Excessive TT-TG of 25 treated with<br />

a. 60 degree slope or 8.7 = 16.3 post op TT-TG<br />

b. 50 degree slope or 12.5 = 12.5 post op TT-TG<br />

d. AMZ-Indications<br />

i. TT-TG - abnormally elevated and distal lateral chondrosis of the patella<br />

without trochlear involvement<br />

ii. Very steep AMZ in severe lateral PF compartment overload (excessive<br />

lateral compression/ tilt)<br />

iii. Straight anterization when TT-TG is within normal limits and the goal is<br />

to decrease PF forces (Fulkerson osteotomy modified for straight<br />

anterization).<br />

e. AMZ-Contraindications<br />

i. Proximal pole, medial, panpatellar chondrosis or concomitant chondrosis<br />

of the trochlea<br />

ii. TT-TG within normal limits (Straight anterization is an option in this<br />

setting)<br />

iii. Pain is not directly related to a biomechanical abnormality that will be<br />

reversed by<br />

f. TTO-Indications<br />

i. Highly controversial


ii. Lyon France school of thought: Patellar alta and trochlear dysplasia are<br />

the main problems to be addressed for patellar instability<br />

1. Distalization of the tuberosity only<br />

iii. US surgeons are divided: patellar instability is treated with good reported<br />

results by:<br />

1. TTO alone (medialization +/- distalization)<br />

2. MPFL repair/ reconstruction alone<br />

3. MPFL surgery concomitant with TTO<br />

iv. TTO biomechanically indicated<br />

1. Normalization of the lateral resultant vector would appear to aid in<br />

the PF management when there is minimal chondrosis (grade 2 or<br />


AMZ<br />

Author Procedure Journal Subjects Follow up Results<br />

Fulkerson<br />

JP (11)<br />

Pidoriano<br />

AJ (5)<br />

Buuck D<br />

(12)<br />

Bellemans<br />

J (13)<br />

Naranja<br />

RJ (14)<br />

AMZ AJSM 1990 30 pts12<br />

pts<br />

AMZ AJSM<br />

1997<br />

AMZ Op Tech Spt<br />

Med<br />

> 2 y F/U>5 y<br />

F/U<br />

89-93% G/E<br />

Advance DJD<br />

subgroup: No E;<br />

75% G<br />

23 pts 10 F/U 87% G/E<br />

distal/lateral<br />

chondrosis<br />

55% G/E medial<br />

chondrosis<br />

poor results with<br />

trochlear<br />

42 knees<br />

in 36 pts<br />

AMZ AJSM 1997 29<br />

patients<br />

Elmslie-Trillat<br />

Maquet<br />

AJSM 1996 55 knees<br />

in 51<br />

patients<br />

chondrosis<br />

8.2 mean F/U 86% G/E<br />

32 mon mean<br />

(25- 44 mon)<br />

74.2 mon (13<br />

– 196 mon)<br />

28 successful<br />

73-84% G/E


PFA<br />

Authors Procedure Journal Subjects Follow up Results<br />

Sisto DJ,<br />

Sarin VK<br />

(15)<br />

Ackroyd<br />

CE, et.al.<br />

(16)<br />

Merchant,<br />

AC (17)<br />

Lonner,<br />

JH (18)<br />

Kooijman<br />

HJ, et.al.<br />

(19)<br />

Custom PFA<br />

(Kinamatch,<br />

Kinamed)<br />

Avon<br />

(Stryker)<br />

LCS based<br />

(DePuy)<br />

Lubinus<br />

(Waldemar<br />

Link) and<br />

Avon<br />

(Stryker)<br />

Richards<br />

(Smith<br />

Nephew)<br />

JBJS<br />

2007<br />

JBJS<br />

2007<br />

CORR<br />

2005<br />

CORR<br />

2004<br />

JBJS<br />

2003<br />

25 PFA<br />

in 22<br />

pts.<br />

109<br />

PFA in<br />

85 pts.<br />

16 PFA<br />

in 16<br />

pts<br />

30 PFA<br />

Lubinus 25<br />

PFA<br />

Avon<br />

56 PFA<br />

in 51<br />

pts.<br />

73m<br />

mean<br />

range; 32 to<br />

119 months<br />

Min 5<br />

yr F/U<br />

F/U<br />

2.75-<br />

6.25<br />

years<br />

4 yr<br />

mean<br />

(2-6) 6<br />

mon<br />

mean<br />

(1-12<br />

mon)<br />

17 y<br />

mean<br />

range<br />

15-21<br />

25/25 G/E<br />

80% success<br />

95.8%<br />

survival<br />

94% G/E<br />

84% G/E<br />

96% G/E<br />

86% G/E<br />

2% loosening


Meniscal Allograft Transplantation<br />

12 th Annual Articular Cartilage Course<br />

Wayne K. Gersoff, M.D.<br />

1. Goal of MAT is to preserve articular cartilage, inhibit<br />

degeneration, and prolong function.<br />

2. Historical Perspective- first MAT done in 1984, since<br />

then thousands have been performed.<br />

3. Basic Science Considerations<br />

Chondroprotective Potential<br />

Healing Potential<br />

Immunoligoc Factors<br />

Histological Analysis<br />

Preservation Techniques<br />

Biomechanical Considerations<br />

4.Tissue Preservation<br />

Fresh- logistically not practical<br />

Freeze Dried – cell death, structural damage<br />

Cryopreservation – cost, allow for slight increase in cell<br />

viability.<br />

Fresh Frozen – slight increase in cell death, minimal<br />

structural damage.<br />

Role of cell viability – limited, meniscus acts as a<br />

biological scaffold.


5. Tissue Preservation<br />

Gamma radiation negative effects- Naal, J. Biomed<br />

Mater. 2008.<br />

Sterilization doesn’t compromise tissue integrity –<br />

McNickle, COOR.2008.<br />

Significant deep freezing can alter meniscus collagen<br />

Gelber. Knee Surg Sports Trauma. 2008.<br />

Proprietary processing techniques<br />

6. Indications : subtotal or total menisectomy; pain secondary to<br />

meniscal deficiency; grade 3 or less chondromalacia; stable<br />

knee without malalignment.<br />

7. Contraindications : RA, Metabolic degenerative disease,<br />

obesity<br />

post-infectious disease, remodeling of femoral condyle.<br />

8. Techniques<br />

Double bone plug<br />

Bone bridge – slot and trough<br />

9. Double Bone Plug<br />

commonly used on medialside<br />

importance of anatomical placement of both horns<br />

maintain configuration<br />

avoids tibial spine debridement<br />

fragility of bone plugs and fixation<br />

10. Bone Bridge<br />

utilized medially or laterally<br />

allow for maintenance of anatomy and architecture


provides excellent bone fixation and healing<br />

Trough – press fit fixation<br />

Slot – interference screw fixation<br />

11. Sizing Considerations<br />

AP and lateral radiograph with radiologic marker – Pollard<br />

Width – tibial eminence to periphery of tibial compartment on<br />

AP radiograph<br />

Accuracy of predictability – McDermott, 2004<br />

MRI of contralateral knee – Prodromos. Arthroscopy. 2007<br />

12.Complications<br />

Arthrofibrosis<br />

Meniscal Detachment<br />

Meniscal Shrinkage<br />

Meniscal Failure<br />

Meniscal Extrusion<br />

13. Pitfalls and Pearls<br />

Appropriate surgical planning<br />

Don’t compromise exposure<br />

Adequate fixation techniques<br />

Presence of meniscal remnant<br />

Accurate positioning of bone<br />

Anterior horn fixation<br />

Placement of pull through suture<br />

14. Rehabilitation<br />

Early protected WB, Full WB at 4-6 weeks<br />

Limited early motion<br />

Similar to delayed meniscal repair


Use of Unloader brace<br />

Early treatment of concerns<br />

15.Concomitant Procedures<br />

Ligamentous Reconstruction<br />

Malalignment Correction<br />

Articular Cartilage Restoration<br />

16. Asymptomatic Post-Menisectomy Knee<br />

Education about signs and symptoms of knee degeneration<br />

45 degree PA radiographs<br />

? yearly bone scans<br />

Articular cartilage tends to rapidly break down, especially in<br />

lateral compartment.<br />

Younger patient- strongly consider early intervention<br />

17. Future Considerations<br />

Tissue Scaffolding<br />

Tissue Engineering<br />

Synthetic Replacements<br />

Allograft – immunologic and technical considerations<br />

Long Term Functional Results<br />

18. Treatment of Meniscal Deficiency and Chondral Defects<br />

19. Symbiotic Realtionship of Meniscus and Articular<br />

Cartilage<br />

Concept of Functional Unit


20.Common Factors in Treatment<br />

Knee Stability<br />

Knee Alignment<br />

Knee Environment<br />

21. MAT and ACI<br />

arthroscopic debridement<br />

arthrotomy and preparation of tibial bone anchor site<br />

meniscal allograft and chondral defect preparation<br />

placement and fixation of meniscal allograft<br />

completion of ACI procedure<br />

22. Applications for 2012<br />

Complex injuries to meniscus will continue and be problematic<br />

Articular cartilage injuries present challenge for athletic<br />

population especially competitive athletes.<br />

Primary goal is not return to competitive sports but restoration<br />

of functional knee.<br />

Challenging population is the adolescent and young adult.


BASIC SCIENCE<br />

NEXT GENERATION EXPERIENCE BMAC & OTHER SCAFFOLDS:<br />

EUROPE AND BEYOND.<br />

Alberto Gobbi,MD, Anup Kumar,MD, GeorgiosKarnatzikos, MD.<br />

Relevant Anatomy<br />

Articular cartilage is a thin layer of specialized connective tissue lining the articulations of<br />

diarthrodial joints. Properties, which are unique to this tissue, enable an almost frictionless joint movement<br />

and afford protection to the underlying bone from excessive load and trauma by dissipating the forces<br />

produced during movement.<br />

The structural organization of articular cartilage can be divided into four major zones: superficial,<br />

middle, deep and calcified [45, 69]. Each zone is distinctly structured with cells and extracellular matrix<br />

(ECM) organized in specific patterns. The cells, known as chondrocytes, make up 1-2% of the total weight<br />

of the articular cartilage. On the other hand, the ECM, which makes up the rest of the cartilage, is generally<br />

composed of type II collagen and proteoglycans.<br />

Biomechanics<br />

Within joints and in particular the knee, there are two types of cartilage: fibrocartilage and hyaline<br />

cartilage. Fibrocartilage is an elastic or “fibrous” cartilage of which the menisci are composed whereas<br />

hyaline cartilage is the tissue that covers the extremities of the bones that make up the joint. Hyaline cartilage<br />

has an extremely important biomechanical function as shock absorber as well as providing frictionless<br />

movement to the joint. Even though this layer of cartilage is only a few millimetres thick it has a significant<br />

capacity to absorb forces as well as distributing load to reduce stress on subchondral bone.<br />

Once this protective layer of articular cartilage is compromised, subsequent trauma and excessive<br />

loading can accelerate the progression of the “wear and tear”. Moreover, significant functional properties are<br />

lost leading to further pathological changes that can involve the surrounding cartilage and subchondral bone<br />

[66, 69].<br />

1


Biology<br />

As early as 1743, Hunter recognized that “articular cartilage lesions don’t heal”; the limited intrinsic<br />

healing potential of articular cartilage is attributed to the presence of few and specialized cells with a low<br />

mitotic activity [38]. As the human body matures, the cell density, which influences the amount of<br />

extracellular matrix produced, declines further, limiting its capacity to regenerate. The reparative capacity of<br />

cartilage is further reduced by the fact that the cartilage is avascular. The absence of a vascular network<br />

prevents access for mesenchymal stem cells and macrophages, which would normally help repairing tissues.<br />

Stem cells are responsible for the formation of new chondrocytes while macrophages remove debris<br />

associated with damaged cartilage. As a result, once injury occurs, surgical intervention may be necessary to<br />

achieve repair of the resulting focal chondral defects to obtain a good functional outcome.<br />

CLINICAL EVALUATION<br />

History<br />

Patients suffering from chondral injuries often find it difficult to recall a specific incident that<br />

triggered their symptoms. Swelling is usually present in the affected knee, which can sometimes be<br />

accompanied by mechanical symptoms like catching and clicking. A high index of suspicion for chondral<br />

injuries should be considered in patients who present with episodes of recurrent swelling in a knee that has<br />

effusion at the time of examination [52]. In addition, Mandelbaum emphasized that chondropenia (a process<br />

of loss of cartilage volume and elevation of contact pressures over time resulting in downward progression<br />

on the dose response curve and the eventual OA formation) as a possible causative factor, should not be<br />

overlooked when evaluating patients who present with these particular knee symptoms [46].<br />

Physical Findings<br />

Several authors have reported specific symptoms synonymous to chondral defects. Brittberg et al<br />

and Ochi et al [10, 11, 61] stressed that knee pain, symptoms of locking, retropatellar crepitus and swelling<br />

are among the prominent findings to look out for. Other authors including Hangody et al [34, 35] mentioned<br />

that instability could also be present. As signs and symptoms elicited during physical examination can mimic<br />

the presentation of other knee pathologies, authors agree that correlation with other diagnostic modalities<br />

should be made routinely to increase the accuracy of diagnosis.<br />

2


Imaging<br />

Among the diagnostic imaging modalities used, MRI has proven to be the most accurate having a<br />

sensitivity which is > 95% [14, 36, 37]. Aside from delineating the extent of the articular cartilage lesions,<br />

subchondral bone and associated ligament or meniscal injuries can also be assessed. The use of fast-spin-<br />

echo (with or without fat suppression) and/or fat-suppressed (or water-selective excitation) spoiled gradient-<br />

echo image for better resolution has been recommended. Signal properties of articular cartilage are<br />

dependent on: MR pulse sequence utilized, cellular composition of collagen, proteoglycans and water,<br />

orientation of collagen in different laminae of cartilage, and effective cartilage pulse sequencing (Potter et al)<br />

[70].<br />

Recently, surgeons used arthroscopy more extensively as a diagnostic tool: the benefit afforded by<br />

direct visualization of the extent of the defect, together with the information provided by MRI significantly<br />

enhances the surgeon’s capacity to precisely plan the appropriate treatment necessary to address the<br />

pathology.<br />

Decision Making, Algorithms and Classification<br />

The Outerbridge classification was the traditional system used but recently a more comprehensive<br />

system (ICRS Classification) has been adopted [10].<br />

Several options exist in the treatment of cartilage lesions; however, integrating all of these into a<br />

comprehensive algorithm has not been easy. Any form of planned treatment should be based on patient<br />

characteristics and expectations, clinical symptoms and type of lesions.<br />

One algorithm proposed by Cole and Miller [17, 51] presents an overview of a surgical-decision<br />

scheme where multiple options are presented for similar lesions without endorsing a specific treatment over<br />

the others.<br />

In general, surgeons agree that parameters such as lesion size, depth and associated issues,<br />

like alignment, ligament and meniscal integrity should be considered when planning treatment for<br />

chondral defects. Furthermore, other factors related to the patient (e.g. age, genetic predisposition,<br />

level of activity, associated pathologies and expectations) should not be overlooked.<br />

3


TREATMENT<br />

Operative<br />

Traditional palliative techniques or newer reparative treatment options have demonstrated variable<br />

results. Lavage and chondroplasty can provide symptomatic pain relief with no actual hyaline tissue<br />

formation. However, these techniques remove superficial cartilage layers, which include collagen fibers that<br />

are responsible for the tensile strength, creating a less functional cartilage tissue [49]. Bone marrow<br />

stimulation techniques, such as subchondral plate drilling or microfracture have been reported to stimulate<br />

production of hyaline-like tissue with variable properties and durability compared to normal cartilage, but it<br />

has been observed in many cases that these techniques tend to produce fibrocartilaginous tissue, which<br />

degenerates with time [29, 31, 53, 73]. Osteochondral autologous transplantation and mosaicplasty can<br />

restore normal cartilage tissue, but they can be applied only to small defects and there are some concerns<br />

regarding donor site morbidity [29, 34]. Autologous chondrocyte implantation, which was first introduced<br />

by Peterson, on the other hand, has been proven to be capable of restoring normal hyaline-like cartilage<br />

tissue, that is mechanically and functionally stable even in athletes at long-term follow up, but this method<br />

showed local morbidity for periosteal harvest and requires two surgical procedures [6, 36, 37, 54, 68].<br />

The apparent complexity of the Peterson periosteal technique and the possible complication of<br />

periosteal patch hypertrophy prompted surgeons to look for alternative techniques to enhance cell delivery<br />

and outcome. At present the most promising technique seems to be tissue engineering, where cells are<br />

combined with scaffolds to pre-form a given tissue. In general, the concept involves cultured autogenous or<br />

allogenous chondrocytes integrated in biodegradable and biocompatible scaffolds. Once cultivated on the<br />

scaffold, the chondrocytes must reacquire and maintain their chondrogenic phenotype in order to synthesize<br />

an extracellular matrix containing type II collagen, glycosaminoglycans and proteoglycans, all of which are<br />

necessary to produce hyaline cartilage.<br />

Scaffolds<br />

The use of a three-dimensional scaffold for autologous chondrocyte culture was developed with the<br />

aim to improve both the biological performance of chondrogenic autologous cells as well as renders the<br />

surgical technique easier, avoiding the use of the periosteal flap [30, 47, 48, 58]. A scaffold that is properly<br />

sized can be positioned directly into the articular defect under arthroscopic guidance; although, some<br />

4


technical limitations prevail, which include treating patellar lesions and posterior portions of femoral<br />

condyles or tibial plateau and could partly be resolved with the development of new arthroscopic tools.<br />

For some years now, different types of scaffolds with different matrices have been tested in animal<br />

models, and in some cases, human trials were carried out to determine the efficacy in facilitating and<br />

promoting cartilage repair. These scaffolds can be divided according to their chemical nature into protein<br />

based polymers (collagen, fibrin and gelatine), carbohydrate polymers (hyaluronan, agarose, polylactic acid,<br />

polyglycolic acid, chitosan and alginate) and other polymers (Teflon, Dacron, carbon fibers, polybutyric<br />

acid, hydroxyapatite). Combinations of these different polymers are also available.<br />

The ideal scaffold should be biocompatible (by not triggering any inflammatory response and not<br />

being cytotoxic), biodegradable by offering a temporary support to cells in order to promote replacement of a<br />

newly synthesized matrix and possibly induce proliferation of the transplanted cells. The matrix should also<br />

be permeable to nutrients and provide firm adhesion to the surrounding cartilage wound edges so, as to<br />

promote integration. Furthermore the scaffold must be reproducible and readily available and versatile for<br />

repair and resurfacing [39].<br />

Hyaluronan (hyaluronic acid, HA) is a naturally occurring and highly conserved<br />

glycosaminoglycans, which is widely distributed in the body. It has proven to be an ideal molecule for tissue<br />

engineering strategies in cartilage repair, given its impressive multi-functional activity through its structural<br />

and biological role [16].<br />

Through the chemical modification of HA, a scaffold can be obtained and may be processed into<br />

stable configurations to produce a variety of biodegradable structures with different physical forms and in-<br />

vivo residence times. Extensive biocompatibility studies have demonstrated the safety of these biomaterials<br />

and their ability to be resorbed in the absence of an inflammatory response [15].<br />

Three-dimensional non-woven scaffolds support the in vitro growth of highly viable chondrocytes<br />

and promote the expression of the original chondrogenic phenotype [12, 13].<br />

Chondrocytes, previously expanded on plastic and seeded into the scaffold produce a characteristic<br />

extracellular matrix rich in proteoglycans and express typical markers of hyaline cartilage, such as collagen<br />

II and aggrecan [1, 32, 55]. When implanted in full-thickness defects of the femoral condyle in rabbits,<br />

chondrocytes regenerated a cartilage-like tissue [32, 33, 75].<br />

5


An important issue to be addressed for the future perspective for cartilage repair is the quality of the<br />

bonding and the integration of the newly formed tissue to the native tissue. Different studies were completed<br />

and all seem to demonstrate the importance to the presence of both cells and the newly synthesized matrix<br />

for achieving a stable healing [65].<br />

Indications<br />

The main indications for cartilage transplantation are symptomatic focal, full thickness cartilage<br />

lesion (ICRS Grade III – IV) in the absence of significant arthritis in physiologically young patients (15 - 55<br />

years). Additional factors to consider include the patient’s motivation and willingness to comply with the<br />

post-implantation rehabilitation regimen. Defect size ranging from 2-12 cm 2 has been shown to be<br />

favourable to regeneration. Osteochondritis Dissecans is not a contraindication for cartilage transplantation<br />

as long as the bone loss does not exceed 8 mm [67].<br />

Second Generation Autologous Chondrocyte Implantation Technique<br />

Autologous Chondrocyte Implantation is carried out through the conventional arthrotomy approach;<br />

however, recent advances in scaffold technology have enabled surgeons to perform this technique in<br />

condylar lesions arthroscopically [2, 3, 26, 27, 42, 43, 48, 58, 74, 77].<br />

The chondrocytes obtained from the biopsy taken during initial arthroscopy are then expanded in<br />

vitro and finally seeded onto a 3-dimensional biomaterial where they will continue to proliferate and re-<br />

differentiate. After proliferation, chondrocytes organize three-dimensionally to stimulate the synthesis of<br />

extracellular matrix molecules and to prevent the loss of cell phenotype. The time frame observed for this<br />

cultivation with respect to ACI is three to four weeks. At the time of graft implantation, the lesion is prepared<br />

using a low-profile cannulated drill maintained in place by a Kirschner guide wire (0.9 mm diameter)<br />

anchored in the bone. A depth of 2 mm is observed while preparing the defect to avoid violating the<br />

subchondral bone plate. Once the defect is completely devoid of fibrotic tissues, the hyaluronic acid patch<br />

containing the cultured chondrocytes is obtained. At this point, the knee joint is then drained of fluid and the<br />

scaffold is loaded in the delivery system instrument using the cannula to position the patch into the defect.<br />

The intrinsic adhesive characteristics of the scaffold assure its stability once positioned in the defect and in<br />

the majority of cases there is no need for further fixation. Graft stability is then verified through a range of<br />

6


knee movement prior to closure of the portals. Otherwise, in large uncontained defects and in the<br />

patellofemoral compartment treated with open surgery, fibrin glue and/or other fixation systems maybe<br />

indicated to keep the graft in place [28].<br />

Several reports from controlled trials in patients operated with the use of these HA scaffolds have<br />

been presented [48, 58]. However, the largest collection of data (more than 4.000 cases in Europe) using the<br />

HA scaffold in clinical practice is represented by a multicenter observational study conducted in Italian<br />

Orthopaedic Centers since 2001 [48, 63, 64].<br />

EVALUATION OF OUTCOME<br />

Since 2001, we have participated in an ongoing observational multicenter investigation to evaluate<br />

the long- term clinical outcomes of the treatment with HA scaffold (HYAFF 11 ® Fidia Advanced<br />

Biopolymers, Abano Terme, Italy).<br />

141 patients with follow up assessments ranging from 2 to 5 years (average follow up time: 38<br />

months) were evaluated. At follow up 91.5% of patients improved according to the international knee<br />

documentation committee subjective evaluation; 76% and 88% of the patients had no pain and mobility<br />

problems respectively assessed by the Euro Qol-EQ5D measure. Furthermore, 95.7% of the patients had<br />

their treated knee normal or nearly normal as assessed by the surgeon; cartilage repair was graded<br />

arthroscopically as normal or nearly normal in 96.4% of the scored knees; the majority of the second – look<br />

biopsies of the grafted site, histologically, were assessed as hyaline-like. A very limited complication rate<br />

was recorded in this study [47].<br />

We also evaluated the patellofemoral full- thickness chondral defects treated with Hyalograft C.<br />

Thirty-two chondral lesions located in the patella and trochlea, were treated. The International Cartilage<br />

Repair Society-International Knee Documentation Committee and EuroQol EQ-5D scores demonstrated a<br />

statistically significant improvement (P < .0001). Objective preoperative data improved from 6/32 (18.8%)<br />

with International Knee Documentation Committee A or B to 29/32 (90.7%) at 24 months after<br />

transplantation. Mean subjective scores improved from 43.2 points preoperatively to 73.6 points 24 months<br />

after implantation. Magnetic resonance imaging studies at 24 months revealed 71% to have an almost normal<br />

cartilage with positive correlation with clinical outcomes. Second-look arthroscopies in 6 cases revealed the<br />

repaired surface to be nearly normal with biopsy samples characterized as hyaline-like in appearance [30].<br />

7


COMPLICATIONS AND SPECIAL CONSIDERATIONS<br />

The most commonly cited complications of first generation autologous chondrocyte implantation are<br />

graft hypertrophy and arthrofibrosis [8, 42, 46, 54, 66]. Graft hypertrophy has been noted to occur between 3<br />

to 9 months post-operatively and it is thought to result from abrasion of the periosteal patch, which<br />

overhangs from the margin of the defect.<br />

Adverse events of second generation ACI are apparently lower than first generation as reported by<br />

Mandelbaum et al [46] and today second generation ACI represents a modern and viable technique for<br />

cartilage full thickness chondral lesion repair but it is still not without problems: aside from the risk of<br />

harvest site morbidity and two surgical procedures, the total cost of the operation, scaffold and process of<br />

chondrocytes culture remain drawbacks of this procedures [6, 26].<br />

IMPROVEMENTS<br />

Very promising results have been shown with ACI to date, however, ACI is a technology that<br />

involves the implantation of an expanded chondrocyte population derived from a cartilage biopsy. These<br />

expansion results in the loss of its phenotypic traits also called “de-differentiation” [9, 44, 57, 79]. This<br />

produces chondrocytes with a decreased capacity to regenerate hyaline cartilage cells [73].<br />

Characterized Chondrocyte Implantation (CCI) is a second-generation ACI procedure that uses<br />

ChondroCelect. ChondroCelect (Ti-Genix NV, Haasrode, Belgium) was developed to limit this loss of<br />

phenotype and is an expanded population of chondrocyte that expresses a marker profile predictive of the<br />

capacity to form stable hyaline like cartilage in vivo in a consistent and reproducible manner.<br />

The chondrogenic character of the cells is confirmed with the ChondroCelect Score (CC Score)<br />

representing a potency assay done after every step of the process. In a mouse model, the CC-score and<br />

histological score showed a high degree of correlation. The CC-score is a range from -6 to +6 (4 positive<br />

and 2 negative genetic markers). Only chondrocytes with a CC-score of > 0 would be used ensuring a high<br />

cartilage-forming capacity.<br />

In a prospective, randomized controlled trial, that compared CCI versus Microfracture as treatment<br />

for single symptomatic cartilage defects of the femoral condyle, CCI produced a superior type of tissue<br />

regenerate. The primary aims of the trial were (a) to demonstrate superiority of CCI over microfracture in<br />

overall quality of structural regeneration of the articular tissue at 12 months post treatment using<br />

8


histomorphometry and Overall Histology Assessment Score; and (b) to demonstrate a clinical outcome as<br />

assessed by Overall Knee Injury and Osteoarthritis Outcome Score (KOOS) at 12-18 months post treatment<br />

that was at least comparable in both treatment groups. For the first time this trial proved that joint surface<br />

repair/regeneration using cell technology produced higher quality regenerate than intrinsic repair. It also<br />

showed clinical outcome similar in both treatments. These results suggest CCI may lead to an improved<br />

long-term clinical outcome [73].<br />

FUTURE DIRECTIONS<br />

In the future the use of autologous mesenchymal stem cells seeded in a temporary scaffold with<br />

growth factors could be an improvement of the currently available techniques.<br />

Mesenchymal Stem Cell<br />

ACI has the potential for repair of the damaged cartilage and, as of today, remains one of the more<br />

promising technologies of tissue engineering. Nevertheless, the chondrocyte being a quiescent cell, the<br />

expansion of the cell population to obtain a sufficient cell number for the surgical procedure appears critical<br />

and in this regard the approach has some limitations: (a) if the damaged surface is particularly wide, the<br />

availability of healthy cartilage to harvest appears limited; (b) in vitro expanded human articular<br />

chondrocytes reduce their chondrogenic potential after 5-6 cell doubling.<br />

Research is currently underway to develop alternatives to this protocol. Recent directions in cartilage<br />

repair are moving towards the possibility to perform one-step surgery; several groups are analysing the<br />

possibility to use mesenchymal stem cells (MSC) with chondrogenic potential and growth factors [19, 20,<br />

21, 22, 26, 44, 60].<br />

MSC have a self-renewal capacity and multi-lineage differentiation potential, which remain of great<br />

interest for scientists involved in cell therapy and tissue engineering.<br />

MSC can be characterized by their cultivation behaviour and their differentiation potential into<br />

adipogenic, osteogenic and chondrogenic cells, as well as form bone, cartilage and fat [32, 37, 40, 55, 79].<br />

Researches are currently exploring the possibility of manipulating stem cells in the laboratory to differentiate<br />

into chondrocytes and which can then be integrated into a synthetic scaffold for later implantation. If MSC<br />

are cultured in the appropriate microenvironment, they can differentiate to chondrocytes and form cartilage.<br />

9


Onset of chondrogenesis requires a chemically defined serum free medium supplemented with<br />

dexamethasone, ascorbic acid and growth factors such as TGF-B [72]. The micromass culture or pellet<br />

culture system is generally considered a good in vitro model of chondrogenesis [44]. Johnstone et al cultured<br />

MSC as pellets at the bottom of a tube for 2 weeks in a specific serum free cocktail medium. Under these<br />

conditions cells organize a cartilaginous matrix by secreting proteoglycans and type II collagen and cells<br />

appear as real chondrocytes embedded in their own matrix lacunae [40].<br />

Ochi et al found that injection of cultured MSCs combined with a bone marrow stimulation<br />

procedure in the chondral defect of rat model can accelerate the regeneration of articular cartilage. They<br />

believed that this cell therapy was a less invasive treatment for the patients with cartilage injury [62].<br />

Wakitani et al have used autologous culture expanded BMSC transplantation for repair of cartilage<br />

defects in osteoarthritic knees. They chose 24 knees of 24 patients with knee O.A. who underwent a high a<br />

high tibial osteotomy. Patients were divided into cell transplanted group and cell free group. After 16 months<br />

follow-up, they concluded that M.S.C. was capable of regenerating a repair tissue for large chondral defects<br />

[78]. Giannini et al [25] presented their one-step surgery procedure using MSC and scaffold.<br />

In order to find a more effective and simple technique for cartilage repair, we have attempted to use<br />

high concentration MSC combined with biologic scaffolds for osteochondral defect repair (Figures 7-06_1-<br />

6). For this operative procedure, we firstly prepared the osteochondral defect with debridement by<br />

arthroscopy or mini-open arthrotomy, and extracted bone marrow from iliac crest. High concentration MSC<br />

was obtained by centrifuge (four to six times the baseline value). Then, we activate the bone marrow<br />

concentrate and produce a sticky clot material. <strong>Final</strong>ly, we paste BMC into the osteo-chondral defect and<br />

cover it with a collagenic membrane. Due to the porous structure of the scaffold, we hypothesize it is easy<br />

for adhesion, proliferation and differentiation of MSC. Preliminary data from our institution and other Italian<br />

authors on MSC implantation with a one-step procedure seem to be promising, showing good clinical<br />

outcomes at early follow up [32].<br />

Plasma Rich in Growth Factors<br />

The plasma fraction just above the red and the white cells is called plasma rich in growth factors<br />

(PRGF). It is a plasma fraction with a high number of platelets; about three times the number of platelets in<br />

blood [71]. In these platelets there is a high density of alpha granules, which contain proteins. PRGF is an<br />

10


autologous preparation rich in growth factors, obtained from platelet-enriched plasma. Studies have been<br />

carried out using PRGF in articular cartilage lesion treatment, and presented the following positive results:<br />

PRGF can increase total G.A.G., collagen II synthesis and decrease degradation; induce chondrogenesis of<br />

MSCs and promote chondrocyte proliferation, differentiation and adhesion [18].<br />

PRGF contains different growth factors, such as PDGF, IGF-1, TGF-B, EGF, bFGF, VEGF, and<br />

others. They regulate key processes involved in tissue repair, including cell proliferation, chemotaxis,<br />

migration, cellular differentiation, and extracellular matrix synthesis [7]. Some growth factors in PRGF show<br />

good activity for chondrogenesis. Barry F et al demonstrated MSC cultured with TGF-β produced<br />

significantly more proteoglycans and collagen II [5]. Fukumoto T et al found that IGF-1 has a synergistic<br />

effect with TGF- β in promoting MSC chondrogenesis [23]. The studies by Stevens MM et al have shown<br />

that bFGF induce M.S.C. proliferation and chondrogenesis [76].<br />

Cugat et al have used PRGF treating chondral defects and have obtained good results. According to<br />

their experiences for other connective tissue repair, they think that PRGF in physiological concentrations is<br />

effective for the recovery of connective tissue. Local treatment is safe and does not alter the systemic<br />

concentrations of these proteins [18].<br />

Kon et al [41] have studied a group of 30 patients with symptomatic degenerative disease of the knee<br />

joints treated with three PRP intra-articular injections weekly; the follow up at 6 months showed positive<br />

effects on the function and symptoms.<br />

We have also attempted to use PRGF combined with a scaffold for osteochondral defects. We<br />

hypothesized that penetration of subchondral plate would allow the release of MSC, which provide the cell<br />

source for cartilage repair. 23 patients with a mean age of 44.3 years, among a group of 50 patients, were<br />

followed-up. We collected pain visual analogue scale (VAS) and KOOS score at pre-treatment, 3 and 6<br />

months post-treatment, and the preliminary results are encouraging. There was a trend towards improvement<br />

in both scores. PRGF can induce MSC chondrogenesis, promote cell proliferation and increase deposition of<br />

ECM.<br />

Recently authors have proven synergistic effects of PRP combined with MSC. Nishimoto et al [59]<br />

suggested that simultaneous concentration of PRP and bone marrow cells (BMC), could play important role<br />

in future regenerative medicine. Milano et al [50] in an in vivo study showed a more effective cartilage repair<br />

11


after microfracture associated to hydro gel scaffold with PRP. <strong>Final</strong>ly, several authors [1, 25, 26, 57, 61, 62,<br />

78, 79] have stated that growth factors could act like as a carrier to fix chondrocytes into cartilage defects<br />

and can be combined with mesenchymal stem cells. Preliminary data are encouraging but further studies on<br />

clinical efficacy will clarify if simultaneous use of PRP and MSC could represents a real solution for<br />

regenerative medicine in cartilage repair.<br />

However, application of growth factor proteins is hampered by their short pharmacological half-life.<br />

This has let to the application of gene transfer to achieve a localized delivery into the defect of therapeutic<br />

gene constructs. Among the most studied candidates, polypeptide growth factors have shown promise to<br />

enhance the structural quality of the repair tissue. These studies show less morbidities and complications<br />

inherent to cartilage surgical techniques by lessening surgical procedures translating to lower cost for the<br />

patient. However, medium-term prospective randomized studies are suggested to confirm these short-term<br />

results.<br />

CONCLUSIONS<br />

A number of viable options have been made available over the years to address problems concerning<br />

cartilage damage and each technique has its advantages and disadvantages. Numerous studies are currently<br />

under way to clarify some of the questions that still remain unanswered regarding the long-term durability of<br />

these procedures and the possible modifications that can still be made to achieve better results.<br />

From an initial arthrotomy approach, some of the techniques can now be entirely performed<br />

arthroscopically. This modification has enabled surgeons to avoid possible intra-operative problems and<br />

decreased operative time. By eliminating the need for an open procedure and harvesting of periosteal flap,<br />

joint trauma is significantly reduced. Complications, such as graft hypertrophy and ossification are also<br />

avoided with the use of this three-dimensional hyaluronic acid scaffold.<br />

At present, not all lesions can be addressed with a single technique. Retropatellar, tibial plateau and<br />

posterior condylar lesions remain a challenge. Development of better instruments and introduction of new<br />

techniques in the near future should be able to solve this dilemma.<br />

Characterized Chondrocyte Implantation is a second-generation ACI procedure and improves<br />

chondrocyte selection to form stable and consistent hyaline cartilage.<br />

12


Biotechnology is progressing at a rapid pace, allowing the introduction of numerous products for<br />

clinical application. The research on the identification or creation of the ideal material for engineering<br />

cartilage substitutes has been very prolific in the last decade. The use of polymers, both natural and<br />

synthetic, that undergo controllable bulk erosion or resorption have been largely investigated as they could<br />

represent a favorable solution for engineering cartilage tissues in vitro or in vivo.<br />

An important issue to be addressed for the future perspective for cartilage repair is the quality of the<br />

bonding and the integration of the newly formed tissue to the native tissue, and several studies seem to<br />

demonstrate the importance of both cells and the new matrix to achieve a stable healing. This possibly would<br />

lead to the generation of tissue-engineered strategies for the repair of complex lesions involving cartilage,<br />

together with the subchondral bone and other structures. Several scaffolds have been tested in animal models<br />

for regenerating cartilage tissue. However, carefully conducted randomised prospective studies for each of<br />

these innovations should be carried out to validate their efficacy for cartilage regeneration.<br />

REFERENCES<br />

1. Aigner J, Tegeler J, Hutzier P et al: Cartilage Tissue Engineering with Novel Non-Woven Structured<br />

Biomaterial Based on Hyaluronic Acid Benzyl Ester. J Biomed Mater Res. 1998; 42: 172-181.<br />

2. Ando W, Tateishi K, Nakamura N et al: Cartilage repair using an in vitro generated scaffold-free<br />

tissue-engineered construct derived from porcine synovial mesenchymal stem cells. Biomaterials.<br />

2007 Dec; 28(36): 5462-70. Epub 2007 Sep 14.<br />

3. Ando W, Tateishi K, Nakamura N et al: In vitro generation of a scaffold-free tissue-engineered<br />

construct (TEC) derived from human synovial mesenchymal stem cells: biological and mechanical<br />

properties and further chondrogenic potential. Tissue Eng Part A. 2008 Dec; 14(12): 2041-9.<br />

4. Anitua E, Sanchez M et al: The potential impact of the preparation rich in growth factors<br />

(PRGF) in different medical fields. Biomaterials 2007; 28:4551-4560.<br />

5. Barry F, Boynton RE, Liu B et al: Chondrogenic differentiation of mesenchymal stem cells from<br />

bone marrow: differentiation-dependent gene expression of matrix components. Exp Cell Res. 2001<br />

Aug 15;268(2):189-200.<br />

6. Behrens P, Bitter T, Kurz B, et al: Matrix-associated autologous chondrocyte<br />

transplantation/implantation (MATC/MACI)-5-year follow –up. The knee. 2006; 13:194-202.<br />

7. Bennett NT, Schultz GS: Growth factors and wound healing: biochemical properties of growth<br />

factors and their receptors. Am J Surg. 1993 Jun;165(6):728-37.<br />

13


8. Bentley G, Biant LC, Carrington RW et al: A Prospective, Randomised Comparison of Autologous<br />

Chondrocyte Implantation Versus Mosaicplasty for Osteochondral Defects in the Knee. J Bone Joint<br />

Surg Br Mar. 2003; 85(2):223-230.<br />

9. Benya, PD, Shaffer JD: Dedifferentiated chondrocytes reexpress the differentiated collagen<br />

phenotype when cultured in agarose gels. Cell. 1982 Aug; 30(1): 215-24.<br />

10. Brittberg M, Winalski C: Evaluation of Cartilage Injuries and Repair. J Bone Joint Surg Am. 2003;<br />

85-A Suppl 3:58-69.<br />

11. Brittberg M, Peterson L, Sjogren-Jansson E et al: Articular Cartilage Engineering with Autologous<br />

Chondrocyte Transplantation. A Review of Recent Developments. J Bone Joint Surg Am. 2003; 85-A<br />

Suppl 3:109-115.<br />

12. Brun P, Abatangelo G, Radice M et al: Chondrocyte Aggregation and Reorganization into Three-<br />

Dimensional Scaffolds. J. Biomed Mater Res. 1999; 46: 337-346.<br />

13. Brun P, Dickinson SC, Abatangelo G et al: Characteristics of repair tissue in second-look and third-<br />

look biopsies from patients treated with engineered cartilage: relationship to symptomatology and<br />

time after implantation. Arthritis Res Ther 2008; 10(6): R132. Epub 2008 Nov 11.<br />

14. Burstein D and Gray M: New MRI Techniques for Imaging Cartilage. J Bone Joint Surg Am. 2003;<br />

85-A Suppl: 70-77.<br />

15. Campoccia D, Doherty P, Radice M et al: Semisynthetic Resorbable Materials from Hyaluronan<br />

Esterification. Biomaterials. 1998; 19: 2101-2127.<br />

16. Chen WJ, Abatangelo G: Functions of Hyaluronan in Wound Repair. Wound Repair Regen. 1999; 7:<br />

79-89.<br />

17. Cole B and Farr J: Putting It All Together. Oper Tech Orthop. 2001; 11(2): 151-154.<br />

18. Cugat R, Carrillo JM, Serra I et al: Articular cartilage defects reconstruction by plasma rich growth<br />

factors. Basic science, clinical repair and reconstruction of articular cartilage defects: current status<br />

and prospects. TIMEO 801-807.<br />

19. Fortier LA, Balkman CE, Sandell LJ et al: Insulin-like growth factor-I gene expression patterns<br />

during spontaneous repair of acute articular cartilage injury. J Orthop Res. 2001 Jul; 19(4): 720-8.<br />

20. Fortier LA, Nixon AJ, Williams J et al: Isolation and chondrocytic differentiation of equine bone<br />

marrow-derived mesenchymal stem cells. Am J Vet Res 1998 Sep; 59(9): 1182-7.<br />

21. Fortier LA, Mohammed HO, Lust G et al: Insulin-like growth factor-I enhances cell-based repair of<br />

articular cartilage. J Bone Joint Surg Br. 2002 Mar; 84(2): 276-88.<br />

22. Fortier L, Potter H, Rickey E et al: Concentrated Bone Marrow Aspirate Improves Full-Thickness<br />

Cartilage Repair Combined to Microfracture in an Equine Model of Extensive Cartilage Loss. Journal<br />

of Bone and Joint Surgery: in Press.<br />

23. Fukumoto T, Sperling JW, Sanyal A et al: Combined effects of insulin-like growth factor-1 and<br />

transforming growth factor-beta1 on periosteal mesenchymal cells during chondrogenesis in vitro.<br />

Osteoarthritis Cartilage. 2003 Jan;11(1):55-64.<br />

14


24. Giannini S, Buda R, Vannini F et al: Arthroscopic autologous chondrocyte implantation in<br />

osteochondral lesions of the talus: surgical technique and results. Am J Sports Med. 2008 May; 36(5):<br />

873-80. Epub 2008 Jan 28.<br />

25. Giannini S, Buda R, Vannini F et al: One-step Bone Marrow-derived Cell Transplantation in Talar<br />

Osteochondral Lesions. Clin Orthop Relat Res. 2009 May;(467): 3307-3320.<br />

26. Gobbi A: Use of autologous MSC and PRP in the treatment of cartilage lesions. Ortopedia e<br />

Reumatologia vol.120; n. 3-4. Nov 2009.<br />

27. Gobbi A, Bathan L: Minimal Invasive Second-Generation Autologous Chondrocyte Implantation. In:<br />

Cole b, Gomoll A (2009) Biologic Joint Reconstruction. SLACK Incorporated, NJ 08086 USA.<br />

28. Gobbi A, Kon E, Berruto M et al: Patellofemoral Full-Thickness Chondral Defects Treated With<br />

Hyalograft- C: A Clinical, Arthroscopic, and Histologic Review Am. J. Sports Med., Nov 2006; 34:<br />

1763 – 1773.<br />

29. Gobbi A, Francisco R, Allegra F et al: Arthroscopic Treatment of Osteochondral Lesions of the<br />

Talus: A Prospective Study of Three Different Techniques. Arthroscopy. 2006 Oct; 22(10): 1085-92.<br />

30. Gobbi A, Kon E, Filardo G et al: Patellofemoral Full-Thickness Chondral Defects Treated with<br />

Second Generation ACI: A Clinical Review at 5 years Follow-up. Am J Sports Med. 2009 Jun; 37(6):<br />

1083-92.<br />

31. Gobbi A, Nunag P, Malinowski K: Treatment of Full Thickness Chondral Lesions of the Knee with<br />

Microfracture in a Group of Athletes. Knee Surg, Sports Traumatol, Arthro 2005 Apr; 13(3): 213-21.<br />

32. Grigolo B, Lisignoli G, Piacentini A et al: Evidence for Redifferentiation of Human Chondrocytes<br />

Grown on a Hyaluronan-based Biomaterial (HYAFF ® 11): Molecular, Immunohistochemical and<br />

Ultrastructural Analysis. Biomaterials. 2002; 23: 1187-1195.<br />

33. Grigolo B, Lisignoli G, Desando G et al: Osteoarthritis treated with M.S.C. on hyaluronan-based<br />

scaffold in rabbit. Tissue Eng Part C Methods. 2009 Dec; 15(4): 647-58.<br />

34. Hangody L, Fules P: Autologous Osteochondral Mosaicplasty for the Treatment of Full-Thickness<br />

Defects of Weight-bearing Joints. J Bone Joint Surg Am. 2003; 85-A Suppl 3:25-32.<br />

35. Hangody L, Kish G, Karpati Z et al: Mosaicplasty for the Treatment of Articular Cartilage Defects:<br />

Application in Clinical Practice. Orthopaedics. 1998; 21:751–756.<br />

36. Henderson I, Francisco R, Oakes B et al: Autologous Chondrocyte Implantation for Treatment of<br />

Focal Chondral Defects of the Knee: A Clinical, Arthroscopic, MRI and Histologic Evaluation at 2<br />

Years. 2005 Jun; 12(3): 209-16. Epub 2004 Nov 14.<br />

37. Henderson IJ, Tuy B, Oakes B et al: Prospective Clinical Study of Autologous Chondrocyte<br />

Implantation and Correlation with MRI at Three and 12 months. J Bone Joint Surg Br. 2003 ;<br />

85:1060-1066.<br />

38. Hunter W: On the Structure and Diseases of Articulating Cartilage. Philos Trans Rsoc Lond B Biol<br />

Sci; 1743; 9:277.<br />

15


39. Hunziker E: Articular Cartilage Repair: Basic Science and Clinical Progress. A Review of the Current<br />

Status and Prospects. Osteoarthritis Cart. 2002 433-463.<br />

40. Johnstone B, Hering TM, Caplan AI et al: In vitro chondrogenesis of bone marrow-derived<br />

mesenchymal progenitor cells. Exp Cell Res. 1998 Jan 10;238(1):265-72.<br />

41. Kon E, Buda R, Filardo G et al: Platelet-rich plasma: intra-articular knee injections produced<br />

favorable results on degenerative cartilage lesions. Knee Surg Sports Traumatol Arthrosc. 2010 Apr;<br />

18(4): 472-9. Epub 2009 Oct 17.<br />

42. Kon E, Gobbi A, Filardo G et al: Arthroscopic Second-Generation A.C.I. Compared with<br />

Microfractures for chondral lesions of the knee. AM J Sports Med 2009; 37:33.<br />

43. Kon E, Filardo G, Delcogliano M, et al: Arthroscopic second-generation autologous chondrocyte<br />

implantation at 48 months follow up. Osteoarthritis and Cartilage. 2007; Vol 15, Supplement B,<br />

22.1:44-45.<br />

44. Mackay AM, Beck SC, Murphy JM et al: Chondrogenic differentiation of cultured human<br />

mesenchymal stem cells from marrow. Tissue Eng. 1998 Winter;4(4):415-28.<br />

45. Mainil-Varlet P, Aigner T, Brittberg M, et al: Histological Assessment of Cartilage Repair. A Report<br />

by the Histology Endpoint Committee of the ICRS. J Bone Joint Surg Am. 2003; 85-A Suppl 2:45-57.<br />

46. Mandelbaum B, Browne JE, Fu F et al: Treatment outcomes of autologous chondrocyte implantation<br />

for full-thickness articular cartilage defects of the trochlea. Am J Sports Med. 2007; 35(6): 915-921.<br />

47. Marcacci M, Berruto M, Gobbi A, Kon E et al: Articular cartilage engineering with Hyalograft C: 3-<br />

year clinical results. Clin Orthop Relat Res. 2005 Jun;(435):96-105.<br />

48. Marcacci M, Zaffagnini S, Kon E et al: Arthroscopic Autologous Chondrocyte Transplantation:<br />

Technical Note. Knee Surg Sports Traumatol Arthrosc 2002; 10:154-159.<br />

49. McKinley BJ, Cushner FD, Scott WN: Debridement Arthroscopy. 10-year Follow-Up. Clin Orthop.<br />

1999; 367:190-194.<br />

50. Milano G, Zarelli D et al: Does Platelet Rich Plasma Injection enhance Cartilage Healing After<br />

Microfractures? An Animal Study. Poster no. 536. 54th Annual Meeting of the Orthopaedic Research<br />

Society.<br />

51. Miller M, Cole B: Atlas of Chondral Injury Treatment. Oper Tech Orthop. 2001; 11(2): 145-150.<br />

52. Miller M, Howard R, Plancher K: Treatment of Chondral Injuries and Defects. In: Surgical Atlas of<br />

Sports Medicine. Pennsylvania: Saunders, 2003: 110-115.<br />

53. Minas T, Gomoll AH, Rosenberger R et al: Increased failure rate of autologous chondrocyte<br />

implantation after previous treatment with marrow stimulation techniques. Am J Sports Med. 2009<br />

May; 37(5): 902-8. Epub 2009 Mar 4.<br />

54. Minas T, Peterson L: Advanced Techniques in Autologous Chondrocyte Transplantation. Clin Sports<br />

Med. 1999 Jan; 18(1): 13-44.<br />

55. Muraglia A, Cancedda R, Quarto R.: Clonal mesenchymal progenitors from human bone marrow<br />

differentiate in vitro according to a hierarchical model. J Cell Sci. 2000 Apr;113 ( Pt 7):1161-6.<br />

16


56. Nakamura N, Miyama T, Engebretsen L et al: Cell-based therapy in articular cartilage lesions of the<br />

knee. Arthroscopy 2009 May; 25(5): 531-52.<br />

57. Nakamura Y, Sudo K, Kanno M et al: Mesenchymal progenitors able to differentiate into osteogenic,<br />

chondrogenic, and/or adipogenic cells in vitro are present in most primary fibroplast like cell<br />

populations. Stem Cells. 2007 Jul; 25(7): 1610-7. (EPUB 2007).<br />

58. Nehrer S, Domayer S, Dorotka R et al: Three-year clinical outcome after chondrocyte transplantation<br />

using a hyaluronan matrix for cartilage repair. European Journal of Radiology. 2006; 57:3-8.<br />

59. Nishimoto S, Oyama T, Matsuda K: Simultaneous concentration of platelets and marrow cells: a<br />

simple and useful technique to obtain source cells and growth factors for regenerative medicine.<br />

Wound Repair Regen. 2007 Jan-Feb; 15(1): 156-62.<br />

60. Nixon AJ, Wilke MM, Nydam DV: Enhanced early chondrogenesis in articular defects following<br />

arthroscopic mesenchymal stem cell implantation in an equine model. J Orthop Res. 2007. Jul; 25(7):<br />

913-25.<br />

61. Ochi M, Adachi N, Nobuto H et al: Articular Cartilage Repair Using Tissue Engineering Technique:<br />

Novel Approach with Minimally Invasive Procedure. Artif Organs. 2004; 28(1): 28-32.<br />

62. Ochi M, Adachi N, Nobuto H et al: Effects of CD44 antibody-or RGDS peptide-immobilized<br />

magnetic beads on cell proliferation and chondrogenesis of mesenchymal stem cells. J Biomed Mater<br />

Res A. 2006 Jun 15; 77(4): 773-84.<br />

63. Pavesio A, Abatangelo G, Borrione A et al: Hyaluronan-based Scaffolds (Hyalograft ® C) in the<br />

Treatment of Knee Cartilage Defects: Preliminary Clinical Findings. Novartis Foundation<br />

Symposium, 2003; 249: 203-217.<br />

64. Pavesio Β, Abatangelo G, Borrione A et al: Hyaluronan-based Scaffolds (Hyalograft ® C) in the<br />

Treatment of Knee Cartilage Defects: Clinical Results. Tissue Engineering in Musculoskeletal<br />

Clinical Practice (AAOS), 2004; 8: 73-83.<br />

65. Peretti GM, Zaporojan V, Spangenberg KM et al: Cell-based bonding of articular cartilage: an<br />

extended study. J Biomed Mater Res, 1; 64A (3): 517-24, 2003.<br />

66. Peterson L, Brittberg M, Kiviranta I et al: Autologous Chondrocyte Transplantation. Biomechanics<br />

and Long-term Durability. Am J Sports Med 2002; 30: 2-12.<br />

67. Peterson L, Minas T, Brittberg M et al: Treatment of Osteochondritis Dissecans of the Knee with<br />

Autologous Chondrocyte Transplantation: Results at Two to Ten Years. J Bone Joint Surg Am. 2003;<br />

85-A Suppl 3:17-24.<br />

68. Peterson L, Vasiliadis HS, Brittberg M et al: Autologous Chondrocyte Implantation: A Long-term<br />

Follow-up. Am J Sports Med. 2010 Feb 24. [Epub/ahead of print].<br />

69. Poole R: What Type of cartilage Repair Are We Attempting to Attain? J Bone Joint Surg Am 2003;<br />

85-A Suppl 2:40-44.<br />

70. Potter H, Foo L et al: MRI and Articular Cartilage. Evaluating Lesions and Postrepair Tissue. In:<br />

Potter H, Foo L et al. (2007) Cartilage Repair Strategies. Humana Press (Springer) Berlin Heidelberg.<br />

17


71. Robert BD: The clinical and laboratory utility of platelet volume parameters. Aus J Med Sci. 1994;<br />

14: 625- 41.<br />

72. Robey PG, Bianco P: The use of adult stem cells in rebuilding the human face. J Am Dent Assoc.<br />

2006Jul; 137(7): 961-72. Review.<br />

73. Saris DBF, Vanlauwe J, Victor J et al: CCI Results in Better Structural Repair When Treating<br />

Symptomatic Cartilage Defects of the Knee in a Randomised Clinical Trial versus Microfracture. Am<br />

J Sports Med. 2008 Feb; 36(2): 235-46.<br />

74. Sgaglione NA, Miniaci A, Gillogly SD et al: Update on Advanced Surgical Techniques in the<br />

Treatment of Traumatic Focal Articular Cartilage Lesions in the Knee. Arthroscopy. 2002; 18 (Suppl<br />

1): 9-32.<br />

75. Solchaga LA, Yoo JU, Lundberg M et al: Hyaluronan-based Polymers in the Treatment of<br />

Osteochondral Defects. J Orthop Res. 2000; 18: 773-780.<br />

76. Stevens MM, Marini RP, Martin I et al: FGF-2 enhances TGF-beta1-induced periosteal<br />

chondrogenesis. J Orthop Res. 2004 Sep;22(5):1114-9.<br />

77. Tateishi K, Ando W, Nakamura N et al: Comparison of human serum with fetal bovine serum for<br />

expansion and differentiation of human synovial MSC: potential feasibility for clinical applications.<br />

Cell Transplant. 2008; 17(5): 549-57.<br />

78. Wakitani S, Imoto K, Yamamoto T et al: Human autologous culture expanded bone marrow<br />

mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis<br />

Cartilage. 2002 Mar;10(3):199-206.<br />

79. Wakitani S, Yokoyama M, Miwa H et al: Influence of fetal calf serum on differentiation of<br />

mesenchymal stem cells to chondrocytes during expansion. J Biosci Bioeng 2008 Jul; 106(1): 46-50.<br />

18


1<br />

STEM CELL BIOLOGY: FACT,<br />

FICTION AND OPPORTUNITY<br />

January 27-28, 2012<br />

C. Thomas Vangsness, JR., M.D.<br />

Professor of Orthopaedic Surgery<br />

Chief of Sports Medicine<br />

Keck School of Medicine<br />

University of Southern California<br />

Los Angeles, California<br />

vangsness@usc.edu<br />

1. What is a Stem Cell?<br />

a. A stem cell is a cell that has the ability to divide (self replicate) for<br />

indefinite periods – often throughout the life of the organism. Under the<br />

right conditions, or given the right signals, stem cells can give rise<br />

(differentiate) to the many different cell types that make up the organisms<br />

(plasticity).<br />

b. Naturally-occurring living cells that haven’t yet differentiated into the<br />

specialized cells.<br />

c. Part of the body’s repair system, and can divide to replenish other cells.<br />

d. Each new cell has the potential to become a cell with a more specialized<br />

function such as a red blood cell, bone marrow cell, muscle cell, nerve<br />

cell, etc.<br />

e. Stem cells have the ability to divide asymmetrically-one portion of the cell<br />

division becomes a differentiated cell while the other becomes another<br />

stem cell.<br />

f. Discovered at University of Wisconsin 1998-James Thomson<br />

2. Difference Between an Embryonic Stem Cell and Adult Stem Cell<br />

a. Embryonic stem cells are isolated from embryos only a few days old.<br />

i. Used to create stem cell “lines” – cultures that can be grown<br />

indefinitely in the laboratory and widely distributed to researchers.<br />

ii. Potential to develop into any type of cell in the body.<br />

b. Adult stem cells are found in many body parts such as cardiac tissue, liver<br />

and bone marrow<br />

c. Mesenchymal stem cells (MSC’s) are found in the bone marrow in<br />

varying quantities. Rare in numbers, approximately 1 in 100,000 cells<br />

(which will vary inversely with age).<br />

d. Scientists are only beginning to understand the mechanisms involved.<br />

e. Embryonic stem cells function to generate new organs and tissues.


2<br />

f. Adult stem cells function to replace cells during the natural course of cell<br />

turnover.<br />

g. Researchers continue to explore where stem cells might reside in the body<br />

and how many different organs in the body contain stem cells.<br />

3. Why are Doctors and Scientists So Excited About Them?<br />

a. Hold enormous promise for helping to understand and treat a wide variety<br />

of diseases and disorders.<br />

b. Research can provide unprecedented insight into cell interactions, cancer,<br />

birth defects and other disorders that develop during cell growth and<br />

transformation.<br />

c. Could become a renewable source of replacement cells and tissues that<br />

could be used to treat illnesses, conditions and disabilities.<br />

4. Are Stem Cells Being Used Successfully to Treat Any Human Disease?<br />

a. Blood-forming stem cells in bone marrow called homatopoietic stem cells<br />

(HSCs) are routinely being used to treat disease.<br />

b. Doctors are transferring these cells as bone marrow transplants for more<br />

than 40 years.<br />

c. Treat: leukemia, lymphoma and several inherited blood disorders.<br />

d. Only adult stem cells have been tested in humans.<br />

e. Tests have taken place in very limited clinical studies.<br />

5. The Controversy<br />

a. The mass of cells created during nuclear transfer is known as a blastocyst,<br />

or an early embryo, which theoretically could be implanted into a uterus to<br />

produce a baby.<br />

b. Groups that oppose abortion have objected to this research on the grounds<br />

that creating an embryo through nuclear transfer, only to destroy it to<br />

obtain the stem cells, is tantamount to destroying a human life. Newer<br />

technologies are moving past this.<br />

c. 1996 Law prohibits tax payer dollars in work that harms an embryo – law<br />

suits and appeals persist.<br />

d. In 2001, President Bush issued an executive order limiting federal funding<br />

of embryonic stem cell research to about 60 stem cells lines obtained from<br />

discarded embryos created in the in vitro process at fertility clinics. The<br />

order does not pertain to privately funded research.<br />

e. Scientists hope to find stem cells in the adult body that have a greater<br />

plasticity or potency – the ability to divide into multiple cell types.


3<br />

6. Media Influence<br />

a. Controversy and confusion commonly present in the media.<br />

b. Issues of Cloning<br />

c. Abortion Debate<br />

-This is rapidly becoming a moot point.<br />

d. False promises of treatments<br />

7. U.S. Technology Leader<br />

a. United States controls most of the world’s stem cell patents.<br />

b. June 2004, there were approximately 5,125 stem cell patents worldwide.<br />

c. 75% of these were held by U.S. companies and academic institutions.<br />

d. Top 30 stem cell corporations only hold close to 10% of all patents.<br />

e. The majority of stem cells patients are controlled by the government and<br />

academia.<br />

f. 5% of all patents are embryonic stem cell related.<br />

g. 2000 and 2004, the United States filed for patents four times as often as<br />

Japan. Australia and England combined.<br />

8. Government Funding<br />

a. 80% of NIH’s funding to 50,000 researches at 2.800 universities, medical<br />

schools and research institution<br />

b. 10% of the NIH’s budget goes to its own laboratories, most of which are<br />

in Bethesda, Maryland and 6,000 scientist.<br />

c. FY 2002-2010 the NIH funded approximately 2.85 billion for human stem<br />

cell research and 4.3 billion for non human stem cell research.<br />

d. September 2010: Obama admisistration expands the number of stem cells<br />

for government funding. Re-expanded to 142 hESC in December 2011.<br />

e. ClinicalTrials.gov<br />

i. Stem Cells -3861<br />

ii. Stem Cells – Orthopaedic Surgery -24<br />

9. State Funding<br />

a. 2004 Propsition 71 in California<br />

i. California Institute for Regenerative Medicine.<br />

ii. 3 Billion (300 Million/year for 10 Years).<br />

b. Several other states are getting involved with much smaller proposals.<br />

10. The Future<br />

a. Continued increase in funding<br />

b. More understainding of the biology of cell to cell interactions and<br />

commumication.<br />

c. Increase in clinical trials<br />

d. Minimal Evidence-Based Medicine to date.


4<br />

11. Most Advanced Stem Cell Companies


12 th International Sports Medicine Fellows Conference<br />

January 27-29, 2012● Carlsbad, California<br />

12:30 PM - 14:30 PM Session 3<br />

Session III<br />

12:30 PM - 13:00 PM Non-Operative Treatment - Viscosupplementation or PRP Alberto Gobbi, MD ITALY<br />

13:00 PM - 13:15 PM<br />

Nutrition and Cartilage Management; Glucosamine and<br />

What Else?<br />

Jason Theodosakis, MD USA<br />

13:15 PM - 13:30 PM Future Approaches to Cartilage: Growth Factors Susan Chubinskaya, PhD USA<br />

13:30 PM - 13:45 PM<br />

Overview - Algorithm for Cartilage Care: Where Do We<br />

Go From Here?<br />

Jason Scopp, MD USA<br />

13:45 PM - 14:00 PM The New Frontier of the Hip Jason Snibbe, MD USA<br />

14:00 PM - 14:15 PM<br />

Product Challenges for Partial Thickness Articular<br />

Cartilage Treatment<br />

Wayne Augé II, MD USA<br />

14:15 PM - 14:30 PM The Business of Cartilage Repair Ralph Gambardella, MD USA<br />

14:30 PM - 14:45 PM Discussion


Introduction<br />

“Non-Operative Treatment – Viscosupplementation or PRP”<br />

Alberto Gobbi, Kenneth Zaslav, Georgios Karnatzikos, Anup Kumar<br />

Articular cartilage damage of the knee is becoming increasingly identified as a source of<br />

functionally limiting injuries in athletes 1, 2 . Cartilage has long been recognized as having<br />

limited intrinsic healing potential 3 by the fact that it is avascular, has very few and<br />

specialized cells with a low mitotic activity and the lack of undifferentiated cells that can<br />

promote tissue repair. Once cartilage is injured it gradually degenerates, leading to<br />

Osteoarthritis (OA) 3, 4 . Many conservative treatment options such as Glucosamine,<br />

Chondroitin Sulfate and other dietary, diacerein and intra-articular and viscosupplementation<br />

have been utilized for the treatment of OA. Platelet-rich Plasma (PRP) intra-articular<br />

injections represent a therapeutic option with promising preliminary clinical results.<br />

Glucosamine, Chondroitin Sulfate and other dietary supplements<br />

Glucosamine (G) 1,500-2,000 mg/day and Chondroitin Sulfate (Cs) 800-1,200 mg/day and<br />

Avocado-Soy Unsaponifiables (ASU) 300-600 mg/day taken together or alone are useful as<br />

adjunct therapies in cartilage disorders 5 . Basic science studies indicate more anti-catabolic<br />

than anabolic effects, and all three probably act as signal modulators of inflammatory and<br />

degredative enzyme pathways. 6-8 Some animal evidence indicates pre-treatment with G, Cs,<br />

or ASU might delay the course of OA after traumatic injury, 9-11 but without human evidence,<br />

prophylactic use of these supplements cannot be recommended. G, Cs and ASU have<br />

NSAID-sparring effects. 12, 13 In acute pain, co-administration of fast acting agents such as<br />

acetaminophen or NSAIDs with G, Cs and/or ASU, followed by prompt removal of the<br />

former, may be advised. Most of the 50+ human clinical trials on G, Cs, or ASU, have<br />

shown positive results either for structure modifying effects or pain/function<br />

improvement. 14-19 20, 21<br />

Importantly, structural benefits were independent of symptom relief.


Viscosupplementation<br />

Viscosupplementation with intra-articular injection of exogenous Hyaluronic acid, has an<br />

integral role in the treatment algorithm of osteoarthritis and cartilage damage.<br />

The healthy human knee contains approximately 2cc of synovial fluid. In the<br />

osteoarthritic knee, the concentration of HA is reduced to one-half to one-third of the<br />

normal value. 22 Endogenous HA is produced by the type B synoviocytes and fibroblasts<br />

of the synovium and its role is multifactorial. 22 It provides joint lubrication and absorbs<br />

shock, while also promoting chondrocyte proliferation/ differentiation. 23-25 HA has also<br />

been shown to inhibit tissue nocioceptors and stimulate endogenous hyaluronan<br />

formation 26 Viscosupplements have both chondroprotective and anti-inflammatory<br />

effects. 27 Chondroprotection occurs through down regulation of the gene expression of<br />

osteoarthritis-associated cytokines and enzymes. 28 The anti-inflammatory effect occurs<br />

by down regulation of TNF-alpha, IL-8 and iNOS in synoviocytes. 27<br />

These chondroprotective effects have driven the use of viscosupplementation in the post-<br />

operative knee. Pain that persists after arthroscopy can be decreased with the use of<br />

Hyaluronic acid injection. It has been shown to result in decreased joint swelling and to be<br />

NSAID sparing. 29 Additionally, the disease modifying effects of HA have been shown to<br />

reduce cartilage degeneration and promote tissue repair after micro-fracture in an animal<br />

model. 30, 31 This occurs by inhibiting the production of nitric oxide and by stabilizing<br />

32, 33<br />

proteoglycan structure.<br />

Platelet-Rich Plasma (PRP)<br />

PRP can be defined as the volume of the plasma fraction from autologous blood with<br />

platelet concentration above baseline (200000 platelets/µl). 34 PRP contains different<br />

growth factors, which regulate key processes involved in tissue repair. 35, 36 The rationale<br />

for topical use of PRP is to stimulate the natural healing cascade and tissue regeneration<br />

by a “supra-physiological” release of platelet- derived factors directly at the site of


treatment. PRP has been successfully used in surgical and outpatient procedures in the<br />

treatment of several musculoskeletal problems. 37-39<br />

While recent published RCT’s using PRP in the Achilles and Rotator cuff tendons have<br />

shown little to no statistical improvement, various authors have used PRP to treat chondral<br />

defects in athletes and obtained good results. 40, 41 We prospectively followed up 50 patients<br />

active in sports with degenerative lesions of the knee 42 . All patients were treated with<br />

two intra-articular injections (one monthly) with autologous PRP (Regen® ACR-C,<br />

RegenLab, Switzerland) (Fig.1a-d). Study revealed that the use of PRP in patients with<br />

chronic degenerative disease of the knee could act to diminish pain and improve symptoms<br />

and quality of life. A prospective randomized study comparing PRP to HMW HA as well<br />

as LMW HA reported superior outcomes at 6 months with PRP injections. 43<br />

Figure 1. PRP preparation: a) blood aspiration b, c) centrifugation of the blood sample<br />

d) fraction of PRP after centrifugation (yellow upper part in tube)


Role of Pulsed Electromagnetic Fields (PEMFs)<br />

Pre-clinical studies have shown that pulsed electromagnetic fields (PEMFs), with specific<br />

physical signal parameters 44 , in vitro favour the proliferation of chondrocytes, stimulate<br />

proteoglycan synthesis and demonstrate an A2A adenosine receptor agonist activity 45-47 . In<br />

vivo, PEMFs prevent degeneration of articular cartilage and down-regulate the synthesis<br />

and release of pro-inflammatory cytokines in the synovial fluid 66-68 . We prospectively<br />

followed up 22 patients treated with PEMFs (I-ONE therapy, IGEA, Carpi, Italy) therapy 50<br />

for 1 year (fig. 2). Patients showed significant improvement in all scores at final follow up<br />

(p < .005).<br />

CONCLUSION<br />

Figure 2: I-ONE PEMFs generator<br />

A number of conservative treatment options have been utilized in order to prevent progression<br />

of OA. PRP represents a user friendly therapeutic application, which is well-tolerated and<br />

shows encouraging preliminary clinical results in active patients with knee OA.


REFERENCES<br />

1. Aroen A, Loken S, Heir S, Alvik E, Ekland A, Granlund OG, Engebretsen L.<br />

Articular cartilage lesions in 993 consecutive knee arthroscopies. Am J Sports Med.<br />

2004;32:211-5.<br />

2. Bartz RL, Laudicina L. Osteoarthritis after sports knee injuries. Clin Sports Med.<br />

2005;24:39-45.<br />

3. Hunter W. On the structure and diseases of articulating cartilage. Philos Trans Rsoc<br />

Lond B Biol Sci. 1743;9:277.<br />

4. Mankin HJ. The response of articular cartilage to mechanical injury. J Bone Joint<br />

Surg Am. 1982;64(3):460-6.<br />

5. Kenneth Zaslav K, McAdams T, Scopp J, Theosadakis J,Mahajan V, Gobbi A. New<br />

Frontiers for Cartilage Repair and Protection. Cartilage January 2012 3: 77S-86S.<br />

6. Iovu M, Dumais G, du Souich P. Anti-inflammatory activity of chondroitin sulfate.<br />

Osteoarthritis Cartilage. 2008;16 Suppl 3:S14-8.<br />

7. Campo GM, Avenoso A, Campo S, D'Ascola A, Traina P, Calatroni A. Chondroitin-<br />

4-sulphate inhibits NF-kB translocation and caspase activation in collagen-induced<br />

arthritis in mice. Osteoarthritis Cartilage. 2008;16(12):1474-83.<br />

8. Piepoli T, Zanelli T, Letari O, Persiani S, Rovati LC, Caselli G. Glucosamine sulfate<br />

inhibits IL-1-stimulated gene expression at concentrations found in humans after oral<br />

intake [abstract]. Arthritis Rheum 2005;52 Suppl 9:S502.<br />

9. Cake MA, Read RA, Guillou B, Ghosh P. Modification of articular cartilage and<br />

subchondral bone pathology in an ovine meniscectomy model of osteoarthritis by<br />

avocado and soya unsaponifiables (ASU). Osteoarthritis Cartilage. 2000;8(6):404-11.<br />

10. Boileau C, Martel-Pelletier J, Caron J, Msika P, Guillou GB, Baudouin C, et al.<br />

Protective effects of total fraction of avocado/soybean unsaponifiables on the structural<br />

changes in experimental dog osteoarthritis: inhibition of nitric oxide synthase and matrix<br />

metalloproteinase-13. Arthritis Res Ther. 2009;11(2):R41. Epub 2009 Mar 16.


11. Uebelhart D, Thonar EJ, Zhang J, Williams JM. Protective effect of exogenous<br />

chondroitin 4,6-sulfate in the acute degradation of articular cartilage in the rabbit.<br />

Osteoarthritis Cartilage. 1998;6 Suppl A:6-13.<br />

12. Conrozier T. [Chondroitin sulfates (CS 4&6): practical applications and economic<br />

impact]. Presse Med. 1998;27(36):1866-8. French.<br />

13. Appelboom T, Schuermans J, Verbruggen G, Henrotin Y et al. Symptoms modifying<br />

effect of avocado/soybean unsaponifiables (ASU) in knee osteoarthritis. A double blind,<br />

prospective, placebo-controlled study. Scand J Rheumatol. 2001;30(4):242-7.<br />

14. Ernst E. Avocado-soybean unsaponifiables (ASU) for osteoarthritis – a systematic<br />

review. Clin Rheumatol. 2003;22(4-5):285-8. Review.<br />

15. Bruyere O, Pavelka K, Rovati LC, Deroisy R, Olejarova M, Gatterova J, et al.<br />

Glucosamine sulfate reduces osteoarthritis progression in postmenopausal women with<br />

knee osteoarthritis: evidence from two 3-year studies. Menopause. 2004;11(2):138-43.<br />

16. Christensen R, Bartels EM, Astrup A, Bliddal H. Symptomatic efficacy of avocado-<br />

soybean unsaponifiables (ASU) in osteoarthritis (OA) patients: a meta-analysis of<br />

randomized controlled trials. Osteoarthritis Cartilage. 2008;16(4):399-408. Review.<br />

17. Towheed TE, Maxwell L, Anastassiades TP, Shea B, Houpt J, Robinson Vet al.<br />

Glucosamine therapy for treating osteoarthritis. Cochrane Database Syst Rev.<br />

2005;(2):CD002946. Review.<br />

18. Hochberg MC. Structure-modifying effects of chondroitin sulfate in knee<br />

osteoarthritis: an updated meta-analysis of randomized placebo-controlled trials of 2-year<br />

duration. Osteoarthritis Cartilage. 2010;18 Suppl 1:S28-31.<br />

19. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG.Effect of glucosamine or chondroitin<br />

sulfate on the osteoarthritis progression: a meta-analysis. Rheumatol Int. 2010;30(3):357-<br />

63.<br />

20. Michel BA, Stucki G, Frey D, De Vathaire F, Vignon E, Bruehlmann P, et al.<br />

Chondroitins 4 and 6 sulfate in osteoarthritis of the knee: a randomized, controlled trial.<br />

Arthritis Rheum. 2005;52(3):779-86.


21. Kahan A, Uebelhart D, De Vathaire F, Delmas PD, Reginster JY. Long-term effects<br />

of chondroitins 4 and 6 sulfate on knee osteoarthritis: the study on osteoarthritis<br />

progression prevention, a two-year, randomized, double-blind, placebo-controlled trial.<br />

Arthritis Rheum. 2009;60(2):524-33.<br />

22. Watterson JR, Esdaile JM. Viscosupplementation: therapeutic mechanisms and<br />

clinical potential in osteoarthritis of the knee. J Am Acad Orthop Surg. 2000;8(5):277-<br />

84.Review.<br />

23. Kawasaki K, Ochi M, Uchio Y, Adachi N, Matsusaki M. Hyaluronic acid enhances<br />

proliferation and chondroitin sulfate synthesis in cultured chondrocytes embedded in<br />

collagen gels. J Cell Physiol. 1999;179(2):142-8.<br />

24. Kujawa MJ, Caplan AI. Hyaluronic acid bonded to cell-culture surfaces stimulates<br />

chondrogenesis in stage 24 limb mesenchyme cell cultures. Dev Biol. 1986;114(2):504-8.<br />

25. Yagishita K, Sekiya I, Sakaguchi Y, Shinomiya K, Muneta T. The effect of<br />

hyaluronan on tendon healing in rabbits. Arthroscopy. 2005;21(11):1330-6.<br />

26. Waddell DD, Bert JM. The use of hyaluronan after arthroscopic surgery of the knee.<br />

Arthroscopy. 2010;26(1):105-11. Review.<br />

27. Greenberg DD, Stoker A, Kane S, Cockrell M, Cook JL. Biochemical effects of two<br />

different hyaluronic acid products in a co-culture model of osteoarthritis. Osteoarthritis<br />

Cartilage. 2006;14(8):814-22.<br />

28. Wang CT, Lin YT, Chiang BL, Lin YH, Hou SM. High molecular weight hyaluronic<br />

acid down-regulates the gene expression of osteoarthritis-associated cytokines and<br />

enzymes in fibroblast-like synoviocytes from patients with early osteoarthritis.<br />

Osteoarthritis Cartilage. 2006;14(12):1237-47.<br />

29. Mathies B. Effects of Viscoseal, a synovial fluid substitute, on recovery after<br />

arthroscopic partial meniscectomy and joint lavage. Knee Surg Sports Traumatol<br />

Arthrosc. 2006; 14(1):32-9.


30. Goldberg VM, Buckwalter JA. Hyaluronans in the treatment of osteoarthritis of the<br />

knee: evidence for disease-modifying activity. Osteoarthritis Cartilage. 2005;13(3):216-<br />

24. Review.<br />

31. Strauss E, Schachter A, Frenkel S, Rosen J. The efficacy of intra-articular hyaluronan<br />

injection after the microfracture technique for the treatment of articular cartilage lesions.<br />

Am J Sports Med. 2009;37(4):720-6.<br />

32. Díaz-Gallego L, Prieto JG, Coronel P, Gamazo LE, Gimeno M, Alvarez AI.<br />

Apoptosis and nitric oxide in an experimental model of osteoarthritis in rabbit after<br />

hyaluronic acid treatment. J Orthop Res. 2005;23(6):1370-6.<br />

33. Hulmes DJ, Marsden ME, Strachan RK, Harvey RE, McInnes N, Gardner DL. Intra-<br />

articular hyaluronate in experimental rabbit osteoarthritis can prevent changes in cartilage<br />

proteoglycan content. Osteoarthritis Cartilage. 2004 ;12(3):232-8.<br />

34. Mazzucco L, Balbo V, Cattana E, Guaschino R, Borzini P. Not every PRP-gel is born<br />

equal. Evaluation of growth factor availability for tissues through four PRP-gel<br />

preparations: Fibrinet, RegenPRP-Kit, Plateltex and one manual procedure. Vox Sang.<br />

2009;97(2):110-8.<br />

35. Bennett NT, Schultz GS. Growth factors and wound healing: biochemical properties<br />

of growth factors and their receptors. Am J Surg. 1993 ;165(6):728-37. Review.<br />

36. Molloy T, Wang Y, Murrell G. The roles of growth factors in tendon and ligament<br />

healing. Sports Med. 2003;33(5):381-94. Review.<br />

37. Anitua E, Sánchez M, Orive G, Andía I. The potential impact of the preparation rich<br />

in growth factors (PRGF) in different medical fields. Biomaterials. 2007;28(31):4551-60.<br />

38. Mishra A, Pavelko T. Treatment of chronic elbow tendinosis with buffered platelet-<br />

rich plasma. Am J Sports Med. 2006;34(11):1774-8.<br />

39. Sampson S, Gerhardt M, Mandelbaum B. Platelet rich plasma injection grafts for<br />

musculoskeletal injuries: a review. Curr Rev Musculoskelet Med. 2008 ;1(3-4):165-74.<br />

40. Ares O, Cugat R. Biological Repair of Knee Injuries in Soccer Players. Techniques in<br />

Knee Surgery. 2010; 9(3): 132-138. doi: 10.1097/BTK.0b013e3181ef5012


41. Kon E, Filardo G, Di Martino A, Marcacci M. Platelet-rich plasma (PRP) to treat<br />

sports injuries: evidence to support its use. Knee Surg Sports Traumatol Arthrosc. 2011<br />

Apr;19(4):516-27. Epub 2010 Nov 17. Review.<br />

42. Gobbi A, Karnatzikos G, Mahajan V, Malchira S. Platelet-rich Plasma treatment in<br />

symptomatic patients with knee Osteoarthritis: Preliminary results in a group of active<br />

patients. Sports Health: A Multidisciplinary Approach, (In Press).<br />

43. Kon E, Mandelbaum B, Buda R, Filardo G, Delcogliano M, Timoncini A, Fornasari<br />

PM, Giannini S, Marcacci M. Platelet-rich plasma intra-articular injection versus<br />

hyaluronic acid viscosupplementation as treatments for cartilage pathology: from early<br />

degeneration to osteoarthritis. Arthroscopy. 2011 Nov;27(11):1490-501. Epub 2011 Aug<br />

10.<br />

44. De Mattei M, Fini M, Setti S, Ongaro A, Gemmati D, Stabellini G, et al.<br />

Proteoglycan synthesis in bovine articular cartilage explants exposed to different low-<br />

frequency low-energy pulsed electromagnetic fields. Osteoarthritis Cartilage.<br />

2007;15(2):163-8.<br />

45. Varani K, Gessi S, Merighi S, Iannotta V, Cattabriga E, Spisani S, et al. Effect of low<br />

frequency electromagnetic fields on A2A adenosine receptors in human neutrophils. Br J<br />

Pharmacol. 2002;136(1):57-66.<br />

46. Varani K, De Mattei M, Vincenzi F, Gessi S, Merighi S, Pellati A, et<br />

al. Characterization of adenosine receptors in bovine chondrocytes and fibroblast-like<br />

synoviocytes exposed to low frequency low energy pulsed electromagnetic fields.<br />

Osteoarthritis Cartilage. 2008;16(3):292-304.<br />

47. De Mattei M, Varani K, Masieri FF, Pellati A, Ongaro A, Fini M, et al. Adenosine<br />

analogs and electromagnetic fields inhibit prostaglandin E(2) release in bovine synovial<br />

fibroblasts. Osteoarthritis Cartilage. 2009 Feb;17(2):252-62.<br />

48. Fini M, Torricelli P, Giavaresi G, Aldini NN, Cavani F, Setti S, et al. Effect of pulsed<br />

electromagnetic field stimulation on knee cartilage, subchondral and epiphyseal


trabecular bone of aged Dunkin Hartley guinea pigs. Biomed Pharmacother. 2008<br />

Dec;62(10):709-15.<br />

49. Benazzo F, Cadossi M, Cavani F, Fini M, Giavaresi G, Setti S, et al. Cartilage repair<br />

with osteochondral autografts in sheep: effect of biophysical stimulation with pulsed<br />

electromagnetic fields. J Orthop Res. 2008 May;26(5):631-42.<br />

50. Gobbi A, Karnatzikos G, Malchira S, Maggi L, Ongaro A. The use of pulsed<br />

electromagnetic fields in symptomatic patients with degenerative cartilage lesions of the<br />

knee: a preliminary report. Journal of Sports and Traumatology (In Press).


Dr. Theo<br />

Nutrition and Cartilage Management:<br />

Glucosamine and What Else?<br />

Jason Theodosakis, MD, MS, MPH, FACPM<br />

“Dr. Dr. Theo” Theo<br />

University of Arizona College of Medicine<br />

Tucson, Arizona<br />

drtheo.com / drjtheo@gmail.com<br />

How Should You Use/Prescribe<br />

Supplement for Joint Health?<br />

• For symptom relief<br />

• To reduce need for acetaminophen/NSAIDs<br />

p<br />

• For structure modification/delay of OA<br />

progression<br />

• As an adjunct to cartilage repair surgeries?<br />

Prescription DMOADs do not exist<br />

for OA, and pharmacologic agents<br />

are known to have serious<br />

adverse events<br />

Some dietary supplements have<br />

DMOAD action, are safe and should<br />

be used as co-first line agents, but<br />

there is a lot of hype and concerns<br />

about product quality and efficacy<br />

1<br />

3<br />

Disclosures<br />

Stock Ownership, Past Speaker: Pfizer<br />

Consultant and/or Grants: Bioiberica,<br />

Ownership/Officer: Supplement Testing Institute -<br />

“Dr. Theo’s ® Dr. Theo s ” Joint health products manufacturer and<br />

seller, sell GS on website<br />

Book Author<br />

Future OA Therapy (Speculation) - Tailor made?<br />

OA Tyype<br />

Dr. Theo<br />

Ia<br />

Ib<br />

IIa<br />

IIb…<br />

Pharmacogenomic Profile<br />

A1 A2 B1 B2…<br />

Optimal Pharmacologic<br />

Therapy<br />

= COX/5-LOX Inh.<br />

= G/CS<br />

= Diacerrhein<br />

= IL-1 RA<br />

=ASU = ASU<br />

= HA injection<br />

= NO NSAID<br />

Other Factors: Age, OA grade, location of lesions, biomechanical factors, etc.<br />

Acetaminophen is not that safe<br />

or effective<br />

• No Significant DMOAD studies published through 12/11<br />

• Several pain/function studies are null, especially long-term 1-4<br />

• Safety is becoming more and more of an issue …<br />

– #1 cause of accidental and intentional acute liver failure in the US 5<br />

– Even short-term use (2 weeks) can elevate LFTs 6<br />

– Even low-dose (>500 mg/day) chronic use is associated with 2x HTN<br />

incidence in middle-aged women 7<br />

– Toxic/Lethal dose is only 2.5x7 /8x of max daily dose (4 gm/day)<br />

Dr. Theo<br />

1- Case JP. Arch Intern Med. 2003;163:169-178<br />

2- Miceli-Richard C. Ann. Rheum. Dis. 2004:923-30<br />

3- Pincus T. Ann Rheum Dis. 2004;63;931-939<br />

4- Herrero Herrero-Beaumont Beaumont G. Arth & Rheum 2007. 56;2<br />

pp 555 555-567. 567.<br />

•<br />

5- Larson AM. Hepatology 2005;42:1364-72<br />

6- Watkins P. JAMA 2006;296:87-93<br />

7- Forman JP. Hypertension. Vol. 46(3), 9/2005, pp 500-507.<br />

8- Hung OL. Emergency Medicine: Comprehensive Study Guide<br />

6 th Ed (2004) Section 14; 171.<br />

4<br />

6<br />

1


NSAIDs are not that safe and offer<br />

only short-term symptom relief<br />

• No Significant DMOAD studies published through 12/11<br />

• All long-term pain/function studies are null 1-4<br />

GI safety benefits of Celebrex ® • GI safety benefits of Celebrex are lost with the addition of<br />

® are lost with the addition of<br />

prophylactic aspirin<br />

• Chronic NSAID usage often requires co-administration of<br />

PPIs<br />

Dr. Theo<br />

1- Case JP. Arch Intern Med. 2003;163:169-178<br />

2- Miceli-Richard C. Ann. Rheum. Dis. 2004:923-30<br />

3- Pincus T. Ann Rheum Dis. 2004;63;931-939<br />

4- Herrero Herrero-Beaumont Beaumont G. Arth & Rheum 2007. 56;2<br />

pp 555-567. 555 567.<br />

Copyright ©2005 American Physiological Society<br />

5- Larson AM. Hepatology 2005;42:1364-72<br />

6- Watkins P. JAMA 2006;296:87-93<br />

7- Forman JP. Hypertension. Vol. 46(3), 9/2005, pp 500-507.<br />

8- Hung OL. Emergency Medicine: Comprehensive Study Guide<br />

6 th Ed (2004) Section 14; 171.<br />

Rationale for Weight Loss<br />

Intra-abdominal fat creates chronic low-grade systemic inflammation<br />

Petersen, A. M. W. et al. J Appl Physiol 98: 1154-1162 (2005)<br />

Mediterranean Diet<br />

Fat as an endocrine<br />

organ (especially<br />

omental fat)<br />

7<br />

9<br />

11<br />

What About Weight Loss?<br />

Effect of weight reduction in obese patients diagnosed<br />

with knee osteoarthritis: a systematic review and<br />

meta-analysis<br />

• 35 trials identified; 4 RCTs (5 comparisons) included (N=454)<br />

• Pooled ES for a weight reduction of ~6 kg:<br />

– pain 0.20 (0 - 0.4)<br />

– disability 0.23 (0.04 - 0.4)<br />

• Disability improved significantly when weight was reduced<br />

>5% or at a rate >0.24% per week<br />

• Clinical efficacy for pain was evident, but not predictable<br />

Christensen, et. al. Annals of the Rheumatic Diseases 2007;66:433-439<br />

About 90% of Arthritis Patients<br />

Use Dietary Supplements – Why?<br />

• “Conventional medicine is not optimally effective<br />

• Drugs frequently have adverse effects<br />

• Herbal medicines are heavily yp promoted, , for example, p , by ythe<br />

popular press, Internet sites, popular books, and celebrities<br />

• Herbal medicines are widely available, usually marketed as food<br />

supplements<br />

• Exaggerated claims are often made regarding their efficacy<br />

• The public tends to view herbs as natural and thus devoid of risks<br />

• Most consumers can afford the extra costs for herbal medicines”<br />

Dr. Theo<br />

Ernst E. Rheum Dis Clin N Am 37 (2011) 95–102<br />

8<br />

10<br />

12<br />

2


What Supplements May Help Gout?<br />

• Lifestyle modification and prescriptions are<br />

quite effective<br />

– Xanthine oxidase inhibitors, Uricosuric agents<br />

– New URAT 1 inhibitor, IV pegloticase<br />

• No major definitive supplement trials<br />

• Vitamin C has most evidence<br />

• Cherries and glucosamine may be of benefit<br />

Dr. Theo<br />

Osteoarthritis - Evolving Definition<br />

“Osteoarthritis is a disorder involving movable joints<br />

characterized by micro- and macro-injury–initiated cell<br />

and extracellular matrix degeneration that activates<br />

Dr. Theo<br />

maladaptive l d ti repair i responses, iincluding l di sterile t il<br />

autoinflammatory pathways of innate immunity, bone<br />

remodeling, and osteophyte formation.”<br />

Virginia Kraus MD, PhD 2011<br />

Nutraceutical Interventions for Cartilage/OA<br />

Dr. Theo<br />

• ASU (Avocado-Soybean Unsaponifiables)* 300-600 mg<br />

• Chondroitin* 800-1,600 mg (≥ USP Grade, single animal origin)<br />

• Glucosamine* 1,500-3,000 mg<br />

• SAM-e (s-adenosylmethionine)* 800-1,600 mg<br />

* Also sold as OTC or Prescription drugs in some countries<br />

13<br />

15<br />

17<br />

Dr. Theo<br />

Trauma<br />

Vitamin C and Serum Uric Acid review article<br />

NF-�B NF-�B<br />

I-�B I-�B<br />

Summary<br />

Median 500 mg C<br />

Reduction -0.35 mg/dl<br />

P = 0.032<br />

NF-kB activation pathway<br />

Grimm et al., J Inflamm 3: 1-5, 2006<br />

• 200-2,000 mg<br />

• Heterogeneous<br />

populations and<br />

research designs<br />

• Uric acid levelsnot<br />

gout attacks<br />

evaluated<br />

Juraschek SP. Arthritis care<br />

& research. 63(9):1295-306,<br />

2011 Sep<br />

Nutraceutical Interventions for Cartilage/OA<br />

Secondary agents<br />

Dr. Theo<br />

• MSM (methylsulfonylmethane) 1,500-6,000 mg<br />

• Anti-inflammatory enzymes*<br />

• Hyaluronic Acid, Oral 80 mg - 240 mg/day<br />

• Omega-3 Fish Oil 2 gm & up of Omega-3, especially EPA<br />

• French Maritime Pine Bark Extract 100 100-150 150 mg<br />

• Herbal derivatives<br />

– Devil’s Claw 1,800 to 3,600 mg<br />

– Boswellia 300 – 3,000 mg<br />

– Rose Hips 2,250 mg<br />

– Curcumin-phosphatidylcholine complex 1,000 mg<br />

• Egg Shell Membrane 500 mg<br />

* Also sold as OTC or Prescription drugs in some countries<br />

14<br />

18<br />

3


Effect Sizes for Pain Improvement in<br />

Major OA Treatments (Es pain, 95%Cl, Level of Evidence)<br />

Acupuncture 0.35 (0.15, 0.55), la<br />

Weight reduction 0.20 (0.06, 0.33), la<br />

Electrotherapy/EMG 0.15 (-0.09, 0.39), la<br />

Glucosamine sulphate 0.40 (0.15, 0.64), la<br />

Chondroitin sulphate p 0.68 (0.43, ( , 0.93), ), la<br />

ASU 0.38 (0.01, 0.76), la<br />

Acetaminophen (paracetamol) 0.15 (0.01, 0.76) la<br />

NSAIDs 0.29 (0.22, 0.35), la<br />

Topical NSAIDs 0.44 (0.27, 0.62), la<br />

Opioids 0.37 (0.23, 0.51), la<br />

IA corticosteroid 0.55 (0.34, 0.75), la<br />

Zhang W, Moskowitz R, Nuki G. OARSI Treatment Guideline Committee ACR 10/2008 Includes studies published up to 1/31/08<br />

ASU - Research Review<br />

• Five, randomized, double-blinded, placebo-controlled<br />

human clinical trials lasting three months to two years in<br />

duration 1-4<br />

• Three studies positive for pain and function<br />

improvement 1-3 and NSAID use reduction 3<br />

improvement 1 3 and NSAID use reduction 3<br />

• Two studies evaluated Joint Space Width (JSW) of hip<br />

on X-Ray 4-5<br />

• Meta-Analysis: “Give ASU a chance for e.g. 3 months;<br />

Knee results>hip” 6<br />

1- Blotman F. Rev Rheum 1997;64(12);825-834<br />

2- Maheu E. Arth & Rheum 1998;41(1):81-91<br />

3- Appelboom T. Scand J Rheum 2001;30:242-7<br />

4- Lequesne M. Arth & Rheum 47:50-58, 2002<br />

5- Maheu E. Ann Rheum Dis 2010;69(Suppl3):150<br />

6- Christensen R. OA&Cart 2008;6,399-408<br />

SAM-e (S-Adenosyl- L-methionine)<br />

• Naturally occurring methyl donor used as a drug in<br />

Europe for treatment of depression, OA and intrahepatic<br />

cholestasis<br />

• Dose used in OA studies = 800 - 1,600 mg<br />

• Symptom, but no disease-modifying data for OA<br />

• Onset of action 8-12 weeks<br />

Dr. Theo<br />

19<br />

21<br />

23<br />

All herbal medicines should be<br />

ceased 2 weeks before surgery<br />

The peri-operative implications of herbal medicines<br />

P. J. Hodges and P. C. A. Kam<br />

Anaesthesia. Volume 57; Page 889 - 9/2002<br />

SAM-e (S-Adenosyl- L-methionine)<br />

• Equivalent effect overall to low-dose NSAIDs or placebo for OA<br />

pain and anti-depressants for mild/moderate depression (but with<br />

less side effects than prescription anti-depressant drugs)*<br />

• Effect size of studies lower than that for glucosamine/chondroitin g<br />

or<br />

ASU<br />

• Synthetic version appears to be less active - common in retail<br />

brands in US<br />

• Too expensive for general use. Reserve for OA/depression combo<br />

Dr. Theo<br />

Dr. Theo<br />

* Agency for Healthcare Research and Quality (AHRQ)<br />

Publication No. 02-E033, August 2002. Agency for<br />

Healthcare Research and Quality, Rockville, MD.<br />

What’s New in<br />

Glucosamine/Chondroitin?<br />

• Once daily & 4-6 month trial for symptom relief<br />

• Symptom relief unrelated to structural effect<br />

• D Dose (1 (1,500 500 mg glucosamine) l i ) may bbe llow<br />

• Should use of 800 mg chondroitin now be<br />

mandatory?<br />

• Chondroitin evidence is very strong at 800 mg, ≥<br />

USP Grade, single animal sourced<br />

22<br />

24<br />

4


Example of what happens in a long-term OA Study<br />

Dr. Theo<br />

Dr. Theo<br />

Meta-Analysis of Chondroitin Sulfate for Structure<br />

Modification<br />

Positive effect size (0.26), highly statistically significant p< 0.0001<br />

Chondroitin can beneficially affect structure independent of symptom modification<br />

Even with the addition of GAIT part II, the effect size was 0.22<br />

and still significant (Hochberg, OARSI 2008)<br />

Hochberg M, et. al. Current Medical Research and Opinion Vol.24, No. 11, 3029-35 (2008) 27<br />

Chondroitin Biologic Activity Variation<br />

• Testing in vitro biologic activity of 10 chondroitin raw<br />

materials on bovine chondrocytes<br />

• About half had either reduced activity or activity not<br />

significantly different from negative control<br />

Int’l Cartilage Repair Society Meeting, San Diego 1/06<br />

25<br />

29<br />

TID Dosing May Lower Glucosamine’s Peak Plasma Concentration<br />

Dr. Theo<br />

[<br />

Peak Plasma<br />

Concentration<br />

C max (µM)<br />

GS 1.5 g/day over 3 days 1 8.9<br />

G•HCL 1.5 g One-Time Dose 2 3.0<br />

G•HCL 0.5 g TID Multiple Dose<br />

over 12 weeks 3<br />

1.2<br />

3,000 mg GS<br />

13.3 µM<br />

Glucosamine IC50 on IL-1b-stimulated gene expression 4<br />

Marker iNOS TNF-a COX-2 MMP-3 IL-1b IL-6 ADAMTS5 NF-kB Subunits<br />

P50 p52 p65 RelB<br />

IC 50 (µM) 13.8 12.8 11.2 10.2 6.2 4.4 2.8 0.4 / 0.6 / 1.0 / 0.3<br />

Dr. Theo<br />

Dr. Theo<br />

1- Persani S. Osteoarthritis and Cart. 13, 1041-1049 (2005)<br />

2- Jackson CG. ACR 2005 meeting Late Breaking Abstract L13<br />

3- Jackson CG. ACR 2006 meeting Poster 352<br />

4- Piepoli et al, Arthritis Rheum 2005; 9 Suppl: 1326<br />

• 69 patients with synovitis, 800 mg CS or Placebo, 6 mo then all<br />

given CS for 6 months more<br />

• Symptoms similar between groups but:<br />

– At 6 months, CS group had less cartilage volume loss in global knee, lateral<br />

compartment, and tibial plateaus (p = 0.030, 0.015 and 0.002)<br />

– At 12 months, CS group lower BML scores in Lat compartment and Lat<br />

femoral condyle (p = 0.035 and 0.044)<br />

Ann Rheum Dis 2011;70:982-89<br />

Glucosamine/Chondroitin Safety<br />

• “Anything that has an effect can have an adverse effect”<br />

Theodosakis 1995<br />

• Case Reports<br />

– INR values may be affected with lower quality chondroitin<br />

– Skin rashes have been reported for shellfish based glucosamine,<br />

especially in those with shellfish anaphylaxis history<br />

• No binding to plasma proteins, no use of Cytochrome<br />

enzymes<br />

• No statistically significant difference has been in any human<br />

controlled or observational trial to date, compared to<br />

placebo, for up to 8 years of assessment<br />

26<br />

28<br />

30<br />

5


What are the chances that<br />

Glucosamine and Chondroitin are<br />

Placebos?<br />

• Most studies show statistically significant effects on<br />

primary or secondary outcome measures such as<br />

pain, ffunction, quality of f life, f joint space narrowing,<br />

cost effectiveness<br />

• In no instance out of > 100 study arms has a<br />

placebo resulted in a statistically significant better<br />

primary or secondary outcome than G/CS<br />

• Odds of this due to chance are astronomical<br />

Dr. Theo<br />

Dr. Theo<br />

Dr. Theo<br />

Chondroitin Effect Size Review<br />

31<br />

Long-term structure<br />

studies that were<br />

positive for<br />

structure<br />

changes, but not for<br />

pain (since subjects<br />

had little pain to<br />

start)<br />

Reichenbach S. Ann<br />

Intern Med.<br />

2007;146:580-590.<br />

Was GAIT 1 Falsely Negative?<br />

• Before even evaluating the effect of the<br />

supplements, the effect of the active comparator<br />

drug (celecoxib) must be similar to all other<br />

celecoxib studies or the study may be void<br />

• 3 ways to determine the effect of the active<br />

comparator drug:<br />

– Study outcome measures X - 40/42 failed<br />

– Clinical relevance of treatment effect<br />

– Effect Sizes<br />

X – Not 15% > Placebo<br />

X X – Not Clinically Relevant or Statistically Significant<br />

33<br />

35<br />

Dr. Theo<br />

Dr. Theo<br />

Why Doesn’t Glucosamine and<br />

Chondroitin “Work” in Some<br />

Patients?<br />

• What does “Work” Mean?<br />

– Less pain? p Improved p activity y tolerance? Less<br />

Stiffness?<br />

– Decreased use of medications?<br />

– Less need for surgery or physical therapy?<br />

– Lower overall cost of care?<br />

Pharmacological response to Chondroitin as a function of<br />

baseline pain intensity and duration of treatment<br />

(data from Reichenbach et al. Ann Intern Med 2007;146:580-90)<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

Responders VAS/Lequesne<br />

Non responders VAS/Lequesne<br />

Non responders WOMAC<br />

20<br />

0 3 6 9 12 15 18 21 24<br />

Duration of treatment (months)<br />

War- Supplements vs Drugs<br />

• Manufacturers hype/skew data<br />

• Studies are designed or performed<br />

improperly to get intended result<br />

– Improper dose, form, or outcomes are used<br />

– Meta-analyses are altered to include<br />

inappropriate studies<br />

– Structure studies are used to negate positive<br />

pain data<br />

32<br />

36<br />

6


Dr. Theo<br />

Some of the Negative (Null) Studies on G/CS are so<br />

flawed that they should not even be included in the<br />

argument against G/CS effectiveness<br />

• Uncontrolled use of NSAIDs, opiates,<br />

acetaminophen<br />

• Insufficient dose of supplements<br />

• Insufficient duration of the study<br />

• Mixed conditions/joints being studied<br />

• Inappropriate outcome measures<br />

No Excuse for having low levels of<br />

Vitamin D<br />

• Work in sun w/o sunscreen and only partially<br />

clothed - problem: your dermatologist<br />

• Measure levels: optimal appears to be about 50<br />

ng/ml (conversion to nmol/L: multiply by 2.5)<br />

• Most people need 1,000-5,000 IU by<br />

supplements<br />

• With low levels, 1,000 IU raises serum by about<br />

10 ng/ml, but less as level rises<br />

Oral HA Products<br />

• 3 main origins of HA raw material<br />

– Low HA concentration (mainly collagens, e.g. BioCell ® )<br />

– High HA concentration extracted from Avian sources (e.g. Hyal-Joint ® )<br />

– Fermented (China, e.g. HyaMax®)<br />

• Evidence (Avian)<br />

– 1 pilot DBPC clinical positive for improved pain, ADLs and QOL<br />

– 2 positive human pain/synovitis studies: effect > acetaminophen<br />

– Equine oral absorption/effect study at human equivalent dose<br />

– Several positive basic science mechanism studies<br />

Dr. Theo<br />

37<br />

39<br />

41<br />

HSA - Health Savings Accounts<br />

FSA - Flexible Spending Accounts<br />

www.irs.gov/pub/irs-pdf/p502.pdf<br />

MSM (Methylsulfonylmethane)<br />

• Naturally occurring sulfur-containing supplement<br />

• Anti-oxidant and anti-inflammatory properties<br />

• In-vitro and animal study- anti-inflammatory @ 6g/d<br />

• 3 pilot studies on OA pain and function<br />

– 1,500 mg/day Indian study1 3 000 mg/day Israel study2 – 3,000 mg/day Israel study2 – 6,000 mg/day US study 3<br />

• Lower doses (e.g. 500 mg) may help offset sulfur<br />

losses in cartilage due to OA and medication use<br />

• Use distilled product to reduce the risk of DMSO or<br />

heavy-metal contaminants<br />

1- Usha, et. al. Clin. Drug Investigations. 24:6;pp.353-63<br />

2- Debi, et. al. OARSI 2007<br />

3- Kim, et. al. OA & Cartilage 14:(2006), 286-294<br />

Omega-3 considerations<br />

• Oxidized oil (rancid) is proinflammatory<br />

• People convert short chain to long chain oils<br />

variably (fish oil may be better than others)<br />

• Cod liver oil - too much Vit A?<br />

• TG vs. EE form<br />

• EPA/DHA ratios<br />

Supplements<br />

7


Study Number of<br />

patients<br />

Pain Joint stiffness Physical<br />

function<br />

Farid et al. 37 -43 % -35 % +52 %<br />

Cisar et al 100 -40 % -40 % *<br />

+22 % *<br />

Farid et al. 37 -43 % -35 % +52 %<br />

Cisar et al 100 -40 % -40 % *<br />

+22 % *<br />

Cisar et al. 100 -40 % -40 % +22 %<br />

Belcaro et al. 156 -55 % -53 % +56 %<br />

Farid et al., Nutr Res, 27:692-697, 2007<br />

Cisar et al. Phytother Res 22: 1087-1092, 2008 (* = 2 month results)<br />

Belcaro et al. Phytother Res, 22: 518-523, 2008<br />

Key Points<br />

• The evidence for most dietary supplements or nutraceuticals<br />

in OA is incomplete<br />

• The four that are OTC or prescription drugs throughout the<br />

world do have good evidence (ASU, glucosamine,<br />

chondroitin, , SAMe) )<br />

• The risk/benefit/cost ratio of the first three is better than that<br />

of other pharmacologic interventions for OA<br />

• Vitamin D levels should be optimized in all patients<br />

• Some herbals like curcumin, boswellia and oral, avianderived<br />

Hyaluronic acid appear to be promising<br />

Dr. Theo<br />

Book - Integrative OA Treatment <strong>Program</strong><br />

Purpose: How-To guide to get patients directly involved in care, integrate all areas of<br />

treatment and reduce the time needed to educate patients. 8th grade reading level.<br />

Nine Steps:<br />

1. Accurate diagnosis, modify<br />

reversible contributors to OA<br />

2. Use glucosamine and<br />

chondroitin (& ASU)<br />

3. Improve biomechanics<br />

4. Low-impact exercise<br />

Dr. Theo<br />

5. Mediterranean diet, lots of fruits &<br />

vegetables, anti-oxidants and<br />

Vitamin D<br />

66. Maintain ideal body weight, weight lose<br />

weight if overweight<br />

7. Treat associated depression<br />

8. Use conventional medicine as<br />

necessary<br />

9. Maintain positive attitude/social<br />

support<br />

45<br />

© Jason Theodosakis, M.D. 1996, 2004 47<br />

Curcumin<br />

• Component of Turmeric from root of Curcuma longa plant<br />

• Absorption in humans after oral consumption of unmodified<br />

curcumin extract nil - even at multi-gram doses1-2 • Good 90 day study on 50 subjects3 expanded to 8-month<br />

DBPC OA study with 100 subjects subjects. Used 11,000 000 mg of<br />

phosphatidylcholine bound curcumin4 • When combined with black pepper extract piperine,<br />

bioavailability increased 20-fold, but piperine is a CYP3A4<br />

cytochrome inhibitor – a concern for drug interactions<br />

Dr. Theo<br />

1- Anand p" et al. Mol. Pharmaceutics: 2007, 4(6), pp. 807-818.<br />

2- Marczylo T., et al. Chemother. Pharmacol. 2007, 60, 171·177.<br />

3- Belcaro G., et al. Panminerva Medica: 2010 Giugno; 52 (2 Suppll). 55·62<br />

4- Belcaro G., et al Altern Med Rev. 2010 Dec;15(4):337-44<br />

44<br />

1- Alt. Med Rev. 15:4, pp 337-344<br />

Key Points - Nutraceuticals<br />

• Regarding Glucosamine/Chondroitin… Most of the<br />

null/negative studies to date are so flawed that they<br />

should provide no evidence against efficacy, but are<br />

being used in such a manner anyway<br />

• ASU/Glucosamine/Chondroitin should be co-firstline<br />

pharmacologic therapy for OA<br />

• The quality, dose (potency), label claim accuracy,<br />

biologic activity, and wild claims of dietary<br />

supplement category for OA needs improvement<br />

Dr. Theo<br />

Appendices<br />

48<br />

46<br />

8


Dr. Theo<br />

1<br />

Reading a Label<br />

Ask:<br />

• Are the right ingredients included<br />

in the proper form and amount?<br />

• Is the the amount amount hidden hidden within within a<br />

“proprietary blend?”<br />

• Labeling law requires listing the<br />

ingredients from the highest to<br />

lowest quantity<br />

Does an Oral Cartilage Treatment Work?<br />

The Seven Stages of Evidence<br />

2<br />

3<br />

4<br />

5<br />

6<br />

7<br />

Is it absorbed?<br />

What’s the mechanism of action?<br />

Clinical/Animal symptom y p studies<br />

Clinical/Animal structure studies<br />

Comparison to known treatments<br />

Is it safe?<br />

Long-term outcome measures<br />

ASU - Research Review<br />

Anti-Catabolic, Anti-Inflammatory mechanisms<br />

• Suppresses TNF-a, IL-1b, COX-2, iNOS, PGE2 and NO production in LPS<br />

activated articular chondrocytes and TNF-a, IL-1b in monocyte/macrophages 1<br />

• Inhibits NFk-B, MAPK-14, c-fos and c-jun expression 1<br />

• Suppresses IL-1b stimulation of stromelysin, IL-6, IL-8 and PGE2 in human<br />

articular chondrocytes in culture 2<br />

• Using RA synoviocytes, ASU lowered levels of VGEF (vascular endothelial<br />

growth factor) while increasing levels of TIMP-1 (both p


45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Effect of ASU on NSAID use<br />

and Pain Reduction<br />

# days on NSAIDs VAS Pain<br />

= P < 0.01<br />

N = 260 subjects<br />

Placebo<br />

ASU 300 mg<br />

ASU 600 mg<br />

Appelboom T, Schuermans J, et. al. Symptoms modifying effect of<br />

ASU in knee osteoarthritis. Scan J. Rheumtol. 2001;30:242-7.<br />

55<br />

Where do you draw the line for<br />

acceptable evidence?<br />

Any kind of study of<br />

any quality with any<br />

dosage of any form<br />

of an ingredient, g ,<br />

performed only by<br />

those with direct<br />

personal gain from a<br />

positive outcome<br />

Dr. Theo<br />

Double-blinded,<br />

placebo-controlled<br />

studies, with animal<br />

and basic science<br />

support. Biologic<br />

plausibility and<br />

dosing<br />

Over-Hyped “Science”<br />

Dr. Theo<br />

Multiple, high quality,<br />

DBPC studies, active<br />

comparator trials,<br />

outcome studies and<br />

review articles by<br />

different, independent<br />

reviewers using the<br />

exact same product<br />

in the exact same<br />

population?<br />

57<br />

59<br />

Outcome Data on G/CS<br />

• CS has been shown to reduce NSAID usage by 63-85% in<br />

(11,000) OA patients. Several weeks of CS allowed for an<br />

elimination of about 50% of refill prescriptions for NSAIDs 1<br />

• GS also lowers cost of OA care 2 and decreased need for<br />

Total Knee Replacement 3<br />

• G/CS have shown beneficial f or promising effects ff in most of f<br />

the proposed biochemical targets/pathways in OA: IL-1b,<br />

TNF-a, NF-kB, MMPs, iNOS/NO, PG & HA synthesis,<br />

oxidation, apoptosis, others. 4,5,6<br />

1- Conrozier T. Presse Med 1998 Nov 21;27(36):1866-8 4- Herrero-Beaumont G. Future Rheumatology (2006):1(4);397-414<br />

2- Reginster JY. Arthritis Rheum 2003;48 (9 suppl):89 5- Bali J-P. Sem Arth Rheum. 2001:3(1);58-68 (review)<br />

3- Pavelka K et al, Arthritis Rheum 2004;50,(9 suppl): 251 6- Volpi N. Curr Drug Targets Immune Endocr Metabol Disord. 2004<br />

Jun;4(2):119-27 (review)<br />

Dr. Theo<br />

56<br />

Dr. Theo<br />

Dr. Theo<br />

Common Marketing Ploys<br />

“Fraud Dosing”<br />

• Fraud implies intentional deception for personal<br />

gain for self (or for harm to another)<br />

• Sophisticated, often Doctorate-level formulators<br />

from most large companies know the exact dose<br />

that corresponds with benefit in the research<br />

• Products with a fraction of the efficacious,<br />

researched dose commonly have, or imply claims<br />

and benefits that are not substantiated<br />

58<br />

60<br />

10


Dr. Theo<br />

Dr. Theo<br />

Dr. Theo<br />

“The Non-Inferiority Game”<br />

• Instead of doing multiple double-blinded, placebo<br />

controlled clinical trials…<br />

• Set-up a clinical study between a new agent and a<br />

known, effective agent<br />

• Manipulate the study to favor the new agent by:<br />

– Hand-picking the population to study<br />

– Fidgeting with the dose or form<br />

– Picking inappropriate time intervals<br />

– Being creative with the analysis and statistics used<br />

Deceptive Marketing?<br />

Over-Hyped “Science”<br />

61<br />

• Too short of a duration<br />

• Only 26 patients per<br />

group started<br />

• NNo Pl Placebo b group<br />

• C + Cs has never<br />

performed so poorly in<br />

any other study<br />

• Efficacy vs Effectiveness<br />

63<br />

“The Absorption Game”<br />

• Some marketers try to confuse the public into<br />

believing that absorption relates precisely to<br />

effect<br />

• Often, there’s a significant price premium for a<br />

small increase in absorption<br />

• Absorption helps determine dose, not effect<br />

Dr. Theo 62<br />

Over-Hyped “Science”<br />

Dr. Theo<br />

64<br />

11


Bone<br />

High<br />

Future Approaches to<br />

Cartilage: Growth Factors.<br />

Is that yes or no?<br />

Susan Chubinskaya, PhD<br />

The Ciba-Geigy Professor<br />

of Biochemistry<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

The Spectrum of Repair<br />

Potential of Musculoskeletal<br />

Tissues<br />

Muscle Tendon Ligament Meniscus<br />

Cartilage<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Low<br />

•Current US Cartilage Repair<br />

Market value ~60M/1B<br />

•Focal Defects can eventually<br />

lead to OA and Total Knee<br />

Arthroplasty (TKA)<br />

•Cartilage C til regeneration ti<br />

represents an opportunity for<br />

early intervention and<br />

maintenance of an active<br />

lifestyle, delaying or avoiding<br />

TKA<br />

Science drives the ship<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Treatment of Cartilage Injuries: A Medical Need!<br />

Osteochondral<br />

Graft<br />

8%<br />

MicroFracture<br />

20%<br />

Cartilage Repair: US<br />

Procedural Share<br />

ACI<br />

1%<br />

Allografts<br />

1%<br />

Debridment &<br />

Lavage<br />

70%<br />

Still we could not reproduce the native<br />

articular cartilage!<br />

Courtesy of Dr. Cole<br />

Natural History of Cartilage Injury & Repair<br />

Spontaneous Repair Integrative Repair Regeneration<br />

Fails to replicate<br />

normal<br />

structure,<br />

composition and<br />

function<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Fails to repair<br />

interface<br />

between repair<br />

tissue &<br />

cartilage<br />

Does not form<br />

new tissue that<br />

duplicates<br />

structure,<br />

composition &<br />

function<br />

Courtesy of Dr. Hurtig<br />

1


Adult repair tissue tissue-- --Can Can it develop<br />

appropriate properties or…the challenge of<br />

an aging but active patient population!<br />

• But adult repair tissue will adapt slowly<br />

• Regional specialization will be limited<br />

• Need for specialization is greater because<br />

bi biomechanical h i l loads l d will ill be b higher hi h<br />

• Repair tissue will be susceptible to re-injury<br />

Sport Fellows Course<br />

Carlsbad, 01-16-2009<br />

IGFs<br />

Cell proliferation<br />

Chondrogenesis<br />

Images-Dr. A. Shimmin-Melbourne<br />

TGF TGF-β Osteogenesis<br />

(42members) ) Chondrogenesis<br />

BMPs Wound Healing<br />

GDFs/CDMPs BMP BMP-2 2 & 7<br />

EGF<br />

EGF<br />

GGrowth th Factors F t<br />

Proliferation of<br />

Mesenchymal, glial,<br />

and epithelial cells<br />

VEGF<br />

Role in tissue<br />

engineering,<br />

MSC proliferation,<br />

vascular system<br />

repair<br />

TGF TGF-α<br />

Potential role in<br />

normal wound<br />

healing<br />

Slide-Courtesy of Dr. Hurtig<br />

PDGF<br />

Proliferation of<br />

Connective tissue<br />

Glial & Smooth<br />

muscle l cells ll<br />

FGFs, FGF-18 FGF 18<br />

Cell proliferation<br />

Chondrogenesis<br />

Osteogenesis<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Synovial environment<br />

The Diamond Model of<br />

Cartilage Repair<br />

Interactions<br />

Chondrogenic Cells Chondrogenic Scaffolds<br />

Host<br />

Mechanical Environment<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Chondrogenic Cells<br />

Diamond Concept<br />

Host<br />

Bone<br />

Active Agents/Growth Factors<br />

Adapted from Giannoudis et al, 2008, Injury<br />

FGF-18 in cartilage repair/animal study<br />

Intra-articular injection j of FGF18 induced<br />

chondrogenesis and promoted repair of cartilage<br />

lesions in rats with meniscal tear-induced OA. Rats<br />

were subjected to meniscal tear and treatment with<br />

either vehicle alone (a–d) or vehicle containing 5.0 μg<br />

of FGF18 (e–h). (a–d) Black arrows indicate sites of<br />

cartilage damage; red arrows indicate chondrophytes;<br />

green arrow in (a) indicates collapse of the<br />

subchondral bone. (e–h) Black arrowheads indicate<br />

sites of chondrogenesis and repair of cartilage<br />

lesions; red arrows represent chondrophytes.<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

2


FGF-18 human trials<br />

Cell therapy with growth<br />

factors<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

OP-1/BMP-7 Repair Models<br />

Soft Tissue Models<br />

Ex Vivo Model<br />

•Cartilage Acute trauma/PTOA model<br />

• Joint Tissue Repair<br />

– Articular Cartilage<br />

In Vitro Studies<br />

• Osteochondral Defect<br />

Models<br />

• Chondral Defect Models<br />

• O Osteoarthritis t th iti M Models d l<br />

•Organ/explant cultures (fresh and<br />

allograft cartilage)<br />

•Isolated chondrocytes in<br />

monolayers (primary, ACI)<br />

•Isolated Chondrocytes in Alginate<br />

• Acute trauma/PTOA model<br />

Beads (primary, ACI)<br />

• Clinical data<br />

– Meniscal Cartilage<br />

Hard Tissue Models<br />

– Tendon/Ligament<br />

•Long Bone Defects<br />

• Intervertebral Disc Repair<br />

•Spinal Fusion<br />

•Cranialfacial Defects<br />

– Injurious compression model •Metal Implant Fixation<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Loeser et al A&R<br />

2003<br />

Chubinskaya et al<br />

O&C 2007<br />

•Dentin Regeneration<br />

•Periodontal Defects<br />

Combined growth factor treatment: OP-1 & IGF-1<br />

Primary normal chondrocytes<br />

Alginate<br />

beads<br />

Safranin O<br />

Type II Col<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

3


ACI chondrocytes<br />

Chubinskaya et al, 2008 JKS<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

•Adult male goats<br />

•14 days follow-up<br />

•histological &<br />

immunohistochemical<br />

analysis<br />

•3.2 mm Ø x 2 mm (deep)<br />

OCD, medial femoral<br />

condyle & trochlea<br />

•Treatment Groups<br />

•100 mg collagen<br />

putty + 250 µg OP-1<br />

•100 mg collagen<br />

putty<br />

•Empty defect<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Osteochondral Defect Model in Goats<br />

(Collaboration with Drs. Oakes and Hurtig)<br />

Day 0 Day 2<br />

Day 3 Day 5 Day 14<br />

Chubinskaya et al, 2004, ICRS<br />

• Adult male dogs<br />

• 6 and 12 weeks follow-up<br />

• Gross and histologic analysis<br />

• Bilateral, 5 mm (diameter) x<br />

5 mm (deep) osteochondral<br />

defects in the medial condyle<br />

• Treatment GGroups<br />

– 100 mg OP-1 collagen putty<br />

(250 µg OP-1)<br />

– 100 mg collagen putty<br />

Untreated<br />

(Collagen<br />

Putty)<br />

– Empty defect<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Cook et al. JBJS 2003, 85-<br />

A(Suppl 3):116-123<br />

Osteochondral Defect Model<br />

OP-1<br />

6 Weeks 12 Weeks<br />

Summary<br />

•No ossification in<br />

any joint;<br />

•Significant<br />

enhancement of both<br />

cartilage and bone<br />

repair in the defects;<br />

•Less fibrocartilage<br />

and an increase in<br />

hyaline cartilage.<br />

Day 0<br />

Placebo Control<br />

Staining with pro-OP-1 antibody<br />

Day 14<br />

LMC LTP LMC LTP<br />

Sport Fellows Course<br />

Carlsbad, 01-15-2011<br />

4


Mature OP OP-1 1<br />

staining<br />

• rhOP-1 remains within collagen<br />

vehicle for two weeks;<br />

• healing and bridging of the defect is<br />

initiated by day 5;<br />

• activation of endogenous OP-1;<br />

14 days post op + OP-1<br />

Chubinskaya et al, 2004, ICRS<br />

Pro-OP-1 staining<br />

• Autocrine and rhOP-1 contribute to<br />

the repair process suggesting that<br />

both have to be considered for growth<br />

Sport factors Fellows therapy Course<br />

Carlsbad, 01-16-2009 RMC LTP RTP<br />

Mosaicplasty Augmentation<br />

Autograft Plug Control at 12 weeks<br />

Autograft Plug with OP-1 Putty at 12 weeks<br />

OP-1 augments both cartilage and bone integration in mosaicplasty<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

• 9 adult sheep<br />

• 3,6 and 12 weeks follow-up<br />

• Bilateral model, two osteochondral<br />

defects in each medial femoral<br />

condyle<br />

• Donor site (5.5 mm x 10 mm) and<br />

mosaicplasty plug recipient site (5.4<br />

mm x 10 mm) on each condyle<br />

• Treatment groups<br />

– Donor hole filled with 75 mg OP-1<br />

collagen putty (200 µg OP-1)<br />

– Donor hole left empty<br />

– Recipient hole with mosaicplasty<br />

plug augmented with 75 mg OP-1<br />

collagen putty (200 µg OP-1)<br />

– Recipient hole with mosaicplasty<br />

plug only<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Shimmin et al. Trans ICRS 4 th<br />

Symposium, No. 16, 2002, Toronto<br />

• Experimental Procedure 24<br />

sheep (1year old, 60 kg)<br />

• Knee 10 mm chondral<br />

defects<br />

– Trochlea<br />

– Condyle<br />

• Continuous 2 week OP-1<br />

delivery via mini-osmotic<br />

mini osmotic<br />

pumps<br />

• Doses: 0, 55 μg and 170<br />

μg<br />

• Arthroscopic monitoring of<br />

repair<br />

• Sacrifice at 3 and 6 months<br />

Jelic et al (2001) Growth<br />

Factors 19:101-113<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Sheep Mosaicplasty<br />

Augmentation Study<br />

Mosaicplasty Augmentation<br />

Donor Holes-Histology at 12 weeks<br />

Untreated<br />

OP-1 Treated<br />

Chondral Defect Repair: OP-1 Sheep Model<br />

Histology, 6 mo<br />

Buffer Treated<br />

OP-1 Treated Summary:<br />

•Significant g cartilage g repair p<br />

was observed with OP-1<br />

delivered into the joint fluid;<br />

•No cartilage repair was<br />

observed in control defects<br />

without OP-1;<br />

•New cartilage is filled with<br />

chondrocytes embedded into<br />

Type II collagen;<br />

•New cartilage was well<br />

bonded with the old<br />

cartilage.<br />

5


Rabbit ACLT model: Protective effect of OP-1<br />

on cartilage in the development of OA<br />

Badlani et al, 2008 OA&C<br />

India Ink<br />

Hayashi et al. Arth Res & Ther 2008<br />

Mini-osmotic pump delivery<br />

Injections of 5% lactose-buffered<br />

rhBMP-7 in PBS. Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

OP-1 PREVENTS DEGENERATIVE CHANGES IN A<br />

CONTUSSIVE IMPACT MODEL OF OA IN SHEEP<br />

Mark Hurtig, Susan Chubinskaya, Jim Dickey, David Rueger<br />

Is OP-1 as a Osteoarthritis-Modifying Peptide?<br />

Time=0 bilateral impact injuries (30 MPa, 6mm diameter x2 )<br />

Time=0 Vehicle control OP-1 Implant injection (340 µg) i.a. injection<br />

Time=1 week Vehicle control OP-1 Implant injection (340 µg) i.a injection<br />

Time=12 weeks Sacrifice<br />

Results: Results<br />

OP OP-1 suppressed leukocyte influx in acute phase<br />

OP OP-1 preserved sGAG content of cartilage<br />

OP OP-1 improved (P< (P


India ink stained area for the medial femoral condyle<br />

Group A Group B Group C<br />

OP-1 Vehicle OP-1 Vehicle OP-1 Vehicle<br />

Mean 11.1 44.8 15.7 30.0 30.0 28.0<br />

T-test p


μCT of impacted cartilage explants<br />

Histology Safranin O staining<br />

Disc degeneration model<br />

Untreated cntr Compressed disc ROP-1<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Sham COP-1<br />

Chubinskaya et al, JOR 2007<br />

Kawakami et al, 2005; 2007<br />

Bajaj et al, ORS<br />

2011<br />

Attenuation levels<br />

at varying depths.<br />

• OP-1 injection induces:<br />

• anabolic<br />

– restoration of the<br />

ECM;<br />

– stimulation of OP-1<br />

synthesis;<br />

• anti-catabolic effects<br />

– downregulates:<br />

• Aggrecanase<br />

• Substance P<br />

• MMP-13<br />

• Interleukin-1β<br />

• TNF-α<br />

Degenerative disc with annular tear<br />

or Contained herniated disc<br />

Intervertebral Disc Degeneration Models<br />

The compressed NP<br />

OP OP-1 OP OP-1 1 injection injection Insertion of 0.8 mm K-wire<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Spring<br />

HUMAN EXPERIENCE WITH INTRA-<br />

ARTICULAR OP-1<br />

more than 50 cases, up to 10 years follow up<br />

Example: 41 yo football player<br />

TIRM, Regensburg, November 2010<br />

Osteochondral fragment retrieved from the joint space<br />

Courtesy of Dr. Shimmin<br />

8


OP-1 OP 1 putty was placed beneath the defect<br />

12 MONTH ARTHROSCOPY<br />

•OP-1 has not done any harm;<br />

•Assisted fixation of the<br />

fragment;<br />

•Improved integration of the<br />

fragment to the surrounding<br />

normal cartilage<br />

Overall Conclusions<br />

• Application of a single growth factor or a combination<br />

of those is a promising approach in cartilage repair<br />

• OP-1 is definitely one of the best candidates so far<br />

for cartilage repair and showed similar efficacy in<br />

various in vivo and ex vivo models, both animal and<br />

human cartilage<br />

• Traditional approaches pp ( (allograft, g , autograft, g , ACI) )<br />

might be optimized by addition of growth factors<br />

• Combination of growth factors and other anticatabolic<br />

agents of various mode of action has a<br />

potential in cartilage repair<br />

• Activation of autocrine anabolic pathways has to be<br />

taken in consideration in designing growth factor<br />

therapy.<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

Therapeutic Application of OP-1<br />

Clinical Studies<br />

• Phase 1, double-blind, randomized, multicenter, placebo-controlled,<br />

single-dose escalation safety study in patients with knee OA<br />

OARSI responder criteria<br />

•All subjects completed the study<br />

•OP-1 groups had more responders<br />

with OARSI criteria than placebo<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

A trend toward greater<br />

symptomatic improvement<br />

than placebo<br />

A higher frequency of<br />

injection site pain<br />

Hunter et al, 2010<br />

Challenges of Growth Factors Therapy<br />

• Which one or which ones?<br />

• Alone or in combination with other agents of a<br />

different mode of action<br />

• Formulations<br />

• Scaffolds/Carriers<br />

• DDoses<br />

• Treatment regimes<br />

• Delivery methods: local vs systemic vs gene<br />

• Time of Intervention<br />

• Possible adverse effects<br />

Sport Fellows Course<br />

Carlsbad, 01-28-2012<br />

9


Charities<br />

giftofhope<br />

Organ&Tissue<br />

Donor Network<br />

TIRM, Regensburg, November Donor’s 2010Families<br />

Collaborators:<br />

Rueger D<br />

Hurtig M<br />

Kildey R<br />

Cole B<br />

Wimmer M<br />

Oegema T<br />

Oakes B<br />

Loeser R<br />

Chubinskaya’s<br />

Chubinskaya s<br />

lab:<br />

Lidder S<br />

Pascual Garrido C<br />

Hakimiyan A<br />

Rappoport L<br />

Yanke A<br />

Segalite D<br />

Stryker Biotech<br />

Bajaj S<br />

Kirk S<br />

Ruta D<br />

Ciba-Geigy Endowed Chair Meininger A<br />

Drs. Cole and Galante Research<br />

Funding<br />

10


Hip the New Frontier: Cartilage and Labral Considerations<br />

International Sports Medicine Fellowship Course<br />

Carlsbad, CA<br />

2012<br />

Michael Gerhardt, MD<br />

Santa Monica Orthopaedic and Sports Medicine Foundation<br />

2020 Santa Monica Blvd, 4 th Floor<br />

Santa Monica, CA 90404<br />

mgerhardt@smog-ortho.net<br />

I. Current Indications for Hip Arthroscopy (1)<br />

a. FAI – accounts for majority of arthroscopic procedures today<br />

i. Labral debridement/repair<br />

ii. Chondroplasty<br />

iii. Osteoplasty – femoroplasty and acetabuloplasty<br />

b. Ligamentum teres injuries (2)<br />

c. Snapping Hip (3-4)<br />

i. Internal<br />

1. Snapping Iliopsoas tendon<br />

ii. External<br />

1. Snapping iliotibial band<br />

2. Snapping glut max<br />

d. Loose Bodies<br />

e. Synovial disease<br />

f. Peritrochanteric disorders (5, 6)<br />

i. Trochanteric bursitis<br />

ii. Abductor injuries of the hip<br />

g. Instability (7)<br />

h. Joint Sepsis<br />

i. Early OA?<br />

j. List is expanding rapidly<br />

i. Subgluteal syndrome (8,9)<br />

ii. Proximal Hamstring tendon injuries<br />

iii. Osteitis pubis?<br />

II. Labral Injuries – Beyond FAI<br />

a. Function of the Labrum<br />

i. Maintains continual contact on femoral head<br />

ii. Augments stability of the joint (10,11)<br />

1. Provides suction seal of femoracetabular joint


2. Maintains negative intrarticular pressure – resists<br />

distractive forces<br />

iii. Disperses Contact stress during weight bearing<br />

iv. Synovial fluid distribution<br />

v. Proprioceptive function<br />

b. Management options for labral injuries (12)<br />

i. Debridement (13, 16)<br />

ii. Labral Repair (14)<br />

1. Primary repair of labral tears<br />

a. Only 4% labral tears amenable to primary repair<br />

(Gerhardt, 2010)<br />

b. Majority of “labral repair” is actually LABRAL<br />

REFIXATION<br />

iii. Labral Reconstruction (15-16)<br />

1. Theory<br />

a. Labrum plays critical role in normal mechanics<br />

of hip<br />

b. Labral deficiency leads to internal derangement<br />

i. Microinstability<br />

ii. Abnormal mechanics of femoroacetabular<br />

joint<br />

iii. Joint preservation depends on labral<br />

integrity<br />

2. Indications<br />

a. Segmental labral deficiency<br />

b. Minimal existing chondral damage<br />

c. Favorable biological and mechanical<br />

environment<br />

3. Clinical studies<br />

a. Literature review<br />

4. Options<br />

a. Autograft<br />

i. Iliotibial band<br />

ii. Gracilis (distal)<br />

iii. Ligamentum teres<br />

b. Allograft<br />

i. Soft tissue donor sites – Hamstring, post<br />

tib, etc<br />

c. Bioscaffolds?<br />

III. Chondral Injuries (16-19)<br />

a. Femoral Head Lesions<br />

i. Types of FH chondral injuries<br />

1. Primary chondral injury<br />

2. Osteochondritis Dessicans (OCD) - rare<br />

3. Lateral Impact Syndrome (20)<br />

4. AVN?


REFERENCES<br />

ii. Management of FH chondral injuries<br />

1. Microfracture<br />

2. Repair?<br />

3. Resurfacing<br />

a. Biologic options<br />

b. Arthroplasty options<br />

b. Acetabular Lesions<br />

i. Types of Acetabular chondral injuries<br />

1. Primary articular cartilage injury<br />

2. Osteochondritis Dessicans (OCD) – very rare!<br />

ii. Management options of acetabular chondral injuries (21-22)<br />

1. Microfracture<br />

2. Repair of “delamination” injuries<br />

a. Suture repair (23)<br />

b. Fibrin Glue augmentation (24)<br />

c. Resection and microfracture (25-26)<br />

d. Microfracture and repair of cartilage flap<br />

3. Resurfacing options (27)<br />

a. ACI<br />

b. Scaffolds<br />

c. OATS<br />

4. Arthroplasty<br />

a. Options<br />

i. Partial – Hemi caps<br />

ii. Hip Resurfacing<br />

iii. Total Hip Replacement<br />

1. Stevens MS, Legay DA, Glazebrook MA, Amirault D. The Evidence for hip<br />

arthroscopy: grading the current indications. Arthroscopy. 2010 Oct;26(10):1370-83<br />

2. Byrd JW, Jones, KS. Traumatic rupture of the ligamentum teres as a source of hip<br />

pain. Arthroscopy. 2004 Apr;20(4):385-91.<br />

3. Ilizaturri VM, Chaidez C, Villegas P, Briseno A, Camacho-Galindo J. Prospective<br />

randomized study of 2 different techniques for endoscopic iliopsoas tendon release in the<br />

treatment of internal snapping hip syndrome. Arthroscopy. 2009 Feb;25(2):159-63. Epub<br />

2008 Oct 10.


4. Voos JE, Rudzki JR, Shindle MK, Martin H, Kelly BT. Arthroscopic anatomy and<br />

surgical techniques for peritrochanteric space disorders in the hip. Arthroscopy. 2007<br />

Nov;23(11):1246.e1-5.<br />

5 Voos JE, Shindle MK, Pruett A, Asnis PD, Kelly BT, Endoscopic Repair of Gluteus<br />

Medius Tendon Tears of the Hip.Am J Sports Med. 2009 Feb 9.<br />

6 Robertson WJ, Gardner MJ, Barker JU, Boraiah S, Lorich DG, Kelly BT. Anatomy and<br />

dimensions of the gluteus medius tendon insertion. Arthroscopy. Feb;24(2):130-6, 2008.<br />

7. Phillippon MJ, Kuppersmith DA, Wolff AB, Briggs KK. Arthroscopic findings<br />

following traumatic hip dislocation in 14 professional athletes. Arthroscopy. 2009<br />

Feb;25(2):169-74.<br />

8. Martin H, Shears S, Palmer I. Evaluation of the Hip. Sports Medicine & Arthroscopy<br />

Review: June 2010;18(2):63-75<br />

9. Martin H. The Endoscopic treatment of sciatic nerve entrapment/deep gluteal<br />

syndrome. Presented at ISHA 2010 Oct 8.<br />

10. Ferguson SJ, Bryant JT, Ganz R, Ito K. The influence of the acetabular labrum on<br />

hip joint cartilage consolidation: a poroelastic finite element model. Journal of<br />

Biomechanics. 2000 Aug;33(8):953-60.<br />

11. Ferguson SJ, Bryant JT, Ganz R, Ito K. An invitro investigation of the acetabular<br />

labral seal in hip joint mechanics. Journal of Biomechanics. 2003;36:171-8.<br />

12. Kelly BT, Weiland DE, Schenker ML, Philippon MJ. Arthroscopic labral repair in the<br />

hip: surgical technique and review of the literature. Arthroscopy. 2006;21:1496-504.<br />

13. Santori N, Villar RN. Acetabular labral tears: result of arthroscopic partial<br />

limbectomy. Arthroscopy. 2000 Jan-Feb;16(1):11-15<br />

14. Larson CM, Giveans MR. Arthroscopic debridement versus refixation of the<br />

acetabular labrum associated with femoroacetabular impingement. Arthroscopy 2009<br />

Apr;25(4):369-76.<br />

15. Phillipon MJ, Briggs KK, Hay CJ, Kuppersmith DA, Dewing CB, Huang MJ.<br />

Arthroscopic labral reconstruction in the hip using iliotibial band autograft: technique and<br />

early outcomes. Arthroscopy 2010 Jun;26(6):750-6.<br />

16. Byrd JW, Jones KS. Hip arthroscopy for labral pathology: prospective analysis with<br />

10-year follow-up. Arthroscopy. 2009 Apr;25(4):365-8.<br />

17. Byrd JW, Jones KS. Prospective analysis of hip arthroscopy with 10-year followup.<br />

CORR. 2010 Mar;468(3):741-6.


18. Philippon MJ, Schenker M, Briggs K, Kuppersmith D. Femoroacetabular<br />

impingement in 45 professional athletes: associated pathologies and return to sport<br />

following arthroscopic decompression. Knee Surgery Sports Traumatology Arthroscopy.<br />

2007 Jul;15(7):908-14.<br />

19. Singh PJ, O’Donnell JM. The outcome of hip arthroscopy in Australian football<br />

league players: a review of 27 hips. Arthroscopy. 2010 Jun;26(6):743-9.<br />

20. Byrd JW. Lateral impact injury: A source of hip pathology. Clin Sports Med. 2001<br />

Oct;20(4):801-15.<br />

21. Beaule PE, O’Neill M, Rakhra. Acetabular labral tears. JBJS 2009 Mar 1;91(3):701-<br />

10.<br />

22. Gdalvitch M, Smith K, Tanzer M. Delamination cysts: a predictor of acetabular<br />

cartilage delamination in hips with a labral tear. CORR. 2009 Apr;467(4):985-91.<br />

23. Sekiya JK, Martin RL, Lesniak BP. Arthroscopic Repair of Delaminated Acetabular<br />

Articular Cartilage in Femoroacetabular Impingement. Orthopedics. September<br />

2009;32(9):692.<br />

24. Tzaveas AP, Villar RN. Arthroscopic repair of acetabular chondral delamination<br />

with fibrin adhesive. Hip International. 2010 Jan-Mar;20(1):115-9.<br />

25. Philippon MJ, Schenker ML, Briggs KK, Maxwell RB. Can microfracture produce<br />

repair tissue in acetabular chondral defects? Arthroscopy. 2008 Jan;24(1):46-50.<br />

26. Crawford K, Philippon MJ, Sekiya JK, Rodkey WG, Steadman JR. Microfracture of<br />

the hip in athletes. Clin Sports Med. 2006 Apr;25(2):327-35<br />

27. Ellander P, Minas T. Autologous Chondrocyte Implantation in the hip: Case report<br />

and technique. Operative Techniques in Sports Medicine. 2008 Oct;16(4)201-6


12th International Sports Medicine Fellows Conference<br />

Comprehensive approaches to articular cartilage repair and hip arthroscopy<br />

Carlsbad, CA January 27-29, 2012<br />

Product Challenges for Partial Thickness Articular Cartilage Treatment<br />

Wayne K. Augé II, MD<br />

Department of Research and Development<br />

NuOrtho Surgical, Inc. at the<br />

Advanced Technology & Manufacturing Center<br />

University of Massachusetts Dartmouth<br />

This presentation will focus on the treatment of partial thickness articular cartilage lesions with<br />

an emphasis on the challenges these lesions create for therapeutic product development.<br />

Traditionally, surgical intervention has been limited to palliative tissue removal because of<br />

volumetric and functional over-resection that enlarge lesion size and eliminate surrounding<br />

healing phenotypes. Viewing partial-thickness lesions as a wound healing problem has enabled<br />

new therapeutic approaches which have been designed to decrease the disease burden associated<br />

with these lesions.


Reimbursement and Coding<br />

Issues for Articular<br />

Cartilage Treatment<br />

Ralph A. Gambardella, M.D.<br />

Kerlan Kerlan-Jobe Jobe Ortho Clinic<br />

Carlsbad Cartilage Course 2011<br />

Linking Industry Practices<br />

and Technologies<br />

Experience gained<br />

from engagements<br />

with other<br />

Orthopaedic<br />

practices,<br />

Ambulatory Surgery<br />

Centers, and<br />

Hospitals<br />

How do we identify pilot opportunities?<br />

Potential<br />

Pilots for the<br />

COE<br />

Internal J&J Competencies<br />

COE Outputs<br />

•Strategic Partnerships with Technology Vendors<br />

•Research papers with repeatable findings for Alumni<br />

Ideas and<br />

concepts gained<br />

from knowledge<br />

partners<br />

(Gartner) and<br />

participation in<br />

industry<br />

technology<br />

summits<br />

Business Center of<br />

Excellence (COE) Concept<br />

The Business COE provides a<br />

mechanism for experimenting p gwith<br />

business concepts and technology to<br />

evaluate the potential for improving the<br />

performance of Orthopaedic practices<br />

SUGICAL<br />

REIMBURSEMENT<br />

� Accounts Receivable Duration<br />

– Driven by y ‘controllable’ activities<br />

� Creating appropriate coding supported by<br />

appropriate documentation<br />

� Time between surgery and submission of a ‘clean<br />

claim’<br />

– Driven by ‘uncontrollable’ activities<br />

� Time between submission of ‘clean claim’ and<br />

receipt of reimbursement<br />

1


TOTALCHART<br />

TECHNOLOGY<br />

A SURGEON’S SURGEON S PARTNER<br />

Enhance accuracy<br />

Enhance Reimbursement<br />

Pilot Timing and Metrics<br />

The cycle time data utilized in this presentation was<br />

captured for Dr. Gambardella during three distinct<br />

periods:<br />

Pilot Purpose<br />

The technology introduced in this study enables<br />

the surgeon to code and dictate via a handheld<br />

device and includes coding logic to ensure the<br />

appropriate use of codes and modifiers modifiers.<br />

This analysis focused on two questions:<br />

1. Will the introduction of the handheld device<br />

impact administrative cycle times?<br />

2. Will these cycle times change as a surgeon<br />

becomes more familiar with the technology?<br />

Summary of Findings – Faster<br />

Claim Submission<br />

Duration between surgery and claim submission<br />

� Manual: 9.9 Days<br />

� Technology enabled: 7.4 Days (a 25% reduction)<br />

Percent of claims submitted with 10 days<br />

� Manual: 72.0%<br />

� Technology enabled: 86.6% (a 20%<br />

improvement)<br />

Percent of claims submitted within one week<br />

� Manual: 49.5%<br />

� Technology enabled: 81.8% (a 65%<br />

improvement)<br />

2


Summary of Findings – Faster<br />

Claim Submission<br />

Duration between surgery and claim submission<br />

� Manual: 21.2 Days<br />

� Technology enabled: 14.0 Days (a 34%<br />

reduction)<br />

Percent of claims submitted with 10 days<br />

� Manual: 59.2%<br />

� Technology enabled: 70.8% (a 12%<br />

improvement)<br />

Percent of claims submitted within one week<br />

� Manual: 32.7%<br />

� Technology enabled: 50.8% (a 80%<br />

improvement)<br />

Summary of Findings – Faster<br />

Reimbursement<br />

Duration between claim submission and<br />

reimbursement<br />

� Manual: 51.2 Days<br />

� Technology enabled: 32.5 Days (a 36%<br />

reduction)<br />

Percent of reimbursements received within 40<br />

days<br />

� Manual: 40.8%<br />

� Technology enabled: 63.1% (a 36%<br />

improvement)<br />

Summary of Findings – Faster<br />

Reimbursement<br />

Duration between claim submission and<br />

reimbursement<br />

� Manual: 62.8 Days<br />

� Technology enabled: 43.7 Days (a 30%<br />

reduction)<br />

Percent of reimbursements received within 40<br />

days<br />

� Manual: 44.1%<br />

� Technology enabled: 49.2% (a 11%<br />

improvement)<br />

Summary of Findings – Fewer<br />

Past Due Accounts<br />

Percentage of claims with reimbursement<br />

cycle > 90 days<br />

�� Manual: 31.2% 31 2%<br />

� Technology enabled: 14.4% (a 54%<br />

reduction)<br />

3


Summary of Findings – Fewer<br />

Past Due Accounts<br />

Percentage of claims with reimbursement<br />

cycle > 90 days<br />

�� Manual: 30.6% 30 6%<br />

� Technology enabled: 18.5% (a 45%<br />

reduction)<br />

ARTICULAR CARTILAGE<br />

SURGICAL CODES<br />

� CPT 29886 OCD drill intact lesion<br />

� CPT 29887 OCD intact lesion with internal<br />

fixation<br />

� CPT 29885 OCD with bone graft with or<br />

without internal fixation<br />

ARTICULAR CARTILAGE<br />

SURGICAL CODES<br />

� CPT 29877 Chondroplasty/Debridement<br />

� CPT 29874 Removal of Loose Body<br />

� CPT 29879 Microfracture<br />

� CPT 29866 Arthroscopic Osteochondral Autograft<br />

� CPT 29867 Arthroscopic Osteochondral Allograft<br />

� CPT 27415 Open Osteochondral Allograft<br />

� CPT 27416 Open Osteochonral Autograft<br />

� CPT 29870 Genzyme Chondrocyte Biopsy<br />

ARTICULAR CARTILAGE<br />

SURGICAL CODES<br />

� CPT 27412 Autologous Chondrocyte<br />

Implantation<br />

�� CPT 29868 Meniscal Transplantation<br />

� CPT 27457 Osteotomy after epiphyseal<br />

closure<br />

� CPT 27418 Anterior Tibial Tubercleplasty<br />

� CPT 29870 Arthroscopy with biopsy<br />

4


ARTICULAR CARTILAGE<br />

SURGICAL CODES<br />

� CPT 27442 Arthrosurface Trochlea<br />

� CPT 27438 Arthrosurface Patella<br />

�� CPT 27446 Unicompartmental Knee<br />

ARTICULAR CARTILAGE<br />

CODES<br />

� Modifiers<br />

59 is used to identify a distinct procedural service<br />

Separate p incision or lesion<br />

Separate compartment<br />

Separate injury<br />

22 is for extraordinary situation ( ie,obesity)<br />

Must document with cover letter<br />

ARTICULAR CARTILAGE<br />

ICD ICD-9 9 CODES<br />

� 717.9 Unspecified internal derangement<br />

� 719.96 Unspecified disorder of joint<br />

�� 732 732.7 7 Osteochonditis Dissecans<br />

� 733.90 Disorder of bone and cartilage<br />

� 733.92 Chondromalacia<br />

� 836.0 Tear medial meniscus<br />

� 836.1 Tear lateral meniscus<br />

ARTICULAR CARTILAGE<br />

REIMBURSEMENT<br />

� Hospital or Surgicenter Problems<br />

– Gl Global b l CContract t t ffor services i<br />

�Differs for different payors<br />

�Differs for different geographic<br />

areas<br />

5


ARTICULAR CARTILAGE<br />

REIMBURSEMENT<br />

� Surgical Implant and Disposable Costs<br />

– Shaver ( $40-100 )<br />

– RF Device ( $112-185 )<br />

– Osteochondral autograft kit ( $380-618 )<br />

– Osteochondral OBI kit ($225 )<br />

– Osteochondral allograft kit (Rental)<br />

– Chondral pins/screws ( $695 )<br />

– Meniscal repair implants ( $95-220 )<br />

ARTICULAR CARTILAGE<br />

REIMBURSEMENT<br />

� DME’s<br />

– Braces<br />

– Cold Therapy<br />

– Pain pumps<br />

� Viscosupplementation<br />

� Medications<br />

� Physical Therapy<br />

ARTICULAR CARTILAGE<br />

REIMBURSEMENT<br />

� IMPLANT MATERIALS<br />

� Osteochondral allografts ($9,950)<br />

� OBI Plugs ($500)<br />

� Meniscal allografts ($4,750)<br />

� ACL allografts ($2,500)<br />

� Osteotomy implants ($795)<br />

� Cell implants<br />

– Carticel ($20,000)<br />

� OP-1<br />

CASE 1<br />

� Chondroplasty Patella $400<br />

� MFC defect (3.0cm) $1,500<br />

�� Meniscal Repair $440<br />

� ACL Allograft $2,500 & $500<br />

TOTAL $6,340<br />

6


CASE 2<br />

� Chondroplasty Patella $400<br />

� ACI MFC $20,000<br />

�� Meniscal Transplant $4,750 $4 750 & $1100<br />

� ACL Allograft $2500 & $500<br />

TOTAL $29,250<br />

CONCLUSIONS<br />

� Get involved early in your career with the<br />

business end of what you do<br />

� Be a leader and embrace industry as your<br />

� Be a leader and embrace industry as your<br />

partner to help improve your practice and<br />

improve your patients outcomes<br />

CONCLUSIONS<br />

Impact to Overall A/R Cycle<br />

�RAG : 30% improvement<br />

– 72.8 days Manual<br />

– 51 51.2 2 d days w/Tech /T h<br />

�JTD : 35.8% improvement<br />

– 72.5 days Manual<br />

– 46.5 days w/Tech<br />

7


General Criteria<br />

for<br />

Cartilage Repair<br />

� Age 15-50 years<br />

�� Di Disabling bli llocalized li d kknee pain i ffor six i months th<br />

which has failed conservative treatment<br />

� An intact meniscus is present<br />

� Lesion must be discrete, single and unipolar<br />

� Lesion is largely contained with near normal<br />

surrounding cartilage (Gr 0,1,2)<br />

General Criteria<br />

for<br />

Cartilage Repair<br />

�� No history of cancer in bones fat or<br />

muscle of affected limb<br />

� Body Mass Index (BMI) of < or = to<br />

30<br />

General Criteria<br />

for<br />

Cartilage Repair<br />

� A normal joint space is present<br />

� No active infection<br />

� No osteoarthritis or inflammation<br />

� Knee is stable with normal mechanical<br />

alignment<br />

� Patient can comply with post-operative<br />

restrictions for wt. bearing<br />

ACI Criteria<br />

� Inadequate response to prior surgery<br />

� Defect size 1.5-10cm sq<br />

�� Cartilage only defect<br />

� No allergy to gentamicin<br />

� Focal, full thickness isolated to<br />

MFC,LFC trochlea<br />

� Defect from acute or repetitive trauma<br />

� ALL General Criteria met<br />

8


Osteochondral<br />

Autograft Criteria<br />

� Arthroscopic examination<br />

� Defect size between 1.0 to 2.5cm sq<br />

� Focal, full thickness isolated to<br />

MFC,LFC or trochlea<br />

� Defect from acute or repetitive<br />

trauma<br />

� ALL General Criteria met<br />

Investigational<br />

Procedures<br />

� ALL procedures on joints other than<br />

the knee<br />

� ANY procedure d th that t ddoes not t meet t<br />

ALL of the criteria<br />

� ALL use of non-autologous synthetic<br />

bone filler materials<br />

� ALL use of minced cartilage<br />

Osteochondral<br />

Allograft Criteria<br />

� Arthroscopic examination<br />

�� Defect size = or > 22.0cm 0cm sq<br />

� Focal, full thickness isolated to<br />

MFC,LFC or trochlea<br />

� Defect from acute or repetitive<br />

trauma<br />

� ALL General Criteria met<br />

9


12 th International Sports Medicine Fellows Conference<br />

January 27-29, 2012● Carlsbad, California<br />

7:30 AM - 7:35 AM Introduction<br />

Session IV: Hip Arthroscopy<br />

7:35 AM - 7:45 AM The Basics - Indications Michael Gerhardt, MD USA<br />

7:45 AM - 8:00 AM<br />

8:00 AM - 8:15 AM<br />

8:15 AM - 8:30 AM<br />

How To Get Started - Patient Assessment, Treatment<br />

Algorithm<br />

ABC's in the OR - Patient Set-Up, Positioning, Central and<br />

Peripheral Compartment Access and Portal Placement<br />

FAI - Making the Diagnosis, Imaging, Management and<br />

Surgical Treatment<br />

Warren Kramer, MD USA<br />

J.W. Thomas Byrd, MD USA<br />

Mark Safran, MD USA<br />

8:30 AM - 8:40 AM Sports Hernia – Diagnosis, Algorithm and Surgical Treatment William Hutchinson, MD USA<br />

8:40 AM - 8:55 AM<br />

8:55 AM - 9:10 AM<br />

Case Presentation - Hip and Groin Injury in the Professional<br />

Athlete<br />

Hip Arthroscopy - Where Have We Been, Where Are We Now<br />

and Where Are We Going?<br />

Michael Gerhardt, MD USA,<br />

Mark Safran, MD USA,<br />

Warren Kramer, MD USA<br />

J.W. Thomas Byrd, MD USA


INTERNATIONAL SPORTS<br />

MEDICINE FELLOWSHIP<br />

SYMPOSIUM 2012<br />

Michael B. Gerhardt, MD<br />

Intrarticular Hip Disorders<br />

�� Commonly misdiagnosed<br />

�� Traditionally athletes told “live with it”<br />

�� Hip Arthroscopy now offers a<br />

minimally invasive treatment option !<br />

Assessment<br />

Assessment<br />

Is the problem truly<br />

the hip joint???<br />

Confounding disorders<br />

• Sports hernia/<br />

hernia/<br />

Pubalgia<br />

• Lumbar disease<br />

• Extrarticular<br />

disorders:<br />

bursitis, tendonitis,<br />

etc<br />

INDICATIONS<br />

Overview of Common<br />

Hip p Injuries j<br />

Michael B. Gerhardt, MD<br />

Team Physician, LA Galaxy and Chivas USA<br />

Team Physician, US Soccer<br />

Assessment<br />

Presentation<br />

�� History of trauma variable<br />

�� Mechanical symptoms common<br />

�� Exacerbating features<br />

1


Favorable Prognostic Indicators<br />

�� History of significant traumatic event<br />

�� Mechanical symptoms<br />

• Intermittent pain/catching<br />

• Sharp or stabbing pain<br />

Current Indications<br />

“The “The big big 3” 3”<br />

�� Labral Pathology<br />

�� Chondral Damage<br />

�� FAI<br />

Current Current Indications<br />

Indications<br />

�� Ligamentum teres rupture<br />

�� Loose Bodies<br />

�� Degenerative Arthritis?<br />

SALT Lesion<br />

Lesion<br />

(Superior Acetab Labral Tear)<br />

Common Lesions of the Hip<br />

Recently…<br />

indications have<br />

expanded…<br />

and the list is<br />

growing rapidly!<br />

Current Current Indications<br />

Indications<br />

�� Synovial disease / Rheumatologic<br />

�� Joint sepsis<br />

2


Current Current Indications<br />

Indications<br />

Capsular laxity / Instability<br />

-Microinstability<br />

Microinstability<br />

-Macroinstability<br />

Macroinstabilityy<br />

Current Current Indications<br />

Indications<br />

The Peritrochanteric Space<br />

�� Trochanteric bursitis<br />

�� Snapping IT band<br />

• The The External External Snapper Snapper<br />

�� Abductor tears – Glut Medius Tears<br />

“Rotator “Rotator Cuff Cuff Tears Tears of of the the Hip” Hip”<br />

Most Common Lesions of the Hip<br />

�� Labral Pathology<br />

�� Articular cartilage injury<br />

�� Femoroacetabular Impingement (FAI)<br />

�� Ligamentum teres rupture<br />

�� Degenerative disease<br />

Current Current Indications<br />

Indications<br />

�� Diagnostic – Unresolved hip pain<br />

�� �� AVN?<br />

AVN?<br />

�� Snapping hip<br />

• Internal snapper – ILIOPSOAS TENDON<br />

Relative Relative Contraindications Contraindications to to<br />

Hip Hip Arthroscopy<br />

Arthroscopy<br />

�� Severe degenerative arthritis<br />

�� Stress Riser – impending fracture<br />

�� Severe obesity<br />

Labral Labral pathology pathology<br />

Etiology<br />

• Most common injury in athletes<br />

• Twisting mechanism common<br />

�� Certain sports pre-dispose<br />

pre dispose<br />

• Macrotrauma ( (tackle tackle) ) vs. Microtrauma<br />

(FAI FAI)<br />

3


Labral Labral pathology pathology<br />

Natural History<br />

• Unknown<br />

• Do labral tears heal?<br />

Labral Labral pathology pathology<br />

�� �� Uncertain<br />

Uncertain<br />

�� Some patients improve with conservative<br />

treatment<br />

Arthroscopic Treatment<br />

• Labral debridement vs repair???<br />

�� �� Labral tissue contains important nociceptive<br />

and neural mechanoreceptors critical to joint<br />

�� Johnson Johnson & & Gerhardt, Gerhardt, 2010 2010<br />

Labral Repair Technique<br />

Hip Specific<br />

Implants,<br />

Instrumentation<br />

and Techniques<br />

rapidly improving!<br />

Cinch Stitch for PushLock use in the hip<br />

Labral Labral pathology pathology<br />

Arthroscopic Treatment<br />

• Goal of surgery<br />

Labral Labral pathology pathology<br />

�� Resect or repair damaged tissue<br />

�� Create stable transition zone at labral-<br />

cartilage junction<br />

�� Preserve as much normal tissue as possible<br />

Arthroscopic Treatment<br />

•Labral Labral debridement vs repair repair???<br />

???<br />

�� Repair is possible and preferable<br />

�� �� Most lesions not repairable<br />

�� Awaiting studies to assess success of<br />

repaired labrum<br />

�� Repair is critical if acetabular pincer lesion<br />

resected<br />

Will discuss controversies later in meeting!<br />

PEEK and Bio-Pushlock<br />

Bio Pushlock<br />

�� Hip specific push-<br />

locks<br />

(2.9 mm Bio,<br />

PEEK, , or<br />

Biocomposite)<br />

�� Pretension repair<br />

�� Knotless Option<br />

�� Good fixation!<br />

4


SutureTaks w/ FiberWire<br />

Bio, PEEK, or<br />

Biocomposite<br />

SutureTaks with<br />

FiberWire<br />

Cinch Stitch Repair<br />

Labral Base refixation Labral Base refixation<br />

Labral Base refixation<br />

Chondral Chondral Injuries Injuries<br />

�� Articular cartilage injuries can be<br />

devastating!<br />

�� Worst W WWorst t prognosis i<br />

�� MRI/MRA low sensitivity<br />

• Often Often not not diagnosed diagnosed until until arthroscopy<br />

arthroscopy<br />

5


Chondral Chondral Injuries Injuries<br />

Common locations<br />

• Femoral head – less common<br />

less common<br />

�� Lateral impact syndrome<br />

• Anterolateral acetabulum<br />

�� Common in impingement syndrome<br />

Chondral Chondral Injuries Injuries<br />

Lateral Impact Syndrome (Byrd, 2001)<br />

• Usually contact sports<br />

• Athlete lands directly on lateral hip<br />

�� �� Violent contact between femoral head and<br />

acetabulum<br />

�� Shear Shear force force creates creates medial medial femoral femoral head head<br />

articular articular cartilage cartilage injury! injury!<br />

Lateral Impact Syndrome Lateral Impact Syndrome<br />

Lateral Impact Syndrome<br />

Chondral Chondral Injuries Injuries<br />

Treatment<br />

• Debridement/chondroplasty<br />

• Microfracture<br />

�� Promising results acetabular defects (Byrd, 2006)<br />

• Resurfacing?<br />

R RResurfacing? f i ?<br />

�� Biologic – ACI?<br />

�� Non Non-biologic biologic<br />

• Arthrosurface?<br />

• Resurfacing Arthroplasties?<br />

• THR?<br />

6


Femoroacetabular Impingement<br />

(FAI)<br />

PINCER TYPE CAM TYPE<br />

Arthroscopic Rim<br />

Trim<br />

Ligamentum Ligamentum Teres Teres Injury Injury<br />

Ligamentum Ligamentum Teres Teres Injury Injury<br />

Acute tear<br />

Arthroscopic treatment<br />

• Debridement<br />

Femoroplasty<br />

�� Excellent results reported<br />

�� �� 96% improvement rate (Byrd 2004)<br />

• Repair – case report (Philippon, 2001)<br />

�� NOT recommended!<br />

Ligamentum Teres Injury<br />

�� Athletes can have acute tear<br />

�� Mechanism<br />

Mechanism<br />

• Twisting/rotating<br />

�� Symptoms<br />

• Vague, non specific groin pain<br />

•+/ +/- mechanical complaints<br />

�� Difficult diagnosis<br />

• Often not confirmed until arthroscopy<br />

Ligamentum Ligamentum Teres Teres Injury Injury<br />

Chronic tear<br />

Loose Bodies<br />

Clearest indication for hip arthroscopy<br />

• Important for future integrity of joint<br />

• Arthroscopic removal superior to open<br />

procedure<br />

7


Loose Loose Bodies Bodies<br />

�� Symptoms<br />

• Catching, locking, giving way<br />

�� Diagnosis<br />

• S Sometimes ti evident id t on plain l i xrays<br />

•MRI MRI<br />

• CT scan excellent for subtle<br />

osteochondral frags<br />

THANK YOU!<br />

Thank You!<br />

Loose Loose Bodies Bodies<br />

�� Always look in peripheral<br />

compartment!!<br />

�� Central compartment only allows<br />

ACCESS TO ~70% of the joint<br />

• Gerhardt, AAOS, 2006<br />

8


Case Report<br />

Warren G. Kramer III, MD<br />

� 23 year old female professional surfer with bilateral hip<br />

pain.<br />

� No specific injury but pain is made worse with surfing.<br />

patient complains of popping sensation in the groin.<br />

� Exam: +FAIR/FABER. +Pain with resisted straight leg raise<br />

(internal and external rotation).<br />

� Failed conservative care (PT, NSAIDs, Rest).<br />

� MRI and xrays were taken.<br />

� Labral tear<br />

Hip Injuries From Surfing<br />

� Pain secondary to FAI‐ CAM or PINCER Type<br />

� Snapping Psoas Tendon and/or Tendonitis<br />

� Proximal Hamstring Tear<br />

� Rectus Femoris Tear<br />

� Ligamentum Teres Tear<br />

� Avulsion of ASIS tensor Fascia Lata Sartorius<br />

� Sports Hernia<br />

� Trochanteric Bursitis<br />

� Osteitis Pubis<br />

� ericah.MOV<br />

Things to look out for…<br />

Clear Cell Chondrosarcoma<br />

Center Edge Angle 37 degrees (right hip)<br />

Center Edge Angle 40 degrees (left hip)<br />

1/23/2012<br />

1


Alpha Angle 95 degrees (right hip)<br />

Alpha Angle 95 degrees (left hip)<br />

(Subtle Anterior Prominence)<br />

Surgical photos of right hip<br />

with evidence of FAI and<br />

s/p osteoplasty<br />

Surgical photos of left hip<br />

with evidence of FAI and<br />

s/p osteoplasty<br />

MRI Left Hip<br />

(Labrum appears normal despite<br />

significant pain/limitation)<br />

MRI Left Hip<br />

(Labrum appears normal despite<br />

significant pain/limitation)<br />

Patient did well post op for 5 months but had<br />

recurrence of left hip groin pain…<br />

� NSAIDs were given, repeat MRI (possible labral re‐tear), U/S<br />

guided cortisone injection into the joint and psoas bursa<br />

� Patient reported some relief with intra‐articular joint/psoas<br />

bursa but continued to have pain that kept her from surfing.<br />

Surgical photos of right hip<br />

with labral tear and repair<br />

Surgical photos of left hip<br />

with labral tear and repair<br />

Surgical photos of left hip showing intact labral repair and<br />

thickened, yellow degeneration of psoas tendon<br />

1/23/2012<br />

2


Patient is 3 months post op and has<br />

returned to surfing without pain.<br />

� Pain after hip arthroscopy is not uncommon.<br />

� Is a psoas tendon release under or over utilized?<br />

� Does repairing the labrum or capsular release near the<br />

psoas cause adhesions around the psoas tendon?<br />

� Why would a 23 year old female have yellowish<br />

degeneration in her psoas tendon?<br />

Gracias!<br />

1/23/2012<br />

3


ABC's in the OR - Patient Set-Up, Positioning,<br />

Central and Peripheral Compartment Access and Portal Placement<br />

J. W. Thomas Byrd, M.D.<br />

info@nsmfoundation.org<br />

12th International Sports Medicine Fellows Conference<br />

Carlsbad, California<br />

January 29, 2012<br />

I. Equipment<br />

A. Fracture table<br />

- Tensiometer for monitoring traction forces intraoperatively is the most<br />

important modification<br />

B. C-arm image intensifier<br />

C. 70 and 30 video-articulated arthroscopes<br />

D. Fluid management system<br />

E. Specialized hip arthroscopy cannula system<br />

1. Extra length cannulas<br />

2. Shortened bridge accommodates extra length cannulas with standard<br />

arthroscope<br />

3. Cannulated obturators<br />

- Allows prepositioning with spinal needle. Cannula/obturator assembly<br />

can be passed over a guide wire initially placed through the needle<br />

F. Extra length slotted cannulas allow passage of curved shaver blades<br />

G. Extra length sturdy hand instruments<br />

- Avoid instruments designed for other endoscopic purposes that might be less<br />

sturdy and at greater risk of breakage<br />

H. Radiofrequency devices exhibit distinct advantages in the hip<br />

1. Maneuverability<br />

2. Ability to effectively ablate tissue despite limits of maneuverability<br />

II. Anesthesia<br />

- Typically performed under general anesthetic. Epidural anesthesia is an appropriate<br />

alternative but requires adequate block for assuring muscle relaxation.<br />

1


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

III. Patient Positioning<br />

A. Placed supine on the fracture table (Figure 1)<br />

B. Heavily padded perineal post, lateralized against the medial thigh of the operative leg<br />

(Figure 2)<br />

1. Lateralizing the post adds a slight transverse component to the traction vector<br />

(Figure 3)<br />

2. Also lessens the likelihood of compression and possible neuropraxia of the<br />

pudendal nerve<br />

C. Operative hip positioned in extension, approximately 25 of abduction, and neutral<br />

rotation<br />

1. Slight flexion might relax the capsule and facilitate distraction, but can place<br />

more traction on the sciatic nerve and draw it closer to the joint, making it<br />

more vulnerable to injury. Thus, significant flexion is avoided during<br />

arthroscopy.<br />

2. Neutral rotation is important during portal placement (Figure 4) although<br />

freedom of rotation during arthroscopy can facilitate visualization.<br />

D. The contralateral extremity is abducted as necessary to accommodate positioning of<br />

the image intensifier between the legs.<br />

- Prior to applying traction to the operative leg, counter force is created by<br />

placing the contralateral extremity under light traction. This stabilizes the<br />

pelvis so that it does not shift as traction is gradually applied to the operative<br />

extremity.<br />

E. Traction is applied to the operative extremity and distraction of the joint confirmed<br />

by fluoroscopy.<br />

1. Typically about 50 pounds is applied. More force may be necessary for a tight<br />

joint, but should be undertaken with caution.<br />

2. Initially, adequate distraction (8-10 mm) may not be readily achieved.<br />

a) Allowing a few minutes for the capsule to accommodate to the tensile<br />

forces often results in relaxation of the capsule (physiologic creep) and<br />

adequate distraction without excessive force.<br />

b) Also, a vacuum phenomenon created by the capsular seal will later be<br />

released when the spinal needle is introduced and the joint is distended<br />

with fluid, which may further facilitate distraction.<br />

F. After confirming the ability to distract the joint, the traction is released until ready to<br />

begin the surgical procedure.<br />

G. Peripheral compartment<br />

1. Inspected after arthroscopy of intraarticular compartment<br />

2. Traction released & hip flexed 45º<br />

- Relaxes capsule & allows access to peripheral compartment<br />

3. Especially useful for loose bodies, synovial disease, & thermal capsulorrhaphy


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

Figure1<br />

Figure 3<br />

Figure 4<br />

Figure 2


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

IV. Portals<br />

- The three standard portals are:<br />

Anterior, Anterolateral, and<br />

Posterolateral (Figures 5 & 6).<br />

Figure 5<br />

Table 1<br />

Distance from Portal to Anatomic Structures<br />

(Based on Anatomic Dissection of Portal Placements in Eight Fresh Cadaver Specimens)<br />

Portals Anatomic Structure Average Range<br />

Anterior Anterior Superior Iliac Spine<br />

a Lateral Femoral Cutaneous Nerve<br />

b Femoral Nerve (level of Sartorius)<br />

(level of Rectus Femoris)<br />

(level of Capsule)<br />

Ascending branch of Lateral Circumflex<br />

Femoral Artery<br />

c Terminal Branch<br />

(cm)<br />

6.3<br />

0.3<br />

4.3<br />

3.8<br />

3.7<br />

3.7<br />

0.3<br />

(cm)<br />

6.0-7.0<br />

0.2-1.0<br />

3.8-5.0<br />

2.7-5.0<br />

2.9-5.0<br />

1.0-6.0<br />

0.2-0.4<br />

Anterolateral Superior Gluteal Nerve 4.4 3.2-5.5<br />

Posterolateral Sciatic Nerve 2.9 2.0-4.3<br />

a<br />

Nerve had divided into three or more branches and measurement was made to the closest<br />

branch.<br />

b<br />

Measurement made at superficial surface of sartorius, rectus femoris, and capsule.<br />

c<br />

Small terminal branch of ascending branch of lateral circumflex femoral artery identified in<br />

three specimens.<br />

Figure 6


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

A. Anterior portal (Figure 7)<br />

1. Positioning<br />

a) Entry site is at the<br />

intersection of a sagittal<br />

line drawn distally<br />

from the ASIS and a<br />

transverse line across<br />

the superior margin of<br />

the greater trochanter<br />

b) Directed approximately<br />

45 cephalad and 30<br />

towards the midline<br />

c) Enters the joint under<br />

the anterior margin of Figure 7<br />

the acetabular labrum<br />

(Position is facilitated by direct arthroscopic visualization)<br />

2. Relationship to extraarticular anatomic structures<br />

a) Penetrates the sartorius and rectus femoris before entering the anterior<br />

capsule<br />

b) At the level of this portal, the lateral femoral cutaneous nerve has<br />

trifurcated<br />

(1) One of these branches will usually lie close to the portal<br />

(2) Most branches lie lateral to the portal<br />

- Moving portal more laterally does not reliably avoid these<br />

branches<br />

- Moving portal medially is ill-advised because of closer<br />

proximity to femoral nerve<br />

(3) Laceration of the LFCN is avoided by utilizing careful technique,<br />

not incising too deeply with the skin incision<br />

(4) Although laceration can be avoided, neuropraxia of one of these<br />

branches may occur due to manipulation of the cannula and<br />

instrumentation from the anterior position (


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

B. Anterolateral (Anterior Paratrochanteric) Portal (Figure 8)<br />

1. Positioning<br />

a) Entry site is over the superior<br />

margin of the greater trochanter at<br />

its anterior border<br />

b) Direction<br />

(1) In the AP fluoroscopic view,<br />

the portal passes immediately<br />

above the greater trochanter<br />

and then close to the superior<br />

surface of the femoral head<br />

to stay underneath the lateral<br />

acetabular labrum.<br />

(2) Accounting for normal<br />

femoral neck anteversion,<br />

with the hip in neutral rotation,<br />

Figure 8<br />

the portal courses parallel to the floor thus entering the hip joint<br />

just anterior to its mid-coronal plane.<br />

2. Relationship to the extraarticular structures<br />

a) The anterolateral portal lies most centrally in the "Safe Zone" for<br />

arthroscopy (Consequently it is the first portal established)<br />

b) The portal penetrates the gluteus medius before entering the lateral<br />

capsule<br />

c) The superior gluteal nerve runs transversely an average of 4.4 cm cephalad to<br />

the portal<br />

C. Posterolateral (Posterior Paratrochanteric) Portal (Figure 9)<br />

1. Positioning<br />

a) Entry site is over the superior<br />

margin of the greater trochanter<br />

at its posterior border<br />

b) Directed slightly cephalad and<br />

anterior (converges toward<br />

anterolateral portal)<br />

c) Enters the joint underneath the<br />

posterolateral margin of the labrum<br />

(Entry location is performed under<br />

direct arthroscopic visualization)<br />

Figure 9


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

2. Relationship to the extraarticular structures<br />

a) The portal pierces the gluteus medius and minimus before entering the<br />

lateral aspect of the capsule posteriorly.<br />

b) Like the anterolateral portal, the superior gluteal nerve averages a<br />

distance of 4.4cm.<br />

c) It enters the capsule superior and anterior to the piriformis tendon.<br />

d) It lies closest to the sciatic nerve at the level of the capsule with an<br />

average distance of 2.9 cm.<br />

(1) Inadvertent external rotation of the hip during portal placement<br />

will move the greater trochanter more posterior relative to the hip<br />

joint. This will unnecessarily cause the posterolateral portal to<br />

pass closer to the sciatic nerve. Consequently, external rotation is<br />

avoided during initial portal placement.<br />

(2) Hip flexion might partially relax the capsule and improve<br />

distraction. However this will place more traction on the sciatic<br />

nerve and may draw it closer to the joint, again placing it at more<br />

risk for inadvertent damage. Thus, inordinate hip flexion during<br />

hip arthroscopy should be avoided.<br />

V. Portal Placement and Normal Arthroscopic Exam<br />

A. Traction is applied to the hip<br />

B. Anterolateral Portal<br />

1. Placed first because it lies most centrally in the “safe zone” for arthroscopy<br />

2. Prepositioning performed with a custom spinal needle (6", 17 gauge)<br />

under fluoroscopic guidance<br />

3. Joint is then distended with saline<br />

4. Important to avoid penetrating the lateral labrum with the needle<br />

a) The needle meets greater resistance penetrating the labrum and can be<br />

felt during placement.<br />

b) After distending the joint, if necessary, the needle can be repositioned<br />

closer to the femoral head, further lessening the likelihood of piercing<br />

the labrum.<br />

5. A guide wire is passed through the needle and the<br />

needle is withdrawn.<br />

6. The cannula/obturator assembly is then passed over the<br />

guide wire into the joint. (Figures10 & 11)<br />

- As the assembly pierces the capsule, it is lifted up<br />

to avoid grazing the articular surface of the<br />

femoral head.<br />

7. The 70 arthroscope is then introduced in the<br />

anterolateral cannula.<br />

Figure 10<br />

a) Use of the 70 scope allows a direct view of where<br />

the anterior and posterolateral portals enter the<br />

joint simply by rotating the lens anteriorly and<br />

posteriorly.<br />

b) If there is a chance that the cannula may still have<br />

pierced the labrum, at this point, excessive<br />

maneuvering of the cannula should be minimized.<br />

Figure 11


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

C. Anterior Portal<br />

1. Prepositioned with spinal needle<br />

a) Facilitated by fluoroscopy<br />

b) Precise intracapsular positioning confirmed by direct arthroscopic view<br />

2. As the cannula/obturator assembly enters the joint, again by utilizing<br />

arthroscopic visualization, the labrum is avoided and the assembly is lifted off<br />

the articular surface of the femoral head.<br />

D. Posterolateral Portal<br />

1. Rotating the arthroscope posteriorly in the anterolateral portal allows viewing<br />

of the entry site for the posterolateral position.<br />

2. Needle placement and introduction of the cannula are then carried out in the<br />

standard fashion.<br />

E. With all three portals established, the inflow is switched to the posterolateral portal.<br />

1. A separate inflow optimizes flow dynamics.<br />

2. With the inflow in the posterolateral cannula, bubbles tend to be flushed out<br />

anteriorly, keeping them from obscuring the view.<br />

F. The arthroscope is then switched to the anterior portal to critique positioning of the<br />

anterolateral cannula.<br />

- Once it is assured that the cannula is free of the labrum, it is then safer to<br />

freely maneuver the instrumentation.<br />

G. Systematic examination of the joint is then carried out from the three interchangeable<br />

cannulas, using both the 70 and 30 arthroscopes.<br />

1. The 70 scope is better for viewing the labrum, periphery of the acetabulum<br />

and femoral head, and the deep recesses of the acetabular fossa and<br />

ligamentum teres.<br />

2. The 30 scope is better for viewing the central portions of the acetabulum and<br />

femoral head and the superior portion of the acetabular fossa.<br />

H. Instrumentation<br />

1. Transverse capsular incisions improve maneuverability of the instruments<br />

within the joint.<br />

- These are created by passing an arthroscopic knife through the cannula<br />

to incise the surrounding capsule.<br />

2. Interchanging flexible cannulas allow passage of curved shaver blades for<br />

maneuvering around the convex surface of the femoral head.<br />

3. Extra length hand instruments can be passed through the cannulas.<br />

4. With careful attention to the orientation, once the portal tracts have been<br />

established, larger hand instruments can be maneuvered free-hand through<br />

these tracts into the joint, such as for retrieving large loose bodies.


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

I. Peripheral compartment<br />

1. Instruments removed, traction released, hip flexed 45º (Figure 12)<br />

2. Arthroscope repositioned from anterolateral portal onto anterior neck of femur<br />

(Figure 13)<br />

- Prepositioned under fluoroscopy with spinal needle, guide wire &<br />

cannulated obturator<br />

3. Ancillary portal established 4-5 cm distally (Figure 14)<br />

- Placed under arthroscopic visualization; prepositioning with needle<br />

4. Peripheral compartment best inspected by:<br />

a) Combination of 30 & 70º scopes<br />

b) Switching between two peripheral portals<br />

c) Rotation of hip<br />

5. Often useful adjunct to arthroscopy of intraarticular compartment<br />

a) Common area for loose bodies to reside, escaping detection when<br />

viewing the intraarticular compartment<br />

b) Allows access to synovium for generous synovectomy<br />

c) Allows access to capsule for effective thermal capsulorrhaphy in cases of<br />

instability due to incompetent capsule<br />

Figure 12 Figure 13<br />

Figure 14


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

VI. Arthroscopic Anatomy<br />

A. Intraarticular compartment (Figure 15)<br />

1. The following structures are reliably viewed:<br />

a) Entire labrum<br />

b) Entire lunate articular<br />

surface of the acetabulum<br />

c) The acetabular fossa<br />

(including ligamentum<br />

teres and pulvinar)<br />

d) Most of the articular<br />

surface of the femoral<br />

head<br />

e) Portions of the transverse<br />

acetabular ligament<br />

f) Capsule and capsular<br />

reflection from the<br />

labrum<br />

2. Structures best viewed from the<br />

anterolateral portal include:<br />

a) Anterior acetabular<br />

labrum<br />

b) Anterior acetabular wall<br />

c) Anterior surface of the<br />

femoral head<br />

Figure 15<br />

3. Structures best visualized from<br />

the posterolateral portal include:<br />

a) Posterior acetabular labrum<br />

b) Posterior acetabular wall<br />

c) Posterior portion of the femoral head<br />

4. Structures best visualized from the anterior portal include:<br />

a) Lateral acetabular labrum and the relation of the lateral two portals<br />

b) Anterior aspect of the transverse acetabular ligament<br />

5. The acetabular fossa and its contents can be viewed with a different<br />

perspective from each portal.<br />

6. Approximately 75% of the weight bearing articular surface of the femoral head<br />

can be viewed, facilitated by intra-operative rotation of the hip.


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

B. Peripheral compartment (Figures 16 & 17)<br />

1. Visualization most reliably achieved for medial, anterior, and lateral portions<br />

a) Peripheral labrum & capsular reflection<br />

b) Peripheral aspect of femoral head<br />

- Facilitated by ROM<br />

c) Femoral neck<br />

d) Medial synovial fold<br />

- Consistent anatomic landmark<br />

e) Zona orbicularis<br />

- Circumferentially oriented coalescence of fibers creating capsular<br />

collar around femoral neck<br />

2. Posterior compartment least well visualized<br />

a) Capsule tighter<br />

b) Femoral capsular attachment more proximal<br />

c) Closer proximity of sciatic nerve<br />

Figure 16<br />

REFERENCES<br />

Byrd JWT: The Supine Position. In Byrd JWT (ed)<br />

Operative Hip Arthroscopy Second Edition, New York,<br />

Springer, 2005, 145-169.<br />

Byrd JWT: Hip Arthroscopy, principles and application:<br />

Smith & Nephew Endoscopy, 160 Dascomb Road, Andover,<br />

MA 01810, 1996.<br />

Byrd JWT: Operative Hip Arthroscopy (video). New<br />

York, Thieme, 1998.<br />

Byrd JWT, Pappas JN, Pedley MJ: Hip Arthroscopy: an<br />

anatomic study of portal placement and relationship to the<br />

extra-articular structures, Arthroscopy, 1995;11(4):418-423.<br />

Byrd, JWT, Chern KY: Traction vs. distension for<br />

distraction of the hip joint during arthroscopy, Arthroscopy,<br />

1997;13(3):346-349.<br />

Figure 17<br />

Byrd JWT: Portal Anatomy. In Byrd JWT (ed) Operative<br />

Hip Arthroscopy Second Edition, New York, Springer,<br />

2005, 110-116.<br />

Byrd JWT: Avoiding the labrum in hip arthroscopy,<br />

Arthroscopy, 2000;16(7):770-773.<br />

Dienst M, Gödde S, Seil R, Hammer D, Kohn D: Hip<br />

arthroscopy without traction: in vivo anatomy of the<br />

peripheral hip joint cavity. Arthroscopy 2001;17(9):924-<br />

931.<br />

Dienst M: Hip arthroscopy without traction. In Byrd JWT<br />

(ed) Operative Hip Arthroscopy Second Edition, New York,<br />

Springer, 2005, 170-188.


ABC’s in the OR<br />

J. W. Thomas Byrd, M.D.<br />

NOTES:


Safran<br />

FAI – Dx & Rx<br />

Page 1<br />

Femoroacetabular Impingement (FAI):<br />

What Is It, Making The Diagnosis, Imaging, Management and<br />

Surgical Treatment<br />

For 12 th Annual International Sports Medicine Fellows Conference<br />

Carlsbad, California<br />

January 29, 2012<br />

MARC R. SAFRAN, MD<br />

Professor, Orthopaedic Surgery<br />

Associate Director, Sports Medicine<br />

Stanford University<br />

1) INTRODUCTION<br />

a. Ganz and Associates Described FAI<br />

i. First described in the Swiss Literature 1995<br />

ii. First described in English Literature 1999<br />

b. Conflict / abutment of the femoral head-neck region against the acetabulum<br />

i. Generally antero-superior acetabulum<br />

ii. Occurs with internal rotation<br />

1. Less internal rotation needed to impinge with<br />

a. Increasing flexion<br />

b. Increasing adduction<br />

2. May impinge in abduction<br />

3. May impinge in flexion<br />

4. Dependent on location of bony dysmorphology<br />

c. Types<br />

i. Cam<br />

1. Loss of Offset at the femoral head neck junction<br />

ii. Pincer<br />

1. Overcoverage of the Femoral Head by the Acetabulum<br />

iii. Combined<br />

1. Most common that there are components of both<br />

NOTES:


Safran<br />

FAI – Dx & Rx<br />

Page 2<br />

2) CAM IMPINGEMENT<br />

a. Loss of Femoral Head-neck offset<br />

b. Result of aspherical head-neck region<br />

i. Femoral head epiphysis protrudes laterally out of a circle outlining femoral head<br />

ii. Pistol Grip deformity<br />

iii. Lateral contour of the femoral head extends in a convex shape to the base of the<br />

femoral neck<br />

iv. Pathophysiology<br />

1. Cam lesion goes under the labrum resulting initially in a labral-chondral<br />

separation.<br />

a. Later intra-substance labral tearing<br />

2. Articular cartilage delamination of acetabulum<br />

3. Predominantly anterosuperior damage<br />

a. Deep and more focal<br />

3) PINCER<br />

a. Relative overcoverage of the femoral head by the acetabulum<br />

b. Several types / causes<br />

i. Coxa Profunda (deep hip)<br />

1. Floor of the Acetabular fossa touches the ilioischial line and center edge<br />

angle >35 degrees<br />

ii. Protrusio (otto pelvis, coxa protrusio)<br />

1. Femoral head crosses the ilioischial line<br />

iii. Retroversion<br />

1. Posterior wall of acetabulum is medial to center of the femoral head<br />

iv. Cranial Retroversion<br />

1. Upper portion of the anterior acetabulum crosses the posterior border<br />

medial to the lateral edge of the acetabulum<br />

2. Figure of 8 or crossing sign.<br />

v. Ossified Labrum<br />

1. Deepens acetabulum functionally<br />

c. Pathophysiology<br />

i. Labrum crushed between the Acetabular rim and femoral head-neck region<br />

1. Crush labrum<br />

2. notching femoral head – neck region<br />

3. Chondral damage shallow but more global<br />

a. Not just antero-superior<br />

4. ContreCoup injury<br />

a. Femoral head-neck levers on Acetabular rim<br />

b. Posterior femoral head Chondral damage<br />

c. Posterior labral and acetabular injury<br />

NOTES:


4) COMPENSATORY ISSUES<br />

a. Concept – some evidence suggesting (Birmingham, et al)<br />

b. Hip with limited range of motion at femoroacetabular joint<br />

c. As try to maintain stride length or hip motion with athletic activities, compensate for<br />

limited femoroacetabular motion<br />

i. Increase stress on other local joints<br />

1. Pubic symphysis<br />

a. Result in osteitis pubis<br />

b. Sports Hernia as try to stabilize pubic symphysis<br />

2. Sacro-Iliac Joint<br />

a. SI Joint Dysfunction<br />

3. Lumbar Spine<br />

a. Facet joint inflammation<br />

b. Disc degeneration / herniation<br />

5) PRESENTATION<br />

a. History<br />

i. Uncommon to have a history of trauma<br />

1. Usually insidious onset<br />

2. Traumatic injury may be the straw that broke the camel’s back<br />

ii. Groin Pain<br />

iii. Aching<br />

iv. Difficulty putting on socks and/or shoes<br />

v. Worse with activity<br />

vi. Worse with prolonged sitting<br />

vii. Pain with<br />

1. Squatting<br />

2. Cutting / Pivoting<br />

3. Sudden Stop / Starts<br />

4. Going up stairs<br />

viii. May report limited range of motion<br />

1. Flexion<br />

2. Internal rotation<br />

3. Adduction<br />

ix. Often reported as a recalcitrant hip flexor strain<br />

x. May have mechanical symptoms<br />

1. Locking<br />

2. Catching<br />

3. Sharp pain<br />

xi. Not uncommon to have had other previous surgeries without success<br />

NOTES:<br />

Safran<br />

FAI – Dx & Rx<br />

Page 3


NOTES:<br />

b. Physical Examination<br />

i. See Hal Martin’s Work for complete Evaluation<br />

ii. My exam<br />

1. Observe Sitting<br />

2. Trendelenberg<br />

3. Gait<br />

4. Hip Flexion<br />

5. Hip Rotation (in 90 degrees flexion)<br />

a. Internal<br />

b. External<br />

6. Impingement Test<br />

a. Internal rotation of hip that is flexed to 90 degrees and adducted<br />

7. Labral Stress (Scour) Test<br />

a. Start in Flexion, Abduction and external rotation<br />

i. Move into adduction, extension and internal rotation<br />

b. Then Start in Flexion, Adduction and internal rotation<br />

i. Move into abduction, extension and external rotation<br />

8. FABER for motion<br />

9. Other Commonly Performed Tests<br />

a. Supine<br />

i. Hesselbach’s Test<br />

ii. Resisted Sit Up<br />

iii. Resisted Straight Leg Raise<br />

iv. Patrick’s Test<br />

v. Log Roll<br />

1. Pain<br />

2. Amount of Rotation<br />

vi. Thomas Test<br />

vii. Hyperextension – External Rotation<br />

1. Posterior Impingement<br />

2. Anterior Instability<br />

viii. McCarthy’s Test<br />

ix. Byrd’s Test for Snapping Hip<br />

1. Variations<br />

x. Posterior Apprehension<br />

xi. Lasegue’s<br />

xii. SI Joint Stress with Leg over table<br />

b. Lateral<br />

i. Tenderness of the Trochanter<br />

ii. Tenderness of the Gluteus Medius<br />

iii. Tenderness of the Piriformis<br />

iv. Ober Test<br />

v. Bicycle Test for Snapping<br />

vi. Pelvic compression<br />

Safran<br />

FAI – Dx & Rx<br />

Page 4


6) IMAGING<br />

a. Plain XR<br />

i. AP Pelvis<br />

1. Coccyx Centered 1-3 cm above Pubic Symphysis<br />

2. Symmetric Obturator Fossae<br />

ii. Lateral of the hip<br />

1. Cross Table<br />

2. Dunn View or Modifications<br />

iii. Pitfalls<br />

1. Frog Lateral good lateral for proximal femur but not acetabulum<br />

2. AP of the Hip is not the Same as an AP of the Pelvis<br />

iv. Additional views<br />

1. AP of the Hip<br />

2. False Profile View<br />

v. Proximal Femur<br />

1. Sphericity<br />

2. Symmetry<br />

3. Version<br />

4. Neck Shaft Angle<br />

5. Notching<br />

vi. Look at Acetabulum<br />

1. Depth<br />

a. Relative to Ilioischial Line<br />

i. Floor of Cotyloid Fossa (profunda)<br />

ii. Femoral head (protrusio)<br />

b. Center-edge angle<br />

2. Acetabular Index<br />

3. Crossing Sign<br />

4. Peri-articular Ossifications<br />

a. Labral Ossification<br />

b. Os Acetabuli<br />

c. Rim Stress Fracture<br />

b. MRI<br />

i. Small field of view ((FOV) MRI better<br />

1. MRI of the hip better than MRI of the Pelvis<br />

ii. Arthrography more sensitive<br />

iii. 3T better than 1.5 T<br />

iv. I prefer adding Anesthetic (Ropivicaine) to see if pain reduced<br />

1. Assure pain coming from inside the joint<br />

c. 3D CT Scan<br />

i. Better assessment of bony morphology<br />

ii. Some get all the time<br />

iii. Risk of fair amount of radiation<br />

NOTES:<br />

Safran<br />

FAI – Dx & Rx<br />

Page 5


d. Alpha Angle<br />

i. Defined on Radial Cut MRI Imaging<br />

1. People use with XR, CT, and non Radial MRI<br />

ii. Angle Made by drawing a line from the Center of the femoral head<br />

1. To the center of a line at the narrowest part of the femoral neck<br />

2. To the point where the femoral head exceeds the a sphere set to the<br />

circumference of the femoral head<br />

iii. Normal less than 50 – 55 degrees<br />

iv. Insurance companies want to see this number<br />

v. Poor inter and intra-observer reliability<br />

vi. Does not correlate with clinical success of the surgery<br />

7) TREATMENT<br />

a. Non operative<br />

i. No proven benefit<br />

ii. Treat compensatory changes / loss of strength<br />

1. Core strengthening<br />

2. Often hip abductor weakness<br />

iii. Do not force ROM<br />

1. Increases impingement<br />

iv. Avoid Deep Squats<br />

v. Recreational athletes – shorten stride length with running / jogging<br />

b. Operative<br />

i. ROM without abutment<br />

1. Eliminate impingement<br />

2. Restore Femoral Head-Neck offset<br />

a. Chielectomy / Femoral Osteoplasty<br />

3. Address Acetabular overcoverage<br />

a. Acetabuloplasty / Acetabular osteoplasty<br />

ii. Address associated pathology<br />

1. Labral<br />

a. Repair<br />

b. Resect<br />

c. Reconstruct ?<br />

2. Chondral<br />

a. Debride<br />

b. Microfracture<br />

c. Repair ?<br />

NOTES:<br />

Safran<br />

FAI – Dx & Rx<br />

Page 6


Safran<br />

FAI – Dx & Rx<br />

Page 7<br />

8) RESULTS<br />

a. Problems with results<br />

i. Most are short term<br />

ii. Published studies to date lack good quality of life score for non-arthritic hips in<br />

younger active patients<br />

1. IHOT<br />

b. Comparative Systematic reviews<br />

i. Matsuda, et al, Arthroscopy, 2011<br />

ii. Botser, et al, Arthroscopy, 2011<br />

c. Systematic Reviews<br />

i. Ng, et al, Am J Sports Med 2010<br />

ii. Clohisy, et al, Clin Orthop, 2010<br />

iii. Bedi, et al, Arthroscopy 2008<br />

d. Results similar<br />

e. Morbidity and Complications less with Arthroscopy<br />

9) CONCLUSION<br />

a. FAI is an important cause of hip pain<br />

i. Associated with Labral Damage<br />

ii. Associated with Chondral Damage<br />

iii. Associated with Early Arthritis<br />

b. 2 Main Types<br />

i. Cam<br />

ii. Pincer<br />

iii. Most people have combination of both to varying degrees<br />

c. Treatment is Usually Surgical<br />

d. Open, Arthroscopic and Combined techniques are safe and effective<br />

i. Long Term Follow Needed<br />

ii. Follow Up using validated outcomes measures for active patients without arthritis<br />

critical<br />

e. Must treat impingement as well as associated labral and Chondral pathology


Definition:<br />

Sports Hernia – Diagnosis, Algorithm and Surgical Treatment<br />

William Hutchinson, MD USA<br />

Sports hernias include basically the area of the umbilicus to the mid thigh and<br />

are without a bulge in the inguinal area. There are many definitions and<br />

names for these so-called sports hernias and none seems to incorporate the<br />

exact problem. These hernias include skin, fat, fascia, muscle, vessels,<br />

tendons, cartilage, bone, and nerves. The nerves seem to be the most<br />

important as they transmit the pain or discomfort from the site of pathology.<br />

Occasionally they are the only thing involved in the so-called sports hernia.<br />

Diagnosis:<br />

There are a myriad of presentations with the various injury involved and to try<br />

to use a simple set of questions is almost impossible with these hernias. An<br />

attempt will be made to outline a set of questions that will help separate the<br />

various types of sports hernias.<br />

Sports Hernias Examples:<br />

Pictures and cases reports will be included to show of the more common<br />

presentations.<br />

Treatment:<br />

An attempt will be made to try to outline some of the various treatments for the<br />

more common sports hernias. These will all be related to those treated by the<br />

general surgeon as opposed to the orthopedic injuries which will be covered<br />

by other presentors.


Michael B. Gerhardt, MD<br />

Player details<br />

� 28 year old male professional soccer<br />

player<br />

� Midfielder for Major League Soccer<br />

franchise in the United States<br />

� Drafted in 2002 after four year college<br />

career in the United States<br />

History of present complaint<br />

� Right hip pain developed in opening<br />

months of MLS season 2009<br />

� Able to complete competition with<br />

conservative measures<br />

� NSAIDs<br />

� Stretching<br />

� Ice<br />

� Modified practice schedule<br />

� Progressed from soccer related pain to<br />

daily pain in most activities<br />

Chief Complaint<br />

Michael B. Gerhardt, MD<br />

Team Physician<br />

LA Galaxy, CD Chivas USA<br />

US Soccer<br />

� Right hip and groin pain<br />

Past surgical history<br />

� Right sports hernia repair 1 year prior*<br />

*2008 Muschaweck repair, Munich,<br />

Germany – successful return to play<br />

4 weeks after surgery<br />

1/23/2012<br />

1


Presenting physical examination<br />

� Pain with impingement maneuver of<br />

right hip<br />

� Flexion, internal rotation<br />

�� Ten degrees internal rotation right side<br />

vs twenty degrees on the left<br />

Ext Flex IR ER Abd<br />

Right -10 10 125 10 60 65<br />

Left -10 10 125 20 60 65<br />

Physical examination<br />

� Positive sports hernia exam<br />

○ Rocker’s test<br />

○ Resisted hip adduction<br />

○ Hesselbach’s test<br />

AP Pelvis Lateral Right Hip<br />

CAM Lesion<br />

1/23/2012<br />

2


CAM Lesion Labral Tear<br />

Summary<br />

� 28 year old male professional soccer<br />

with new onset HIP PAIN and<br />

RECURRENT GROIN PAIN<br />

� Hip pain secondary to CAM impingement<br />

and labral tear<br />

� Groin pain secondary to recurrent sports<br />

hernia<br />

� Failed prolonged conservative treatment<br />

� Desires return to professional soccer<br />

Labral tear Synovitis<br />

Management – Phase I<br />

� Right hip arthroscopy (November 2009)<br />

� Findings<br />

� Primarily CAM impingement<br />

�� Anterolateral labral fraying<br />

� Grade II articular cartilage defect at<br />

anterolateral acetabulum<br />

� Partial tear of ligamentum teres<br />

� Synovitis<br />

1/23/2012<br />

3


Ligamentum teres partial tear CAM Lesion<br />

What was done<br />

� Labral debridement<br />

� Femoroplasty<br />

� Chondroplasty<br />

� SSynovectomy t<br />

� Ligamentum teres debridement<br />

Cam lesion – requiring<br />

femoroplasty<br />

Labral tear and chondral<br />

delamination – consistent<br />

with cam FAI<br />

Postoperative AP Pelvis<br />

1/23/2012<br />

4


Postoperative Lateral<br />

Management – Phase II<br />

� Right groin sports hernia repair<br />

(December 2009 - 3 weeks following<br />

hip arthroscopy)<br />

�� Findings<br />

� Posterior inguinal deficiency (transversalis<br />

fascia)<br />

� Ilioinguinal nerve entrapment<br />

Rehabilitation<br />

� No crutches – weight bearing as<br />

tolerated<br />

� Gentle progressive stretching program<br />

�� Stationary bike starting POD #7<br />

� Hip arthroscopy protocol continues<br />

Rehabilitation<br />

� Partial weight bearing 14 days on<br />

crutches<br />

� Continuous passive motion for two<br />

weeks<br />

� Rehabilitation inititated at 14 days<br />

What was done<br />

� Sports hernia repair<br />

� Posterior ing wall imbrication (no mesh)<br />

� Ilioinguinal neurolysis<br />

Current Status<br />

� Rehabilitated with MLS team staff<br />

� Trainer<br />

� Strength and Conditioning coach<br />

�� Returned to full competition March 2010<br />

MLS season opener (5 months)<br />

1/23/2012<br />

5


Relationship of Hip Injuries and<br />

Sports Hernia<br />

� Dual diagnosis<br />

� Likely relationship between hip<br />

morphology and development of hip and<br />

groin injuries<br />

� VERY COMMON PROBLEM IN<br />

SOCCER PLAYERS THROUGHOUT<br />

THE WORLD!!!<br />

� Area of ongoing research – FIFA funded<br />

Sports Hernia<br />

Current research<br />

-Professional soccer<br />

players<br />

-Incidence of xray<br />

abnormalities – FAI<br />

-Correlation with<br />

previous sports hernia<br />

**Prospective look at development<br />

of FAI symptoms and/or sports<br />

hernia<br />

Scope of the Problem<br />

� Harris, Dietz, Gerhardt, and<br />

Mandelbaum<br />

� Identifies relationship between<br />

RADIOGRAPHIC abnormalities of the hip<br />

and prevalence of HIP AND GROIN<br />

INJURIES in the professional soccer athlete<br />

Sports Hernia<br />

Common duality<br />

Sports Hernia +<br />

Hip Disorder<br />

� Theory<br />

1. Pre-existing FAI<br />

2. Limited Hip ROM<br />

3. Compensatory stress groin/pelvis<br />

4. Development of Sports Hernia<br />

Scope of the Problem<br />

� Harris, Dietz, Gerhardt, and<br />

Mandelbaum<br />

� “The Prevalence of Radiographic<br />

Abnormalities in Elite Soccer Players” y<br />

� Radiographs of 95 elite level soccer players<br />

� Abnormal radiographs<br />

○ 72% men<br />

○ 50% female<br />

Labral Tears in young population<br />

� Ganz et al. CORR 2003<br />

� Femoroacetabular impingement as the primary<br />

cause of labral tears in the nondysplastic hip<br />

� CAM effect<br />

○ Anterosuperior acetabular cartilage and labral shearing<br />

○ Anterosuperior acetabular cartilage and labral shearing<br />

� Pincer effect<br />

○ Labrum caught between acetabulum and femoral neck<br />

○ Peripheral and circumferential labral degeneration<br />

1/23/2012<br />

6


Bedi et al. The Management of Labral Tears and FAI of the hip in the<br />

Young, Active Patient. Arthroscopy 2008 Return to play after hip<br />

arthroscopy<br />

� Phillipon et al AJSM 2009<br />

� 28 professional hockey players<br />

� Acetabular rim trimming, femoral neck<br />

osteoplasty, p y and labral repair p<br />

� Average age 27 years old<br />

� Return to skating/drills at 3.8 months<br />

� Average of 94 games played at 2 years<br />

postop<br />

Is it the labrum or FAI?<br />

� Philippon et al. AJSM 2007<br />

� 37 patients for revision hip arthroscopy<br />

� Persistent pain 21 mos after index<br />

procedure p<br />

� 36/37 had evidence of FAI on xray that was<br />

not/inadequately addressed<br />

� Underlying FAI pathology treatment is critical<br />

in treating labral pathology<br />

Sports Hernia<br />

History<br />

Current research<br />

US Men’s National<br />

Soccer Team<br />

-37.5% sports<br />

hernia surgery<br />

-12.5% bilateral<br />

-66% dual diagnosis<br />

>>sports hernia + FAI morphology<br />

Return to play after hip<br />

arthroscopy<br />

� Bharam Clin Sports Med 2006<br />

� Average return to play for labral surgery<br />

� No impingement surgery performed<br />

� Golf: 6 weeks<br />

� Hockey: 10 weeks<br />

� Baseball and soccer: 12 weeks<br />

Sports Hernia + FAI<br />

Conclusion<br />

*FAI and Sports<br />

Hernia common<br />

in in elite elite athletes athletes<br />

*Addressing BOTH<br />

diagnoses may be<br />

indicated<br />

*May result in lower<br />

recurrence rates<br />

1/23/2012<br />

7


Thank You<br />

1/23/2012<br />

8

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!