Coverage Policy Manual
Policy #: 2010023
Category: Medicine
Initiated: April 2010
Last Review: February 2024
  Orthopedic Applications of Stem Cell Therapy

Description:
Mesenchymal stem cells (MSCs) have the capability to differentiate into a variety of tissue types, including various musculoskeletal tissues. Potential uses of MSCs for orthopedic applications include treatment of damaged bone, cartilage, ligaments, tendons and intervertebral discs.
 
MSCs are multipotent cells (also called stromal multipotent cells) that possess the ability to differentiate into various tissues including organs, trabecular bone, tendon, articular cartilage, ligaments, muscle, and fat. MSCs are associated with the blood vessels within bone marrow, synovium, fat, and muscle, where they can be mobilized for endogenous repair as occurs with healing of bone fractures. Tissues such as cartilage, tendon, ligaments, and vertebral discs show limited capacity for endogenous repair because of the limited presence of the triad of functional tissue components: vasculature, nerves, and lymphatics. Orthobiologics is a term introduced to describe interventions using cells and biomaterials to support healing and repair. Cell therapy is the application of MSCs directly to a musculoskeletal site. Tissue engineering techniques use MSCs and/or bioactive molecules such as growth factors and scaffold combinations to improve the efficiency of repair or regeneration of damaged musculoskeletal tissues (Goldberg, 2017).
 
Bone marrow aspirate is considered the most accessible source and, thus, the most common place to isolate MSCs for the treatment of musculoskeletal disease. However, harvesting MSCs from bone marrow requires a procedure that may result in donor-site morbidity. Also, the number of MSCs in bone marrow is low, and the number and differentiation capacity of bone marrow-derived MSCs decreases with age, limiting their efficiency when isolated from older patients.
 
In vivo, the fate of stem cells is regulated by signals in the local 3-dimensional microenvironment from the extracellular matrix and neighboring cells. It is believed the success of tissue engineering with MSCs will also require an appropriate 3-dimensional scaffold or matrix, culture conditions for tissue-specific induction, and implantation techniques that provide appropriate biomechanical forces and mechanical stimulation. The ability to induce cell division and differentiation without adverse effects, such as the formation of neoplasms, remains a significant concern. Given that each tissue type requires different culture conditions, induction factors (signaling proteins, cytokines, growth factors), and implantation techniques, each preparation must be individually examined.
 
 
Regulatory Status
The U.S. Food and Drug Administration (FDA) regulates human cells and tissues intended for implantation, transplantation, or infusion through the Center for Biologics Evaluation and Research, under Code of Federal Regulation, Title 21, parts 1270 and 1271. MSCs are included in these regulations.
 
Concentrated autologous MSCs do not require approval by the U.S. Food and Drug Administration (FDA). No products using engineered or expanded MSCs have been approved by the FDA for orthopedic applications.
 
The following products are examples of commercialized demineralized bone matrix (DBM) products. They are marketed as containing viable stem cells. In some instances, manufacturers have received communications and inquiries from the FDA related to the appropriateness of their marketing products that are dependent on living cells for their function. The following descriptions are from the product literature.
 
    • Allostem® (AlloSource) is a partially demineralized allograft bone seeded with adipose-derived MSCs.
    • Map3® (RTI Surgical) contains cortical cancellous bone chips, DBM, and cryopreserved multipotent adult progenitor cells (MAPC®).
    • Osteocel Plus® (NuVasive) is a DBM combined with viable MSCs that have been isolated from allogeneic bone marrow.
    • Trinity Evolution Matrix™ (Orthofix) DBM combined with viable MSCs that have been isolated from allogeneic bone marrow.
    • Other products contain DBM alone and are designed to be mixed with bone marrow aspirate:
        • Fusion Flex™ (Wright Medical) is a dehydrated moldable DBM scaffold (strips and cubes) that will absorb autologous bone marrow aspirate;
        • Ignite® (Wright Medical) is an injectable graft with DBM that can be combined with autologous bone marrow aspirate.
 
A number of DBM combination products have been cleared for marketing by the FDA through the 510(k) process. FDA product code: MQV.
 
A sample of these products are listed below. Some are specifically labeled for mixing with bone marrow aspirate.
Demineralized Bone Matrix Products Cleared by FDA
    • Vitoss® Bioactive Foam Bone Graft Substitute, matrix type Type I bovine collagen (mix with autologous MSCs) is manufactured by Stryker and received clearance November 2008 (K083033)
    • NanOss BVF-E, matrix type Nanocrystalline hydroxyapatite, is manufactured by Pioneer Surgical and received clearance August 2008 (K081558)
    • OrthoBlast® II Demineralized bone matrix putty and paste, matrix type Human cancellous bone chips, is manufactured by SeaSpine and received clearance September 2007 (K070751)
    • CopiOs® Bone Void Filler (sponge and powder disc), matrix type Type I bovine dermal collagen (mix with autologous MSCs) is manufactured by Kensey Nash and received clearance May 2007 (K071237)
    • DBX® Demineralized bone matrix putty, paste and mix, matrix type Processed human bone and sodium hyaluronate (mix with autologous MSCs) is manufactured by Musculoskeletal Transplant Foundation and received clearance December 2006 (K053218)
    • Integra MOZAIK™ Osteoconductive Scaffold-Putty, matrix type Human cancellous bone (mix with autologous MSCs) is manufactured by IsoTis OrthoBiologics and received clearance December 2006 (K062353)
    • Formagraft™ Collagen Bone Graft Matrix, matrix type Bovine fibrillary collagen (mix with autologous MSCs) is manufactured by R and L Medical and received clearance May 2005 (K050789)
    • DynaGraft® II Gel and Putty, matrix type Processed human bone particles, is manufactured by IsoTis Orthobiologics and received clearance March 2005 (K040419)
 
In 2020, the FDA updated their guidance on "Regulatory Considerations for Human Cells, Tissues, and Cellular and Tissue-Based Products: Minimal Manipulation and Homologous Use" (FDA, 2020).
 
Human cells, tissues, and cellular and tissue-based products (HCT/P) are defined as human cells or tissues that are intended for implantation, transplantation, infusion, or transfer into a human recipient. If an HCT/P does not meet the criteria below and does not qualify for any of the stated exceptions, the HCT/P will be regulated as a drug, device, and/or biological product and applicable regulations and premarket review will be required.
 
An HCT/P is regulated solely under section 361 of the PHS Act and 21 CFR Part 1271 if it meets all of the following criteria:
 
"1) The HCT/P is minimally manipulated;
2) The HCT/P is intended for homologous use only, as reflected by the labeling, advertising, or other indications of the manufacturer’s objective intent;
3) The manufacture of the HCT/P does not involve the combination of the cells or tissues with another article, except for water, crystalloids, or a sterilizing, preserving, or storage agent, provided that the addition of water, crystalloids, or the sterilizing, preserving, or storage agent does not raise new clinical safety concerns with respect to the HCT/P; and
4) Either:
i) The HCT/P does not have a systemic effect and is not dependent upon the metabolic activity of living cells for its primary function; or
ii) The HCT/P has a systemic effect or is dependent upon the metabolic activity of living cells for its primary function, and: a) Is for autologous use; b) Is for allogeneic use in a first-degree or second-degree blood relative; or c) Is for reproductive use."
 
The FDA does not consider the use of stem cells for orthopedic procedures to be homologous use.
 
 
Related Policies
2009049 - Platelet-Rich Plasma (Autologous Growth Factors) Orthopedic Applications
 
There are no specific CPT or HCPCS codes that describe all the services included in the orthopedic application of mesenchymal stem cell therapy.

Policy/
Coverage:
Effective August 2017
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
The use of stem cell therapy in the treatment of avascular necrosis (AVN) of the femoral head, or the use of mesenchymal stem cell therapy for any orthopedic application, does not meet primary coverage criteria for effectiveness. This technology is currently being studied in ongoing clinical trials.
 
For contracts without primary coverage criteria the use of stem cell therapy in the treatment of avascular necrosis (AVN) of the femoral head, or the use of mesenchymal stem cell therapy for any orthopedic application is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
 
The use of allograft bone products containing viable stem cells, including but not limited to demineralized bone matrix with stem cells, does not meet primary coverage criteria for effectiveness.
 
For contracts without primary coverage criteria, use of allograft bone products containing viable stem cells, including but not limited to demineralized bone matrix with stem cells, is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
 
The use of allograft or synthetic bone graft substitutes that must be combined with autologous blood or bone marrow does not meet primary coverage criteria for effectiveness. This technology is currently being studied in ongoing clinical trials.
 
For contracts without primary coverage criteria, the use of allograft or synthetic bone graft substitutes that must be combined with autologous blood or bone marrow is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective Prior to August 2017:
 
The use of stem cell therapy in the treatment of avascular necrosis (AVN) of the femoral head, or the use of mesenchymal stem cell therapy for any orthopedic application, does not meet primary coverage criteria for effectiveness. This technology is currently being studied in ongoing clinical trials.
 
For contracts without primary coverage criteria the use of stem cell therapy in the treatment of avascular necrosis (AVN) of the femoral head, or the use of mesenchymal stem cell therapy for any orthopedic application is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective Prior to August 2016
The use of mesenchymal stem cell therapy for any orthopedic application does not meet primary coverage criteria for effectiveness.  This technology is currently being studied in ongoing clinical trials.  
 
For contracts without primary coverage criteria the use of mesenchymal stem cell therapy for any orthopedic application is considered investigational.  Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Allograft bone products containing viable stem cells, including but not limited to demineralized bone matrix (DBM) with stem cells, does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.
 
For members with contracts without primary coverage criteria, allograft bone products containing viable stem cells, including but not limited to demineralized bone matrix (DBM) with stem cells is considered investigational for all orthopedic applications. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective prior to April 2013
The use of mesenchymal stem cell therapy for any orthopedic application does not meet primary coverage criteria for effectiveness.  This technology is currently being studied in ongoing clinical trials.  
 
For contracts without primary coverage criteria the use of mesenchymal stem cell therapy for any orthopedic application is considered investigational.  Investigational services are an exclusion in the member benefit certificate.  

Rationale:
At this time, the literature consists almost entirely of review articles describing the potential of stem cell therapy for orthopedic applications in humans, along with basic science experiments on sources of mesenchymal stem cells (MSCs), regulation of cell growth and differentiation, and development of scaffolds (Deans, 2009).  Authors of these reviews indicate that the technology is in an early stage of development. In a literature search of the MEDLINE database in March 2010, use of cultured MSCs in humans was identified in only a few centers (U.S. and Asia).
 
Wakitani and colleagues (2002) first reported use of expanded MSCs for repair of cartilage defects.  Cells from bone marrow aspirate of 12 patients with osteoarthritic knees were culture expanded, embedded in collagen gel, transplanted into the articular cartilage defect and covered with autologous periosteum at the time of high tibial osteotomy. Clinical improvement was not found to be different between the experimental group and a group of 12 control patients who underwent high tibial osteotomy alone. Wakitani (2007) has since published several cases of patients treated for isolated cartilage defects, with clinical improvement reported at up to 27 months.  However, most of the defects appear to have been filled with fibrocartilage, and in the absence of controlled studies, the benefit of this procedure in comparison with established alternatives such as microfracture is unclear.
 
A 2010 article from Centeno et al. describes the use of percutaneously injected culture-expanded MSCs from the iliac spine in 226 patients.  Following harvesting, cells were cultured with autologous platelet lysate and re-injected under fluoroscopic guidance into peripheral joints (n=213) or intervertebral discs (n=13). Follow-up for adverse events at a mean of 10.6 months showed 10 cases of probable procedure-related complications (injections or stem cell related), all of which were considered to be self-limited or treated with simple therapeutic measures. Serial MRIs from a subset of patients showed no evidence of tumor formation at a median follow-up of 15 months. The efficacy of these procedures was not reported.
 
A search of ClinicalTrials.gov in March 2010 identified a number of trials on use of MSCs for orthopedic indications from both within and outside the U.S. The following is a sample of some of the larger studies:
 
A Phase I/II randomized, placebo controlled, double blind study of 2 doses of Chondrogen™ (Osiris Therapeutics) or a placebo intra-articular injection following meniscectomy in 60 patients is listed as completed in 2008 (NCT00225095). Chondrogen™ is a preparation of adult MSCs in a solution containing hyaluronic acid. Three-year follow-up of Chondrogen™ versus placebo injections is listed as a separate study (NCT00702741).
 
Medipost is sponsoring a randomized, open-label, multi-center phase III clinical trial to compare the efficacy and safety of Cartistem® and microfracture in patients with knee articular cartilage injury or defect (NCT01041001). MSCs will be isolated from umbilical cord blood and cultured, mixed with semi-solid polymer, and administered in the cartilage tissue lesion by orthopedic surgery. The study has an estimated enrollment of 104 patients with completion in 2011.
 
Orthofix is sponsoring a radiographic and clinical study evaluating a novel allogeneic, cancellous, bone matrix containing viable stem cell (Trinity Evolution Matrix,) in subjects undergoing tibiotalar arthrodesis with or without hind foot fusion (NCT00988338). The study has an estimated enrollment of 100 patients with completion in 2012.
 
NCT00885729 is a phase I randomized, single blind, active control trial of MSCs compared with chondrocytes to heal articular cartilage defects in 50 patients. The study is sponsored by an academic medical center in Norway. Both MSCs and chondrocytes will be delivered in a commercially available scaffold (not described). The estimated study completion date is 2018.
 
The American Association of Orthopaedic Surgeons (AAOS) states that stem cell procedures in orthopaedics are still at an experimental stage; most musculoskeletal treatments using stem cells are performed at research centers as part of controlled clinical trials, and results of studies in animal models provide proof-of-concept that in the future, similar methods could be used to treat osteoarthritis, nonunion of fractures, and bone defects in humans.
 
Overall, the literature suggests a technology that is at a very early stage of development, with the vast majority of studies focused on development of methods for tissue engineering along with preliminary testing in animal models. A number of clinical trials are in progress, but are not expected to be completed for several years. Current evidence on procedures using autologous bone marrow derived MSCs for orthopedic indications in humans consists of several case reports/case series, with insufficient data to evaluate health outcomes.
 
2012 Update
This policy is being updated with a literature search through July 2012.  There was no new information identified that would prompt a change in the coverage statement. A summary of the relevant literature is as follows.
 
A 2011 report from Wakitani et al. was a follow-up safety study of 31 of the 41 patients (3 patients had died and 5 had undergone total knee arthroplasty) who had received MSCs for articular cartilage repair in their clinics between 1998 and 2008 (Wakitani, 2011). Patients who could not return to clinic were interviewed by telephone to determine if there were any abnormalities in the operated joints. At a mean of 75 months (range, 5 to 137) since the index procedure, no tumors or infections were identified. Function was not reported. In the absence of controlled studies, the benefit of this procedure in comparison with established alternatives such as microfracture is unclear.
 
Another study from Asia compared the efficacy of bone marrow-derived MSCs and autologous chondrocyte implantation (ACI) in 36 matched patient pairs (Nejadnik, 2010). Thirty-six consecutive patients with at least 1 symptomatic chondral lesion on the femoral condyle, trochlea, or patella were matched with 36 cases of ACI performed earlier, based on lesion sites and 10-year age intervals. At baseline, the MSC group had a slightly (non-significantly) greater lesion size (4.6 cm2 vs. 3.6 cm2, respectively). The grade of the lesion was comparable for the 2 groups (30-33% grade 4). Autologous MSCs were cultured from 30 mL of bone marrow from the iliac crest and tested to confirm that the cultured cells were MSCs. The cultured chondrocytes or MSCs were implanted beneath a periosteal patch. Concomitant procedures included patella realignment, high-tibial osteotomy, partial meniscectomy, and anterior cruciate ligament reconstruction. Clinical outcomes were measured pre-operatively and at 3, 6, 12, 18, and 24 months’ after operation using the International Cartilage Repair Society (ICRS) Cartilage Injury Evaluation Package. The Evaluation Package included questions from the Short-Form-36 (SF-36) Health Survey, International Knee Documentation Committee (IKDC) subjective knee evaluation form, Lysholm knee scale, and Tegner activity level scale. Both treatments improved patients’ scores over the 2-year follow-up by mixed effect models. There was no significant difference between groups for any of the measures except for Physical Role Functioning on the SF-36, which showed a greater improvement over time in the MSC group. Although scatter plots were provided, neither the absolute improvement nor the proportion of patients with clinically significant improvement was reported. This study is also limited by the small number of patients.
In 2009, Giannini et al. reported a one-step procedure for transplanting bone marrow-derived cells for Type II (>1.5 cm2, <5 mm deep) osteochondral lesions of the talus in 48 patients (Giannini, 2009).  The mean age of the patients was 29 years. Fifteen of the patients had been previously treated by microfracture (n=8), debridement (n=5), or autologous chondrocyte transplantation (n=2). A total of 60 mL-bone marrow aspirate was collected from the iliac crest. The bone marrow-derived cells were concentrated in the operating room and implanted with a scaffold (collagen powder or hyaluronic acid membrane) and platelet gel. Twenty-two patients underwent associated surgical procedures (osteophytectomy, synovectomy, loose body extraction, or calcaneal osteotomy). Active and passive ankle motions were advised beginning the day after surgery; no weight bearing was recommended for 6 weeks. Patients were evaluated at 6, 12, 18, and 24 months after surgery with standard radiographs and magnetic resonance imaging (MRI). At 24-month follow-up, the mean American Orthopaedic Foot and Ankle Society (AOFAS) score had changed from 64.4 to 91.4, representing a meaningful improvement. The AOFAS score at follow-up was affected by the area of the lesion and previous surgeries. MRI showed newly formed tissue at the lesion site in all patients. Histologic evaluation in the first 3 patients (asymptomatic) showed regenerated tissue in various degrees of remodeling, although none showed entirely hyaline cartilage. Two additional patients were reoperated due to hypertrophic regenerated tissue. Integration with the healthy cartilage was reported to be complete in all 5 patients with a smooth transition zone.
 
The use of mesenchymal stem cell therapy for orthopedic applications continues to be studied in the following clinical trials:
    • A Phase I/II randomized, placebo controlled, double blind study of 2 doses of Chondrogen™ (Osiris Therapeutics) or a placebo intra-articular injection following meniscectomy in 60 patients is listed as completed in 2008 (NCT00225095). Chondrogen™ is a preparation of adult MSCs in a solution containing hyaluronic acid. Three-year follow-up of Chondrogen™ versus placebo injections is listed as a separate study (NCT00702741).
    • Medipost is sponsoring a randomized, open-label, multicenter Phase III clinical trial to compare the efficacy and safety of Cartistem® and microfracture in patients with knee articular cartilage injury or defect (NCT01041001). MSCs will be isolated from umbilical cord blood and cultured, mixed with semi-solid polymer, and administered in the cartilage tissue lesion by orthopedic surgery. The study has an estimated enrollment of 104 patients with completion in 2011. Preliminary results of this study were presented at the annual meeting of the American Academy of Orthopaedic Surgeons in February 2012.
    • Orthofix is sponsoring a radiographic and clinical study evaluating a novel allogeneic, cancellous, bone matrix containing viable stem cells (Trinity Evolution Matrix) in subjects undergoing tibiotalar arthrodesis with or without hind-foot fusion (NCT00988338). The study has an estimated enrollment of 100 patients with completion in 2012.
    • NCT00885729 is a Phase I randomized, single-blind, active control trial of MSCs compared with chondrocytes to heal articular cartilage defects in 50 patients. The study is sponsored by an academic medical center in Norway. Both MSCs and chondrocytes will be delivered in a commercially available scaffold (not described). The estimated study completion date is 2018.
    • The National University of Malaysia is sponsoring a randomized controlled trial of intra-articular MSC injection versus hyaluronic acid in patients with osteoarthritis (NCT01459640). The study has an estimated enrollment of 50 patients with completion in 2014.
 
2013 Update
This policy is being updated with a literature review through March 2013. The following is a summary of the key literature identified.
 
Cartilage Defects
 
Caritilage Defects: Adipose-derived MSCs
In 2012, Koh et al. reported a retrospective analysis of the injection of adipose-derived MSCs and platelet-rich plasma (PRP) into arthroscopically-debrided knees of 25 patients with osteoarthritis (Koh, 2012). Results were compared with a randomly selected group of patients who had previously undergone arthroscopic debridement and PRP injections without stem cells. Although there was a trend for greater improvement in the MSC group, at final follow-up there was no significant difference between the MSC and control groups in clinical outcomes (Lysholm, Tegner, visual analog score). Use of PRP is discussed separately in policy No 2009049.
 
Caritilage Defects: MSCs from Peripheral Blood
A 2013 report from Asia described a small randomized controlled trial with autologous peripheral blood MSCs for focal articular cartilage lesions (Saw, 2013). Fifty patients with grade 3 and 4 lesions of the knee joint underwent arthroscopic subchondral drilling followed by 5 weekly injections of hyaluronic acid. Half of the patients were randomly allocated to receive injections of peripheral blood stem cells or no further treatment. There were baseline differences in age between the groups, with a mean age of 38 years for the treatment group compared to 42 for the control group. The peripheral blood stem cells were harvested after stimulation with recombinant human granulocyte colony-stimulating factor, divided in vials, and cryopreserved. At 6 months after surgery, hyaluronic acid and MSC were re-administered over 3 weekly injections. At 18 months after surgery, second look arthroscopy on 16 patients in each group showed significantly higher histological scores (by about 10%) for the MSC group (1,066 vs. 957 by independent observers) while blinded evaluation of magnetic resonance imaging (MRI) showed a higher morphologic score (9.9 vs. 8.5). There was no difference in International Knee Documentation Committee (IKDC) scores between the 2 groups at 24 months after surgery. It is uncertain how differences in patient age at baseline may have affected the response to subchondral drilling.
 
Conclusions-The evidence base on MSCs for cartilage repair is increasing, although as of March 2013 only one study was identified that was randomized. This small randomized study, which is limited by group differences in age at baseline, is also the only comparative study to show an improvement in histological and morphologic outcomes. No study to date has shown an improvement in functional outcomes following treatment with MSCs for cartilage repair.
 
Fusion and Non-union
There is limited evidence on the use of allografts with stem cells for fusion of the extremities or spine or for the treatment of non-union. One retrospective series from 2009 was identified on the use of Trinity Evolution Matrix MSC bone allograft for revision surgery of the foot and ankle (Rush, 2009). Twenty-three patients were included who had undergone revision foot and/or ankle surgery for residual malunion, non-union, or significant segmental bone loss. Patients were followed to the point of radiographic and clinical union, which occurred at a median of 72.5 days for 21 of the 23 patients (91.3%).
 
Osteonecrosis
Two randomized comparative trials from Asia have been identified that evaluated the use of MSCs for osteonecrosis of the femoral head.
 
Osteonecrosis: MSCs Expanded from Bone Marrow
In 2012, Zhao et al. reported a randomized trial that included 100 patients (104 hips) with early stage femoral head osteonecrosis treated with core decompression and expanded bone marrow MSCs versus core decompression alone (Zhao, 2012)  At 60 months after surgery, 2 of the 53 hips (3.7%) treated with MSCs progressed and underwent vascularized bone grafting, compared with 10 of 44 hips (23%) in the decompression group who progressed and underwent either vascularized bone grafting (n=5) or total hip replacement (n=5). The MSC group also had improved Harris Hip Scores compared with the control group on independent evaluation (data presented graphically). The volume of the lesion was also reduced by treatment with MSCs.
 
Osteonecrosis: MSCs Concentrated from Bone Marrow
Another small trial randomized 40 patients (51 hips) with early stage femoral head osteonecrosis to core decompression plus concentrated bone marrow MSCs or core decompression alone (Sen, 2012).  Blinding of assessments in this small trial was not described. Harris Hip Score was significantly improved in the MSC group (scores of 83.65 and 82.42) compared with core decompression (scores of 76.68 and 77.39). Kaplan-Meier analysis showed improved hip survival in the MSC group (mean of 51.9 weeks) compared to the core decompression group (mean of 46.7 weeks). There were no significant differences between the groups in the radiographic assessment or MRI results.
 
Conclusions-Two small studies from Asia have compared core decompression alone versus core decompression with MSCs in patients with osteonecrosis of the femoral head. Both studies reported improvement in the Harris Hip Score in patients treated with MSCs, although it was not reported whether the patients or investigators were blinded to the treatment group. Hip survival was significantly improved following treatment with either expanded or concentrated MSCs. The effect appears to be larger with expanded MSCs compared to concentrated MSCs. Additional studies with a larger number of patients are needed to permit greater certainty regarding the effect of this treatment on health outcomes.
 
Ongoing Clinical Trials
A search of online site: ClinicalTrials.gov in March 2013 identified a number of trials on use of MSCs for orthopedic indications from both within and outside the U.S. The following is a sample of some of the larger studies:
    • A Phase I/II randomized, placebo controlled, double blind study of 2 doses of Chondrogen™ (Osiris Therapeutics) or a placebo intra-articular injection following meniscectomy in 60 patients is listed as completed in 2008 (NCT00225095). Chondrogen™ is a preparation of adult MSCs in a solution containing hyaluronic acid. Three-year follow-up of Chondrogen™ versus placebo injections is listed as a separate study (NCT00702741). The status of this trial is unknown.
    • Medipost is sponsoring a randomized, open-label, multicenter Phase III clinical trial to compare the efficacy and safety of Cartistem® and microfracture in patients with knee articular cartilage injury or defect (NCT01041001). MSCs will be isolated from umbilical cord blood and cultured, mixed with semi-solid polymer, and administered in the cartilage tissue lesion by orthopedic surgery. The study is listed as completed as of April 2012 with an enrollment of 104 patients. Preliminary results of this study were presented at the annual meeting of the American Academy of Orthopaedic Surgeons in February 2012. As of March 2013, no peer-reviewed publications from this trial have been identified.
    • Medipost is sponsoring a 60-month follow-up study (NCT01626677) of the patients who participated in the Phase III trial of Cartistem® (NCT01041001). The study has an estimated enrollment of 103 patients with completion in May 2015.
    • NCT00885729 is a Phase I randomized, single-blind, active control trial of MSCs compared with chondrocytes to heal articular cartilage defects in 50 patients. The study is sponsored by an academic medical center in Norway. Both MSCs and chondrocytes will be delivered in a commercially available scaffold (not described). The estimated study completion date is 2018.
    • The National University of Malaysia is sponsoring a randomized controlled trial of intra-articular MSC injection versus hyaluronic acid in patients with osteoarthritis (NCT01459640). The study has an estimated enrollment of 50 patients with completion in 2014.
    • Three series are listed with Trinity Evolution Matrix for foot and ankle surgery, anterior cervical discectomy and fusion (ACDF), and posterior or transforaminal lumbar interbody fusion (PLIF or TLIF). All 3 studies are listed as ongoing but not recruiting subjects.
 
2014 Update
A search of the MEDLINE database conducted through March 2014 did not reveal any new information that would prompt a change in the coverage statement. The following is a summary of the key identified literature.
 
In December 2013 (after the systematic review by Filardo et al was published), Wong et al reported an RCT of cultured MSCs in 56 patients with osteoarthritis who underwent medial opening-wedge high tibial osteotomy and microfracture of a cartilage lesion (Wong, 2013). Bone marrow was harvested at the time of microfracture and the MSCs were isolated and cultured. After 3 weeks, the cells were assessed for viability and delivered to the clinic, where patients received an intra-articular injection of MSCs suspended in hyaluronic acid (HA), or for controls, intra-articular injection of HA alone. The primary outcome was the International Knee Documentation Committee (IKDC) score at 6 months, 1 year, and 2 years. Secondary outcomes were the Tegner and Lysholm scores through 2 years and the Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) scoring system by MRI at 1 year. All patients completed the 2-year follow-up. After adjusting for age, baseline scores, and time of evaluation, the group treated with MSCs showed significantly better scores on the IKDC (mean difference 7.65 on 0-100 scale, p=0.001), Lysholm (mean difference 7.61 on 0-100 scale, p=0.02), and Tegner (mean difference 0.64 on a 0-10 scale, p=0.02). Blinded analysis of MRI results found higher MOCART scores in the MSC group. The group treated with MSCs had a higher proportion of patients who had complete cartilage coverage of their lesions (32% vs 0%), greater than 50% cartilage cover (36% vs 14%) and complete integration of the regenerated cartilage (61% vs 14%). This study is ongoing and recruiting additional patients.
 
In 2014, Vangsness et al reported an industry-sponsored Phase I/II randomized, double-blind, multicenter study (NCT00225095, NCT00702741) of cultured allogeneic MSCs (Chondrogen™, Osiris Therapeutics) injected into the knee after partial meniscectomy (Vangsness, 2014). The 55 patients in this U.S. study were randomized to intra-articular injection of either 50 x 106 allogeneic MSCs, 150 x 106 allogeneic MSCs in HA, or HA vehicle control at 7 to 10 days after meniscectomy. The cultured MSCs were derived from bone-marrow aspirates from unrelated donors. At 2-year follow-up, 3 patients in the low-dose MSC group had significantly increased meniscal volume measured by MRI (with an a priori determined threshold of at least 15%) compared with none in the control group and none in the high-dose MSC group. There was no significant difference between the groups in the Lysholm Knee Scale. On subgroup analysis, patients with osteoarthritis who received MSCs had a significantly greater reduction in pain at 2 years compared with patients who received HA alone. This appears to be a posthoc analysis and should be considered preliminary. No serious adverse events were thought to be related to the investigational treatment.
 
Ongoing and Unpublished Clinical Trials
A search of online site ClinicalTrials.gov in March 2014 identified a number of trials on use of MSCs for orthopedic indications from both within and outside the U.S. The following is a sample of some of the larger studies:
 
    • Medipost is sponsoring a randomized, open-label, multicenter Phase III clinical trial to compare the efficacy and safety of Cartistem® and microfracture in patients with knee articular cartilage injury or defect (NCT01041001). MSCs will be isolated from umbilical cord blood and cultured, mixed with semisolid polymer, and administered in the cartilage tissue lesion by orthopedic surgery. The study is listed as completed as of April 2012 with an enrollment of 104 patients. Preliminary results of this study were presented at the annual meeting of the American Academy of Orthopaedic Surgeons in February 2012. As of March 2014, no peer-reviewed publications from this trial have been identified.
    • Medipost is sponsoring a 60-month follow-up study (NCT01626677) of the patients who participated in the Phase III trial of Cartistem® (NCT01041001). The study has an estimated enrollment of 103 patients with completion in May 2015.
    • NCT00885729 is a Phase I randomized, single-blind, active control trial of MSCs compared with chondrocytes to heal articular cartilage defects in 50 patients. The study is sponsored by an academic medical center in Norway. Both MSCs and chondrocytes will be delivered in a commercially available scaffold (not described). The estimated study completion date is 2018.
    • The National University of Malaysia is sponsoring an RCT of intra-articular MSC injection versus HA in patients with osteoarthritis (NCT01459640). The study has an estimated enrollment of 50 patients with completion in 2014. The status of this study is unknown.
    • Three large series are listed with Trinity Evolution Matrix (Orthofix) for foot and ankle surgery, anterior cervical discectomy and fusion, and posterior or transforaminal lumbar interbody fusion. All 3 studies are listed as ongoing but not recruiting subjects.
    • NCT01413061 is a randomized comparative trial of Allostem® (AlloSource) versus autologous bone marrow aspirate in subtalar arthrodesis procedures. The study has an estimated enrollment of 136 patients with completion expected in 2016.
    • Five large multicenter series are posted for Osteocel® Plus (NuVasive) covering 5 different approaches to lumbar and cervical spinal fusion (ie, transformaminal, anterior, posterior, lateral). All 5 studies are listed as completed. No publications from these trials have been identified and no results have been posted.
 
2015 Update
A literature search conducted through May 2015 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Adipose-Derived MSCs
The literature on adipose-derived MSCs for articular cartilage repair comes from 2 different research groups in Korea. One of the groups appears to have been providing this treatment as an option for patients for a number of years. They compare outcomes of this new add-on treatment with those of patients who only received other cartilage repair procedures.
 
In 2014, Koh and colleagues reported results of an RCT that evaluated cartilage healing after high tibial osteotomy (HTO) in 52 patients with osteoarthritis of the medial compartment (Koh, 2014). Patients were randomly assigned via sealed envelopes to HTO with application of platelet-rich plasma (PRP) or HTO with application of PRP plus MSCs. (Use of PRP is considered investigational, see Policy No. 2.01.16 for further information.) MSCs from adipose tissue were obtained through liposuction from the buttocks. The tissue was centrifuged and the stromal vascular fraction mixed with PRP for injection. A total of 44 patients completed second look arthroscopy and 1 and 2 year clinical follow-up. The primary outcomes were the Knee Injury and Osteoarthritis Outcome Score (KOOS, five subscales with 0-100 scale), the Lysholm score (0-100 scale), and a VAS pain scale (0-100 scale). There were statistically significant differences for PRP only versus PRP+MSC on 2/5 KOOS subscales; pain (74±5.7 vs. 81.2±6.9, p<0.001) and symptoms (75.4±8.5 vs. 82.8±7.2, p=0.006). There were also statistically significant differences on the final pain score for the PRP only versus PRP+MSC groups (16.2±4.6 vs. 10.2±5.7, p<0.001), but the final Lysholm score was not significantly different between the PRP only and PRP+MSC groups (80.6±13.5 vs. 84.7±16.2, all respectively, p=0.36). Articular cartilage healing was rated as improved with MSCs following video review of second-look arthroscopy; blinding of this measure is unclear. There are a number of limitations of this study, including the small sample size the short duration of follow-up, and significant improvements on only some of the outcomes. All of the significant differences in outcomes were modest in magnitude, and as a result there is uncertainty regarding the clinical significance of the findings.
 
A 2014 retrospective review from Kim and colleagues reported clinical outcomes and MRI results from 49 patients who had undergone marrow stimulation with or without MSCs at their institution (Kim, 2014). The use of MSCs in addition to microfracture was determined by patient choice, and there was an overlap of 26 patients between this report and their 2013 publication above (Koh, 2014). This analysis also found modest but statistically significant improvements in clinical outcomes for the MSC group compared to microfracture alone. Blinded ratings with the Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) scale resulted in a score of 49.4 for the conventional group and 62.1 for the MSC group (p=0.037).
 
Another group reported a Phase I/II trial of intra-articular injection of adipose-derived MSCs for the treatment of osteoarthritis of the knee (Jo, 2014). Phase I was a dose escalation study in 9 patients and Phase II assessed efficacy of the highest dose in 9 patients. The study of 18 patients was approved by the Korean Food and Drug Administration. Procedures included liposuction, arthroscopy of the knee one week later with MSC injection through the portal, MRI at 3 and 6 months, and second-look arthroscopy with punch biopsy at 6 months. Intent-to-treat analysis showed a 39% improvement in WOMAC at 6 months after injection and a 45% improvement in VAS. Arthroscopy showed a decrease in size of the cartilage defect and an increase in the volume of cartilage. Histology showed thick, hyaline-like cartilage regeneration. Additional study is needed with a larger sample size, sham-treated controls, and longer follow-up.
 
MSCs from Synovial Tissue
Akgun and colleagues reported a small (N=14), though without major bias, investigator-blinded RCT that compared matrix-induced autologous MSCs from synovial tissue versus matrix-induced autologous chondrocyte implantation (MACI) (Akgun, 2015). Both chondrocytes from cartilage and MSCs from synovia were harvested in an arthroscopic procedure, expanded in culture, and then cultured on a collagen membrane for 2 days. Implantation was performed with the construct trimmed to the size and shape of the defect and placed with the cells facing the subchondral bone. Rehabilitation was the same for the 2 groups, with continuous passive motion (CPM) for at least 1 hour daily and non-weight bearing for the first 6 weeks. The 2 groups were similar at baseline, and all patients completed the evaluations through 24 months. Outcomes on the KOOS subscales and Tegner activity score were statistically better in the MSC group, although it is not clear if the difference observed would be considered clinically significant, with differences of around 6 on the 100 point KOOS subscales and 0.6 on the 10 point Tegner. The results of this small pilot study do suggest that cartilage repair with matrix-induced MSCs from synovial tissue may result in outcomes that are at least as good as MACI, warranting additional study in a larger sample. It should also be noted that neither of these procedures is approved for use in the U.S.
 
In summary, a small RCT found that MSCs from synovial tissue and cultured on collagen resulted in outcomes that were at least as good as those following MACI.  The literature on adipose-derived MSCs includes a Phase I/II study with cultured MSCs and an RCT from a separate group in Asia that have been using uncultured MSCs as an adjunctive procedure in clinical practice. Comparisons between patients who have and have not received uncultured adipose-derived MSCs shows modest improvement in health outcomes that are of uncertain clinical significance. Potential for bias from non-blinded use of a novel procedure on subjective outcome measures is also a limitation of these studies. The Phase I/II study of cultured MSCs from adipose tissue shows promising results for this technology. Additional study in a larger sample of patients with longer follow-up is needed to evaluate the long term efficacy and safety of the procedure. FDA approval for this method has also not been obtained.
 
Spinal Fusion
There is limited evidence on the use of allografts with stem cells for fusion of the extremities or spine or for the treatment of nonunion, although several large observational studies are ongoing (see Table 1). In 2014, Eastlack and colleagues reported outcomes from a series of 182 patients who were treated with anterior cervical discectomy and fusion using Osteocel Plus in a PEEK cage and anterior plating (Eastlack, 2014). At 24 months, 74% of patients (180 out of 249 levels treated) were available for follow-up. These patients had significant improvements in clinical outcomes; 87% of levels achieved solid bridging and 92% of levels had range of motion less than 3º. With 26% loss to follow-up at 24 months and lack of a standard of care control group, interpretation of these results is limited.
 
Ongoing and Unpublished Clinical Trials
A 2014 review on FDA regulations of adult stem cell therapies for sports medicine identified over 45 ongoing clinical trials on this topic (Chirba, 2015). Some of the currently ongoing and unpublished trials that might influence this policy are listed in below. Many are observational studies with commercially available products (Cartistem®, AlloStem®, Trinity Evolution™, and Osteocel® Plus).
 
Ongoing
  • (NCT01626677 / NCT01041001a) Randomized, Open-Label Multi-Center and Phase 3 Clinicl Trial to Compare the Efficacy and Safety of Cartistem® and Microfracture in Patients with Knee Articular Cartilage Injury or Defect / Long Term Follow-Up Study of CARTISTEM® Versus Microfracture; planned enrollment 104; completion date May 2015.
  • (NCT00885729) Mesenchymal Stem Cells in a Clinical Trial to Heal Articular Cartilage Defects; planned enrollment 50; completion date 2018.
  • (NCT01413061a) Study of Subtalar Arthrodesis Using AlloStem® Versus Autologous Bone Graft; planned enrollment 140; completion date February
 
Unpublished
  • (NCT00965380a) A Radiographic and Clinical Study Evaluating a Novel Allogeneic, Cancellous, Bone Matrix Containing Viable Stem Cells (Trinity Evolution™ Viable Cryopreserved Cellular Bone Matrix) in Posterior Lumbar or Transforaminal Lumbar Interbody Fusion (PLIF or TLIF); planned enrollment 200; completion date June 2014.
  • (NCT00988338a) A Radiographic and Clinical Study Evaluating a Novel Allogeneic, Cancellous, Bone Matrix Containing Viable Stem Cells (Trinity Evolution™ Matrix) in Subjects Undergoing Foot and Ankle Fusion; planned enrollment 106; completion date January 2013.
  • (NCT00951938a) A Radiographic and Clinical Study Evaluating a Novel Allogeneic, Cancellous, Bone Matrix Containing Viable Stem Cells (Trinity Evolution™ Viable Cryopreserved Cellular Bone Matrix) in Patients Undergoing Anterior Cervical Discectomy and Fusion; planned enrollment 200; completion date August 2012.
  • (NCT0094853a) Osteocel® Plus in extreme Lateral Interbody Fusion (XLIF®): Evaluation of Radiographic and Patient Outcomes; planned enrollment 104; completion date October 2012.
  • (NCT00948831a) Osteocel® Plus in Anterior Lumbar Interbody Fusion (ALIF): Evaluation of Radiographic and Patient Outcomes
  
2016 Update
A literature search conducted through July 2016 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Allogeneic Bone Marrow
In 2015, Vega and colleagues reported a small phase I/II RCT of 30 patients with osteoarthritis unresponsive to conventional treatments (Vega, 2015). The MSC-treated group received intra-articular injection of expanded allogeneic bone marrow MSCs from healthy donors and the control group received an intra-articular injection of hyaluronic acid (HA). Follow-up using standard outcome measures was performed at 3, 6, and 12 months after injection. In the MSC-treated group, pain scores (VAS and WOMAC [Western Ontario and McMasters Universities Arthritis Index]) decreased significantly between baseline and the 12-month follow-up, whereas pain scores in the control group did not improve significantly. A significant improvement in cartilage in the MSC group was supported by T2 magnetic resonance imaging (MRI). It was not reported if the patients or assessors were blinded to treatment in this phase I/II study. Additional study in a larger number of patients in a phase III trial with blinding of patients and assessors is needed.
 
Centeno and colleagues reported a multi-center registry of patients treated with autologous stem cells, bone marrow concentrate, and platelet-rich plasma (Centeno, 2015).  This report focused on 102 patients (115 shoulders) diagnosed with either osteoarthritis of the shoulder or rotator cuff tears. Patients were treated with a protocol that included a hypertonic dextrose solution (prolotherapy) injection to create an inflammatory response several days prior to the bone marrow concentrate injection. The bone marrow concentrate injection included platelet-rich plasma and platelet lysate. Both DASH (disabilities of the arm, shoulder, and hand score) and numeric pain scores (NPS) decreased by about 50%, although the absolute decrease in the NPS was a very modest 0.9. Interpretation of these results is limited by the lack of a placebo control and blinding, subjective outcome measures, and the multiple treatments used; although it is acknowledged that neither prolotherapy nor PRP appear to have efficacy on their own. Additional study with randomized and placebo-controlled trials is needed to evaluate this treatment protocol.
 
Lu and colleagues published a systemic review on stem cell therapy for the treatment of early stage avascular necrosis of the femoral head (Lau, 2014). Avascular necrosis (AVN) of the femoral head (FH) is believed to be caused by a multitude of etiologic factors and is associated with significant morbidity in younger populations. Eventually, the disease progresses and results in FH collapse. A key focus is on early disease management aimed at joint preservation by preventing or delaying progression. The use of stem cells (SC) for the treatment of AVN of the FH has been proposed. We undertook a systematic review of the medical literature examining the use of SC for the treatment of early stage (precollapse) AVN of the FH, in both pre-clinical and clinical studies. Data collected included: Pre-clinical studies - model of AVN, variety and dosage of SC, histologic and imaging analyses. Clinical studies - study design, classification and etiology of AVN, SC dosage and treatment protocol, incidence of disease progression, patient reported outcomes, volume of necrotic lesion and hip survivorship. In pre-clinical studies, the use of SC uniformly demonstrated improvements in osteogenesis and angiogenesis, yet source of implanted SC was variable. In clinical studies, groups treated with SC showed significant improvements in patient reported outcomes; however hip survivorship was not affected. Discrepancies regarding dose of SC, AVN etiology and disease severity were present. Routine use of this treatment method will first require further research into dose and quality optimization as well as confirmed improvements in hip survivorship.
 
2017 Update
A literature search conducted through July 2017 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Shapiro and colleagues reported on the results of a prospective, single-blind, placebo-controlled trial assessing 25 patients with bilateral knee pain from bilateral osteoarthritis (Shapiro, 2017). Patients were randomized to BMAC into 1 knee and to saline placebo into the other. Fifty-two milliliters of bone marrow was aspirated from the iliac crests and concentrated in an automated centrifuge. The resulting BMAC was combined with platelet-poor plasma for an injection into the arthritic knee and was compared with a saline injection into the contralateral knee, thereby using each patient as his or her own control. Safety outcomes, pain relief, and function as measured by Osteoarthritis Research Society International (OARSI) measures and a visual analog scale (VAS) score were tracked initially at 1 week, 3 months, and 6 months postprocedure. Study patients experienced a similar relief of pain in both BMAC- and saline-treated arthritic knees. This trial is important because some previously reported studies have used injection of high-molecular weight HA derivatives as an active comparator. HA derivatives have been used for viscosupplementation in arthritic knee joints. However, a systematic review, prepared for the Agency for Healthcare Research and Quality on effectiveness of HA in the treatment of degenerative joint disease in knee joints, reported that no conclusions can be drawn from available literature on avoidance or delay of total knee replacement with the use of HA (Newberry, 2015). Guideline updates on viscosupplementation are included in that section of this evidence review.
 
Whitehouse and colleagues published a report on techniques of in vitro expansion of autologous-derived MSCs and a case series of first-in-human implantation to treat meniscal defects in 5 patients (Whitehouse, 2017). The regulatory framework in the U.K. allows cell manipulation and requires immunohistochemical documentation of the presence and volume of mesenchymal cells. Over the first 12 months postprocedure, 3 of the 5 patients were reported to have clinical symptom relief, which persisted through 24 months. MRI scans showing lack of meniscal displacement were the only other postoperative assessment. The 2 patients who failed to obtain symptom relief at 6 and 12 month had repeat arthroscopic procedures with meniscectomy.
 
A prospective, clinical, and radiographic 12-month outcomes study of patients undergoing single-level anterior cervical discectomy and fusion (ACDF) for symptomatic cervical degenerative disc disease using a novel viable allogeneic stem cell and cancellous bone matrix (Trinity Evolution) was reported using comparison to historical controls (Vanichkachorn, 2016).
The ACDF procedure was performed using the polyetheretherketone (PEEK) interbody spacer and bone graft substitute (Trinity Evolution) in 31 patients at multiple clinical sites. At 6 and 12 months, the primary end point of radiographic fusion was evaluated as determined by independent radiographic review and the fusion rate was 78.6% at 6 months and 93.5% at 12 months. Secondary endpoints included function as assessed by Neck Disability Index scores, and neck and arm pain as assessed by individual VAS scores. Neck function and neck and arm pain were reported as significantly improved at both 6 and 12 months postprocedure. Reported adverse events included carpal tunnel syndrome, minor pain, numbness, permanent and/or unresolved pain, and swelling. Independent medical adjudication of the 26 adverse events occurring in 31 patients found that no adverse events definitely or probably related to Trinity Evolution. However, 5 adverse events were found to be possibly related to Trinity Evolution with 3 events of mild severity and 2 of moderate severity.
 
A similar study involving several of the same investigators and clinical sites reported on the clinical and radiographic evaluation of an allogeneic bone matrix containing stem cells (Trinity Evolution Viable Cellular Bone Matrix) in patients undergoing 2-level ACDF (Peppers, 2017). This study involved 40 patients exposed to the ACDF and bone graft substitute procedure at 2 adjacent disc levels. A panel blinded to clinical outcomes reviewed 12-month dynamic motion plain radiographs and thin-cut computed tomography with multiplanar reconstruction. At 12 months, the per-subject and per-level fusion rates were 89.4% and 93.4%, respectively. The clinical function assessments using Neck Disability Index and VAS scores were reported to have improved from baseline.
 
A 2015 prospective, multicenter, open-label clinical trial using a cryopreserved, donor mesenchymal cell scaffold (Trinity Evolution) was performed in subjects undergoing foot and/or ankle arthrodesis with the surgeons preferred technique (Jones, 2015). A total of 103 subjects were prospectively enrolled at 10 participating sites. No restrictions were placed on the diagnosis, which included arthritis (primary osteoarthritis, posttraumatic osteoarthritis, and rheumatoid), deformity, neuropathy (Charcot and diabetic), revision surgery, and degenerative joint disease, and arthrodesis was performed in 171 joints. The per-protocol population consisted of 92 patients at 6 months and 76 patients at 12 months, with 153 and 129 total arthrodeses, respectively. The primary end point was fusion at 6 months, as assessed from computed tomography scans and standard radiographs by an independent radiology consultant. At 6 months fusion rate was 68.5% in 81.1% of joints. American Orthopaedic Foot and Ankle Society Hindfoot Scale scores for disability improved over time.
 
ONGOING AND UNPUBLISHED CLINICAL TRIALS
A search of ClinicalTrials.gov in July 2017 did not identify any ongoing or unpublished trials that would likely influence this review.
 
2018 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2018. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
MSCs Expanded From Bone Marrow
 
Autologous Bone Marrow for Treatment of Osteoarthritis
A 2017 systematic review by Borakati et al included 13 studies assessed patients with osteoarthritis who were treated with MSCs or with a control treatment that was identical other than the inclusion of MSCs (ie, studies using chondrogenic cellular therapy as control were not included) (Borakati, 2017). Pain assessment results were noted for each of the controlled studies, resulting in a pooled standardized mean difference of -1.27 (95% confidence interval, -1.95 to -0.58) in favor of the group treated with MSCs. Reviewers reported a Z-statistic effect size of 3.62, again in favor of the groups treated with MSCs (p<0.001); although they noted high heterogeneity across controlled studies (I2=92%). Additionally, 34 uncontrolled studies (n=737 patients) were summarized and evaluated qualitatively: reviewers noted consistent cartilage regrowth and reduction of pain following treatment with MSCs in these studies; however, because pain medication was often given concurrently, interpretation of the latter outcome is limited.
 
Adipose-Derived MSCs
In 2014, another group reported a phase 1/2 trial of intra-articular injection of adipose-derived MSCs for the treatment of osteoarthritis of the knee (Jo, 2014). Phase 1 was a dose-escalation study of 9 patients and phase 2 assessed efficacy of the highest dose in another 9 patients. The study of 18 patients was approved by the Korean Food and Drug Administration. Procedures included liposuction, arthroscopy of the knee 1 week later with MSC injection through the portal, MRI at 3 and 6 months, and second-look arthroscopy with punch biopsy at 6 months. Intention-to-treat analysis showed a 39% improvement in Western Ontario and McMaster Universities Arthritis Index score and a 45% improvement in VAS score at 6 months postinjection. The knee section of the Knee Society clinical rating system showed significant increases in the low-dose group (91%) and in the high-dose group (50%), compared with baseline (p=0.025 and p<0.001, respectively). The function section of Knee Society clinical rating System was also significantly improved for the low-dose group (39%; p=0.020). Arthroscopy showed a significant decrease in the size of the cartilage defect: in four of the domains assessed (medial femoral and tibial condyles and lateral femoral and tibial condyles), decreases in defect ranged from 40% to 51%. There was also an increase in the volume of cartilage in the medial femoral condyle for both high- and low-dose groups, although this change was only significant in the low-dose patients, who showed a 27% increase (p=0.26). For the high-dose group and other domains, changes in cartilage volume was insignificant over 6 months. Histology showed thick, hyaline-like cartilage regeneration. The results of 2-year follow-up are reported below, as are the limitations of both phases of the trial.
  
The 2-year follow-up of this phase 1/2 trial was published by Jo et al in 2017; functional outcomes were assessed by Western Ontario and McMaster Universities Osteoarthritis Index, Knee Society clinical rating System, KOOS, and VAS; almost exclusively, the high-dose group showed significantly improved scores at 2 years compared with baseline and 1-year follow-up, while the medium- and low-dose groups showed insignificant changes or deterioration from 1-year to 2-year follow-up (Jo, 2017). In the high-dose group, the Western Ontario and McMaster Universities Osteoarthritis Index score decreased from baseline 54.2 to 16.0 at 1-year follow-up; after 2 years, the score was 19.0, which reflected the tendency of high-dose patients to improve early in treatment and remain at a stable level of improvement. Those in the medium- and low-dose groups, on the other hand, were more likely to show signs of deterioration after 2 years, a finding supported by structural measures (eg, no significant changes in cartilage defects were reported for these patients, as assessed by MRI). In the high-dose group, measurement of the medial femoral defect showed a 49.4% decrease after 2 years (p=0.005), and measurement of lateral tibial condyles defect showed a 64.4% decrease (p=0.037); additionally, no adverse events were reported, prompting investigators to call for more randomized trials of the treatment. Among the trial’s limitations were a lack of a control group and the absence of data at 1 year for both MRI results and patients receiving a medium stem cell dose. Another factor potentially influencing results was the use of arthroscopic exploration with lavage.
 
2019 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2019. No new literature was identified that would prompt a change in the coverage statement.
 
2020 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2020. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Emadedin et al reported a triple-blind placebo-controlled phase 1/2 trial of expanded MSCs in 47 patients with OA of the knee (Emadedin, 2018). Compared to the placebo group, the MSC group showed statistically significant improvements in WOMAC pain and function subscales but not VAS. The WOMAC stiffness subscale improved to a similar extent in the 2 groups. Minimum Clinically Important Improvement and Patient Acceptable Symptom State were not significantly different between the 2 groups. Study limitations included the short duration of follow-up, statistical analysis, and lack of information regarding use of analgesic medications.
 
Another phase 1/2 RCT of expanded MSCs was reported by Lamo-Espinosa et al in 30 patients with OA of the knee (Lamo-Espinosa, 2016 and Lamo-Espinosa, 2018). A Two doses of MSCs (10x106, 100x106) were administered with HA and compared to injection of HA alone. VAS scores were significantly decreased in both MSC groups compared to baseline throughout the 12 months of follow-up, while the decrease in VAS in the control group was not statistically significant. Similarly, total WOMAC scores were statistically decreased only in the high dose group at 12 months. Four-year follow-up was available for 27 of the 30 participants. Two patients in the control group and 1 patient in the low dose group had undergone total knee arthroplasty. VAS scores group were higher than at baseline in the HA control but remained low in the 2 MSC groups. WOMAC scores at the long-term follow-up showed a similar course.
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
A systematic review and meta-analysis by Maheshwer et al identified 25 studies with 439 participants that used MSCs for treatment of OA (Maheshwer, 2021). Although 13 studies were considered level I RCTs by the authors (range of 7 to 40 participants), low quality RCTs would normally be downgraded to level II. Meta-analysis suggested improvement in self-reported function, but only in patients who underwent concomitant surgery, and there was no significant improvement in pain. Few studies reported on cartilage quality. Most of the studies were rated as poor or fair quality. Conclusions are limited due to substantial variability in MSC source, preparation, and concentration in the current literature.
 
A more focused systematic review and meta-analysis of 6 RCTs (203 patients) that evaluated cultured MSCs for OA was reported by Kim et al (Kim, 2020). Four of the studies used bone-marrow derived MSCs, 1 used adipose-derived cells and the other cultured placental cells. Only 2 of the 6 studies were rated as low risk of bias. Pain outcomes measured with VAS and WOMAC pain scales were improved at 6 to 12 months, but there was no significant improvement in measures of WOMAC function or cartilage measured by magnetic resonance imaging.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2022. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Wiggers et al conducted a systematic review of RCTs evaluating autologous mesenchymal stem cell therapy on patient-reported outcome measures and disease severity (Wiggers, 2021). Fourteen RCTs were identified in searches conducted through December 2020. Meta-analysis was precluded because most of the original trial data were not available for pooling and due to heterogeneity across studies. A total of 408 patients with knee osteoarthritis received MSC therapy derived from bone marrow, adipose tissue, or activated peripheral blood. After 1 year, 19 of 26 (73%) clinical outcome measures improved with MSCs compared with control. In the MSC group, patients improved by 1.8 to 4.4 points on the Visual Analogue Scale (0 to 10) and 18 to 32 points on the Knee Osteoarthritis Outcome Score (0 to 100). Four studies showed better disease severity on imaging after MSC compared with control at 1 year. Although the reviewers found a positive effect of autologous MSC therapy compared with control treatments, the certainty of the evidence was rated low to very low due to high risk of bias in the included studies (e.g., 10 of 14 RCTs were at high risk of bias on all outcomes) and high heterogeneity in the source, method of preparation, and dosage of injected stem cells in included RCTs.
 
Lim et al reported on a RCT of 114 patients with large, full-thickness cartilage defects (International Cartilage Repair Society grade 4) treated with either a composite of umbilical cord-derived MSCs plus 4% hyaluronate (MSC-HA) or microfracture (Lim, 2021). The study consisted of a 48-week phase 3 clinical trial and a 5-year follow-up study. Of 114 patients randomized, 89 completed the phase 3 trial (78.1%), and 73 were enrolled in the follow-up study (64.0%). The primary outcome, proportion of participants with cartilage restoration equivalent to at least 1 grade improvement on the ICRS Macroscopic Cartilage Repair Assessment at 48-week arthroscopic evaluation, was 97.7% (42/43) in the MSC-HA group and 71.7% (33/46) in the microfracture group (odds ratio, 16.55; 95% CI, 2.06 to 133.03; p=.001). Both groups had significantly improved patient-reported pain scores (VAS pain, WOMAC, and IKDC scores) at 48 weeks versus baseline, but there was no significant difference between the 2 groups at this timepoint. From 36 to 60 months after intervention, the significant improvements from baseline were maintained in the MSC-HA group, whereas the improvements in VAS pain and WOMAC deteriorated in the microfracture group. This study had several limitations. There was no intervention group that received MSC alone, the comparator (microfracture) is not considered the standard of care for large, full-thickness cartilage defects, surgeons and participants were not blinded to treatment outcome, and there was high loss to follow-up. These limitations, along with a lack of improvement in patient-reported outcomes in the intervention group at 48 weeks, preclude drawing conclusions about the effectiveness of umbilical cord blood-derived MSCs in this population; higher quality evidence from RCTs is needed.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through July 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Jin et al also conducted a more focused systematic review and meta-analysis of 6 RCTs (N=452) that evaluated intra-articular MSC injection in patients undergoing high tibial osteotomy (HTO) (Jin, 2022). Results demonstrated that there were no significant differences in the International Knee Documentation Committee (IKDC) score and KOOS Pain and Symptoms subscales in patients who underwent HTO with or without the MSC injection. However, patients who received MSC injection had significantly greater improvements in Lysholm scores (mean difference, 2.55; 95% CI, 0.70 to 4.40; p=.007), and greater proportions of International Cartilage Regeneration and Joint Preservation Society (ICRS) grade 1 (p=.03) and grade 2 (p=.02) cartilage repair in the medial femoral condyle and grade 2 cartilage repair in the tibial plateau (p=.04).
 
More recently, Zaffagnini et al reported on results of an RCT that evaluated a single intra-articular injection of microfragmented adipose tissue or PRP in patients (N=118) with knee OA (Zaffagnini, 2022). The primary outcomes were the IKDC subjective score and the KOOS pain subscore at 6 months. Overall, both treatments provided significant improvements from baseline in clinical outcomes, with no significant differences found between treatment groups. The IKDC scores significantly improved from baseline to 6 months, from 41.1 ± 16.3 to 57.3 ± 18.8 with microfragmented adipose tissue, and from 44.8 ± 17.3 to 58.4 ± 18.1 with PRP. The improvement in the KOOS pain subscore from baseline to 6 months was 58.4 ± 15.9 to 75.8 ± 17.4 with microfragmented adipose tissue and 63.5 ± 17.8 to 75.5 ± 16.1 with PRP. As a secondary outcome, more patients in the microfragmented adipose tissue group with moderate/severe knee OA reached the minimal clinically important difference for the IKDC score at 6 months compared with the PRP group (75.0% vs 34.6%, respectively; p=.005).
 
2024 Update
Annual policy review completed with a literature search using the MEDLINE database through January  2024. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Multiple autologous and allogeneic cell-based therapies were compared with gold-standard corticosteroid injection in 475 adults with OA of the knee in a single-blind phase 3 RCT (Tables 3 through 6) (Mautner, 2023). Patients were randomized to 1of 2 autologous cell therapies (bone marrow aspirate concentrate [BMAC] or stromal vascular fraction), allogeneic umbilical cord-derived MSCs, or intra-articular corticosteroid injection; the co-primary endpoints were changes from baseline in VAS and Knee injury and Osteoarthritis Outcome Score pain scores at 12-month follow-up. No significant differences in pain scores were noted in comparisons between corticosteroid injection and any of the cell therapy arms.
 
A double-blind phase 3 RCT comparing a single intra-articular injection of autologous adipose tissue-derived MSCs with placebo in patients with knee OA (N=261) was reported on (Kim, 2023). Patients meeting American College of Rheumatology criteria for Kellgren-Lawrence grade 3 knee OA who had 100 mm VAS pain scores 50 and WOMAC functional impairment scores 40despites >3 months of non-operative treatment were eligible for enrollment. All patients underwent abdominal subcutaneous lipoaspiration 3 weeks prior to assigned study injection (1:1 randomization to 1x108 autologous adipose tissue-derived MSCs[n=131] or a mixture of saline with autologous serum [n=130]). The co-primary endpoints were change in 100 mm VAS pain score and WOMAC function score from baseline to 6 months. In the primary analysis, patients assigned to adipose tissue-derived MSCs experienced significantly greater improvements than those assigned to placebo in both VAS pain score (25.2±24.6 vs 15.5 ±23.7; p=.004) and WOMAC function score (21.7 ±18.6 vs 14.3 ±19.2; p=.002) from baseline to 6 months. Six-month changes in patient-reported outcomes (KOOS, 36-Item Short Form Health Survey Score, and International Knee Documentation Committee subjective knee score) also reflected significant improvements in patients who received adipose tissue-derived MSCs compared with those who received placebo. Study limitations include that while patients were required to have received prior non-operative therapy for at least 3 months, specific prior treatments were not reported; it is unclear whether the use of a placebo comparator was more appropriate than an active comparator in this setting.

References: Whitehouse MR, Howells NR, Parry MC, et al.(2017) Repair of torn avascular meniscal cartilage using undifferentiated autologous mesenchymal stem cells: from in vitro optimization to a first-in-human study. Stem Cells Transl Med. Apr 2017;6(4):1237-1248. PMID 28186682

A, Mafi R, Mafi P, et al.(2017) A systematic review and meta-analysis of clinical trials of mesenchymal stem cell therapy for cartilage repair. Curr Stem Cell Res Ther. Sep 15 2017. PMID 28914207

Akgun I, Unlu MC, Erdal OA, et al.(2015) Matrix-induced autologous mesenchymal stem cell implantation versus matrix-induced autologous chondrocyte implantation in the treatment of chondral defects of the knee: a 2-year randomized study. Arch Orthop Trauma Surg. Feb 2015;135(2):251-263. PMID 25548122

American Academy of Orthopaedic Surgeons.(2007) Stem Cells and orthopaedics. Your Orthopaedic Connection 2007. http://orthoinfo.aaos.org/topic.cfm?topic=A00501.

Borakati A, Mafi R, Mafi P, et al.(2017) A systematic review and meta-analysis of clinical trials of mesenchymal stem cell therapy for cartilage repair. Curr Stem Cell Res Ther. Sep 15 2017. PMID 28914207

Centeno CJ, Al-Sayegh H, Bashir J, et al.(2015) A prospective multi-site registry study of a specific protocol of autologous bone marrow concentrate for the treatment of shoulder rotator cuff tears and osteoarthritis. J Pain Res. 2015;8:269-276. PMID 26089699

Centeno CJ, Schultz JR, et al.(2010) Safety and complications reporting on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther, 2010; 5(10:81-93.

CH, Chai JW, Jeong EC, et al.(2017) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a 2-year follow-up study. Am J Sports Med. Oct 2017;45(12):2774-2783. PMID 28746812

Chirba MA, Sweetapple B, Hannon CP, et al.(2015) FDA regulation of adult stem cell therapies as used in sports medicine. J Knee Surg. Feb 2015;28(1):55-62. PMID 25603042

Deans TL, Elisseeff JH.(2009) Stem cells in musculoskeletal engineered tissue. Curr Opin Biotechnol 2009; 20(5):537-44.

Deans TL, Elisseeff JH.(2009) Stem cells in musculoskeletal engineered tissue. Curr Opin Biotechnol, 2009; 20(5):537-44.

Eastlack RK, Garfin SR, Brown CR, et al.(2014) Osteocel plus cellular allograft in anterior cervical discectomy and fusion: evaluation of clinical and radiographic outcomes from a prospective multicenter study. Spine (Phila Pa 1976). Oct 15 2014;39(22):E1331-1337. PMID 25188591

Emadedin M, Labibzadeh N, Liastani MG, et al.(2018) Intra-articular implantation of autologous bone marrow-derived mesenchymal stromal cells to treat knee osteoarthritis: a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy. 2018 Oct;20(10):1238-1246. PMID 30318332

Filardo G, Madry H, Jelic M et al.(2013) Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics. Knee Surg Sports Traumatol Arthrosc 2013 [Epub ahead of print].

Giannini S, Buda R, Cavallo M et al.(2010) Cartilage repair evolution in post-traumatic osteochondral lesions of the talus: from open field autologous chondrocyte to bone-marrow-derived cells transplantation. Injury 2010; 41(11):1196-203.

Giannini S, Buda R, Vannini F et al.(2009) One-step bone marrow-derived cell transplantation in talar osteochondral lesions. Clin Orthop Relat Res 2009; 467(12):3307-20.

Goldberg A, Mitchell K, Soans J, et al.(2017) The use of mesenchymal stem cells for cartilage repair and regeneration: a systematic review. Mar 09 2017;12(1):39. PMID 28279182

Jin L, Yang G, Men X, et al.(2022) Intra-articular Injection of Mesenchymal Stem Cells After High Tibial Osteotomy: A Systematic Review and Meta-analysis. Orthop J Sports Med. Nov 2022; 10(11): 23259671221133784. PMID 36452339

Jo CH, Chai JW, Jeong EC, et al.(2017) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a 2-year follow-up study. J Sports Med. Oct 2017;45(12):2774-2783. PMID 28746812

Jo CH, Lee YG, Shin WH, et al.(2014) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. May 2014;32(5):1254-1266. PMID 24449146

Jones CP, Loveland J, Atkinson BL, et al.(2015) Prospective, multicenter evaluation of allogeneic bone matrix containing viable osteogenic cells in foot and/or ankle arthrodesis. Foot Ankle Int. Oct 2015;36(10):1129-1137. PMID 25976919

Kim KI, Lee MC, Lee JH, et al.(2023) Clinical Efficacy and Safety of the Intra-articular Injection of Autologous Adipose-Derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Phase III, Randomized, Double-Blind, Placebo-Controlled Trial. Am J Sports Med. Jul 2023; 51(9):2243-2253. PMID 37345256

Kim SH, Djaja YP, Park YB, et al.(2020) Intra-articular Injection of Culture-Expanded Mesenchymal Stem Cells Without Adjuvant Surgery in Knee Osteoarthritis: A Systematic Review and Meta-analysis. Am J Sports Med. Sep 2020; 48(11): 2839-2849. PMID 31874044

Kim YS, Lee HJ, Choi YJ, et al.(2014) Does an injection of a stromal vascular fraction containing adipose-derived mesenchymal stem cells influence the outcomes of marrow stimulation in osteochondral lesions of the talus? A clinical and magnetic resonance imaging study. Am J Sports Med. Oct 2014;42(10):2424-2434. PMID 25106781

Koh YG, Choi YJ.(2012) Infrapatellar fat pad-derived mesenchymal stem cell therapy for knee osteoarthritis. Knee 2012; 19(6):902-7.

Koh YG, Kwon OR, Kim YS, et al.(2014) Comparative outcomes of open-wedge high tibial osteotomy with platelet-rich plasma alone or in combination with mesenchymal stem cell treatment: a prospective study. Arthroscopy. Nov 2014;30(11):1453-1460. PMID 25108907

Lamo-Espinosa JM, Mora G, Blanco JF et al.(2016) Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: multicenter randomized controlled clinical trial (phase I/II). J Transl Med. 2016 Aug;14(1). PMID 27565858

Lamo-Espinosa JM, Mora G, Blanco JF, et al.(2018) Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: long-term follow-up of a multicenter randomized controlled clinical trial (phase I/II). J Transl Med. 2018 Jul;16(1). PMID 30064455

Lau RL, Perruccio AV, Evans HM, et al.(2014) Stem cell therapy for the treatment of early stage avascular necrosis of the femoral head: a systematic review. BMC Musculoskelet Disord. 2014 May 16;15:156. doi: 10.1186/1471-2474-15-156.

Lim HC, Park YB, Ha CW, et al.(2021) Allogeneic Umbilical Cord Blood-Derived Mesenchymal Stem Cell Implantation Versus Microfracture for Large, Full-Thickness Cartilage Defects in Older Patients: A Multicenter Randomized Clinical Trial and Extended 5-Year Clinical Follow-up. Orthop J Sports Med. Jan 2021; 9(1): 2325967120973052. PMID 33490296

Maheshwer B, Polce EM, Paul K, et al.(2021) Regenerative Potential of Mesenchymal Stem Cells for the Treatment of Knee Osteoarthritis and Chondral Defects: A Systematic Review and Meta-analysis. Arthroscopy. Jan 2021; 37(1): 362-378. PMID 32497658

Mautner K, Gottschalk M, Boden SD, et al.(2023) Cell-based versus corticosteroid injections for knee pain in osteoarthritis: a randomized phase 3 trial. Nat Med. Nov 02 2023. PMID 37919438

Nejadnik H, Hui JH, Feng Choong EP et al.(2010) Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med 2010; 38(6):1110-6.

Newberry SJ, Fitzgerald JD, Maglione MA, et al.(2015) Systematic Review for Effectiveness of Hyaluronic Acid in the Treatment of Severe Degenerative Joint Disease (DJD) of the Knee (AHRQ Technology Assessment). Rockville, MD: Agency for Healthcare Research and Quality; 2015.

Peppers TA, Bullard DE, Vanichkachorn JS, et al.(2017) Prospective clinical and radiographic evaluation of an allogeneic bone matrix containing stem cells (Trinity Evolution(R) Viable Cellular Bone Matrix) in patients undergoing two-level anterior cervical discectomy and fusion. J Orthop Surg Res. Apr 26 2017;12(1):67. PMID 28446192

Rush SM, Hamilton GA, Ackerson LM.(2009) Mesenchymal stem cell allograft in revision foot and ankle surgery: a clinical and radiographic analysis. J Foot Ankle Surg 2009; 48(2):163-9.

Saw KY, Anz A, Siew-Yoke Jee C et al.(2013) Articular Cartilage Regeneration With Autologous Peripheral Blood Stem Cells Versus Hyaluronic Acid: A Randomized Controlled Trial. Arthroscopy 2013; 29(4):684-94.

Sen RK, Tripathy SK, Aggarwal S et al.(2012) Early results of core decompression and autologous bone marrow mononuclear cells instillation in femoral head osteonecrosis: a randomized control study. J Arthroplasty 2012; 27(5):679-86.

Shapiro SA, Kazmerchak SE, Heckman MG, et al.(2017) A prospective, single-blind, placebo-controlled trial of bone marrow aspirate concentrate for knee osteoarthritis. Am J Sports Med. Jan 2017;45(1):82-90. PMID 27566242

U.S. Food & Drug Administration (FDA).(2020) Regulatory Considerations for Human Cells, Tissues, and Cellular and Tissue-Based Products: Minimal Manipulation and Homologous Use. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/regulatory-considerations-human-cells-tissues-and-cellular-and-tissue-based-products-minimal. Accessed December 29, 2020.

U.S. Food and Drug Administration(2008) Assuring safety and efficacy of stem-cell based products. http://www.fda.gov/BiologicsBloodVaccines/ScienceResearch/BiologicsResearchAreas/ucm127182.htm

U.S. Food and Drug Administration.(2008) Vaccines, Blood & Biologics. Enforcement actions. Untitled letter. www.fda.gov/.../ComplianceActivities/Enforcement/UntitledLetters/ucm091991.htm

Vangsness CT, Jr., Farr J, 2nd, Boyd J et al.(2014) 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):90-8.

Vanichkachorn J, Peppers T, Bullard D, et al.(2016) A prospective clinical and radiographic 12-month outcome study of patients undergoing single-level anterior cervical discectomy and fusion for symptomatic cervical degenerative disc disease utilizing a novel viable allogeneic, cancellous, bone matrix (trinity evolution) with a comparison to historical controls. Eur Spine J. Jul 2016;25(7):2233-2238. PMID 26849141

Vega A, Martin-Ferrero MA, Del Canto F, et al.(2015) Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. Aug 2015;99(8):1681-1690. PMID 25822648

Wakitani S, Imoto K, et al.(2002) Human autologous culture expanded bone marrow mesenchymal cell transplantation for repari of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage, 2002; 10(3):199-206.

Wakitani S, Nawata M, et al.(2007) Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. J Tissue Eng Regen Med, 2007; 1(1):74-9.

Wakitani S, Okabe T, Horibe S et al.(2011) Safety of autologous bone marrow-derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months. J Tissue Eng Regen Med 2011; 5(2):146-50.

Wiggers TG, Winters M, Van den Boom NA, et al.(2021) Autologous stem cell therapy in knee osteoarthritis: a systematic review of randomised controlled trials. Br J Sports Med. Oct 2021; 55(20): 1161-1169. PMID 34039582

Wong KL, Lee KB, Tai BC et al.(2013) 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):2020-8.

Zaffagnini S, Andriolo L, Boffa A, et al.(2022) Microfragmented Adipose Tissue Versus Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: A Prospective Randomized Controlled Trial at 2-Year Follow-up. Am J Sports Med. Sep 2022; 50(11): 2881-2892. PMID 35984721

Zhao D, Cui D, Wang B et al.(2012) Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow-derived and cultured mesenchymal stem cells. Bone 2012; 50(1):325-30.


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.