Coverage Policy Manual
Policy #: 2000053
Category: Medicine
Initiated: October 2000
Last Review: February 2024
  HDC & Hematopoietic Stem Cell Support-Autoimmune Diseases, Including Multiple Sclerosis

Description:
Most patients with autoimmune disorders respond to conventional drug therapies; however, conventional drug therapies are not curative and a proportion of patients suffer from autoimmune diseases that range from severe to recalcitrant to rapidly progressive. It is in this group of patients with a severe autoimmune disease that alternative therapies have been sought, including hematopoietic cell transplantation (HCT).
 
Autoimmune Disease Treatment
Immune suppression is a common treatment strategy for many autoimmune diseases, particularly rheumatic diseases (e.g., rheumatoid arthritis [RA], systemic lupus erythematosus [SLE], scleroderma). Most patients with autoimmune disorders respond to conventional therapies, which consist of anti-inflammatory agents, immunosuppressants, and immunomodulating drugs; however, conventional drug therapies are not curative, and a proportion of patients suffer from autoimmune diseases that range from severe to recalcitrant to rapidly progressive. It is for this group of patients with a severe autoimmune disease that alternative therapies have been sought, including hematopoietic cell transplantation (HCT). The primary concept underlying the use of HCT for these diseases is this: ablating and “resetting” the immune system can alter the disease process by inducing a sustained remission that possibly leads to cure (Nikolov, 2008).
 
Hematopoietic Stem-Cell Transplantation
Hematopoietic stem-cell transplantation is a procedure in which hematopoietic stem cells are intravenously infused to restore bone marrow and immune function in cancer patients who receive bone marrow-toxic doses of cytotoxic drugs with or without whole-body radiation therapy. Hematopoietic stem cells from the bone marrow may be obtained from the transplant recipient (autologous HCT) or from a donor (allogeneic HCT [allow-HCT]). They can be harvested from bone marrow, peripheral blood, or umbilical cord blood shortly after delivery of neonates.
 
Immunologic compatibility between infused hematopoietic stem cells and the recipient is not an issue in autologous SCT. In allogeneic stem cell transplantation, immunologic compatibility between donor and patient is a critical factor for achieving a successful outcome. Compatibility is established by typing of human leukocyte antigens (HLA) using cellular, serologic, or molecular techniques. The term HLA refers to the gene complex expressed at the HLA A, B, and DR (antigen-D related) loci on each arm of chromosome 6. An acceptable donor will match the patient at all or most of the HLA loci.
 
Conditioning for Hematopoietic Cell Transplantation
Conventional Conditioning
The conventional (“classical”) practice of allo-HCT involves administration of cytotoxic agents (e.g., cyclophosphamide, busulfan) with or without total body irradiation at doses sufficient to cause bone marrow ablation in the recipient. The beneficial treatment effect of this procedure is due to a combination of the initial eradication of malignant cells and subsequent graft-versus-malignancy (GVM) effect mediated by non-self-immunologic effector cells. While the slower GVM effect is considered the potentially curative component, it may be overwhelmed by existing disease in the absence of pretransplant conditioning. Intense conditioning regimens are limited to patients who are sufficiently medically fit to tolerate substantial adverse effects. These include opportunistic infections secondary to loss of endogenous bone marrow function and organ damage or failure caused by cytotoxic drugs. Subsequent to graft infusion in allo-HCT, immunosuppressant drugs are required to minimize graft rejection and graft-versus-host disease (GVHD), which increases susceptibility to opportunistic infections.
 
The success of autologous HCT is predicated on the potential of cytotoxic chemotherapy, with or without radiotherapy, to eradicate cancerous cells from the blood and bone marrow. This permits subsequent engraftment and repopulation of the bone marrow with presumably normal hematopoietic stem cells obtained from the patient before undergoing bone marrow ablation. Therefore, autologous HCT is typically performed as consolidation therapy when the patient’s disease is in complete remission. Patients who undergo autologous HCT are also susceptible to chemotherapy-related toxicities and opportunistic infections before engraftment, but not GVHD.
 
Reduced-Intensity Conditioning Allogeneic Hematopoietic Cell Transplantation
Reduced-intensity conditioning (RIC) refers to the pretransplant use of lower doses of cytotoxic drugs or less intense regimens of radiotherapy than are used in traditional full-dose myeloablative conditioning treatments. Although the definition of RIC is variable, with numerous versions employed, all regimens seek to balance the competing effects of relapse due to residual disease and non-relapse mortality. The goal of RIC is to reduce disease burden and to minimize associated treatment-related morbidity and non-relapse mortality in the period during which the beneficial GVM effect of allogeneic transplantation develops. Reduced-intensity conditioning regimens range from nearly total myeloablative to minimally myeloablative with lymphoablation, with intensity tailored to specific diseases and patient condition. Patients who undergo RIC with allo-HCT initially demonstrate donor cell engraftment and bone marrow mixed chimerism. Most will subsequently convert to full-donor chimerism. In this review, the term reduced-intensity conditioning will refer to all conditioning regimens intended to be nonmyeloablative.
 
Regulatory Status
The U.S. Food and Drug Administration 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. Hematopoietic stem cells are included in these regulations.

Policy/
Coverage:
Effective March 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Autologous hematopoietic cell transplantation as a treatment of systemic sclerosis/scleroderma meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness when all the following conditions are met:
 
    • adult patients <60 years of age; AND
    • maximum duration of condition of 5 years; AND
    • modified Rodnan Scale Scores >15; AND
    • history of < 6 months treatment with cyclophosphamide; AND
    • no active gastric antral vascular ectasia; AND
    • internal organ involvement defined by one of the following:
 
        • Cardiac: abnormal electrocardiogram; OR
        • Pulmonary: diffusing capacity of carbon monoxide (DLCo) <80% of predicted value; decline of forced vital capacity (FVC) of >10% in last 12 months; pulmonary fibrosis; ground glass appearance on high resolution chest CT; OR
        • Renal: scleroderma-related renal disease
 
    • do not have any exclusion criteria for organ involvement listed below (if patient meets any of the criteria below they should not be considered for autologous HCT):
 
        • Cardiac: left ventricular ejection fraction <50%; tricuspid annular plane systolic excursion <1.8 cm; pulmonary artery systolic pressure >40 mm Hg; mean pulmonary artery pressure >25 mm Hg
        • Pulmonary: DLCo <40% of predicted value; FVC <45% of predicted value
        • Renal: creatinine clearance <40 ml/minute
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Autologous hematopoietic cell transplantation as a treatment of systemic sclerosis/scleroderma not meeting the above criteria 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 language, autologous hematopoietic cell transplantation as a treatment of systemic sclerosis/scleroderma not meeting the above criteria is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Autologous or allogeneic hematopoietic cell transplantation does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes as a treatment of autoimmune diseases, including, but not limited to, the following:
 
    • Multiple sclerosis
    • Systemic lupus erythematosus
    • Juvenile idiopathic or rheumatoid arthritis
    • Chronic inflammatory demyelinating polyneuropathy
    • Type 1 diabetes
 
For members with contracts without primary coverage criteria, autologous or allogeneic hematopoietic cell transplantation is considered investigational as a treatment of autoimmune diseases, including, but not limited to, the following:
 
    • Multiple sclerosis
    • Systemic lupus erythematosus
    • Juvenile idiopathic or rheumatoid arthritis
    • Chronic inflammatory demyelinating polyneuropathy
    • Type 1 diabetes
 
Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Allogeneic transplant after previous high dose chemotherapy with autologous stem cell support does not meet primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.
 
For contracts without primary coverage criteria language, allogeneic transplant after previous high dose chemotherapy with autologous stem cell support is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective October 2019-March 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Autologous hematopoietic cell transplantation as a treatment of systemic sclerosis/scleroderma meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness if ALL of the following conditions are met:
 
    • adult patients <60 years of age; AND
    • maximum duration of condition of 5 years; AND
    • modified Rodnan Scale Scores >15; AND
    • history of < 6 months treatment with cyclophosphamide; AND
    • no active gastric antral vascular ectasia; AND
    • internal organ involvement defined by one of the following:
 
            • Cardiac: abnormal electrocardiogram; OR
            • Pulmonary: diffusing capacity of carbon monoxide (DLCo) <80% of predicted value; decline of forced vital capacity (FVC) of >10% in last 12 months; pulmonary fibrosis; ground glass appearance on high resolution chest CT; OR
            • Renal: scleroderma-related renal disease
 
    • do not have any exclusion criteria for organ involvement listed below (if patient meets any of the criteria below they should not be considered for autologous HCT):
 
            • Cardiac: left ventricular ejection fraction <50%; tricuspid annular plane systolic excursion <1.8 cm; pulmonary artery systolic pressure >40 mm Hg; mean pulmonary artery pressure >25 mm Hg
            • Pulmonary: DLCo <40% of predicted value; FVC <45% of predicted value
            • Renal: creatinine clearance <40 ml/minute
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Autologous or allogeneic hematopoietic cell transplantation does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness as a treatment of autoimmune diseases, including, but not limited to, the following:
 
    • Multiple sclerosis
    • Systemic lupus erythematosus
    • Juvenile idiopathic or rheumatoid arthritis
    • Chronic inflammatory demyelinating polyneuropathy
    • Type 1 diabetes
 
For members with contracts without primary coverage criteria, autologous or allogeneic hematopoietic cell transplantation is considered investigational as a treatment of autoimmune diseases, including, but not limited to, the following:
 
    • Multiple sclerosis
    • Systemic lupus erythematosus
    • Juvenile idiopathic or rheumatoid arthritis
    • Chronic inflammatory demyelinating polyneuropathy
    • Type 1 diabetes
 
Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Allogeneic transplant after previous high dose chemotherapy with autologous stem cell support does not meet primary coverage criteria that there be scientific evidence of effectiveness..
 
For contracts without primary coverage criteria language, allogeneic transplant after previous high dose chemotherapy with autologous stem cell support is considered investigational.  Investigational services are exclusions in the member certificate of coverage.
 
Tandem transplants are specific exclusions in most member benefit contract.
 
Effective January 2011 to October 2019
 
High dose chemotherapy with allogeneic or autologous stem cell support for the treatment of autoimmune diseases, including but not limited to juvenile idiopathic and rheumatoid arthritis, SLE, systemic sclerosis (scleroderma) and type 1 diabetes mellitus does not meet primary coverage criteria that there be scientific evidence of effectiveness.
 
For contracts without primary coverage criteria language, high dose chemotherapy with allogeneic or autologous stem cell support for the treatment of autoimmune diseases, including but not limited to juvenile idiopathic and rheumatoid arthritis, SLE, systemic sclerosis (scleroderma) and type 1 diabetes mellitus is considered investigational.  Investigational services are exclusions in the member certificate of coverage.
 
Tandem transplants are specific exclusions in most member benefit contract.
 
Allogeneic transplant after previous high dose chemotherapy with autologous stem cell support does not meet primary coverage criteria that there be scientific evidence of effectiveness..
 
For contracts without primary coverage criteria language, allogeneic transplant after previous high dose chemotherapy with autologous stem cell support is considered investigational.  Investigational services are exclusions in the member certificate of coverage.
 
Effective October 2000 to December 2010
High dose chemotherapy with allogeneic or autologous stem cell support for the treatment of autoimmune diseases, including but not limited to arthritis, SLE and systemic sclerosis (scleroderma) does not meet primary coverage criteria for effectiveness.
 
For contracts without primary coverage criteria language, high dose chemotherapy with allogeneic or autologous stem cell support for the treatment of autoimmune diseases, including but not limited to arthritis, SLE and systemic sclerosis (scleroderma) is considered investigational and is not covered.  Investigational services are an exclusion in the member certificate of coverage.
 
Tandem transplants are not covered based on a specific exclusion in the member benefit contract.
 
Allogeneic transplant after previous high dose chemotherapy with autologous stem cell support is not covered.

Rationale:
2006 Update
An updated literature search performed in February 2006 revealed no evidence from randomized clinical trials that would alter the policy statement. However, Burt and colleagues recently published the results of the largest single-center series available in the United States.  Between April 1997 through January 2005, they enrolled 50 patients (mean age 30 +/- 10.9 years [SD], 43 women, 7 men) with systemic lupus erythematosus (SLE) refractory to standard immunosuppressive therapies and either organ- or life-threatening visceral involvement in a single-arm trial. All subjects had at least 4 of 11 American College of Rheumatology criteria for SLE and required more than 20 mg/d of prednisone or its equivalent despite use of cyclophosphamide. Patients underwent autologous SCT following a lymphoablative conditioning regimen. Two patients died after mobilization, yielding a treatment-related mortality of 4% (2/50). After a mean follow-up of 29 months (range, 6 months to 7.5 years) overall 5-year survival was 84%, and the probability of disease-free survival was 50%. Several parameters of SLE activity (described in the 2001 Assessment ) improved, including renal function, SLE disease activity index (DAI) score, antinuclear antibody, anti-ds DNA, complement, and carbon monoxide diffusion lung capacity. The investigators suggest these results justify a randomized trial comparing immunosuppression plus autologous SCT versus continued standard of care.
 
An editorial by Petri and Brodsky that accompanied the article by Burt and colleagues concurred that randomized clinical trials are needed to determine whether this treatment approach improves outcomes when compared with conventional therapies.
 
One phase III randomized, open-label, active control clinical trial, “Scleroderma: Cyclophosphamide or Transplantation” (SCOT, NCT00114530, NIAID No. SCSSc-01) sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) is currently recruiting patients in the United States. Two other NIAID-sponsored phase II trials are planned but not yet open for patient recruitment. The first is a randomized, active control, open-label trial, Lupus Immunosuppressive/Immunomodulatory Therapy or Stem Cell Transplant (LIST, NCT00230035, NIAID No. SCSLE-01). The second is a nonrandomized, open-label uncontrolled trial, High-Dose Immunosuppression and Autologous Transplantation for Multiple Sclerosis Study (HALT MS, NCT00288626, NIAID No. SCMS2). Other phase III, randomized, clinical trials are being conducted in Europe for scleroderma in the Autologous Stem Cell Transplantation International Scleroderma (ASTIS) trial, for multiple sclerosis (ASTIMS), and for rheumatoid arthritis (ASTIRA). While several phase I/II studies have been completed, longer-term follow-up is needed before conclusions can be reached from study findings. Therefore, the policy statement is unchanged.
 
2007 Update
An updated literature search performed in July 2007 revealed no evidence from randomized, clinical trials that would alter the policy statement.
 
The European Group for Blood and Marrow Transplantation (EBMT) autoimmune diseases working party database reported new data from a retrospective survey of 178 patients with MS who underwent autologous SCT following one of several different preparative regimens.  Overall, at median follow-up of about 42 months, the disease remained stable or improved in 63% of cases and worsened in 37%. Autologous SCT was associated with significantly better progression-free survival in a subset of younger patients (i.e., younger than 40 years of age) affected by severe, progressive multiple sclerosis (MS) who received autologous SCT within 5 years from diagnosis compared to those older than 40 years. The authors suggest that autologous SCT could be considered after failure of conventional treatments in patients with rapidly progressing MS. However, they caution that the role of autologous SCT in the treatment of refractory MS needs to be established through prospective randomized, controlled trials. Several editorials concur with the view that the role of autologous SCT is not established in MS or other autoimmune diseases.  
 
A review article on randomized trials of autologous SCT for autoimmune diseases discusses the relative merits of lymphoablative versus myeloablative SCT regimens, concurring that randomized, controlled trials are needed to confirm benefits observed with this therapy in case reports and uncontrolled series.  The randomized trials outlined in the 2006 update to this policy remain in progress. The policy statement is unchanged.
 
2008 Update
The trials outlined in the 2006 update in this policy remain in progress, with the exception of the LIST study (NCT00230035, NIAID No. SCSLE-01), which was terminated due to lack of accrual.
 
An additional study recruiting patients is a phase II/III randomized, open-label multicenter study of high-dose immunosuppressive therapy followed by autologous SCT versus high-dose pulse cyclophosphamide in patients with severe systemic sclerosis (NCT00545038).
 
Therefore, the policy statement is unchanged.
 
2011 Update
Multiple Sclerosis
Currently, multiple sclerosis is the most common autoimmune disease for which autologous HSCT is being studied (Pasquini, 2010).  Following initial promising clinical experience, more than 350 consecutive cases have been reported by the EBMT over the last decade (Pasquini, 2010).  Most patients who underwent autologous HSCT for MS in the early studies had secondary progressive MS, and relatively fewer had relapsing remitting disease, with a Kurtzke Expanded Disability Status Scale (EDSS) of 3.0–9.5 at the time of HSCT (Pasquini, 2010). Improvements in supportive care and patient selection have contributed to improved outcomes, with a significant reduction in treatment-related mortality to 1.3% seen during 2001–2007(Pasquini, 2010).  It is now generally accepted that administering HSCT relatively early in the course of the disease to reduce inflammation before irreversible neuronal damage occurs is important. Current studies target MS patients with active disease and worsening disability, as evidenced clinically by relapse, change in EDSS, and/or inflammatory activity seen on magnetic resonance imaging (MRI) and who have failed at least one approved first-line immunomodulatory MS therapy for enrollment. Follow-up of several years will be needed to evaluate outcomes of these clinical trials.
 
A recent review summarizes the experience with HSCT and MS (Atkins, 2010).  A small number of patients have undergone autologous HSCT for the rare malignant form of MS, which is characterized by very active inflammatory disease with high relapse rates leading to a rapid progression of disabilities from the onset (Fagius, 2009) (Kimiskidis, 2008) (Mancardi. 2005).  These patients had persistent disease activity despite numerous different treatments. All but one patient were relapse-free without the need for ongoing immunosuppression after autologous HSCT with up to 66 months of follow-up. One patient experienced a mild relapse that improved with conventional treatment. All of the patients had remarkable improvement in their functional abilities.
 
The majority of patients who have undergone autologous HSCT have had poor prognosis MS, which manifests as frequent relapses or the early onset of the secondary progressive (SPMS) phase of the illness within 3 to 5 years of diagnosis (Atkins, 2010).  Studies are mainly case series that report the outcomes of autologous HSCT in MS patients with ongoing disease activity that is refractory to conventional disease-modifying agents. There has not been a “standard” transplant regimen, and different mobilization and conditioning regimens have been used throughout the published series. Clinical relapses were reported following autologous HSCT in one series, but overall, there has been an absence of ongoing acute episodic inflammatory disease activity in most reports. Evidence of ongoing chronic disease activity was seen in 14–76% of cases in the different series, with median follow-up between 1.5 to 3 years. Although the frequency of progression seems to be similar to what might be expected from historical controls, in many of the transplant studies, between 5% and 60% of patients actually had significant and sustained improvement in their disability score, and MS progression-free survival seems to level off with increasing length of follow-up after autologous HSCT, a change from the expected natural history of progressive disabilities increasing with time.
 
Burt and colleagues have transplanted 21 patients with relapsing-remitting MS with ongoing relapses during treatment with interferon (Burt, 2009).  The conditioning regimen was nonmyeloablative. With a median follow-up of 37 months, 16 patients remained free of relapse, whereas 17 of the 21 patients had a 1-point or greater improvement in their EDSS score.
 
The EBMT Autoimmune Diseases Working Party database reported new data from a retrospective survey of 178 patients with MS who underwent autologous HSCT following one of several different preparative regimens (Saccardi, 2006).  Overall, at median follow-up of about 42 months, the disease remained stable or improved in 63% of cases and worsened in 37%. Autologous HSCT was associated with significantly better progression-free survival in a subset of younger patients (i.e., younger than 40 years of age) affected by severe, progressive MS who received autologous HSCT within 5 years from diagnosis compared to those older than 40 years. The authors suggest that autologous HSCT could be considered after failure of conventional treatments in patients with rapidly progressing MS.
 
Clinical Trials
 
A Phase III randomized trial (Stem Cell Therapy for Patients With Multiple Sclerosis Failing Interferon A Randomized Study) is recruiting participants to study the effect of autologous peripheral blood HSCT in patients with relapsing MS versus FDA-approved standard of care. Primary endpoint is disease progression. Patients will be followed for 5 years after randomization. Estimated enrollment is 110, and estimated study completion date is January 2012 (NCT00273364).
 
The Phase II randomized ASTIMS trial mentioned in an earlier update was terminated due to difficulty in accruing patients and lack of funds.
 
The HALT-MS Study (mentioned in an earlier update) is ongoing. Estimated enrollment is 25, and estimated study completion date is January 2015 (NCT00288626).
 
The Canadian MS-BMT Phase II study is to determine the effect of autologous HSCT on early-stage MS. Estimated enrollment is 24. Enrollment completed July 2009.
 
Systemic Sclerosis/Scleroderma
Vonk and colleagues reported the long-term results of 28 patients with severe diffuse cutaneous systemic sclerosis who underwent autologous HSCT from 1998 to 2004 (Vonk, 2008).  There was 1 transplant-related death and 1 death due to progressive disease, leaving 26 patients for evaluation. After a median follow-up of 5.3 years (range, 1–7.5), 81% (n=21/26) of the patients demonstrated a clinically beneficial response. Skin sclerosis was measured with a modified Rodnan skin score, and a significant (i.e., greater than 25%) decrease (i.e., improvement) was achieved in 19 of 26 patients after 1 year and 15/16 after 5 years. At inclusion into the study, 65% of patients had significant lung involvement; all pulmonary function parameters remained stable after transplant at 5 and 7 years’ follow-up. Analyzing World Health Organization (WHO) performance status, which reflects the effect of HSCT on the combination of functional status, skin, lung, heart, and kidney involvement, the percentage of patients with a performance score of 0 increased to 56% compared to 4% at baseline. Estimated survival at 5 years was 96.2% (95% confidence interval [CI]: 89–100%) and at 7 years was 84.8% (95% CI: 70.2–100%) and event-free survival, (survival without mortality, relapse, or progression of systemic sclerosis resulting in major organ dysfunction) was 64.3% (95% CI: 47.9–86%) at 5 years and 57.1% (95% CI: 39.3–83%) at 7 years. For comparison, an international meta-analysis published in 2005 estimated the 5-year mortality rate in patients with severe systemic sclerosis at 40% (Ioannidis, 2005).
 
Nash and colleagues reported the long-term follow-up of 34 patients with diffuse cutaneous systemic sclerosis with significant visceral organ involvement who were enrolled in a multi-institutional pilot study between 1997 and 2005 and underwent autologous HSCT (Nash, 2007).  Of the 34 patients, 79% survived 1 year and were evaluable for response (there were 8 transplant-related deaths and 4 systemic sclerosis-related deaths). Seventeen of the 27 (63%) evaluable patients had sustained responses at a median follow-up of 4 years (range, 1-8 years). Skin biopsies showed a statistically significant decrease in dermal fibrosis compared with baseline (p less than 0.001) and, in general, lung, heart, and kidney function remained stable. Overall function as assessed in 25 patients by the modified Health Assessment Questionnaire Disability Index showed improvement in 19, and disease response was observed in the skin of 23 of 25 and lungs of 8 of 27 patients. Estimated overall and progression-free survival were both 64% at 5 years.
 
Clinical Trials
 
ASTIS is a Phase III randomized, international, multicenter trial. A total of 155 patients with diffuse systemic sclerosis will be randomized to high-dose immunoablation and autologous HSCT or pulsed cyclophosphamide. Outcome measures include survival and prevention of major organ failure, safety, and quality of life. As of September 2009, 151 patients had been accrued .
 
A Phase II randomized, open-label study is recruiting patients with systemic scleroderma to undergo either autologous peripheral blood HSCT or receive pulsed cyclophosphamide. Primary outcome measures are time to treatment failure and improvement in skin score of at least 25% or a 10% improvement in lung function or in cardiac tests. Estimated enrollment is 60, with an estimated study completion date of September 2012 (NCT00278525).
 
The SCOT trial (mentioned in an earlier update) is ongoing.  Estimated enrollment is 114 patients with an estimated study completion date of June 2016 (NCT00114530).
 
Systemic Lupus Erythematosus
 
Burt and colleagues published the results of the largest single-center series of this treatment in systemic lupus erythematosus (SLE) available in the U.S. (Burt, 2006)  Between April 1997 through January 2005, they enrolled 50 patients (mean age: 30 [standard deviation: +/- 10.9 years], 43 women, 7 men) with SLE refractory to standard immunosuppressive therapies and either organ- or life-threatening visceral involvement in a single-arm trial. All subjects had at least 4 of 11 American College of Rheumatology criteria for SLE and required more than 20 mg per day of prednisone or its equivalent in spite of use of cyclophosphamide. Patients underwent autologous SCT following a lymphoablative conditioning regimen. Two patients died after mobilization, yielding a treatment-related mortality of 4% (2/50). After a mean follow-up of 29 months (range, 6 months to 7.5 years) overall 5-year survival was 84%, and the probability of disease-free survival was 50%. Several parameters of SLE activity (described in the 2001 TEC Assessment [4]) improved, including renal function, SLE disease activity index (DAI) score, antinuclear antibody, anti-ds DNA, complement, and carbon monoxide diffusion lung capacity. The investigators suggest these results justify a randomized trial comparing immunosuppression plus autologous SCT versus continued standard of care.
 
Clinical Trials
 
Three nonrandomized, open-label Phase II trials are recruiting patients, ongoing or completed studying the effectiveness of autologous HSCT in patients with SLE: one with an estimated enrollment of 9 and study completion date of April 2018 (NCT00076752), another with an estimated enrollment of 30 and study completion date of April 2014 (NCT00750971), and a third with an estimated enrollment of 50 and study completion date of May 2008 (NCT00271934).
 
Juvenile Arthritis
A review article by Saccardi et al. summarizes the experience thus far with juvenile idiopathic and rheumatoid arthritis as follows (Saccardi, 2008): More than 50 patients with juvenile idiopathic arthritis have been reported to the EBMT Registry. The largest cohort study initially used 1 conditioning regimen, and thereafter, a modified protocol. Overall drug-free remission rate was approximately 50%. Some late relapses have been reported, and only partial correction of growth impairment has been seen. The frequency of HSCT for rheumatoid arthritis has decreased significantly since 2000, due to the introduction of new biologic therapies. Most patients who have undergone HSCT have had persistence or relapse of disease activity within 6 months of transplant.
 
Clinical Trials
 
No Phase II or III clinical trials were identified on ClinicalTrials.gov using HSCT in juvenile idiopathic or rheumatoid arthritis.
 
Type 1 Diabetes Mellitus
Couri and colleagues reported the results of a prospective Phase I/II study of autologous HSCT in 23 patients with type 1 diabetes mellitus (age range, 13-31 years) diagnosed in the previous 6 weeks by clinical findings with hyperglycemia and confirmed by measurement of serum levels of antiglutamic acid decarboxylase antibodies (Couri, 2009).  Enrollment was November 2003-April 2008, with follow-up until December 2008.
 
After a mean follow-up of 29.8 months (range, 7-58 months) following autologous nonmyeloablative HSCT, C-peptide levels increased significantly (C-peptide is a measure of islet cell mass and an increase after HSCT indicates preservation of islet cells) and the majority of patients achieved insulin independence with good glycemic control (Couri, 2009).  Twenty patients without previous ketoacidosis and not receiving corticosteroids during the preparative regimen became insulin free. Twelve patients maintained insulin independence for a mean of 31 months (range, 14-52 months), and 8 patients relapsed and resumed low-dose insulin. In the continuously insulin-independent group, HbA1c levels were less than 7.0% and mean area under the curve (AUC) C-peptide levels increased significantly from 225.0 (standard error [SE], 75.2) ng/mL per 2 hours pretransplantation to 785.4 (SE, 90.3) ng/mL per 2 hours at 24 months post-transplantation (p less than 0.001) and to 728.1 (SE: 144.4) ng/mL per 2 hours at 36 months (p=0.001). In the transiently insulin-independent group, mean AUC of C-peptide levels also increased from 148.9 (SE: 75.2) ng/mL per 2 hours pretransplantation to 546.8 (SE: 96.9) ng/mL per 2 hours at 36 months (p=0.001), which was sustained at 48 months. In this latter group, 2 patients regained insulin independence after treatment with sitagliptin (Januvia®), which was associated with an increase in C-peptide levels. There was no transplant-related mortality.
 
Clinical Trials
 
Two Phase I/II and one Phase II trials are recruiting patients with type 1 diabetes mellitus for autologous HSCT (NCT00315133, NCT01121029, NCT00807651). One Phase II and one Phase II/III trial are ongoing or recruiting patients with type 2 diabetes mellitus for autologous HSCT (NCT00644241, NCT01065298).
 
Other Autoimmune Diseases
Phase II/III protocols are being developed for Crohn’s disease and chronic inflammatory demyelinating polyneuropathy. For the remaining autoimmune diseases (including immune cytopenias, relapsing polychondritis, and others), the numbers are too small to draw conclusions, with further Phase I/II pilot studies proceeding (Tyndall, 2009).
 
Summary
Initial studies focused on using HSCT as salvage therapy for end-stage treatment of refractory autoimmune diseases. More recent experience has better helped to define which patients are most likely to benefit from HSCT, and the field has shifted to the use of HSCT earlier in the disease course before irreversible organ damage and to the use of safer and less intense nonmyeloablative conditioning regimens.
 
The experience with HSCT and autoimmune disorders has been predominantly with autologous transplants, and a number of published clinical reports with follow-up have demonstrated the safety and in some patients (particularly those with systemic sclerosis, SLE, and MS) the impact of HSCT in selected autoimmune diseases.
 
Although some of the initial results have been promising, this field continues to evolve. Many trials (randomized and nonrandomized) are currently recruiting or ongoing comparing the use of HSCT to conventional therapy for most of the diseases addressed in this policy; the results of these trials will further define the role of HSCT in the management of these diseases.
 
2012 Update
A literature search was conducted through November 2012.  There was no new literature identified that would prompt a change in the coverage statement. A summary of the key identified literature is included below.
 
Multiple Sclerosis
Fassas and colleagues reported the long-term results of a phase I/II study conducted in a single center which investigated the effect of HSCT in the treatment of MS (Fassas, 2011). The authors reported on the clinical and MRI outcomes of 35 patients with aggressive MS treated with HSCT after a median follow-up period of 11 (range 2-15) years. Disease PFS at 15 years was 44% for patients with active central nervous system (CNS) disease and 10% for those without (p=0.01); median time to progression was 11 years (95% CI 0-22) and 2 years (0-6). Improvements by 0.5-5.5 (median 1) Expanded Disability Status Scale (EDSS) points were observed in 16 cases lasting for a median of 2 years. In 9 of these patients, EDSS scores did not progress above baseline scores. Two patients died, at 2 months and 2.5 years, from transplant-related complications. Gadolinium-enhancing lesions were significantly reduced after mobilization but were maximally and persistently diminished post-HSCT. The authors concluded that HSCT should be reserved for aggressive cases of MS, still in the inflammatory phase of the disease, and for the malignant form, in which it can be life-saving, and that HSCT can result in PFS rates of 25% and can have an impressive and sustained effect in suppressing disease activity on MRI.
 
Shevchenko and colleagues reported the results of a prospective phase II open-label single-center study which analyzed the safety and efficacy of autologous HSCT with reduced-intensity conditioning regimen in 95 patients with different types of MS (Shevchenko, 2012). The patients underwent early, conventional, and salvage/late transplantation. The efficacy was evaluated based on clinical and quality of life outcomes. No transplantation-related deaths were observed. All of the patients, except one, responded to the treatment. At long-term follow-up (mean 46 months), the overall clinical response in terms of disease improvement or stabilization was 80%. The estimated PFS at 5 years was 92% in the group after early transplant versus 73% in the group after conventional/salvage transplant (p = 0.01). No active, new, or enlarging lesions in magnetic resonance imaging were registered in patients without disease progression.
 
All patients who did not have disease progression were off therapy throughout the post-transplantation period. HSCT was accompanied by a significant improvement in quality of life with statistically significant changes in the majority of quality of life parameters (p < 0.05).
 
Mancardi and colleagues reported their experience with 74 consecutive patients with MS treated with autologous HSCT with an intermediate intensity conditioning regimen in the period from 1996 to 2008 (Mancardi, 2012). Clinical and magnetic resonance imaging outcomes were reported. The median follow-up period was 48.3 months (range = 0.8-126). Two patients (2.7%) died from transplant-related causes. After 5 years, 66% of patients remained stable or improved. Among patients with a follow-up longer than 1 year, eight out of 25 subjects with a relapsing-remitting course (31%) had a 6-12 months confirmed Expanded Disability Status Scale improvement > 1 point after HSCT as compared with one out of 36 (3%) patients with a secondary progressive disease course (p = 0.009). Among the 18 cases with a follow-up longer than 7 years, eight (44%) remained stable or had a sustained improvement while 10 (56%), after an initial period of stabilization or improvement with a median duration of 3.5 years, showed a slow disability progression.
 
Bowen and colleagues reported the long-term safety and effectiveness of high-dose immunosuppressive therapy followed by autologous HSCT in advanced MS (Bowen, 2012). Neurological examinations, brain magnetic resonance imaging and cerebrospinal fluid (CSF) for oligoclonal bands (OCB) were serially evaluated. There were 26 patients with a mean Expanded Disability Status Scale (EDSS) of 7.0; 17 with secondary progressive MS, 8 with primary progressive, and 1 with relapsing/remitting. Median follow up was 48 months after HSCT. The 72-month probability of worsening 1.0 EDSS point was 0.52 (95% confidence interval, 0.30-0.75). Five patients had an EDSS at baseline of 6.0; four of them had not failed treatment at last study visit. OCB in CSF persisted with minor changes in the banding pattern. Four new or enhancing lesions were seen on MRI, all within 13 months of treatment. In this population with high baseline EDSS, a significant proportion of patients with advanced MS remained stable for as long as 7 years after transplant. Non-inflammatory events may have contributed to neurological worsening after treatment. HSCT may be more effective in patients with less advanced relapsing/remitting MS.
 
Systemic Sclerosis/Scleroderma
Henes and colleagues reported on their experience with autologous HSCT for systemic sclerosis in 26 consecutive patients scheduled for HSCT between 1997 and 2009 (Henes, 2012). The major outcome variable was the response to treatment (reduction of modified Rodnan skin score [mRSS] by 25%) at 6 months. Secondary endpoints were transplant-related mortality and PFS. At 6 months, significant skin and lung function improvement of the mRSS was achieved in 78.3% of patients. The overall response rate was 91%, as some patients improved even after month 6. Three patients died between mobilization and conditioning treatment, 2 due to severe disease progression and 1 whose death was considered treatment-related. Seven patients experienced a relapse during the 4.4 years of follow up. PFS was 74%. Four patients died during follow up and the most frequent causes of death were pulmonary and cardiac complications of systemic sclerosis. The authors concluded that autologous HSCT resulted in significant improvement in most patients with systemic sclerosis.
 
2013 Update
A search of the MEDLINE database through November 2013 did not reveal any new literature that would prompt a change in the coverage statement.
   
 
2014 Update
A literature search conducted through September 2014 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The results of the Autologous Stem Cell Transplantation International Scleroderma (ASTIS) trial (ISRCTN54371254) were published in June 2014 (van Laar, 2014). ASTIS was a phase 3 RCT conducted in 10 countries at 29 centers with access to a EBMT-registered transplant facility. A total of 156 patients were recruited between March 2001 and October 2009. Individual patients were eligible if they were between 18 and 65 years of age; had diffuse cutaneous systemic sclerosis according to American Rheumatism Association criteria, with maximum duration of 4 years; minimum modified Rodnan skin score (mRSS) of 15 (range, 0-51 with higher scores indicating more sever skin thickening); and, involvement of heart, lungs, or kidneys. Patients were randomly allocated to receive high-dose chemotherapy (intravenous cyclophosphamide 200 mk/kg over 4 consecutive days and intravenous rabbit antithymocyte globulin 7.5 mg/kg total dose over 3 consecutive days) followed by CD34+ selected autologous HSCT support (n = 79) or 12 monthly treatments with intravenous pulsed cyclophosphamide (750 mg/m2). Median follow-up was 5.8 years (interquartile range, 4.1-7.8 years). The primary end point was event-free survival, defined as the time in days from randomization until the occurrence of death due to any cause or the development of persistent major organ failure (heart, lung, kidney). Main secondary endpoints included TRM, toxicity and disease-related changes in mRSS, organ function, body weight, and QoL scores. The internal validity (risk of bias) of ASTIS was assessed according to the United States Preventive Services Task Force (USPSTF) criteria for randomized trials. The study was rated as “poor” quality according to this framework because it has 2 fatal flaws: outcome assessment was not masked to patients or assessors, and 18 of 75 (24%) of the control group discontinued intervention because of death, major organ failure, adverse events, or nonadherence. Furthermore, the article states that crossover was allowed after the second year, but whether any patients did so and were analyzed as such is not mentioned. Finally, the authors report that the use of unspecified concomitant medications or other supportive care measures were allowed at the discretion of the investigators, adding further uncertainty to the results.
 
A total of 53 primary endpoint events were recorded: 22 in the HSCT group (19 deaths and 3 irreversible organ failures; 8 patients died of treatment-related causes in the first year, 9 of disease progression, 1 of cerebrovascular disease, 1 of malignancy) and 31 in the control group (23 deaths and 8 irreversible organ failures [7 of whom died later]; 19 patients died of disease progression, 4 of cardiovascular disease, 5 of malignancy, 2 of other causes). The data show patients treated with HSCT experienced more events in the first year but appeared to have better long-term event-free survival than the controls, as the Kaplan- Meier curves for overall survival cross at about 2 years after treatment with overall survival at that time estimated at 85%. According to data from the Kaplan-Meier curves, at 5 years, overall survival was an estimated 66% in the control group and about 80% the HSCT group (p-value unknown). Time-varying hazard ratios (modeled with treatment x time interaction) for event-free survival were 0.35 (95% CI, 0.15- 0.74) at 2 years and 0.34 (95% CI, 0.16-0.74) at 4 years, supporting a benefit of HSCT versus pulsed cyclophosphamide. Severe or life-threatening grade 3 or 4 adverse events were reported in 51 (63%) of the HSCT group compared with 30 (37% by intention-to-treat, p=0.002).
 
Ongoing and Unpublished Clinical Trials
A review of the online site ClinicalTrials.gov in September 2014 (and prior years) identified the following clinical studies in progress.
 
A phase 3 randomized trial (Stem Cell Therapy for Patients With Multiple Sclerosis Failing Interferon A Randomized Study) is recruiting participants to study the effect of autologous peripheral blood HSCT in patients with relapsing MS versus U.S. Food and Drug Administration (FDA) - approved standard of care. Primary endpoint is disease progression. Patients will be followed for 5 years after randomization.  Estimated enrollment is 110, and estimated study completion date is December 2017 (NCT00273364).
 
The phase 2 randomized ASTIMS trial evaluating autologous HSCT in severe cases of MS was terminated due to difficulty in accruing patients and lack of funds.
 
The High-Dose Immunosuppression and Autologous Transplantation for Multiple Sclerosis (HALT-MS) Study is a phase 2 nonrandomized, uncontrolled trial to determine the effectiveness of autologous HSCT for the treatment of poor prognosis (relapsing-remitting or secondary progressive) MS. The primary outcome measure is time to treatment failure. Estimated enrollment is 25, and estimated study completion date is September 2015 (NCT00288626).
 
The Canadian MS-BMT phase 2 study is to determine the effect of autologous HSCT on early-stage MS. Estimated enrollment is 24. Enrollment completed July 2009.
 
The SCOT trial is a randomized phase 2 study comparing HSCT and pulsed cyclophosphamide. Primary outcome measure is the global rank composite score at 54 months postrandomization (which includes measures of event-free survival, death, lung function, and skin score). Crossover to the HSCT arm is not allowed. The trial is still recruiting, with an estimated enrollment of 114 patients with an estimated study completion date of June 2016 (NCT00114530). As of September 2014, SCOT has not been published in full-length form.
 
Three nonrandomized, open-label phase 2 trials are recruiting patients, ongoing or completed studying the effectiveness of autologous HSCT in patients with SLE: one with an estimated enrollment of 9 and study completion date of October 2013 (NCT00076752), another with an estimated enrollment of 30 and study completion date of April 2014 (NCT00750971), and a third with an estimated enrollment of 52 and study completion date of April 2012 (NCT00271934).
 
No phase 2 or 3 clinical trials were identified using HSCT in juvenile idiopathic or RA.
 
One nonrandomized, phase 2 clinical trial is recruiting patients to study nonmyeloablative autologous HSCT in patients with CIDP (NCT00278629). It is estimated the study will be complete in December 2014.
 
Three phase 1/2 and 2 phase 2 trials are recruiting patients with type 1 diabetes mellitus for autologous HSCT (NCT00315133, NCT01121029, NCT00807651, NCT01341899, NCT1285934). The status of 1 phase 2 and 1 phase 2/3 trial for patients with type 2 diabetes mellitus for autologous HSCT is unknown.
(NCT00644241, NCT01065298)
 
ASTIS trial results suggest high-dose chemotherapy with autologous HSCT may improve survival among patients with diffuse cutaneous systemic sclerosis compared to pulsed intravenous cyclophosphamide. However, analysis of the internal validity of the trial using USPSTF criteria showed fatal flaws and a poor study rating due to attrition in the control group that could have skewed the survival curve to show better survival for HSCT recipients compared to controls. The investigators acknowledge this limitation in addition to stating that the unblinded outcome assessments may have influenced results, and wide confidence intervals for some secondary outcomes indicated less certainty about those results. An accompanying editorial concurs that autologous HSCT to treat systemic sclerosis requires further study before it should be offered to patients in routine clinical practice (Khanna, 2014).
 
2016 Update
A literature search conducted through September 2016 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Multiple Sclerosis
Only 1 RCT evaluating HSCT for treatment of MS has been published (Mancardi, 2015), but this trial did not report on clinical outcomes. No controlled trials with contemporaneous control groups were identified that reported clinical endpoints such as overall survival, progression-free survival and disability status as their primary outcome. The RCT by Mancardi and colleagues was originally designed as a phase III study reporting on disability progression. However, due to low patient enrollment, the protocol was amended as a phase II study with the primary outcome of cumulative number of new T2 MRI lesions in the 4 years after treatment. Eligibility for the trial was secondary progressive or relapsing-remitting MS, a documented worsening during the last year and lack of response to conventional therapy. A total of 21 patients were randomized to autologous HSCT (n=9) or medical therapy (mitoxantrone) (n=12). Follow-up data were not available on 4 patients; missing data was imputed in the intention to treat analysis of the primary outcome. The median number of new T2 MRI lesions was 2.5 in the HSCT group and 8 in the conventional therapy group (rate ratio [RR]: 0.21, 95% confidence interval [CI]: 0.10 to 0.48, p=0.000016). Among secondary outcomes, the annualized relapse rate was significantly lower in the HSCT group (0.19) than the conventional therapy group (0.6), but there was no statistically significant difference between groups in the rate of disease progression or change in disability status.
 
A 2015 publication reported on 64 patients participating in this study who had at least 36 months follow-up (median 62 months) (Shevchenko, 2015). (Another 35 patients had shorter follow-up and the remainder were lost to follow-up). Thirty of the 64 patients (47%) improved at least 0.5 point on the EDSS scale compared to baseline. Among the other patients, 29 (45%) were stable and 5 (7%) experienced worsening disease.
 
A 2015 single-center case series by Burt and colleagues reported on 151 patients, 123 with relapsing-remitting MS and 28 with secondary progressive MS (Burt, 2015). Patients were treated with non-myeloablative HSCT between 2003 and 2014. Six patients were not included in the outcome analysis. The remaining 145 patients were followed for a median of 2 years (range: 6 months to 5 years). There were no treatment-related deaths. The primary outcome was change in the EDSS. A decrease of at least 1.0 points was considered significant improvement and an increase of at least 1.0 points was considered significant progression. There was statistically significant improvement in the EDSS score for the group as a whole compared with the pretransplant mean score of 4.0 decreasing to a mean EDSS score of 2.5 at 3, 4 and 5 years. In a post hoc analysis, patients most likely to have statistically significant improvements in EDSS were those with relapsing-remitting MS, with duration of disease of 10 years or less and those without sustained fever during HSCT.
 
A 2014 multicenter case series by Burman and colleagues reported on 48 patients with aggressive relapsing-remitting MS, defined as disease with high relapse frequency, and who failed conventional therapy (Burman, 2013). Patients underwent autologous HSCT. At the 5-year follow-up, relapse-free survival was 87% and the EDSS score progression-free survival (EDSS deterioration of less than 0.5 points) was 77%. The rate of disease-free survival (no relapses, no new MRI lesions and no EDSS progression) was 68%.
 
Type 1 Diabetes Mellitus
Several case series were identified evaluating autologous HSCT in patients with new-onset type 1 diabetes; there were no published comparative studies. In the series, although a substantial proportion of patients tended to become insulin free after HSCT, remission rates were high. In 2015, Xiang and colleagues published data on 128 patients age 12 to 35 years who had been diagnosed with type 1 diabetes no longer than 6 weeks before study enrollment (Xiang, 2015). After a mean follow-up of 28.5 months (range, 15-38 months), 71 patients (55%) were considered to be insulin free. These patients had a mean remission period of 14.2 months (SD=6.1 months). The other 57 patients (45%) were insulin dependent. The latter group includes 27 patients with no response to treatment and another 30 patients who relapsed after a transient remission period. Adverse events included ketoacidosis and renal dysfunction (1 patient each); there was no transplant-related mortality. In multiple logistic regression analysis, factors independently associated with becoming insulin free after autologous HSCT were younger age at onset of diabetes, lower tumor necrosis factor (TNF-􀄮) and higher fasting C-peptide.
 
A 2015 case series by Snarski and colleagues reported on 24 patients with a diagnosis of type 1 diabetes within 6 weeks of enrollment who underwent autologous HSCT (Snarski, 2015). Patients had a mean age of 26.5 years (range, 18-34 years). After treatment, 20 of 23 patients (87%) went into remission of diabetes, defined as being insulin free with normoglycemia for at least 9.5 months. The median time of remission was 31 months (range, 9.5-80 months). Mean insulin doses remained significantly lower than baseline doses at 2 and 3 years, but the insulin doses returned to pre-HSCT levels at years 4 and 5. Among patients remaining in follow-up at the time of data analysis for publication (n=20), 4 (20%) remained insulin free. Adverse events include neutropenic fever in 12 patients (50%). There were 4 cases of sepsis, including a fatal case of Pseudomonas aeruginosa sepsis. There was also 1 case of pulmonary emphysema after insertion of a central venous catheter.
 
2017 Update
A literature search conducted through September 2017 did not reveal any new information that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
Results from the High-Dose Immunosuppression and Autologous Transplantation for Multiple Sclerosis (HALT-MS) trial was published in 2017 by Nash and colleagues (Nash, 2017). The trial evaluated 24 patients with MS who were treated with high-dose immunosuppression and autologous HCT. The median follow-up was 62 months (range, 12-72 months). Outcomes were PFS (91%; 90% CI, 75% to 97%), clinical relapse-free survival (87%; 90% CI, 69% to 95%), and MRI activity-free survival (86%; 90% CI, 68% to 95%). Patients experienced high rates of adverse events: 92% had grade 3 and 100% had grade 4 adverse events. The majority of adverse events occurred between the start of conditioning to day 29 in the trial.
 
Systematic Reviews
van Laar and colleagues conducted a systematic review of evidence on the use of HCT for treating poor-prognosis systemic sclerosis (van Laar, 2015). They identified 3 RCTs comparing HCT with the standard of care (cyclophosphamide): a phase 2 trial and a completed phase 3 trial, both of which are described in the Randomized Controlled Trial section, plus a phase 3 trial (Scleroderma: Cyclophosphamide or Transplantation [SCOT]). SCOT has been completed and results presented at a 2016 American College of Rheumatology conference. Results have not been published at the time of this update. Reviewers concluded that there is evidence HCT can result in significant improvements in skin thickness and functional outcomes. However, HCT is associated with serious toxicities that can be fatal. Additional trials are needed to assess how to reduce toxicity and to determine which patients with scleroderma would benefit most from HCT.
 
A review by Milanetti and colleagues summarized 8 phase 1 and 2 clinical studies using autologous HCT to treat systemic sclerosis (Milanetti, 2011). The number of patients in each study ranged from 6 to 57, with the proportion of patients across the studies achieving a 25% decrease in the Rodnan Skin Score (RSS) ranged from 60% to 100%. Pooled analyses were not conducted.
 
JUVENILE IDIOPATHIC OR RHEUMATOID ARTHRITIS
A 2008 review article by Saccardi and colleagues on HCT for autoimmune diseases has summarized the experience with juvenile idiopathic arthritis and rheumatoid arthritis as follows (Saccardi, 2008). More than 50 patients with juvenile idiopathic arthritis have been reported to the EBMT Registry. The largest cohort study initially used a single conditioning regimen and, thereafter, a modified protocol. Overall drug-free remission rate was approximately 50%. Some late relapses have been reported, and only partial correction of growth impairment has been seen. The frequency of HCT for rheumatoid arthritis has decreased significantly since 2000, due to the introduction of new biologic therapies. Most patients who have undergone HCT have had persistence or relapse of disease activity within 6 months of transplant.
 
Cantu-Rodriguez and colleagues published a study of 16 patients with type 1 diabetes who received a less toxic conditioning regimen and transplantation (Cantu-Rodriguez, 2016). The outpatient procedures were completed without severe complications. At the 6-month follow-up, 3 (19%) were nonresponders, 6 (37%) partially independent from insulin, and 7 (44%) were completely independent of insulin. Hemoglobin A1c levels decreased by a mean of -2.3% in the insulin-independent group.
 
Xiang and colleagues published data on 128 patients ages 12 to 35 years who had been diagnosed with
type 1 diabetes no more than 6 weeks before study enrollment (Xiang, 2015). After a mean follow-up of 28.5 months (range, 15-38 months), 71 (55%) patients were considered to be insulin-free. These patients had a mean remission period of 14.2 months. The other 57 (45%) patients were insulin-dependent. The latter group included 27 patients with no response to treatment and another 30 patients who relapsed after a transient remission period. Adverse events included ketoacidosis and renal dysfunction (1 patient each); there was no transplant-related mortality. In multiple logistic regression analysis, factors independently associated with becoming insulin-free after autologous HCT were younger age at onset of diabetes, lower tumor necrosis factor α levels, and higher fasting C-peptide levels.
 
ONGOING AND UNPUBLISHED CLINICAL TRIALS
Some currently unpublished trials that might influence this review are listed below:
 
Ongoing:
(NCT02516124) Autologous Stem Cell Transplantation for Progressive Systemic Sclerosis: a Prospective Non-interventional Approach Across Europe (NISSC) for the Autoimmune Diseases Working Party of the EBMT; planned enrollment 50; projected completion date December 2017
 
(NCT01445821) Randomized Study of Different Non-myeloablative Conditioning Regimens with Hematopoietic Stem Cell Support in Patients with Scleroderma (ASSIST-IIb); planned enrollment 160; projected completion date September 2018
 
(NCT02225795) A Pilot Study of Autologous Stem Cell Transplantation with Posttransplant Cyclophosphamide for Children and Young Adults with Refractory Crohn’s Disease; planned enrollment 15; projected completion date December 2019
 
(NCT02674217) Outpatient Hematopoietic Grafting in Patients with Multiple Sclerosis Employing Autologous Non-cryopreserved Peripheral Blood Stem Cells: a Feasibility Study; planned enrollment 200; projected completion date December 2019
 
(NCT03113162) Evaluation of the Safety and Efficacy of Reduced-Intensity Immunoablation and Autologous  hematopoietic Stem Cell Transplantation (AHSCT) in Multiple Sclerosis; planned enrollment 15; projected date May 2020
 
2018 Update
A literature search was conducted through September 2018.  There was no new information identified that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
American Society for Blood and Marrow Transplantation
The American Society for Blood and Marrow Transplantation published consensus guidelines on the use of HCT to treat specific conditions in and out of the clinical trial setting (ASBMT, 2015).
 
2020 Update
Annual policy review completed with a literature search using the MEDLINE database through September 2020. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Burt et al reported a randomized controlled trial of nonmyeloablative HCT compared to continued disease-modifying therapy (DMT) on disease progression for patients with relapsing-remitting MS (Burt, 2019). Between 2005 and 2016, with final follow-up in 2018, 110 patients with relapsing remitting multiple sclerosis (RRMS) were randomized to receive HCT plus cyclophosphamide and antithymocyte globulin (n = 55) or DMT of higher efficacy or a different class than DMT taken in the previous year (n = 55). To be eligible, the participants had to have at least 2 relapses with DMT in the prior year and an Expanded Disability Status Score (EDSS) of 2.0 to 6.0 (EDSS score range 0–10, with 10 = worst neurological disability). The primary end point of the study was disease progression, defined as an EDSS score increase of 1.0 point (minimally clinically important difference, 0.5) after 1 year on 2 evaluations 6 months apart. Three patients in the HCT group and 34 patients in the DMT group experienced disease progression, with a median follow-up of 2 years (mean = 2.8 years). Too few events in the HCT group prevented calculation of time to progression, but it was 24 months (interquartile range = 18–48 months) in the DMT group (hazard ratio [HR] = 0.07; 95% CI: 0.02–0.24). For the HCT group, the proportion of patients with disease progression was 1.92% (95% CI: 0.27%–12.9%) at 1 year and 2 years, and by 4 and 5 years it was 9.71% (95% CI: 3.0%–28.8%). Disease progression for the DMT group was 24.5% (95% CI: 14.7%–39.1%) at 1 year, and 75.3% (95% CI: 60.4%–87.8%) by year 5. In the HCT group, the mean EDSS score decreased from a baseline of 3.38 to 2.36 at 1 year. In the DMT group, mean EDSS score increased from 3.31 to 3.98 at one year. Between-group difference in change in scores was -1.7 (95% CI: -2.03 to -1.29; p <.001). The results of the study suggest nonmyeloablative HCT is superior to DMT in prolonging time to disease progression in patients with RRMS. Study limitations included sample size, option to cross over from DMT to HCT mid-study and the exclusion of other chemotherapy drugs used in the DMT group.
 
Ge et al reported a systematic review and meta-analysis to assess progression-free survival (PFS) and disease activity-free survival, as well as transplant-related mortality (TRM) and overall deaths, after autologous HCT for MS (Ge, 2019). The authors identified 18 eligible studies with a total of 732 participants. Pooled estimated PFS was 75%. Low- and intermediate-intensity treatments had higher PFS than high-intensity treatments. In addition, RRMS benefited from autologous HCT more than other types of MS subtypes. Patients with gadolinium-enhancing lesions at baseline responded better to autologous HCT. Overall, 9 transplant-related deaths occurred, and estimated TRM was greater with the use of high-intensity treatment regimens and in studies conducted before 2006. Twenty-seven patients died during follow-up; primarily of infection or pneumonia. Several limitations of the meta-analysis include possible publication bias, a lack of RCTs, and differences in autologous HCT procedures, patient characteristics, and duration of follow-up across studies.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through February 2022. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Boffa et al performed a retrospective cohort study of 210 patients in Italy with relapsing/remitting, secondary progressive, or primary progressive MS treated with HCT between 1997 and 2019 (Boffa, 2021). RFS at 5 and 10 years after transplant was 82.9% (95% CI, 76.6% to 89.2%) and 71.2% (95% CI, 61.8% to 80.6%), respectively.
 
Burt et al performed a retrospective cohort study of 414 patients with RRMS and 93 patients with newly diagnosed secondary-progressive MS treated with HCT at a single center in the US between 2003 and 2019. (Burt, 2021). RFS at 5 years for patients with RRMS and secondary-progressive MS was 80.1% and 98.1%, respectively.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through February 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Nabizadeh et al conducted a systematic review and meta-analysis on the use of autologous HCT in patients with MS (Nabizadeh, 2022). Fifty studies, including 7 RCTs, with a total of 4831 patients were included. The pooled estimated PFS was 73% (95% confidence interval [CI], 69% to 77%; I2= 89.89%). There was a significant decrease in Expanded Disability Status Scale (EDSS) score after treatment (standardized mean difference [SMD], -0.48; 95% CI, -0.75 to -0.22), and the annualized relapse rate (ARR) was decreased relative to the pretreatment period (SMD, -1.58; 95% CI, -2.34 to -0.78). However, the analysis found a higher incidence of TRM after autologous HCT versus other disease-modifying therapies when evaluating long-term outcome measures; the analysis considered an endpoint of all TRM at the end of a 5-year follow-up duration. Limitations of the meta-analysis include possible publication bias, minimal number of RCTs, lack of studies focusing on specific subtypes of MS, high heterogeneity between included studies, and unspecified duration of follow-up across studies.
 
Higashitani et al conducted a systematic review and meta-analysis of survival outcomes of HCT in patients with systemic sclerosis (Higashitani, 2022). There were 22 studies included (3 RCTs; 19 observational cohorts). The pooled frequency of transplant-related death (N=700) was 6.30% (95% CI, 4.21 to 8.38). However, the authors note that the estimated frequency of treatment-related deaths has been declining over the last decade.
 
Bruera et al conducted a systematic review of autologous HCT for the treatment of systemic sclerosis (Bruera, 2022). There were 3 RCTs (N=125) included with 3 different transplant modalities (non-myeloablative non-selective; non-myeloablative selective; myeloablative selective) and the comparator in all studies was cyclophosphamide. No study demonstrated an overall mortality benefit of autologous HCT when compared with cyclophosphamide; however, non-myeloablative selective HCT demonstrated OS benefits (using Kaplan-Meier curves) at 10 years and myeloablative selective HCT demonstrated OS benefits at 6 years. Event-free survival was improved with non-myeloablative selective HCT at 48 months (HR, 0.34; 95% CI, 0.16 to 0.74; moderate-certainty evidence) compared with cyclophosphamide; there was no improvement in EFS with myeloablative selective HCT at 54 months (HR, 0.54; 95% CI, 0.23 to 1.27; moderate-certainty evidence). All HCT transplant modalities reported improvement of mRSS compared with cyclophosphamide; however, there was low-certainty evidence that these modalities of HCT improved patient physical function.
 
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.

CPT/HCPCS:
38240Hematopoietic progenitor cell (HPC); allogeneic transplantation per donor
38241Hematopoietic progenitor cell (HPC); autologous transplantation
38242Allogeneic lymphocyte infusions

References: . Mancardi GL, Sormani MP, Gualandi F, et al.(2015) Autologous hematopoietic stem cell transplantation in multiple sclerosis: a phase II trial. Neurology. Mar 10 2015;84(10):981-988. PMID 25672923

Alexander T, Bondanza A, Muraro PA, et al.(2015) SCT for severe autoimmune diseases: consensus guidelines of the European Society for Blood and Marrow Transplantation for immune monitoring and biobanking. Bone Marrow Transplant. Feb 2015;50(2):173-180. PMID 25387090

Atkins H.(2010) Hematopoietic SCT for the treatment of multiple sclerosis. Bone Marrow Transplant 2010; Aug 9 [Epub ahead of print]

Autologous Stem cell Transplantation International Multiple Sclerosis website (undated). Available online at: http://www.astims.org/termination-trial-ASTIMS.html . Last accessed September 2010.

Boffa G, Massacesi L, Inglese M, et al.(2021) Long-Term Clinical Outcomes of Hematopoietic Stem Cell Transplantation in Multiple Sclerosis. Neurology. Jan 20 2021. PMID 33472915

Bowen JD, Kraft GH, Wundes A et al.(2012) Autologous hematopoietic cell transplantation following high-dose immunosuppressive therapy for advanced multiple sclerosis: long-term results. Bone Marrow Transplant 2012; 47(7):946-51.

Bruera S, Sidanmat H, Molony DA, et al.(2022) Stem cell transplantation for systemic sclerosis. Cochrane Database Syst Rev. Jul 29 2022; 7(7): CD011819. PMID 35904231

Burman J, Iacobaeus E, Svenningsson A, et al.(2014) Autologous hematopoietic stem cell transplantation for aggressive multiple sclerosis: the Swedish experience J Neurol Neurosurg Psychiatry. Oct 2014;85(10):1116-1121. PMID 24554104

Burt RK, Balabanov R, Burman J, et al.(2019) Effect of nonmyeloablative hematopoietic stem cell transplantation vs continued disease-modifying therapy on disease progression in patients with relapsing-remitting multiple sclerosis: a randomized clinical trial. JAMA. 2019 Jan 15;321(2):165-174. PMID: 30644983

Burt RK, Balabanov R, Han X, et al.(2015) Association of nonmyeloablative hematopoietic stem cell transplantation with neurological disability in patients with relapsing-remitting multiple sclerosis. JAMA. Jan 20 2015;313(3):275-284. PMID 25602998

Burt RK, Han X, Quigley K, et al.(2021) Real-world application of autologous hematopoietic stem cell transplantation in 507 patients with multiple sclerosis. J Neurol. Oct 11 2021. PMID 34633525

Burt RK, Loh Y, Cohen B et al.(2009) Autologous non-myeloablative haematopoietic stem cell transplantation in relapsing-remitting multiple sclerosis: a phase I/II study. Lancet Neurol 2009; 8(3): 244–53.

Cantu-Rodriguez OG, Lavalle-Gonzalez F, Herrera-Rojas MA, et al.(2016) Long-term insulin independence in type 1 diabetes mellitus using a simplified autologous stem cell transplant. J Clin Endocrinol Metab. May 2016;101(5):2141-2148. PMID 26859103

Couri CE, Oliveira MC, Stracieri AB et al.(2009) C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA 2009; 301(15):1573-9.

European Group for Blood and Marrow Transplantation Clinical Trials website (undated). Available online at: http://www.ebmt.org/ClinicalTrials/TrialDetail.aspx?code=ASTIS . Last accessed September 2010.

Fagius J, Lundgren J, Oberg G.(2009) Early highly aggressive MS successfully treated by hematopoietic stem cell transplantation. Mult Scler 2009; 15(2):229–37.

Fassas A, Kimiskidis VK, Sakellari I et al.(2011) Long-term results of stem cell transplantation for MS: a single-center experience. Neurology 2011; 76(12):1066-70.

Ge F, Lin H, Li Z et al.(2019) Efficacy and safety of autologous hematopoietic stem-cell transplantation in multiple sclerosis: a systematic review and meta-analysis. Neurol. Sci. 2019 Mar;40(3). PMID 30535563

Henes JC, Schmalzing M, Vogel W et al.(2012) Optimization of autologous stem cell transplantation for systemic sclerosis -- a single-center longterm experience in 26 patients with severe organ manifestations. J Rheumatol 2012; 39(2):269-75.

Higashitani K, Takase-Minegishi K, Yoshimi R, et al.(2022) Benefits and risks of Hematopoietic Stem Cell Transplantation for Systemic Sclerosis: A Systematic Review and Meta-Analysis. Mod Rheumatol. Mar 12 2022. PMID 35285885

Ioannidis JP, Vlachoyiannopoulos PG, Haidich AB et al.(2005) Mortality in systemic sclerosis: an international meta-analysis of individual patient data. Am J Med 2005; 118(1):2–10.

Khanna D, Georges GE, Couriel DR.(2014) Autologous hematopoietic stem cell therapy in severe systemic sclerosis: ready for clinical practice? JAMA. Jun 25 2014;311(24):2485-2487. PMID 25058081

Kimiskidis V, Sakellari I, Tsimourtou V et al.(2008) Autologous stem-cell transplantation in malignant multiple sclerosis: a case with a favorable long-term outcome. Mult Scler 2008; 14(2):278–83.

Majhail NS, Farnia SH, Carpenter PA, et al.(2015) Indications for autologous and allogeneic hematopoietic cell transplantation: guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. Nov 2015;21(11):1863-1869. PMID 26256941

Mancardi GL, Murialdo A, Rossi P et al.(2005) Autologous stem cell transplantation as rescue therapy in malignant forms of multiple sclerosis. Mult Scler 2005; 11(3):367–71.

Mancardi GL, Sormani MP, Di Gioia M et al.(2012) Autologous haematopoietic stem cell transplantation with an intermediate intensity conditioning regimen in multiple sclerosis: the Italian multi-centre experience. Mult Scler 2012; 18(6):835-42.

Milanetti F, Abinun M, Voltarelli JC et al.(2010) Autologous hematopoietic stem cell transplantation for childhood autoimmune disease. Pediatr Clin North Am 2010; 57(1):239-71.

Milanetti F, Bucha J, Testori A, et al.(2011) Autologous hematopoietic stem cell transplantation for systemic sclerosis. Curr Stem Cell Res Ther. Mar 2011;6(1):16-28. PMID 20955159

Nabizadeh F, Pirahesh K, Rafiei N, et al.(2022) Autologous Hematopoietic Stem-Cell Transplantation in Multiple Sclerosis: A Systematic Review and Meta-Analysis. Neurol Ther. Dec 2022; 11(4): 1553-1569. PMID 35902484

Nash RA, Hutton GJ, Racke MK, et al.(2015) High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for relapsing-remitting multiple sclerosis (HALT-MS): a 3-year interim report. JAMA Neurol. Feb 2015;72(2):159-169. PMID 25546364

Nash RA, Hutton GJ, Racke MK, et al.(2017) High-dose immunosuppressive therapy and autologous HCT for relapsing-remitting MS. Neurology. Feb 28 2017;88(9):842-852. PMID 28148635

Nash RA, McSweeney PA, Crofford LJ et al.(2007) High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: long-term follow-up of the US multicenter pilot study. Blood 2007; 110(4):1388-96.

Nikolov NP, Pavletic SZ.(2008) Technology Insight: hematopoietic stem cell transplantation for systemic rheumatic disease. Nat Clin Pract Rheumatol. Apr 2008; 4(4): 184-91. PMID 18285764

Pasquini MC, Griffith LM, Arnold DL et al.(2010) Hematopoietic stem cell transplantation for multiple sclerosis: collaboration of the CIBMTR and EBMT to facilitate international clinical studies. Biol Blood Marrow Transplant 2010; 16(8):1076-83.

Saccardi R, Di Gioia M, Bosi A.(2008) Haematopoietic stem cell transplantation for autoimmune disorders. Curr Opin Hematol 2008; 15(6):594-600.

Saccardi R, DiGioia M, Bosi A.(2008) Haematopoietic stem cell transplantation for autoimmune disorders. Curr Opin Hematol. Nov 2008;15(6):594-600. PMID 18832930

Saccardi R, Kozak T, Bocelli-Tyndall C et al.(2006) Autologous stem cell transplantation for progressive multiple sclerosis: update of the European Group for Blood and Marrow Transplantation autoimmune diseases working party database. Mult Scler 2006; 12(6):814-23.

Sanders DB, Wolfe GI, Benatar M, et al.(2016) International consensus guidance for management of myasthenia gravis: Executive summary. Neurology. Jul 26 2016;87(4):419-425. PMID 27358333

Shevchenko JL, Kuznetsov AN, Ionova TI et al.(2012) Autologous hematopoietic stem cell transplantation with reduced-intensity conditioning in multiple sclerosis. Expl Hematol 2012 [Epub ahead of print].

Shevchenko JL, Kuznetsov AN, Ionova TI, et al.(2015) Long-term outcomes of autologous hematopoietic stem cell transplantation with reduced-intensity conditioning in multiple sclerosis: physician's and patient's perspectives. Ann Hematol. Jul 2015;94(7):1149-1157. PMID 25711670

Snarski E, Milczarczyk A, Halaburda K, et al.(2015) Immunoablation and autologous hematopoietic stem cell transplantation in the treatment of new-onset type 1 diabetes mellitus: long-term observations. Bone Marrow Transplant. Dec 7 2015. PMID 26642342

Sullivan KM, Muraro P, Tyndall A.(2010) Hematopoietic cell transplantation for autoimmune disease: updates from Europe and the United States. Biol Blood Marrow Transplant 2010; 16(1 suppl):S48-56.

Tyndall A, Gratwohl A.(2009) Adult stem cell transplantation in autoimmune disease. Curr Opin Hematol 2009; 16(4):285-91.

van Laar JM, Farge D, Sont JK, et al.(2014) Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA. Jun 25 2014;311(24):2490-2498. PMID 25058083

van Laar JM, Naraghi K, Tyndall A.(2015) Haematopoietic stem cell transplantation for poor-prognosis systemic sclerosis. Rheumatology (Oxford). Dec 2015;54(12):2126-2133. PMID 25953700

Vonk MC, Marjanovic Z, van den Hoogen FH et al.(2008) Long-term follow-up results after autologous haematopoietic stem cell transplantation for severe systemic sclerosis. Ann Rheum Dis 2008; 67(1):98-104.

Xiang H, Chen H, Li F, et al.(2015) Predictive factors for prolonged remission after autologous hematopoietic stem cell transplantation in young patients with type 1 diabetes mellitus. Cytotherapy. Nov 2015;17(11):1638- 1645. PMID 26318272

Xiang H, Chen H, Li F, et al.(2015) Predictive factors for prolonged remission after autologous hematopoietic stem cell transplantation in young patients with type 1 diabetes mellitus. Cytotherapy. Nov 2015;17(11):1638-1645. PMID 26318272


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.