Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®): preclinical and clinical trial in osteoarthritis of the knee joint

Pawan Kumar Gupta, Anoop Chullikana, Mathiyazhagan Rengasamy, Naresh Shetty, Vivek Pandey, Vikas Agarwal, Shrikant Yeshwant Wagh, Prasanth Kulapurathu Vellotare, Devi Damodaran, Pachaiyappan Viswanathan, Charan Thej, Sudha Balasubramanian, Anish Sen Majumdar, Pawan Kumar Gupta, Anoop Chullikana, Mathiyazhagan Rengasamy, Naresh Shetty, Vivek Pandey, Vikas Agarwal, Shrikant Yeshwant Wagh, Prasanth Kulapurathu Vellotare, Devi Damodaran, Pachaiyappan Viswanathan, Charan Thej, Sudha Balasubramanian, Anish Sen Majumdar

Abstract

Background: Osteoarthritis (OA) is a common and debilitating chronic degenerative disease of the joints. Currently, cell-based therapy is being explored to address the repair of damaged articular cartilage in the knee joint.

Methods: The in vitro differentiation potential of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®) was determined by differentiating the cells toward the chondrogenic lineage and quantifying sulfated glycosaminoglycan (sGAG). The mono-iodoacetate (MIA)-induced preclinical model of OA has been used to demonstrate pain reduction and cartilage formation. In the clinical study, 60 OA patients were randomized to receive different doses of cells (25, 50, 75, or 150 million cells) or placebo. Stempeucel® was administered by intra-articular (IA) injection into the knee joint, followed by 2 ml hyaluronic acid (20 mg). Subjective evaluations-visual analog scale (VAS) for pain, intermittent and constant osteoarthritis pain (ICOAP), and Western Ontario and McMaster Universities Osteoarthritis (WOMAC-OA) index-were performed at baseline and at 1, 3, 6, and 12 months of follow-up. Magnetic resonance imaging of the knee was performed at baseline, and at 6 and 12 months follow-up for cartilage evaluation.

Results: Stempeucel® differentiated into the chondrogenic lineage in vitro with downregulation of Sox9 and upregulation of Col2A genes. Furthermore, Stempeucel® differentiated into chondrocytes and synthesized a significant amount of sGAG (30 ± 1.8 μg/μg GAG/DNA). In the preclinical model of OA, Stempeucel® reduced pain significantly and also repaired damaged articular cartilage in rats. In the clinical study, IA administration of Stempeucel® was safe, and a trend towards improvement was seen in the 25-million-cell dose group in all subjective parameters (VAS, ICOAP, andWOMAC-OA scores), although this was not statistically significant when compared to placebo. Adverse events were predominant in the higher dose groups (50, 75, and 150 million cells). Knee pain and swelling were the most common adverse events. The whole-organ magnetic resonance imaging score of the knee did not reveal any difference from baseline and the placebo group.

Conclusion: Intra-articular administration of Stempeucel® is safe. A twenty-five-million-cell dose may be the most effective among the doses tested for pain reduction. Clinical studies with a larger patient population are required to demonstrate a robust therapeutic efficacy of Stempeucel® in OA.

Trial registration: Clinicaltrials.gov NCT01453738 . Registered 13 October 2011.

Keywords: Cell therapy; Mesenchymal stromal cells; Osteoarthritis.

Figures

Fig. 1
Fig. 1
CONSORT flow chart showing the number of patients randomized, followed-up, and analyzed. M million cells, Cell Stempeucel®, mITT modified intention to treat
Fig. 2
Fig. 2
Quantification of gene expression and sGAG. Quantitative mRNA expression of a SOX9 and b Col2A in the control (white bar) and chondrogenically differentiated Stempeucel® (black bar) by real-time PCR analysis (n = 6). c Sulfated glycosaminoglycan (sGAG) content in the control (white bar) and chondrogenically differentiated Stempeucel® (black bar) by DMMB dye-binding assay. The sGAG values were normalized to the DNA content in the control and chondrogenically differentiated Stempeucel® (n = 6). Results are represented as mean with SEM
Fig. 3
Fig. 3
Effect of intra-articular injection of Stempeucel® on pain reduction and cartilage repair in an osteoarthritic rat model. a The effect of Stempeucel® on pain reduction at week 0 (before cell injection), and at weeks 4, 8, and 12 after cell injection. Data are presented as mean ± SEM. *P < 0.05, ***P < 0.001 versus hyaluronic acid (HA)-treated group. b Photomicrographs of representative joint sections of femoral condyle stained with H&E at 4 (a–e), 8 (f–j), and 12 weeks (k–o) after Stempeucel® treatment. Osteoarthritic changes, such as loss of chondrocytes (*), loss of cartilage (vertical arrow), and fibrillation (thin arrow) are evident in vehicle-treated and HA-treated joints. Proliferation of chondrocytes (thick arrow), regeneration, and repair of cartilage tissue was evident in Stempeucel®-treated groups. Scale bars = 100 μm, magnification 10×. H high dose of Stempeucel®, L Low dose of Stempeucel®, MIA mono-iodoacetate, ns not significant
Fig. 4
Fig. 4
Histological evaluation of Safranin-O stained joint sections. a Photomicrographs of representative joint specimens of femoral condyle stained with Safranin-O at 4 (a–e), 8 (f–j), and 12 weeks (k–o) after Stempeucel® treatment. Loss of articular surface, roughening of cartilage and reduced staining of Safranin-O (thin arrow) were observed in vehicle- and HA-treated joints. Strongly stained Safranin-O-positive cartilage (thick arrow) with increased numbers of chondrocytes was seen in the Stempeucel®-treated groups. Scale bars = 100 μm, magnification 10×. b Sulfated glycosaminoglycan (GAG) fraction intensity was measured from histological images of Safranin-O-stained sections at weeks 4, 8, and 12. The intensity of Safranin-O staining is represented graphically, and the data are represented as mean ± SEM. At 12 weeks, the Stempeucel®-treated groups (both low (L) and high (H) dose) showed a significant improvement in the sGAG content compared to the disease control (mono-iodoacetate; MIA) and hyaluronic acid (HA)-treated groups. *P < 0.05, *** P < 0.001. ns not significant
Fig. 5
Fig. 5
Visual analog scale values. Data are presented as mean ± SD. 1 M, 3 M, 6 M, and 12 M = 1, 3, 6, and 12 months, respectively; C1 cohort 1, C2 cohort 2, M million cells, P placebo, VAS visual analog scale
Fig. 6
Fig. 6
WOMAC results. WOMAC a, b composite, c, d pain, e, f stiffness, and g, h physical function (PF) results are shown for cohorts 1 (a, c, e and g) and 2 (b, d, f, and h). Data are presented as mean ± SD. 1 M, 3 M, 6 M, and 12 M = 1, 3, 6, and 12 months, respectively; C1 cohort 1, C2 cohort 2, M million cells, P placebo, WOMAC Western Ontario and McMaster Universities
Fig. 7
Fig. 7
ICOAP results. ICOAP a, b total, c, d constant pain, and e, f intermittent pain results are shown for cohorts 1 (a, c, and e) and 2 (b, d, and f). Data are presented as mean ± SD. 1 M, 3 M, 6 M, and 12 M = 1, 3, 6, and 12 months, respectively; C1 cohort 1, C2 cohort 2, ICOAP intermittent and constant osteoarthritis pain, M million cells, P placebo

References

    1. Murray C, Lopez A. The global burden of disease: a comprehensive assessment of mortality and disability from disease, injuries and risk factors in 1990 and projected to 2020. Boston: Harvard University Press; 1996.
    1. Osteoarthritis: care and management. . Accessed 8 May 2016.
    1. Hochberg MC, Altman RD, April KT, Benkhalti M, Guyatt G, McGowan J, et al. American College of Rheumatology 2012 recommendations for the use of nonpharmacologic and pharmacologic therapies in osteoarthritis of the hand, hip, and knee. Arthritis Care Res. 2012;64(4):465–474. doi: 10.1002/acr.21596.
    1. Jordan KM, Arden NK, Doherty M, Bannwarth B, Bijlsma JWJ, Dieppe P, et al. EULAR Recommendations 2003: an evidence based approach to the management of knee osteoarthritis: report of a Task Force of the Standing Committee for International Clinical Studies Including Therapeutic Trials (ESCISIT) Ann Rheum Dis. 2003;62(12):1145–1155. doi: 10.1136/ard.2003.011742.
    1. Schrama JC, Espehaug B, Hallan G, Engesæter LB, Furnes O, Havelin LI, et al. Risk of revision for infection in primary total hip and knee arthroplasty in patients with rheumatoid arthritis compared with osteoarthritis: a prospective, population-based study on 108,786 hip and knee joint arthroplasties from the Norwegian Arthroplasty Register. Arthritis Care Res. 2010;62(4):473–479. doi: 10.1002/acr.20036.
    1. Knutsen G, Drogset JO, Engebretsen L, Grøntvedt T, Isaksen V, Ludvigsen TC, et al. A randomized trial comparing autologous chondrocyte implantation with microfracture. J Bone Jt Surg. 2007;89(10):2105–2112.
    1. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med. 1994;331(14):889–895. doi: 10.1056/NEJM199410063311401.
    1. Von der Mark K, Gauss V, von der Mark H, Müller P. Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature. 1977;267(5611):531–532. doi: 10.1038/267531a0.
    1. Veronesi F, Giavaresi G, Tschon M, Borsari V, Nicoli Aldini N, Fini M. Clinical use of bone marrow, bone marrow concentrate, and expanded bone marrow mesenchymal stem cells in cartilage disease. Stem Cells Dev. 2013;22(2):181–192. doi: 10.1089/scd.2012.0373.
    1. Vangsness CT, Farr J, Boyd J, Dellaero DT, Mills CR, LeRoux-Williams M. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy: a randomized, double-blind, controlled study. J Bone Joint Surg Am. 2014;96(2):90–98. doi: 10.2106/JBJS.M.00058.
    1. Vega A, Martín-Ferrero MA, Del Canto F, Alberca M, García V, Munar A, et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. 2015;99(8):1681–1690. doi: 10.1097/TP.0000000000000678.
    1. Gupta PK, Das AK, Chullikana A, Majumdar AS. Mesenchymal stem cells for cartilage repair in osteoarthritis. Stem Cell Res Ther. 2012;3:25. doi: 10.1186/scrt116.
    1. Koh Y-G, Jo S-B, Kwon O-R, Suh D-S, Lee S-W, Park S-H, et al. Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthroscopy. 2013;29(4):748–755. doi: 10.1016/j.arthro.2012.11.017.
    1. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells Dayt Ohio. 2014;32(5):1254–1266. doi: 10.1002/stem.1634.
    1. Emadedin M, Aghdami N, Taghiyar L, Fazeli R, Moghadasali R, Jahangir S, et al. Intra-articular injection of autologous mesenchymal stem cells in six patients with knee osteoarthritis. Arch Iran Med. 2012;15(7):422–428.
    1. Mamidi MK, Nathan KG, Singh G, Thrichelvam ST, Mohd Yusof NAN, Fakharuzi NA, et al. Comparative cellular and molecular analyses of pooled bone marrow multipotent mesenchymal stromal cells during continuous passaging and after successive cryopreservation. J Cell Biochem. 2012;113(10):3153–3164. doi: 10.1002/jcb.24193.
    1. Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, et al. A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. J Transl Med. 2013;11:143. doi: 10.1186/1479-5876-11-143.
    1. Guingamp C, Gegout-Pottie P, Philippe L, Terlain B, Netter P, Gillet P. Mono-iodoacetate-induced experimental osteoarthritis. A dose-response study of loss of mobility, morphology, and biochemistry. Arthritis Rheum. 1997;40(9):1670–1679. doi: 10.1002/art.1780400917.
    1. Pomonis JD, Boulet JM, Gottshall SL, Phillips S, Sellers R, Bunton T, et al. Development and pharmacological characterization of a rat model of osteoarthritis pain. Pain. 2005;114(3):339–346. doi: 10.1016/j.pain.2004.11.008.
    1. Vermeirsch H, Biermans R, Salmon PL, Meert TF. Evaluation of pain behavior and bone destruction in two arthritic models in guinea pig and rat. Pharmacol Biochem Behav. 2007;87(3):349–359. doi: 10.1016/j.pbb.2007.05.010.
    1. Di Cesare ML, Micheli L, Zanardelli M, Ghelardini C. Low dose native type II collagen prevents pain in a rat osteoarthritis model. BMC Musculoskelet Disord. 2013;14:228. doi: 10.1186/1471-2474-14-228.
    1. Schmitz N, Laverty S, Kraus VB, Aigner T. Basic methods in histopathology of joint tissues. Osteoarthritis Cartilage. 2010;18(Suppl 3):113–116. doi: 10.1016/j.joca.2010.05.026.
    1. Pritzker KPH, Gay S, Jimenez SA, Ostergaard K, Pelletier J-P, Revell PA, et al. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthritis Cartilage. 2006;14(1):13–29. doi: 10.1016/j.joca.2005.07.014.
    1. Gerwin N, Bendele AM, Glasson S, Carlson CS. The OARSI histopathology initiative—recommendations for histological assessments of osteoarthritis in the rat. Osteoarthritis Cartilage. 2010;18(3):24–34. doi: 10.1016/j.joca.2010.05.030.
    1. Moyer JT, Priest R, Bouman T, Abraham AC, Donahue TLH. Indentation properties and glycosaminoglycan content of human menisci in the deep zone. Acta Biomater. 2013;9(5):6624–6629. doi: 10.1016/j.actbio.2012.12.033.
    1. Villegas DF, Hansen TA, Liu DF, Donahue TLH. A quantitative study of the microstructure and biochemistry of the medial meniscal horn attachments. Ann Biomed Eng. 2008;36(1):123–131. doi: 10.1007/s10439-007-9403-x.
    1. Peterfy CG, Guermazi A, Zaim S, Tirman PFJ, Miaux Y, White D, et al. Whole-Organ Magnetic Resonance Imaging Score (WORMS) of the knee in osteoarthritis. Osteoarthritis Cartilage. 2004;12(3):177–190. doi: 10.1016/j.joca.2003.11.003.
    1. Stubendorff JJ, Lammentausta E, Struglics A, Lindberg L, Heinegård D, Dahlberg LE. Is cartilage sGAG content related to early changes in cartilage disease? Implications for interpretation of dGEMRIC. Osteoarthritis Cartilage. 2012;20(5):396–404. doi: 10.1016/j.joca.2012.01.015.
    1. Wu L, Leijten JCH, Georgi N, Post JN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng Part A. 2011;17(9-10):1425–1436. doi: 10.1089/ten.tea.2010.0517.
    1. Sato M, Uchida K, Nakajima H, Miyazaki T, Guerrero AR, Watanabe S, et al. Direct transplantation of mesenchymal stem cells into the knee joints of Hartley strain guinea pigs with spontaneous osteoarthritis. Arthritis Res Ther. 2012;14:R31. doi: 10.1186/ar3735.
    1. Mokbel AN, El Tookhy OS, Shamaa AA, Rashed LA, Sabry D, El Sayed AM. Homing and reparative effect of intra-articular injection of autologus mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord. 2011;12:259. doi: 10.1186/1471-2474-12-259.
    1. Nam HY, Karunanithi P, Loo WCP, Naveen SV, Chen HC, Hussin P, et al. The effects of staged intra-articular injection of cultured autologous mesenchymal stromal cells on the repair of damaged cartilage: a pilot study in caprine model. Arthritis Res Ther. 2013;15:R129. doi: 10.1186/ar4309.
    1. Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48(12):3464–3474. doi: 10.1002/art.11365.
    1. Suhaeb AM, Naveen S, Mansor A, Kamarul T. Hyaluronic acid with or without bone marrow derived-mesenchymal stem cells improves osteoarthritic knee changes in rat model: a preliminary report. Indian J Exp Biol. 2012;50(6):383–390.
    1. Black LL, Gaynor J, Adams C, Dhupa S, Sams AE, Taylor R, et al. Effect of intraarticular injection of autologous adipose-derived mesenchymal stem and regenerative cells on clinical signs of chronic osteoarthritis of the elbow joint in dogs. Vet Ther. 2008;9(3):192–200.
    1. Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27(12):1675–1680. doi: 10.1002/jor.20933.
    1. Van Buul GM, Siebelt M, Leijs MJC, Bos PK, Waarsing JH, Kops N, et al. Mesenchymal stem cells reduce pain but not degenerative changes in a mono-iodoacetate rat model of osteoarthritis. J Orthop Res. 2014;32(9):1167–1174. doi: 10.1002/jor.22650.
    1. Guo W, Wang H, Zou S, Gu M, Watanabe M, Wei F, et al. Bone marrow stromal cells produce long-term pain relief in rat models of persistent pain. Stem Cells Dayt Ohio. 2011;29(8):1294–1303. doi: 10.1002/stem.667.
    1. Rock KL, Kono H. The inflammatory response to cell death. Annu Rev Pathol. 2008;3:99–126. doi: 10.1146/annurev.pathmechdis.3.121806.151456.
    1. Centeno CJ, Schultz JR, Cheever M, Freeman M, Faulkner S, Robinson B, et al. Safety and complications reporting update on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2011;6(4):368–378. doi: 10.2174/157488811797904371.
    1. Davatchi F, Abdollahi BS, Mohyeddin M, Shahram F, Nikbin B. Mesenchymal stem cell therapy for knee osteoarthritis. Preliminary report of four patients. Int J Rheum Dis. 2011;14(2):211–215. doi: 10.1111/j.1756-185X.2011.01599.x.
    1. Nejadnik H, Hui JH, Feng Choong EP, Tai B-C, Lee EH. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med. 2010;38(6):1110–1116. doi: 10.1177/0363546509359067.
    1. Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002;10(3):199–206. doi: 10.1053/joca.2001.0504.
    1. Wong KL, Lee KBL, Tai BC, Law P, Lee EH, Hui JHP. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020–2028. doi: 10.1016/j.arthro.2013.09.074.
    1. Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman MM. Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells, platelet lysate and dexamethasone. Ann Transplant. 2008;9:246–251.
    1. Saether EE, Chamberlain CS, Leiferman EM, Kondratko-Mittnacht JR, Li WJ, Brickson SL, et al. Enhanced medial collateral ligament healing using mesenchymal stem cells: dosage effects on cellular response and cytokine profile. Stem Cell Rev. 2014;10(1):86–96. doi: 10.1007/s12015-013-9479-7.
    1. Koh Y-G, Choi Y-J, Kwon S-K, Kim Y-S, Yeo J-E. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2015;23(5):1308–1316. doi: 10.1007/s00167-013-2807-2.
    1. Koh Y-G, Choi Y-J. Infrapatellar fat pad-derived mesenchymal stem cell therapy for knee osteoarthritis. Knee. 2012;19(6):902–907. doi: 10.1016/j.knee.2012.04.001.
    1. Ruiz M, Cosenza S, Maumus M, Jorgensen C, Noël D. Therapeutic application of mesenchymal stem cells in osteoarthritis. Expert Opin Biol Ther. 2015;1–10.
    1. Orozco L, Munar A, Soler R, Alberca M, Soler F, Huguet M, et al. Treatment of knee osteoarthritis with autologous mesenchymal stem cells: a pilot study. Transplant J. 2013;95(12):1535–1541. doi: 10.1097/TP.0b013e318291a2da.
    1. Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D. Regeneration of meniscus cartilage in a knee treated with percutaneously implanted autologous mesenchymal stem cells. Med Hypotheses. 2008;71(6):900–908. doi: 10.1016/j.mehy.2008.06.042.
    1. Huang JI, Kazmi N, Durbhakula MM, Hering TM, Yoo JU, Johnstone B. Chondrogenic potential of progenitor cells derived from human bone marrow and adipose tissue: a patient-matched comparison. J Orthop Res. 2005;23(6):1383–1389. doi: 10.1016/j.orthres.2005.03.008.1100230621.
    1. Frenkel SR, Toolan B, Menche D, Pitman MI, Pachence JM. Chondrocyte transplantation using a collagen bilayer matrix for cartilage repair. J Bone Joint Surg (Br) 1997;79(5):831–836. doi: 10.1302/0301-620X.79B5.7278.
    1. Iwasa J, Engebretsen L, Shima Y, Ochi M. Clinical application of scaffolds for cartilage tissue engineering. Knee Surg Sports Traumatol Arthrosc. 2009;17(6):561–577. doi: 10.1007/s00167-008-0663-2.
    1. Kim YS, Choi YJ, Suh DS, Heo DB, Kim YI, Ryu J-S, et al. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43(1):176–185. doi: 10.1177/0363546514554190.
    1. CARTISTEM®, mesenchymal stem cells for treatment of osteoarthritis—LifeMap Discovery. . Accessed 13 Nov 2015.
    1. Rush University Medical Center announces human clinical trial for Cartistem. . Accessed 14 Nov 2015.
    1. Berkoff DJ, Miller LE, Block JE. Clinical utility of ultrasound guidance for intra-articular knee injections: a review. Clin Interv Aging. 2012;7:89–95.
    1. Lynch JA, Roemer FW, Nevitt MC, Felson DT, Niu J, Eaton CB, et al. Comparison of BLOKS and WORMS scoring systems part I. Cross sectional comparison of methods to assess cartilage morphology, meniscal damage and bone marrow lesions on knee MRI: data from the osteoarthritis initiative. Osteoarthritis Cartilage. 2010;18(11):1393–1401. doi: 10.1016/j.joca.2010.08.017.
    1. Felson DT, Lynch J, Guermazi A, Roemer FW, Niu J, McAlindon T, et al. Comparison of BLOKS and WORMS scoring systems part II. Longitudinal assessment of knee MRIs for osteoarthritis and suggested approach based on their performance: data from the Osteoarthritis Initiative. Osteoarthritis Cartilage. 2010;18(11):1402–1407. doi: 10.1016/j.joca.2010.06.016.

Source: PubMed

3
Subscribe