Coverage Policy Manual
Policy #: 2011058
Category: Medicine
Initiated: August 2011
Last Review: February 2024
  Autologous Stem-cell Therapy to Treat Peripheral Arterial Disease

Description:
Peripheral arterial disease is a common atherosclerotic syndrome associated with significant morbidity and mortality. Critical limb ischemia (CLI) is the end stage of lower-extremity PAD in which severe obstruction of blood flow results in ischemic pain at rest, ulcers, and a significant risk for limb loss. Use of autologous stem cells freshly harvested and allogeneic stem cells are reported to have a role in the treatment of PAD.
 
Peripheral arterial disease (PAD) is a common atherosclerotic syndrome that is associated with significant morbidity and mortality. A less-common cause of PAD is Buerger disease, also called thromboangiitis obliterans, which is a nonatherosclerotic segmental inflammatory disease that occurs in younger patients and is associated with tobacco use. Development of PAD is characterized by narrowing and occlusion of arterial vessels and eventual reduction in distal perfusion. Critical limb ischemia is the endstage of lower extremity PAD in which severe obstruction of blood flow results in ischemic pain at rest, ulcers, and a significant risk for limb loss.
 
Two endogenous compensating mechanisms may occur with occlusion of arterial vessels, capillary growth (angiogenesis) and development of collateral arterial vessels (arteriogenesis). Capillary growth is mediated by hypoxia-induced release of chemo- and cytokines such as vascular endothelial growth factor (VEGF) and occurs by sprouting of small endothelial tubes from pre-existing capillary beds. The resulting capillaries are small and cannot sufficiently compensate for a large, occluded artery. Arteriogenesis with collateral growth is, in contrast, initiated by increasing shear forces against vessel walls when blood flow is redirected from the occluded transport artery to the small collateral branches, leading to an increase in the diameter of pre-existing collateral arterioles.
 
The mechanism underlying arteriogenesis includes the migration of bone marrow-derived monocytes to the perivascular space. The bone marrow-derived monocytes adhere to and invade the collateral vessel wall. It is not known if the expansion of the collateral arteriole is due to the incorporation of stem cells into the wall of the vessel or to cytokines released by monocytic bone marrow cells that induce the proliferation of resident endothelial cells. It has been proposed that bone marrow-derived monocytic cells may be the putative circulating endothelial progenitor cells. Notably, the same risk factors for advanced ischemia (diabetes, smoking, hyperlipidemia and advanced age) are also risk factors for a lower number of circulating progenitor cells.
 
Use of autologous stem cells freshly harvested and allogeneic stem cells are reported to have a role in the treatment of PAD. Stem cells can be administered in a variety of routes, derived from different progenitors, and be grouped with different co-factors, many of which are being studied in order to determine the best clinical option for patients. The primary outcome in stem cell therapy trials regulated by the U.S. Food and Drug Administration (FDA) is amputation-free survival, defined as time to major amputation and/or death from any cause. Other outcomes for critical limb ischemia include the Rutherford criteria for limb status, healing of ulcers, the Ankle-Brachial Index (ABI), transcutaneous oxygen pressure, and pain-free walking. The ABI measures arterial segmental pressures on the ankle and brachium and indexes ankle systolic pressure against brachial systolic pressure (normative range, 0.95 to 1.2 mm Hg).
 
Regulatory Status
Six point-of-care concentrations of bone marrow aspirate have been cleared by the FDA through the 510(k) process and are summarized below.
 
FDA Approved Point-of-Care Concentration of Bone Marrow Aspirate Devices:
 
  • The SmarktPReP2® Bone Marrow Aspirate Concentrate System, SmarktPReP Platelet Concentration System, manufactured by Harvest Technologies in Lakewood, CO, cleared on 12/06/2010 (K103340)
  • MarrowStim Concentration System (MSC system), manufactured by Biomet Biologics, Inc. in Warsaw, IN cleared 12/18/2009 (BK090008)
  • PureBMC SupraPhysiologic Concentrating System, manufactured by EmCyte Corporation® in Fort Myers, Florida, cleared on 5/30/2019 (K183205)
  • Arthrex Angel System Kit, manufactured by Arthrex, Inc. in Naples, Florida, cleared on 5/23/2018 (BK180180)
  • Magellan® Autologous Platelet Separator System, manufactured by Arteriocyte Medical Systems (Medtronic) in Memphis, TN, cleared 11/09/2004 (BK040068)
  • BioCUE Platelet Concentration Kit, manufactured by Biomet Biologics, Inc. in Warsaw, IN, cleared on 5/26/2010 (BK1000027)
  • ART BMC System, manufactured by SpineSmith Holdings, LLC in Austin, TX
  • PXP® System, manufactured by ThermoGenesis Corp. in Rancho Cordova, CA, cleared on 07/10/2008 (K081345)
 
 
Coding
Beginning in July 2011, there are specific CPT category III codes for this therapy:
 
0263T: Intramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; complete procedure including unilateral or bilateral bone marrow harvest
 
0264T: Intramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; complete procedure excluding bone marrow harvest
 
0265T: Intramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; unilateral or bilateral bone marrow harvest only for intramuscular autologous bone marrow cell therapy.
 
The CPT codes were constructed to allow reporting of the complete procedure and harvesting by a single physician (code 0263T) or separate reporting when the cell harvesting and therapy injections are performed by separate physicians (0264T and 0265T).

Policy/
Coverage:
Effective July 2021
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Treatment of peripheral arterial disease, including critical limb ischemia, with injection or infusion of cells concentrated from bone marrow aspirate does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.
 
For members with contracts without primary coverage criteria, treatment of peripheral arterial disease, including critical limb ischemia, with injection or infusion of cells concentrated from bone marrow aspirate is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective Prior to July 2021
Treatment of peripheral arterial disease, including critical limb ischemia, with injection or infusion of cells concentrated from bone marrow aspirate is currently being studied in clinical trials and therefore does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.
 
For contracts without primary coverage criteria, treatment of peripheral arterial disease, including critical limb ischemia, with injection or infusion of cells concentrated from bone marrow aspirate is considered investigational as this therapy is currently being studied in Phase III clinical trials to determine effectiveness.  Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
 

Rationale:
This policy was created based on a review of literature identified through a search of the MEDLINE database through April 2011. Controlled studies and the larger case series are summarized below.
 
In 2002, the Therapeutic Angiogenesis by Cell Transplantation (TACT) study investigators published results of a pilot study and a small double-masked trial with 22 patients who were treated with bone marrow-mononuclear cells by intramuscular injection into the gastrocnemius of one leg and peripheral blood-mononuclear cells in the other leg as a control (randomized order) (Tateishi-Yuyama, 2002).  Patients qualified for marrow implantation if they had bilateral chronic limb ischemia, including rest pain, non-healing ischemic ulcers, or both, and were not candidates for nonsurgical or surgical revascularization. Seventeen patients (85%) had been previously treated with percutaneous angioplasty, bypass graft, or both. The patients had resting ankle-brachial index (ABI) less than 0.6 in both limbs. Patients with poorly controlled diabetes mellitus or with evidence of malignant disorder during the past 5 years were excluded from the study. About 500 mL of bone marrow cells were aspirated from the ileum, separated, and concentrated to a final volume of about 30 mL. About 3 hours after marrow aspiration the cells were implanted by intramuscular injection into the gastrocnemius. Follow-up with ABI, transcutaneous oxygen pressure (TcO2) and pain-free walking time was performed every week for 4 weeks and every 4 months thereafter. Two patients discontinued the study after randomization due to clinical worsening before 4 weeks. At 4 weeks after treatment, ABI, TcO2, and rest pain were significantly improved in legs injected with bone marrow-mononuclear cells, compared with those injected with peripheral blood-mononuclear cells. For example, ABI increased by 0.1 in the leg treated with bone marrow-mononuclear cell and by 0.02 with peripheral blood-mononuclear cells. TcO2 improved by 17.4 mm Hg with bone marrow-mononuclear cells and by 4.6 mm Hg with peripheral blood-mononuclear cells. Rest pain in legs treated with bone marrow-mononuclear cells was resolved in 16 of 20 patients, while pain in legs treated with peripheral blood-mononuclear cells remained in 17 of 20 patients. These improvements were sustained at 24 week follow-up. Digital subtraction angiography showed a marked increase in the number of visible collateral vessels in 60% of legs treated with bone marrow cells. No adverse events were reported.
 
The 2008 TACT report by the same multicenter group of investigators assessed the 3-year safety and clinical outcomes of intramuscular implantation of bone marrow-mononuclear cells in a series of 74 patients with critical limb ischemia due to atherosclerotic peripheral arterial disease (PAD) and 41 patients with thromboangiitis obliterans (TAO; Buerger’s disease) (Matoba, 2008). The ischemic limbs were not candidates for surgical or nonsurgical revascularization. Twenty-six patients (23%) had a previous bypass operation. Bone marrow cells were aspirated from the ileum, and the mononuclear cells sorted and concentrated to a final volume of 40 mL. The cells were implanted by intramuscular injection into the foot. Patients were followed every week for 4 weeks and at 6, 12, 24, and 36 months thereafter. The overall survival, amputation-free interval, adverse events, ABI, TcO2, pain scale, ulcer size, and pain-free walking distance were evaluated at each time point. Overall patient survival and amputation-free interval were defined as the primary endpoints of this study. Three-year overall survival rates were 80% in patients with atherosclerotic PAD and 100% for patients with TAO, and the median follow-up time of surviving patients was 25 months (range, 0.8 to 69 months). The 3-year amputation-free rate was 60% in atherosclerotic PAD and 91% in patients with TAO. Of the 24 amputations in patients with PAD, 83% occurred within 6 months. Multivariate analysis indicated that the severity of ischemic pain at baseline and prior repeated bypass surgery were the major determinants that negatively affected the amputation-free interval of the therapy. The ABI and transcutaneous oxygen pressure value did not significantly change, but there was a significant improvement in the leg pain scale (from 6 to 2), ulcer size (from 3.5 cm2 to 0), and pain-free walking distance (from about 25 meters to 100 meters) at 6 months.
 
Amann et al. reported a pilot study of autologous bone marrow cell transplantation in 51 consecutive patients with impending major amputation due to end-stage critical limb ischemia in 2009 (Amann, 2009). Forty-five patients (88%) had undergone a mean of two unsuccessful attempts of operative and/or percutaneous revascularization of the ischemic limb. Six patients (12%) were technically not amenable to revascularization. Critical limb ischemia was confirmed if there was angiographic proof of arterial occlusion and one of the following criteria was fulfilled: ABI less than 0.6 or TcO2 less than 30 mm Hg. Major amputation (above the ankle) had been recommended to 46 of the 51 patients (90%) by the treating vascular surgeons. For the first 12 subjects, 450-500 mL bone marrow was aspirated under general anesthesia and processed by the Ficoll method. For the remaining subjects, 240 mL bone marrow was aspirated under sedation and processed using an automated bedside density gradient centrifugation method. The final treating volume (55-85 mL) was adjusted with plasma depending on the area to be treated (whole leg, calf only, or foot), based on the localization and extent of the arterial occlusions. In addition, if a wound was present, 4-10 injections of bone marrow concentrate were given into the wound bed and the wound perimeter. Patients were seen monthly up to 6 months and at least in half-year intervals after. Minimum follow-up was 6 months, and the mean follow-up was 411 days (range 175 to 1,186 days). No patients were lost to follow-up. Improvement in perfusion and subsequent limb salvage was achieved in 30/51 patients (59%) at 6 months and 27/51 (53%) at last follow-up (mean of 411 days). Seventeen minor amputations (6 forefoot and 11 toe) were performed in the 30 patients with 24-week limb salvage. Complete wound healing was achieved in 15 of 21 patients with ischemic wounds. Perfusion, measured at 6 months with ABI and TcO2, increased in patients with limb salvage and did not change in patients who eventually underwent major amputation. Patients with limb salvage improved from a mean Rutherford category of 4.9 at baseline to 3.3 at 6 months. Analgesic consumption was reduced by 62%. Total walking distance improved in non-amputees from a median of 0 to 40 meters at 24 weeks. No unexpected long-term adverse events occurred.
 
In 2010, Prochazka and colleagues reported a randomized study of 96 patients with critical limb ischemia and foot ulcer (Prochazka, 2010). Patient inclusion criteria were critical limb ischemia as define by ABI equal to or less than 0.4, ankle systolic pressure equal to or less than 50 mm Hg or toe systolic pressure equal to or less than 30 mm Hg, and failure of basic conservative and revascularization treatment (surgical or endovascular). The patients were randomized into treatment with bone marrow concentrate (n=42) or standard medical care (n=54). The primary endpoints were major limb amputation during 120 days and degree of pain and function at 90- and 120-day follow-up. At baseline, the control group had a higher proportion of patients with diabetes (98.2% vs. 88.1%), hyperlipidemia (80.0% vs. 54.8%) and ischemic heart disease (76.4% vs. 57.1% - all respectively). In addition, the control group had a higher proportion of patients with stage DIII (deep ulcers with osteitis) University of Texas Wound Classification (72% vs. 40%, respectively). For the 42 patients in the treatment group, there was a history of 50 revascularization procedures; 46 of 54 patients in the control group had a history of revascularization procedures. Forty-two of the 42 patients in the bone-marrow group finished 90 days of follow-up and 37 of 54 patients in the control group finished 120 days of follow-up. The reason for different times of follow-up for the primary outcome measure is unclear. Five patients in the bone-marrow group and 8 in the control group died of causes unrelated to the therapy during follow-up. At follow-up, the frequency of major limb amputation was 21% in patients treated with bone marrow concentrate and 44% in controls. Secondary endpoints were performed only in the group treated with bone marrow concentrate. In the treatment group with salvaged limbs, toe pressure and toe brachial index increased from 22.66 to 25.63 mm Hg and from 0.14 to 0.17, respectively. Interpretation of this study is limited by unequal baseline measures, lack of blinding, different periods of follow-up, different loss to follow-up and different measures at follow-up for the 2 groups.
 
Ongoing Clinical Trials
The design of the BONe Marrow Outcome Trial in Critical Limb Ischaemia (BONMOT-CLI) trial was reported in 2008 (Amann, 2008). It is an investigator-initiated, randomized, double-blinded, placebo-controlled multicenter study at 4 sites in Germany that assesses the therapeutic value of bone marrow cell-induced angiogenesis and arteriogenesis in severe, limb-threatening ischemia. Ninety patients with no option for revascularization or after failed revascularization will be randomized to 40 injections into the ischemic limb with a concentrate of autologous bone marrow cells or to sham bone marrow aspiration and 40 placebo injections. The combined primary endpoint is major amputation or persisting critical limb ischemia (no improvement) over 3 months. Secondary endpoints are death, changes in perfusion, quality of life, walking distance, minor amputations, wound healing, collateral density and cancer incidence. Post study follow-up is 2 years.
 
JUVENTAS (Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation) is a randomized, double-blind, placebo-controlled trial in the Netherlands (Sprengers, 2010). The clinical effects of repeated intra-arterial infusion of bone marrow mononuclear cells will be investigated in 110-160 patients with critical limb ischemia. Patients will receive repeated intra-arterial infusion of bone marrow-mononuclear cells or placebo into the common femoral artery. The primary outcome measure is the rate of major amputation after 6 months. Secondary endpoints include minor amputation, number and extent of leg ulcers, resolution of rest pain, perfusion, change in quality of life, and change in clinical status. Functional characteristics of the bone marrow-mononuclear cells will also be studied, and the bone marrow-mononuclear cell dysfunction will be related to clinical outcome.
 
A search of ClinicalTrials.gov in April 2011 identified a number of ongoing trials with hematopoietic stem cell/bone marrow concentrate for peripheral arterial disease, including:
 
  • A manufacturer-sponsored U.S. multicenter Phase II study on the use of autologous bone marrow cells for the treatment of critical limb ischemia due to PAD (NCT00468000). The double-blind study is expected to enroll 150 patients, randomized into two patient groups. The treatment group will receive intramuscular injections of Aastrom Biosciences TRC autologous bone marrow cell product into the affected limb; the control group will receive intramuscular injections with an electrolyte solution (without cells). The estimated completion date is listed as March 2011.
 
  • A manufacturer-sponsored Phase III trial with bone marrow aspirate concentrate with the SmartPReP2® system for the treatment of critical limb ischemia (NCT01245335). This is a U.S. pivotal randomized double-blind safety and efficacy trial comparing bone marrow aspirate concentrate with placebo injection into ischemic tissue of the lower extremity in 210 patients. The study start date is listed as January 2011; the expected study completion date is June 2014.
 
Summary
Based on initial evidence from case series and small randomized trials, injection of bone marrow concentrate may hold promise as a treatment for critical limb ischemia due to peripheral arterial disease. However, well-designed and well-conducted randomized controlled trials are needed to evaluate the health outcomes of this procedure. A number of trials are in progress, including several large randomized double-blind placebo controlled trials. Results from these trials are needed to adequately evaluate the impact on net health outcome of this procedure. Further information on the safety and durability of the treatment is also needed.
 
2012 Update
A literature search was conducted through September 2012.  There was no new information identified that would prompt a change in the coverage statement.
 
Results from the multi-center PROVASA trial (Intraarterial Progenitor Cell Transplantation of Bone Marrow Mononuclear Cells for Induction of Neovascularization in Patients with Peripheral Arterial Occlusive Disease) were reported in 2011 (Walter, 2011). In this double-blind Phase II trial, 40 patients with critical limb ischemia who were not candidates or had failed to respond to interventional or surgical procedures were randomized to intra-arterial administration of bone marrow-derived mononuclear cells (BM-MNC) or placebo. The cell suspension included hematopoietic, mesenchymal, and other progenitor cells. After 3 months, both groups were treated with BM-MNC in an open-label phase. Twelve patients received additional treatment with BM-MNC between 6 and 18 months. The primary outcome measure, a significant increase in the ABI at 3 months, was not achieved (from 0.66 at baseline to 0.75 at 3 months). Limb salvage and amputation-free survival rates did differ between the groups. There was a significant improvement in ulcer healing (ulcer area 1.89 cm2 vs. 2.89 cm2) and reduced pain at rest (improvement of about 3 vs. 0.05) following intra-arterial BM-MNC administration. This is the only randomized controlled trial to report intra-arterial administration of BM-MNC.
Interim analysis results from 46 patients was reported from the RESTORE-CLI study in 2011 (Powell, 2011).  The RESTORE-CLI is a manufacturer-sponsored U.S. multicenter Phase II study on the use of autologous bone marrow cells for the treatment of critical limb ischemia due to PAD(NCT00468000).  The authors report in the interim analysis that intramuscular injections of autologous bone marrow-derived cells are safe for the treatment of patients with lower extremity critical limb ischemia.  The interim results of 46 of 86 patients indicated an improvement in amputation-free survival and time to first occurrence of treatment failure compared to placebo.  According to the authors, a larger Phase III trial is planned based on these interim results.
 
A search of online site ClinicalTrials.gov in October 2012 identified a number of ongoing trials with hematopoietic stem cell/bone marrow concentrate for peripheral arterial disease, including a manufacturer-sponsored Phase III trial with bone marrow aspirate concentrate with the SmartPReP2® system for the treatment of critical limb ischemia (NCT01245335). This is a U.S. pivotal randomized double-blind safety and efficacy trial comparing bone marrow aspirate concentrate with placebo injection into ischemic tissue of the lower extremity in 210 patients. The study start date is listed as January 2011; the expected study completion date is June 2014.
 
2013 Update
A literature search was conducted using the MEDLINE database through September 2013.  No new information was identified that would prompt a change in the coverage statement. The following is a summary of the key identified literature.
 
A 2011 Cochrane review identified 2 small studies with a total of 57 patients that met the review’s inclusion criteria for local intramuscular transplantation of autologous mononuclear cells (monocytes) for critical limb ischemia (CLI) (Moazzami, 2011). Studies were excluded that used mesenchymal stem cells (MSCs) or bone marrow aspirate. In one of the studies, intramuscular injection of bone marrow-derived mononuclear cells was compared with standard conservative treatment. In the second study, peripheral blood derived mononuclear cells were collected following injections of granulocyte colony-stimulating factor and transplanted by intramuscular injections. Both studies showed a significant reduction in amputations with treatment with monocytes, but larger randomized controlled trials are needed to adequately evaluate the effect of treatment with greater certainty.
 
In 2012, Liu et al. reported a meta-analysis of 6 randomized trials (333 patients) that evaluated mononuclear cell transplantation in patients with CLI (Liu, 2012). Cell therapy was found to decrease the incidence of amputation in patients with CLI with an odds ratio (OR) of 0.37. The rate of amputation free survival was increased in patients with Rutherford class 5 CLI (OR: 3.28) but was not significantly different in patients with Rutherford class 4. Following is a description of some of the randomized controlled trials (RCTs) that were included in the meta-analysis.
 
In 2011, Benoit et al. reported an FDA-regulated double-blind pilot RCT of 48 patients with CLI who were randomized in a 2:1 ratio to bone marrow concentrate using the SmartPReP system or iliac crest puncture with intramuscular injection of diluted peripheral blood (Benoit, 2011). At 6-month follow-up, the difference in the percentage of amputations between the cell therapy group and controls (29.4% vs. 35.7%, respectively) did not achieve statistical significance. In a subgroup analysis of patients with tissue loss at baseline (Rutherford 5), intramuscular injection of bone marrow concentrate resulted in a lower amputation rate than placebo (39.1% vs. 71.4%, respectively). Power analysis indicated that 210 patients would be needed to achieve 95% power in a planned pivotal trial.
 
Expanded Monocytes and Mesenchymal Stem Cells
Interim and final results from the industry-sponsored Phase II randomized double-blind placebo-controlled RESTORE-CLI trial, which utilized cultured and expanded monocytes and MSCs derived from bone marrow aspirate (ixmyelocel-T), were reported by Powell et al. in 2011 and 2012 (Powell, 2011; Powell, 2012). Seventy-two patients with CLI received ixmyelocel-T (n=48) or placebo with sham bone marrow aspiration (n=24) and were followed for 12 months. There was a 40% reduction in any treatment failure (due primarily to differences in doubling of total wound surface area and de novo gangrene), but no significant difference in amputations at 12 months.
 
A 2011 study by Lu et al. was a randomized double-blind safety and feasibility study of 41 patients with bilateral diabetic CLI and foot ulcer who were injected intramuscularly with expanded bone marrow MSCs or bone marrow-derived monocytes in one limb and normal saline in the other limb (Lu, 2011). At 24 weeks after treatment, outcomes (painless walking time, ankle-brachial index, transcutaneous oxygen pressure, and magnetic resonance angiography) were significantly improved in both experimental groups compared to injection with normal saline. Outcomes on some outcome measures were modestly improved for treatment with MSCs compared to mononuclear cells. Ulcer healing at 24 weeks occurred in 100% of experimental limbs with a faster rate of healing in the MSC-treated limbs. No cell-treated limbs underwent amputation, compared to 6 of 37 control limbs.
 
Ongoing Clinical Trials
A 2012 update on clinical trials evaluating the effect of biologic therapy in patients with critical limb ischemia (CLI) describes several products that are currently in Phase II or Phase III trials (Powell, 2012). The U.S. Food and Drug Administration (FDA) recommends that the primary efficacy endpoint in a Phase III CLI trial should be amputation-free survival. When the probability of this outcome is combined with the comorbid burden of CLI patients and variable natural history, large numbers of patients (about 500) may be needed to evaluate clinical outcomes (Powell, 2012).
The design of the BONe Marrow Outcome Trial in Critical Limb Ischaemia (BONMOT-CLI) trial was reported in 2008 (Amann, 2008). It is an investigator-initiated, randomized, double-blinded, placebo-controlled multicenter study at 4 sites in Germany that assesses the therapeutic value of bone marrow cell-induced angiogenesis and arteriogenesis in severe, limb-threatening ischemia. Ninety patients with no option for revascularization or after failed revascularization will be randomized to 40 injections into the ischemic limb with a concentrate of autologous bone marrow cells or to sham bone marrow aspiration and 40 placebo injections. The combined primary endpoint is major amputation or persisting critical limb ischemia (no improvement) over 3 months. Secondary endpoints are death, changes in perfusion, quality of life, walking distance, minor amputations, wound healing, collateral density and cancer incidence. Post study follow-up is 2 years.
JUVENTAS (Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation) is a randomized, double-blind, placebo-controlled trial in the Netherlands (Sprengers, 2010). The clinical effects of repeated intra-arterial infusion of bone marrow mononuclear cells will be investigated in 110-160 patients with critical limb ischemia. Patients will receive repeated intra-arterial infusion of bone marrow-mononuclear cells or placebo into the common femoral artery. The primary outcome measure is the rate of major amputation after 6 months. Secondary endpoints include minor amputation, number and extent of leg ulcers, resolution of rest pain, perfusion, change in quality of life, and change in clinical status. Functional characteristics of the bone marrow-mononuclear cells will also be studied, and the bone marrow-mononuclear cell dysfunction will be related to clinical outcome.
A search of online site ClinicalTrials.gov in April 2013 and a 2012 review by Powell, (Powell, 2012) identified a number of ongoing trials with concentrated or expanded stem cells for peripheral arterial disease, including:
  • A manufacturer-sponsored U.S. multicenter Phase III study on the use of autologous bone marrow cells for the treatment of critical limb ischemia due to PAD. Completion of this study of ixmyelocel-T (REVIVE) is expected mid-2015 (NCT01483898).
  • A manufacturer-sponsored Phase III trial with bone marrow aspirate concentrate with the SmartPReP2® system for the treatment of critical limb ischemia (NCT01245335). This is a U.S. pivotal randomized double-blind safety and efficacy trial comparing bone marrow aspirate concentrate with placebo injection into ischemic tissue of the lower extremity in 210 patients. The study start date is listed as January 2011; the expected study completion date is June 2014.
  • A manufacturer-sponsored sham-controlled Phase III trial of Biomet Biologic’s MarrowStim P.A.D. kit™ (NCT01049919) is currently recruiting. The study has an estimated enrollment of 152 patients with completion expected May 2014.
  • A Phase I study of placenta-derived mesenchymal-like stromal cells (PLX-PAD) by Pluristem has been completed (NCT00951210) A Phase II study in patients with CLI is expected to begin in 2013. A Phase II study of PLX-PAD for the treatment of intermittent claudication (NCT01679990) is currently recruiting with a target enrollment of 150 patients and completion in December 2015.
 
2014 Update
A literature search conducted through June 2014 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
A 2013 meta-analysis by Teraa et al. included 12 randomized controlled trials with a total of 510 patients
with critical limb ischemia (Teraa, 2013). Eight of the trials had fewer than 50 patients. Meta-analysis of all of the trials showed significant improvements with bone marrow-derived cell therapy on both subjective and objective intermediate endpoints (pain score, pain-free walking distance, ankle-brachial index, and
transcutaneous oxygen measurements) and on amputation rates (relative risk [RR] of .58). Overall, there
were 38 amputations in the experimentally-treated limbs compared to 62 amputations for control limbs.
However, when only placebo-controlled trials were included, no effect on major amputation rates was
identified (RR of .78 to .92).
 
In 2013, Poole et al reported results of a phase II double-blind, placebo-controlled study of granulocytemacrophage colony-stimulating factor (GM-CSF) in 159 patients with intermittent claudication due to peripheral arterial disease (Poole, 2013). Patients were treated with subcutaneous injections of GM-CSF or placebo 3 times a week for 4 weeks. The primary outcome, peak treadmill walking time at 3 months, increased by 109 seconds (from 296 to 405 seconds) in the GM-CSF group and by 56 seconds (from 308 to 376 seconds) in the placebo group (p=.08). Changes in the physical functioning subscore of the SF-36 and distance score of the walking impairment questionnaire (WIQ) were significantly better in patients treated with GM-CSF. However, there were no significant differences between the groups in the ankle brachial index, the WIQ distance and speed scores, claudication onset time, or mental or physical
component scores of the SF-36. Post-hoc exploratory analysis found a that patients with a greater than
100% increase in progenitor cells (CD34+/CD133+) had a significantly greater increase in peak walking
time than patients who had less than 100% increase in progenitor cells (131 seconds vs 60 seconds).
 
An ongoing and unpublished clinical trial; PROPEL (Progenitor cell release plus exercise to improve functional performance in peripheral artery disease) is a placebo-controlled trial of a combination of 12 weeks of treadmill training plus granulocytemacrophage colony stimulating factor (GM-CSF) (Domanchuk, 2013)).  A total of 240 patients will be randomized to 1 of 4 arms of the study: GM-CSF plus supervised treadmill exercise; GM-CSF plus attention control; placebo plus supervised exercise; placebo plus attention control. The primary outcome is change in distance for the six-minute walk at 12 weeks. Secondary outcomes include change in brachial artery flow-mediated dilation, change in maximal treadmill walking time, and change in circulating CD34+ cells.
 
A search of online site ClinicalTrials.gov in April 2014 and reviews by Powell in 2012 and Bartel in
2013 (Bartel, 2013) identified a number of ongoing trials with concentrated or expanded stem cells for peripheral arterial disease, including:
  • A manufacturer-sponsored U.S. multicenter Phase III study on the use of autologous expanded   bone marrow cells for the treatment of critical limb ischemia due to PAD. This study has completed enrollment of 41 patients and completion of this study of ixmyelocel-T (REVIVE) is expected mid-2015 (NCT01483898). This study was last verified in July 2013.
  • A manufacturer-sponsored Phase III trial with bone marrow aspirate concentrate with the SmartPReP2® system for the treatment of critical limb ischemia (NCT01245335). This is a U.S. pivotal randomized double-blind safety and efficacy trial comparing bone marrow aspirate concentrate with placebo injection into ischemic tissue of the lower extremity in 210 patients. The study start date is listed as January 2011; the expected study completion date is June 2014
November 2016.
  • A manufacturer-sponsored sham-controlled Phase III trial of Biomet Biologic’s MarrowStim P.A.D. kit™ (NCT01049919) is currently recruiting. The study has an estimated enrollment of 152 patients with completion expected May 2014 2015.
  • A Phase I study of placenta-derived mesenchymal-like stromal cells (PLX-PAD) by Pluristem has been completed (NCT00951210). The study had an enrollment of 12, the results have not been posted. A Phase II study in patients with CLI is expected to begin in 2013. A Phase II study of PLX-PAD for the treatment of intermittent claudication (NCT01679990) is currently recruiting with a target enrollment of 150 patients and completion in December 2015.
 
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.
 
A 2011 Cochrane review, updated in 2014, identified 2 small studies with a total of 57 patients that met the review’s inclusion criteria for local intramuscular transplantation of autologous mononuclear cells (monocytes) for critical limb ischemia (CLI) (Moazzami, 2011; 2014).  Studies were excluded that used mesenchymal stem cells (MSCs) or bone marrow aspirate. In one of the studies, intramuscular injection of bone marrow-derived mononuclear cells (BM-MNCs) was compared with standard conservative treatment. In the second study, peripheral blood-derived mononuclear cells were collected following injections of granulocyte-macrophage colony-stimulating factor (GM-CSF) and transplanted by intramuscular injections. Both studies showed a significant reduction in amputations with treatment with monocytes, but larger randomized controlled trials (RCTs) are needed to adequately evaluate the effect of treatment with greater certainty. No additional studies were found for the 2014 update of this systematic review.
 
Intra-Arterial Injection
JUVENTAS (Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial
Supplementation) is a randomized double-blind, placebo-controlled trial from Europe (NCT00371371) (Teraa, 2015). This foundation-supported trial evaluated the clinical effects of repeated intra-arterial infusion of BMMNCs in 160 patients with non-revascularizable critical limb ischemia. Patients received repeated intraarterial infusion of BM-MNCs or placebo (autologous peripheral blood erythrocytes) into the common femoral artery. The primary outcome measure, the rate of major amputation after 6 months, was not significantly different between the two groups (19% for BM-MNCs vs 13% controls). Secondary outcomes of quality of life, rest pain, ABI, and transcutaneous oxygen pressure improved to a similar extent in both groups, reinforcing the need for a placebo control in this type of trial.
 
Practice Guidelines and Position Statements
The European Society of Cardiology published 2011 guidelines on the diagnosis and treatment of PADs (Tendera, 2011). The guidelines did not recommend for or against stem cell therapy for PAD. The guidelines provided the following information: Stem cell and gene therapy for revascularization is a novel therapy that is currently being evaluated to stimulate neovascularization. For autologous cell transplantation, bone marrow and peripheral blood are rich sources of stem and progenitor cells. At this time, it is unclear which of the many different cell types is the most promising. At present, angiogenic gene and stem cell therapy are still being investigated, and it is too early to make firm recommendations.
   
2016 Update
A literature search conducted through April 2016 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
In 2015, Peeters Weem and colleagues reported a meta-analysis of 10 double-blind placebo-controlled RCTs on bone marrow-derived cell therapy in CLI (Peeters Weem, 2015). There were a total of 499 patients included in the metaanalysis, with a range of 10-160 patients per study. The studies varied in the route of administration (2 intra-arterial and 8 intramuscular) and in the cell type used (BM-MNCs, MSCs, Ixmyocel-T, CD34+ cells or CD133+ cells). Many of the studies were considered to be pilot or phase 2 studies and were rated as low study quality. Meta-analysis found no significant differences between the experimental and control groups for the primary outcomes of major amputation rate (relative risk [RR]: 0.91), survival (RR: 100), or amputation free survival (RR:1.03). There were modest improvements compared to placebo in the ankle brachial index, transcutaneous oxygen, and pain score, with a mean decrease of 1.3 in the cell therapy group and 0.6 in the controls. There was no difference between the percentage of healed ulcers.
 
Skora and colleagues reported on an intramuscular injection with a combination BM-MNCs and gene therapy with a vascular endothelial growth factor (VEGF) plasmid was tested in a European RCT with 32 patients (Skora, 2015). Controls in this study were treated pharmacologically and were therefore the groups were not blinded to treatment. Several objective measures were improved in the BM-MNC group but not in the control group. These included the ankle-brachial index, development of collateral vessels measured with angiography, and healing of ischemic ulcers. Amputations were performed in 25% of patients in the BM-MNC group and 50% of patients in the control group.
 
2018 Update
A literature search was conducted through June 2018.  There was no new information identified that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
PRACTICE GUIDELINES AND POSITION STATEMENTS
 
American Heart Association and American College of Cardiology
The 2016 guidelines from the American Heart Association and American College of Cardiology provided recommendations on the management of patients with lower-extremity peripheral arterial disease (PAD), including surgical and endovascular revascularization for critical limb ischemia (CLI), (AHA and ACC, 2017). Stem cell therapy for PAD was not addressed.
 
European Society of Cardiology
The 2011 European Society of Cardiology guidelines on the diagnosis and treatment of PAD did not recommend for or against stem cell therapy for PAD. However, in 2017, updated guidelines, published in collaboration with the European Society of Vascular Surgery, stated: “Angiogenic gene and stem cell therapy are still being investigated with insufficient evidence in favour of these treatments.” The current recommendation is that stem cell/gene therapy is not indicated in patients with chronic limb-threatening ischemia (class of recommendation: III; level of evidence: B), (ESC, 2017).
 
2019 Update
A literature search was conducted through June 2019.  There was no new information identified that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
In a meta-analysis of RCTs, Xie et al reviewed published evidence evaluating the safety and efficacy of autologous stem cell therapy in critical limb ischemia (Xie, 2018). Cell therapy increased the probability of angiogenesis (relative risk=5.91, confidence interval [CI]: 2.49-14.02, p<0.0001), ulcer healing (relative risk=1.73, CI: 1.45-2.06, p<0.00001), and a reduction in amputation rates (relative risk=0.59, CI: 0.46-0.76, p<0.0001). Compared with the control group, significant improvement in the cell therapy group was also seen in ankle-brachial index (mean difference=0.13, CI=0.11-0.15, p<0.00001), transcutaneous oxygen tension (mean difference=12.22, CI=5.03-19.41, p=0.0009), and pain-free walking distance (mean difference=144.84, CI=53.03-236.66, p=0.002).
 
Gupta et al evaluated the efficacy and safety of intramuscular adult human bone marrowderived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeutics Research, Bangalore, India) in a phase II prospective, open-label dose-ranging study (Gupta, 2017). Ninety patients were nonrandomly allocated to 3 groups: 1 million cells/kg body weight (n=36), 2 million cells/kg body weight (n=36), and standard of care (SOC; n=18). Compared with the SOC group, greater reduction in rest pain and healing of ulcers were see in the 2 million cells/kg body weight group (0.3 units per month [standard error (SE): 0.13], CI: -0.55 to -0.05, p=0.0193 and 11.0% decrease in size per month [SE: 0.05%], CI: 0.80-0.99, p=0.0253, respectively) and in the 1 million cells/kg body weight group (0.23 per month [SE: 0.13], CI: -0.49 to 0.03, p=0.081 and 2.0% decrease in size per month [SE: 0.06%], CI: 0.87-1.10, p=0.6967, respectively). Limitations of this study included the geographically and ethnically homogenous cohort and a lack of clearly defined methods for cohort selection. Additionally, patients in the cell administration groups had lower ankle-brachial pressure index values and larger ulcers indicating potential investigator bias to allocate more severe patients to the treatment groups.
 
Horie et al reported an RCT of 107 patients with PAD characterized as Buerger disease that evaluated the efficacy and safety of GM-CSF-mobilized peripheral blood mononuclear cell transplantation compared with SOC (Horie, 2018). Participants were randomized to guideline-based SOC or SOC plus intramuscular weight based peripheral blood mononuclear cell administration. After disease progression or completion of 1-year follow-up, 17 patients in the control group underwent the cell therapy. Furthermore, 21 patients underwent revascularization after completion of the protocol treatment period or after discontinuation of the study (12 in the cell therapy group, 9 in the control group; 18 patients underwent percutaneous transluminal angioplasty, 2 had bypass surgery, and 1 had thrombectomy). Serious adverse events occurred in 20% of the cell therapy group compared with 11.3% of the control group (p=0.28). Leukopenia, alkaline phosphatase elevation, and hyperuricemia were determined to be adverse events related to GM-CSF administration. This study was limited a small number of advanced cases (Fontaine stage IV cases (20.4%)), a high-risk group of hemodialysis patients and by the high number of patients who did not complete treatment (cell therapy group: 38.5%; control group: 50.9%).
 
2020 Update
A literature search was conducted through June 2020.  There was no new information identified that would prompt a change in the coverage statement.  
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through June 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
In 2019, a Global Vascular Guideline on management of chronic limb-threatening ischemia summarized the available literature on therapeutic angiogenesis for various etiologies (Conte, 2019). The guideline was a joint venture of the Society for Vascular Surgery, the European Society for Vascular Surgery, and the World Federation of Vascular Societies. Based on a moderate level of evidence, the guideline recommended that therapeutic angiogenesis in patients with chronic limb-threatening ischemia should be limited to the context of a clinical trial (strong recommendation). The authors noted that Phase 3 clinical trials are planned or underway so additional data may be forthcoming in the future.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through June 2022. No new literature was identified that would prompt a change in the coverage statement.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through June 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Pu et al included 12 RCTs (N=630) in a meta-analysis of patients with atherosclerosis obliterans (the most common type of PAD) (Pu, 2022). Autologous cell implantation was compared with placebo or standard care in all studies. A single injection of cell products was administered in all but 1 study in which injections were repeatedly administered. Follow-up periods ranged from 1 to 12 months. The analysis found improvements in total amputation (RR, 0.64; 95% CI, 0.47 to 0.87; p=.004; I2, 12%), major amputation (RR, 0.69; 95% CI, 0.50 to 0.94; p=.02; I2, 12%), and ABI (mean difference, 0.08; 95% CI, 0.02 to 0.13; p=.004; I2, 84%). Death and ulcer size were not improved with cell therapy. Findings of this analysis are applicable only to patients with no other therapy options. The analysis is limited by the small sample size in each trial (range, 10 to 160 patients) and heterogeneity in cell therapy methods (e.g., dosage, cell type, route of administration).
 
Moazzami et al published a Cochrane review of 4 RCTs (N=176) in patients with CLI who were treated with autologous bone marrow mononuclear cells (BM-MNCs) (Moazzami, 2022). It was uncertain if amputations were lower (4 studies; RR, 0.52; 95% CI, 0.27 to 0.99), and mortality was not reduced with BM-MNCs (3 studies; RR, 1.0; 95% CI, 0.15 to 6.63). Data were limited by risk of bias, imprecision, and inconsistency.
 
Dubsky et al compared standard therapy with BM-MNC in patients with CLI and diabetic foot (Dubsky, 2022). Forty patients with no-option chronic limb-threatening ischemia and no available treatment options were randomized to no treatment or BM-MNC for 12 weeks. Transcutaneous oxygen pressure (a marker of wound healing) had greater improvement in the BM-MNC group compared with no treatment (difference, 21.8 mm Hg; p=.034). There were more healed ulcers at 12 weeks in the BM-MNC group (31.3% vs. 0%; p=.48). The amputation rate and amputation-free survival was not different between groups. Although short-term improvements in outcomes were seen in this trial, the trial is limited by its small sample size, lack of placebo comparator, and single-center design.
 
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.
 
Use of autologous stem cells freshly harvested and allogeneic stem cells are reported to have a potential role in the treatment of PAD (Hussain, 2018). Stem cells can be administered in a variety of routes, derived from different progenitors, and be grouped with different co-factors, many of which are being studied in order to determine the best clinical option for patients. The primary outcome in stem cell therapy trials regulated by the U.S. Food and Drug Administration (FDA) is amputation-free survival, defined as time to major amputation and/or death from any cause. Other outcomes for critical limb ischemia include the Rutherford criteria for limb status, healing of ulcers, the Ankle-Brachial Index (ABI), transcutaneous oxygen pressure, and pain-free walking. The ABI measures arterial segmental pressures on the ankle and brachium and indexes ankle systolic pressure against brachial systolic pressure(normative range, 0.95 to 1.2 mm Hg).
 
Two endogenous compensating mechanisms may occur with occlusion of arterial vessels: capillary growth (angiogenesis) and development of collateral arterial vessels (arteriogenesis) (Krishna, 2015). Capillary growth is mediated by the hypoxia-induced release of chemokines and cytokines such as vascular endothelial growth factor and occurs by sprouting of small endothelial tubes from preexisting capillary beds. The resulting capillaries are small and cannot sufficiently compensate for a large occluded artery. Arteriogenesis with collateral growth is, in contrast, initiated by increasing shear forces against vessel walls when blood flow is redirected from the occluded transport artery to the small collateral branches, leading to an increase in the diameter of preexisting collateral arterioles.
 
The mechanism underlying arteriogenesis includes the migration of bone marrow-derived monocytes to the perivascular space. The bone marrow-derived monocytes adhere to and invade the collateral vessel wall. It is not known if the expansion of the collateral arteriole is due to the incorporation of stem cells into the wall of the vessel or to cytokines released by monocytic bone marrow cells that induce the proliferation of resident endothelial cells. It has been proposed that bone marrow-derived monocytic cells may be the putative circulating endothelial progenitor cells. Notably, the same risk factors for advanced ischemia (diabetes, smoking, hyperlipidemia, advanced age) are also risk factors for a lower number of circulating progenitor cells.
 
Peripheral arterial disease (PAD) is a common atherosclerotic syndrome associated with significant morbidity and mortality (Kullo, 2016). Aless common cause of PAD is Buerger disease (also called thromboangiitis obliterans), which is a nonatherosclerotic segmental inflammatory disease that occurs in younger patients and is associated with tobacco use (Liew, 2015). The development of PAD is characterized by narrowing and occlusion of arterial vessels and eventual reduction in distal perfusion. Critical limb ischemia is the end stage of lower-extremity PAD in which severe obstruction of blood flow results in ischemic pain at rest, ulcers, and a significant risk for limb loss.

CPT/HCPCS:
0263TIntramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; complete procedure including unilateral or bilateral bone marrow harvest
0264TIntramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; complete procedure excluding bone marrow harvest
0265TIntramuscular autologous bone marrow cell therapy, with preparation of harvested cells, multiple injections, one leg, including ultrasound guidance, if performed; unilateral or bilateral bone marrow harvest only for intramuscular autologous bone marrow cell therapy

References: Aboyans V, Ricco JB, Bartelink MEL, et al.(2017) 2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS): Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteriesEndorsed by: the European Stroke Organization (ESO)The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS). Eur Heart J. Aug 26 2017. PMID 28886620

Amann B, Ludemann C, Ruckert R et al.(2008) Design and rationale of a randomized, double-blind, placebo-controlled phase III study for autologous bone marrow cell transplantation in critical limb ischemia: the BONe Marrow Outcomes Trial in Critical Limb Ischemia (BONMOT-CLI). Vasa 2008; 37(4):319-25.

Amann B, Luedemann C, Ratei R et al.(2009) Autologous bone marrow cell transplantation increases leg perfusion and reduces amputations in patients with advanced critical limb ischemia due to peripheral artery disease. Cell Transplant 2009; 18(3):371-80.

Bartel RL, Booth E, Cramer C et al.(2013) From bench to bedside: review of gene and cell-based therapies and the slow advancement into phase 3 clinical trials, with a focus on Aastrom's Ixmyelocel-T. Stem Cell Rev 2013; 9(3):373-83.

Benoit E, O'Donnell TF, Jr., Iafrati MD et al.(2011) The role of amputation as an outcome measure in cellular therapy for critical limb ischemia: implications for clinical trial design. J Transl Med 2011; 9:165.

Conte MS, Bradbury AW, Kolh P, et al.(2019) Global vascular guidelines on the management of chronic limb-threatening ischemia. J Vasc Surg. Jun 2019; 69(6S): 3S-125S.e40. PMID 31159978

Domanchuk K, Ferrucci L, Guralnik JM et al.(2013) Progenitor cell release plus exercise to improve functional performance in peripheral artery disease: the PROPEL Study. Contemp Clin Trials 2013; 36(2):502-9.

Dubský M, Husáková J, Bem R, et al.(2022) Comparison of the impact of autologous cell therapy and conservative standard treatment on tissue oxygen supply and course of the diabetic foot in patients with chronic limb-threatening ischemia: A randomized controlled trial. Front Endocrinol (Lausanne). 2022; 13: 888809. PMID 36105404

Fadini GP, Agostini C, Avogaro A.(2010) Autologous stem cell therapy for peripheral arterial disease meta-analysis and systematic review of the literature. Atherosclerosis 2010; 209(1):10-7.

Gupta PK, Krishna M, Chullikana A, et al.(2017) Administration of Adult Human Bone Marrow-Derived, Cultured, Pooled, Allogeneic Mesenchymal Stromal Cells in Critical Limb Ischemia Due to Buerger's Disease: Phase II Study Report Suggests Clinical Efficacy. Stem Cells Transl Med. Mar 2017;6(3):689-699. PMID 28297569

Horie T, Yamazaki S, Hanada S, et al.(2018) Outcome From a Randomized Controlled Clinical Trial- Improvement of Peripheral Arterial Disease by Granulocyte Colony-Stimulating Factor-Mobilized Autologous Peripheral-Blood-Mononuclear Cell Transplantation (IMPACT). Circ J. Jul 25 2018;82(8):2165-2174. PMID 29877199

Hussain MA, Al-Omran M, Creager MA, et al.(2018) Antithrombotic Therapy for Peripheral Artery Disease: Recent Advances. J Am CollCardiol. May 29 2018; 71(21): 2450-2467. PMID 29793635

Jonsson TB, Larzon T, Arfvidsson B et al.(2012) Adverse events during treatment of critical limb ischemia with autologous peripheral blood mononuclear cell implant. Int Angiology 2012; 31(1):77-84.

Krishna SM, Moxon JV, Golledge J.(2015) A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int J Mol Sci. May 18 2015; 16(5): 11294-322. PMID 25993296

Kullo IJ, Rooke TW.(2016) CLINICAL PRACTICE. Peripheral Artery Disease. N Engl J Med. Mar 03 2016; 374(9): 861-71. PMID 26962905

Lawall H, Bramlage P, Amann B.(2011) Treatment of peripheral arterial disease using stem and progenitor cell therapy. J Vasc Surg 2011; 53(2):445-53.

Liew NC, Lee L, Nor Hanipah Z, et al.(2015) Pathogenesis and Management of Buerger's Disease. Int J Low Extrem Wounds. Sep 2015;14(3): 231-5. PMID 26264874

Liu FP, Dong JJ, Sun SJ et al.(2012) Autologous bone marrow stem cell transplantation in critical limb ischemia: a meta-analysis of randomized controlled trials. Chin Med J 2012; 125(23):4296-300.

Lu D, Chen B, Liang Z et al.(2011) Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract 2011; 92(1):26-36.

Matoba S, Tatsumi T, Murohara T et al.(2008) Long-term clinical outcome after intramuscular implantation of bone marrow mononuclear cells (Therapeutic Angiogenesis by Cell Transplantation [TACT] trial) in patients with chronic limb ischemia. Am Heart J 2008; 156(5):1010-8.

Moazzami B, Mohammadpour Z, Zabala ZE, et al.(2022) Local intramuscular transplantation of autologous bone marrow mononuclear cells for critical lower limb ischaemia. Cochrane Database Syst Rev. Jul 08 2022; 7(7): CD008347. PMID 35802393

Moazzami K, Majdzadeh R, Nedjat S.(2011) Local intramuscular transplantation of autologous mononuclear cells for critical lower limb ischaemia. Cochrane Database Syst Rev 2011; (12):CD008347.

Moazzami K, Moazzami B, Roohi A, et al.(2014) Local intramuscular transplantation of autologous mononuclear cells for critical lower limb ischaemia. Cochrane Database Syst Rev. 2014;12:CD008347. PMID 25525690

Peeters Weem SM, Teraa M, de Borst GJ, et al.(2015) Bone Marrow derived Cell Therapy in Critical Limb Ischemia: A Meta-analysis of Randomized Placebo Controlled Trials. Eur J Vasc Endovasc Surg. Oct 9 2015. PMID 26460286

Peeters Weem SM, Teraa M, den Ruijter HM, et al.(2015) Quality of Life After Treatment with Autologous Bone Marrow Derived Cells in No Option Severe Limb Ischemia. Eur J Vasc Endovasc Surg. Oct 26 2015. PMID 26511056

Poole J, Mavromatis K, Binongo JN et al.(2013) Effect of progenitor cell mobilization with granulocyte-macrophage colony-stimulating factor in patients with peripheral artery disease: a randomized clinical trial. JAMA 2013; 310(24):2631-9.

Poole J, Mavromatis K, Binongo JN et al.(2013) Effect of progenitor cell mobilization with granulocytemacrophage colony-stimulating factor in patients with peripheral artery disease: a randomized clinical trial. JAMA 2013; 310(24):2631-9.

Powell RJ, Comerota AJ, Berceli SA et al.(2011) Interim analysis results from the RESTORE-CLI, a randomized, double-blind multicenter phase II trial comparing expanded autologous bone marrow-derived tissue repair cells and placebo in patients with critical limb ischemia. J Vasc Surg 2011; 54(4):1032-41.

Powell RJ, Marston WA, Berceli SA et al.(2012) Cellular therapy with Ixmyelocel-T to treat critical limb ischemia: the randomized, double-blind, placebo-controlled RESTORE-CLI trial. Mol Ther 2012; 20(6):1280-6.

Powell RJ.(2012) Update on clinical trials evaluating the effect of biologic therapy in patients with critical limb ischemia. J Vasc Surg 2012; 56(1):264-6.

Prochazka V, Gumulec J, Jaluvka F et al.(2010) Cell therapy, a new standard in management of chronic critical limb ischemia and foot ulcer. Cell Transplant 2010; 19(11):1413-24.

Prochazka V, Gumulec J, Jaluvka F et al.(2010) Cell therapy, a new standard in management of chronic critical limb ischemia and foot ulcer. Cell Transplantation 2010; 19(11):1413-24.

Pu H, Huang Q, Zhang X, et al.(2022) A meta-analysis of randomized controlled trials on therapeutic efficacy and safety of autologous cell therapy for atherosclerosis obliterans. J Vasc Surg. Apr 2022; 75(4): 1440-1449.e5. PMID 34788653

Skora J, Pupka A, Janczak D, et al.(2015) Combined autologous bone marrow mononuclear cell and gene therapy as the last resort for patients with critical limb ischemia. Arch Med Sci. Apr 25 2015;11(2):325-331. PMID 25995748

Sprengers RW, Moll FL, Teraa M et al.(2010) Rationale and design of the JUVENTAS trial for repeated intra-arterial infusion of autologous bone marrow-derived mononuclear cells in patients with critical limb ischemia. J Vasc Surg 2010; 51(6):1564-8.

Tateishi-Yuyama E, Matsubara H, Murohara T et al.(2002) Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet 2002; 360(9331):427-35.

Tendera M, Aboyans V, Bartelink M, et al.(2011) ESC Guidelines on the Diagnosis and Treatment of Peripheral Artery Diseases. Eur Heart J. 2011; 32(22):2851-2906. PMID 21873417

Teraa M, Sprengers RW, Schutgens RE, et al.(2015) Effect of Repetitive Intra-Arterial Infusion of Bone Marrow Mononuclear Cells in Patients With No-Option Limb Ischemia: The Randomized, Double-Blind, Placebo- Controlled Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation (JUVENTAS) Trial. Circulation. Mar 10 2015;131(10):851-860. PMID 25567765

Teraa M, Sprengers RW, van der Graaf Y et al.(2013) Autologous bone marrow-derived cell therapy in patients with critical limb ischemia: a meta-analysis of randomized controlled clinical trials. Ann Surg 2013; 258(6):922-9.

Teraa M, Sprengers RW, van der Graaf Y et al.(2013) Autologous bone marrow-derived cell therapy in patients with critical limb ischemia: a meta-analysis of randomized controlled clinical trials. Ann Surg 2013; 258(6):922-9.

Valentine EA, Ochroch EA.(2017) 2016 American College of Cardiology/American Heart Association guideline on the management of patients with lower extremity peripheral artery disease: perioperative implications. J Cardiothorac Vasc Anesth. Oct 2017;31(5):1543-1553. PMID 28826846

Walter DH, Krankenberg H, Balzer JO et al.(2011) Intraarterial administration of bone marrow mononuclear cells in patients with critical limb ischemia: a randomized-start, placebo-controlled pilot trial (PROVASA). Circulation Cardiovascular interventions 2011; 4(1):26-37.

Xie B, Luo H, Zhang Y, et al.(2018) Autologous Stem Cell Therapy in Critical Limb Ischemia: A Meta-Analysis of Randomized Controlled Trials. Stem Cells Int. 2018;2018:7528464. PMID 29977308


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.