CADTH Health Technology Review

Autologous Hematopoietic Cell Transplant for Patients With Multiple Sclerosis

Rapid Review

Authors: Khai Tran, Hannah Loshak

Abbreviations

AE

adverse event

AHSCT

autologous hematopoietic stem cell transplantation

ALZ

alemtuzumab

ARR

annualized relapse rate

ASBMT

American Society for Blood and Marrow Transplantation

CI

confidence interval

CNS

central nervous system

DMT

disease-modifying therapy

EBMT

European Society for Blood and Marrow Transplantation

EDSS

Expanded Disability Status Scale

Gd

gadolinium

IR

incidence rate

MS

multiple sclerosis

MSFC

Multiple Sclerosis Functional Composite

MTX

mitoxantrone

NEDA

no evidence of disease activity

NRS

Neurologic Rating Scale

PML

progressive multifocal leukoencephalopathy

PPMS

primary-progressive multiple sclerosis

RCT

randomized controlled trial

RRMS

relapsing-remitting multiple sclerosis

SF-36 QoL

Short Form 36 (health survey) quality of life

SPMS

secondary-progressive multiple sclerosis

Key Messages

Context and Policy Issues

Multiple sclerosis (MS) is a chronic inflammatory autoimmune disorder of the central nervous system (CNS) that is more common in women than in men by a factor of approximately 3 to 1.1 One in every 400 Canadians live with MS and more than 4,000 people are diagnosed with MS every year.1

MS is heterogeneous in clinical course and can be classified into 4 subtypes: clinically isolated syndrome, relapsing-remitting MS (RRMS), primary-progressive MS (PPMS), and secondary-progressive MS (SPMS).2 Clinically isolated syndrome is a first episode of inflammation of the CNS that is a characteristic of MS but does not meet the criteria for the diagnosis of MS.2 The diagnosis of MS has been revised by the McDonald Criteria published in 2017 by the International Panel on the Diagnosis of Multiple Sclerosis.3 Biomarkers for MS on MRI are gadolinium (Gd)-enhancing lesions and new T2 or enlarging T2 lesions, which are indicators of ongoing CNS inflammation.4 Approximately 85% to 90% of MS patients have RRMS, which is characterized by clearly defined periods of neurologic symptoms (relapses) alternating with periods of partial or complete recovery (remissions).5 Relapses occur when the immune system attacks the myelin sheet of the nerve fibres, causing an acute inflammation of the CNS.5 Patients with RRMS will eventually transition to SPMS, which is characterized by a progressive worsening of neurologic function over time.2,6,7 Some patients develop PPMS, characterized by progressive disability from the onset of symptoms independent of relapses.2

There are currently more than a dozen of disease-modifying therapies (DMTs) for MS approved in Canada that are grouped into 3 categories: injectable medications (e.g., interferon beta-1a, interferon beta-1b, glatiramer acetate, ofatumumab, peginterferon beta-1a), oral medications (e.g., teriflunomide, fingolimod, cladribine, siponimod, dimethyl fumarate, ozanimod), and infused medications (e.g., alemtuzumab, ocrelizumab, natalizumab).8 These drugs have different mechanisms of action in modulating or suppressing the immune system, thereby reducing the disease activity and slowing down the rate of disability development.9 However, some patients with aggressive MS still show no improvement or have significant side effects after treatment with 1 or more approved DMTs.9

Autologous hematopoietic stem cell transplantation (AHSCT) may be considered as a suitable immune reconstitution therapy for some MS patients at the early stage of disease when the inflammation is predominant and the disability level is low.10 AHSCT has been used for the treatment of MS for more than 2 decades.10 With improving techniques and expertise of transplantation, together with properly selected patients, the efficacy and safety of AHSCT have improved over time.10 AHSCT procedures consists of 5 main stages such as mobilization of hematopoietic stem cells using growth factor and chemotherapy, collection of stem cells from peripheral blood, conditioning chemotherapy to suppress immune system, reinfusion of stem cells, and support with blood products and antibiotics as well as follow-up with regular blood tests and medication.11 The conditioning regimens are classified based on the grade of intensity: high intensity, intermediate intensity, and low intensity.10,12 The intermediate intensity is further divided into myeloablative and non-myeloablative, depending on the regimens, and have been used most commonly in MS.12 High-intensity regimens have been associated with serious adverse events (AEs), while low-intensity regimens have been less efficacious.12

The aim of this report is to review the clinical effectiveness and cost-effectiveness of AHSCT for the treatment of MS. The report also reviews the evidence-based guidelines regarding the use of AHSCT for MS. This report will support a CADTH Early Assessment Bulletin on MS.

Research Questions

  1. What is the clinical effectiveness of AHSCT for treatment of MS?

  2. What is the cost-effectiveness of AHSCT for treatment of MS?

  3. What are the evidence-based guidelines regarding AHSCT for individuals with MS?

Methods

Literature Search Methods

A limited literature search was conducted by an information specialist on key resources including MEDLINE and Embase via Ovid, the Cochrane Database of Systematic Reviews, the international HTA database, the websites of Canadian and major international health technology agencies, as well as a focused internet search. The search strategy comprised both controlled vocabulary, such as the National Library of Medicine’s MeSH (Medical Subject Headings), and keywords. The main search concepts were autologous hematopoietic cell transplant and MS. Search filters were applied to limit retrieval to health technology assessments, systematic reviews, meta-analyses, or network meta-analyses, any types of clinical trials or observational studies, economic studies, or guidelines. Comments, newspaper articles, editorials, conference abstracts, and letters were excluded. Where possible, retrieval was limited to the human population. The search was also limited to English-language documents published between January 1, 2015 and March 24, 2021.

Selection Criteria and Methods

One reviewer screened citations and selected studies. In the first level of screening, titles and abstracts were reviewed and potentially relevant articles were retrieved and assessed for inclusion. The final selection of full-text articles was based on the inclusion criteria presented in Table 1.

Table 1: Selection Criteria

Criteria

Description

Population

Individuals with RRMS or other subtypes of MS

Intervention

AHSCT also known as AHCT and formerly known as autologous bone marrow transplant

Comparator

Q1 and Q2: Standard of care (e.g., DMT); different AHSCT regimen(s) (e.g., myeloablative vs. non-myeloablative).

Q3: Not applicable

Outcomes

Q1: Clinical effectiveness: (e.g., disability, progression free survival, quality of life, pain; safety

Q2: Cost-effectiveness

Q3: Recommendations regarding treatment eligibility and suitability of candidates to receive AHSCT

Study designs

HTA, SR, RCT, non-randomized studies, economic evaluations, evidence-based guidelines

AHCT = autologous hematopoietic cell transplant; AHSCT = autologous hematopoietic stem cell transplant; DMT = disease-modifying therapy; HTA = health technology assessment; MS = multiple sclerosis; RCT = randomized controlled trial; RRMS = relapsing-remitting MS: SR = systematic review.

Exclusion Criteria

Articles were excluded if they did not meet the selection criteria outlined in Table 1, or were published before 2015. Studies that involved allogenic donor stem cells or mesenchymal stem cells were excluded. Economic evaluations without conducting cost-effectiveness analysis were excluded. Guidelines with unclear methodology were also excluded.

Critical Appraisal of Individual Studies

The included publications were critically appraised by 1 reviewer using the following tools as a guide: the Downs and Black checklist13 for randomized and non-randomized studies, and the Appraisal of Guidelines for Research & Evaluation (AGREE) II instrument14 for guidelines. Summary scores were not calculated for the included studies; rather, the strengths and limitations of each included publication were described narratively.

Summary of Evidence

Quantity of Research Available

A total of 464 citations were identified in the literature search. Following the screening of titles and abstracts, 423 citations were excluded and 41 potentially relevant reports from the electronic search were retrieved for full-text review. No potentially relevant publications were retrieved from the grey literature search for full-text review. Of these potentially relevant articles, 33 publications were excluded for various reasons and 8 publications met the inclusion criteria and were included in this report. These comprised 2 randomized controlled trials (RCTs), 4 non-randomized studies, and 2 evidence-based guidelines. Appendix 1 presents the PRISMA15 flow chart of the study selection. Additional references of potential interest are provided in Appendix 5.

Summary of Study Characteristics

The detailed characteristics of the included primary studies16-21 (Table 2) and the European Society for Blood and Marrow Transplantation (EBMT) guideline12 and the American Society for Blood and Marrow Transplantation (ASBMT)22 guideline (Table 3) are presented in Appendix 2.

Study Design

Of the 6 included primary studies, 2 were RCTs16,17 and 4 were retrospective cohort studies.18-21 Both RCTs (1 phase III16 and 1 phase II17) were open-label, multi-centre, parallel, 1:1 trials. Both reported sample size calculations to detect a clinically relevant treatment effect. The results in the phase III trial16 were analyzed using a per-protocol population and not the intention-to-treat population. Adjustment for multiple comparisons were not performed in both RCTs. None of the retrospective cohort studies reported a sample size calculation. One retrospective cohort study20 used multivariate analysis to account for differences in some baseline characteristics between groups, while the other 3 retrospective cohort studies18,19,21 did not identify and adjust for covariates in their analyses.

Both included guidelines12,22 were developed to update their previous guidelines on the recommendations of AHSCT on MS treatment. Evidence was identified from literature searches. The recommendations in both guidelines were developed by a panel of experts in AHSCT and MS. The EBMT guideline12 rated the evidence as I, II, or III based on the health benefits, side effects, and risks of AHSCT, as compared with non-cell transplant options. The strength of recommendations in the EBMT guideline12 were classified as S (standard of care), CO (clinical option), D (developmental), or GNR (generally not recommended) based on the level of evidence obtained. The ASBMT guideline22 did not rate the evidence or the strength of its recommendations. The method of recommendations development and evaluation was not clearly stated in the ASBMT guideline.22

Country of Origin

The included primary studies were conducted by authors from US,16 Italy,17,20,21 and Sweden.18,19 The included guidelines were conducted by authors from countries in Europe,12 and from the US and Canada.22

Patient Population

Patients in 416,19-21 out of 6 primary studies were exclusively RRMS. The phase II RCT17 included patients with progressive MS (67%) and RRMS (33%). One study18 had a patient population consisting mainly of RRMS with a small percentage of patients with PPMS (1.2%) and SPMS (6.1%). The sample sizes of 6 included studies ranged from 21 to 271. The mean age of patients in the included studies was approximately 35 years, with the percent of females ranging from 60% to 79%. The mean disease duration ranged from 6 to 11 years and the mean baseline Expanded Disability Status Scale (EDSS) score (which measures the progression or deterioration of MS) ranged from 2.5 to 6. The EDSS score ranges from 0 to 10 in 0.5-point increments from no neurologic disability (0) to worst neurologic disability (10).

The target populations in both guidelines12,22 are patients diagnosed with defined MS. The intended users of both guidelines are health care workers in the MS field working with transplant teams or considering the referral of patients.

Interventions and Comparators

AHSCT was the intervention in all the included studies classified as myeloablative17,18,20,21 or non-myeloablative,16,18,19 based on the conditioning regimens. The comparators were DMT,16,21 mitoxantrone (MTX),17 and alemtuzumab (ALZ).18-20

The intervention considered in the evidence-based guidelines12,22 was AHSCT for MS.

Outcomes

The outcomes considered in the included primary studies16-21 could be classified as clinical outcomes, MRI outcomes, and safety outcomes. The clinical outcomes consisted of disease progression (defined as an increase in EDSS score of at least 1 point on 2 evaluations 6 months apart after at least 1 year of treatment), clinical relapse (a neurologic symptom lasting more than 24 hours; not associated with infection, fever, or heat intolerance), the Neurologic Rating Scale (NRS; range 0 to 100 in 1-point increments from worst [0] to no [100] disability; minimal clinically important difference, 10), the Short Form-36 quality of life (SF-36 QoL; range, 1 to 100; higher scores indicate more favourable health state), and the Multiple Sclerosis Functional Composite (MSFC) score (which incorporates the timed 25-ft walk test, the Nine-Hole Peg Test [a measure of arm function], and the Paced Auditory Serial Addition Test [scored as the total number correct out of 60 possible, and data shown are % of correct answers]). The MRI outcomes were MRI T2-weighted lesion volume (reported as % change from baseline), new T2 MRI lesions, Gd-enhancing lesions on T1-weighted MRI. No evidence of disease activity (NEDA) was a composite outcome defined as no progression, no relapses, and no new or enlarging lesions on MRI. The safety outcomes included mortality, early AEs (within 100 days after treatment), late AEs (after 100 days of treatment), and serious AEs. The follow-up period was 3 years in 5 studies17-21 and 5 years in 1 study.16

Both guidelines12,22 considered the efficacy and safety outcomes of AHSCT in MS in the development of recommendations. The EBMT guideline12 also considered the resources implications and other issues relevant to the implementation of AHSCT.

Summary of Critical Appraisal

All included primary studies16-21 were explicit in reporting (i.e., clearly described the objective of the study, the main outcomes, the characteristics of the participants, the interventions, differences in baseline characteristics between groups, and the main findings of the study). All studies16-21 provided estimates of the random variability (e.g., standard deviation or 95% confidence interval) in the data of the main outcome and actual P values for main outcomes. As 2 RCTs16,17 and 4 retrospective cohort studies18-21 with relatively small sample sizes, it was not applicable to determine if the participants were representative of the entire population from which they were recruited. However, the treatment settings were representative of the treatment received by most of the patients. Both RCTs16,17 were open-label, as it was not possible to perform blinding of participants, personnel, or outcome assessors. Allocation concealment was not reported in the RCTs.16,17 Sample size was determined in both RCTs16,17 but not in the retrospective cohort studies.18-21 The intervention and comparator groups in all included studies had the same follow-up. Appropriate statistical tests were used to assess the main outcomes, which were accurately measured. Three18,19,21 of 4 retrospective cohort studies did not identify or conduct any adjustment for potential confounders in the analyses from which the main findings were drawn. The findings in those studies were therefore considered as crude and less reliable. Overall, all studies, except for the phase III RCT,16 were of limited methodological quality.

Both included guidelines12,22 were explicit in scope and purpose (i.e., objectives, health questions, and populations) and had clear presentation (i.e., specific and unambiguous recommendations, different options for management of the condition or health issue, and easy to find key recommendations). Regarding stakeholder involvement, the guidelines clearly defined target users and the development groups; however, it was unclear if the views and preferences of the MS populations were sought. For rigour of development, both guidelines12,22 did not report details of systematic searches for evidence, criteria for selecting evidence, and methods of formulating the recommendations. Both guidelines considered health benefits, side effects, and risks in formulating the recommendations, were peer-reviewed before publication, and provided a procedure for updating. For applicability, the EBMT guideline12 was explicit in terms of facilitators and barriers to application, advice and/or tools on how the recommendations can be put into practice, resource (cost) implications, and monitoring and or auditing criteria. For editorial independence, it was unclear if the funding bodies had any influence over the content of the guidelines. The competing interests of the guideline development group members were reported. Overall, both included guidelines appeared to be of acceptable methodological quality.

Summary of Findings

The main findings and authors’ conclusions of the primary studies16-21 (Table 6) and the recommendations of the guidelines12,22 (Table 7) are presented in Appendix 4.

Clinical Effectiveness of AHSCT

Disease Progression

In the phase III RCT,16 the proportions of patients with disease progression at 1, 3, and 5 years of follow-up were significantly lower in the AHSCT group (1.9%, 5.2%, and 9.7%, respectively) compared to those in the DMT group (24.5%, 62.5%, and 75.3%, respectively); P < 0.001. The median time to disease progression of the AHSCT group could not be calculated because of too few events and the median time to disease progression in the DMT group was 24 months (range, 18 to 48 months) (hazard ratio = 0.07; 95% confidence interval [CI], 0.02 to 0.24; P < 0.001). The between-group difference in mean change in EDSS score from baseline to 1 year was −1.7; 95% CI, −2.03 to −1.29; P < 0.001. The phase II RCT17 with a small sample size (N = 21) reported no significant difference between the AHSCT and MTX groups in the proportions of patients with disease progression, or in the EDSS change. The unobserved differences between groups in disease progression were likely due to the low power of this trial. A retrospective cohort study by Zhukovsky et al.19 found that the EDSS changes from baseline to 1, 2, and 3 years of follow-up in the AHSCT group showed significant improvement compared to those in the ALZ group (P < 0.0001). Another retrospective cohort study by Boffa et al.20 found that AHSCT promoted significant EDSS improvement compared with ALZ (P = 0.035). The proportion of patients with confirmed improvement in EDSS scores was significantly higher in the AHSCT group compared to the DMT groups (44.4% versus 6.1%; P = 0.013).21

Clinical Relapse

In the phase III RCT,16 the proportions of patients with clinical relapse in the AHSCT group were significantly lower compared to those in the DMT group at all time points of follow-up (P < 0.001). The annualized relapse rate (ARR) reported in the phase II RCT17 was significantly lower in the AHSCT group compared to the MTX group (0.19 versus 0.60; rate ratio = 0.36; 95% CI, 0.15 to 0.88; P = 0.026). A retrospective study by Zhukovsky et al.19 also reported a significantly lower ARR in the AHSCT group compared to the ALZ group (0.04 versus 0.1; P = 0.03). Similarly, 2 other retrospective cohort studies also showed a significantly lower ARR in the AHSCT group compared to the ALZ group (0.05 versus 0.35; P = 0.02)20 and to DMT (0.0 versus 0.67; P < 0.0001).21

Neurologic Rating Scale

In the phase III RCT,16 the mean change in NRS score from baseline to 1 year in the AHSCT group was + 8.8 (improved) and that in the DMT group was –1.6 (worsened). The between-group difference in mean change in NRS score was + 11.2; 95% CI, + 8.08 to + 14.29; P = 0.001, indicating significant improvement in favour of the AHSCT group compared to the DMT group.

SF-36 QoL

In the phase III RCT,16 the quality of life in the AHSCT group was improved ( + 19.5) but worsened in the DMT group (−3.4). The between-group difference in mean change in SF-36 QoL score from baseline to 1 year was + 23; 95% CI, + 17.6 to + 28.9; P < 0.001.

Multiple Sclerosis Functional Composite

In the phase III RCT,16 the mean change in MSFC score from baseline to 1 year in the AHSCT group was + 0.32 (improved) and in the DMT group was −0.31 (worsened). The between-group difference was + 0.51; 95% CI, + 0.28 to + 0.72; P < 0.001. There were significant improvements in the timed 25-feet walk and the 9-Hole Peg Test in the AHSCT group compared to the DMT group (P < 0.001). However, there was no significant difference in the Paced Auditory Serial Addition Test score between groups (P = 0.61).

MRI outcomes

In the phase III RCT,16 the mean change in MRI T2-weighted lesion volume from baseline to 1 year in the AHSCT group was −31.7% (improved) and in the DMT group was + 34.3% (worsened). The between-group difference was −66% (95% CI, −70.6 to −61.3); P < 0.001. The phase II RCT17 showed that the number of new T2 lesions over 4 years was significantly reduced in the AHSCT group compared to the MTX group (rate ratio = 0.21; 95% CI, 0.10 to 0.48; P = 0.00016). The result remained significant in all sensitivity analyses. No patients in the AHSCT group had Gd-enhancing lesions compared to 56% of the MTX patients, who had at least 1 Gd lesions during 4 years of follow-up (P = 0.029).17 A retrospective cohort study by Boffa et al.20 showed that AHSCT significantly reduced the risk of MRI activity compared to the ALZ (MRI-activity-free survival: 85% versus 59%; hazard ratio = 0.13; 95% CI, 0.03 to 0.59; P = 0.009).

No Evidence of Disease Activity

The post-hoc analysis of the phase III RCT16 showed that the proportions of patients reaching NEDA at 1, 3, and 5 years after randomization were significantly higher in the AHSCT group (98.1%, 90.3%, and 78.5%, respectively) compared to the DMT group (20.8%, 5.9%, and 3%, respectively); P > 0.001. Three retrospective cohort studies19-21 also found similar results in that the proportions of patients with NEDA were significantly higher in the AHSCT group compared to the ALZ group19,20 or DMT group21 at the end of follow-up.

Safety

Four studies16,17,19,21 reported no deaths occurring in either intervention group. One study18 reported 1 death in the AHSCT group due to suicide (incidence rate [IR] per 1,000 person-years = 1.7; 95% CI, 0.0 to 9.6) and 4 deaths in the ALZ group (2 suicides, 1 heart attack, 1 cytomegalovirus reactivation) (IR per 1,000 person-years = 8.6; 95% CI, 2.3 to 22.0).

The phase III RCT16 reported that no AHSCT patients developed Common Toxicity Criteria grade 4 nonhematopoietic toxicities such as myocardial infarction, embolism, dialysis, sepsis, or need for pressor support. There were also no patients in the AHSCT group transferred to intensive care units; or who received parenteral nutrition, surgery, or other disabling or potential life-threatening events. Of the 52 study participants in the AHSCT group, few patients had inpatient grade 3 AE infections: there was 1 case (1.9%) of Clostridium difficile diarrhea, 1 case (1.9%) of Escherichia coli urinary tract infection, and 1 case (1.9%) of culture-negative pneumonia. Post-transplantation infections were mainly upper respiratory tract infections that occurred in both groups (16 cases [31%] in AHSCT group and 15 cases [29%] in DMT group). There was no progressive multifocal leukoencephalopathy (PML) caused by John Cunningham virus infections in either group. The rate of infection per patient-year was 0.19 in the AHSCT group and 0.23 in the DMT group. No statistical comparison between groups was given.

In the phase II RCT,17 early adverse events (occurring < 100 days after treatment) associated with AHSCT included grade 3 AEs, which included febrile neutropenia, leukopenia, anemia, platelet count decrease, amenorrhea, leukopenia; and grade 4 AEs, which included leukopenia, anemia, and platelet count decrease. In the MTX group, grade 3 AEs that occurred included neutrophil count decrease, amenorrhea, leukopenia, and lymphocyte count decrease; and grade 4 AEs that occurred included neutrophil count decrease. Severe AEs that occurred in the AHSCT group were sepsis, prolonged hospitalization, systemic candidiasis, cytomegalovirus reactivation, and engraftment failure. None of the patients in the MTX group had severe AEs. All AEs in the AHSCT group were resolved without sequelae. The IR of these AEs and statistical comparisons between groups were not provided.

A retrospective cohort study by Alping et al.18 found that thyroid disease occurred more often for AHSCT (IR per 1,000 person-years = 34; 95% CI, 18 to 56) compared to the non-induction therapy group (natalizumab, dimethyl fumarate, rituximab, fingolimod) (IR = 5.3; 95% CI, 3.9 to 7.1), but less frequent than ALZ (IR = 109; 95% CI, 75 to 154). The non-thyroid autoimmune disease was rare, and the IRs were comparable between groups. The incidence of any infection was highest among the AHSCT group (IR = 275; 95% CI, 213 to 350) compared to ALZ group (IR = 56; 95% CI, 34 to 87) and the non-induction therapy group (IR = 52; 95% CI, 47 to 58), but dropped to the level near to the ALZ and the non-induction therapy groups within the first year. Common infections found after treatment with AHSCT and ALZ were herpes infections and bacterial sepsis. Systemic antibiotics were given to all patients after AHSCT and to most patients after ALZ. No cases of PML were detected.

A retrospective cohort study by Zhukovsky et al.19 reported that 70% of patients treated with AHSCT developed early Common Toxicity Criteria grade 3 AEs or higher in which febrile neutropenia (58%) was the most common. None of the patients in the ALZ group had grade 3 AEs or higher. Late grade 3 AEs occurred in 1 patient (1.4%) treated with AHSCT (Lyme neuroborreliosis) and 5 patients (6.7%) treated with ALZ (4 cases [5.3%] of immune mediated thrombocytopenia, 1 case [1.3%] of breast cancer). Thyroid disease was the most common autoimmune adverse event that occurred in both groups: 20% in the AHSCT group and 47% in the ALZ group; P = 0.005. Herpes zoster infection developed in 5.8% in the AHSCT group and 6.7% in the ALZ group as late infection.

A retrospective cohort study by Boffa et al.20 reported that serious infections including neutropenic fever (64%) and sepsis (32%) were commonly diagnosed as early AEs in the AHSCT group. Late AEs such as autoimmune disorders, especially thyroid disorders, were more common in the ALZ group (28%) compared to the AHSCT group (4%).

A retrospective cohort study by Mariottini et al.21 found no life-threatening complications including PML after AHSCT. All infections and viral reactivations occurred within 1 year from transplantation. One patient in the AHSCT group (9%) developed autoimmune thyroiditis.

Cost-Effectiveness of AHSCT

No comparative cost-effectiveness studies of AHSCT were identified; therefore, no summary can be provided.

Guidelines

The EBMT guideline12 recommends that AHSCT should be offered to patients with RRMS who have high clinical and MRI inflammatory disease activity despite the use of 1 or more lines of approved DMTs. Patients should be younger than 45 years, with a EDSS score of 5.5, at most, and have a disease duration of less than 10 years. The strength of recommendation was categorized as S (standard of care) based on level I evidence. The guideline recommends that AHSCT should be considered for patients with “aggressive” MS who develop severe disability in the previous 12 months, even before failing a full course of DMT. The guideline suggests that SPMS, PPMS, and MS pediatric patients with documented evident disability progression in the previous 12 months should be considered for AHSCT, preferably in a prospective clinical trial. The strength of these recommendations was categorized as CO based on level II evidence. Details of the levels of evidence and strengths of recommendation are presented in the footnotes of Table 3 in Appendix 2.

The ASBMT guideline22 endorses AHSCT as “standard of care, with clinical evidence available” for patients with relapsing forms of MS (such as RRMS or progressive MS) who are refractory to treatment and have prognostic factors indicating a high risk of disability. This includes a clinical relapse and MRI activity, despite treatment with high efficacy MDTs. The strength of recommendation and the level of evidence were not given.

Limitations

The evidence from this review was derived from the findings of 2 RCTs16,17 and 4 retrospective cohort studies18-21 with relatively small sample sizes. Both RCTs were open-label by design because of the nature of the interventions that might result in a high risk of performance and detection biases. The study design of the phase III RCT16 allowed DMT patients to cross over to receive AHSCT after 1 year of follow-up in the event of treatment failure, resulting in up to 61% of DMT patients crossing over and thus limiting accurate comparisons of longer-term outcomes between groups. Two newer potent monoclonal antibodies — ocrelizumab and ALZ — were not included in the DMT group because of their unavailability or a safety concern that may result in a higher efficacy of AHSCT than their comparators.16 The phase III RCT used a per-protocol population in its analysis, which did not account for patients who were lost to follow-up after treatment. Adjustment for multiple comparisons were not performed in both RCTs. The results of the retrospective non-randomized studies18-21 might be affected by selection bias because of the nature of the study design. None of the retrospective cohort studies reported sample size calculations. There were significant differences between groups in certain patient characteristics, such as age, disease duration, ARR, EDSS, MS type, and MRI activity at baseline. Patients in the ASHCT group were younger and had longer disease duration and higher disease activity compared to those in the control group. Three studies18,19,21 did not attempt to adjust for confounders in the analyses. As most patients in the included studies were of RRMS type, evidence for the clinical effectiveness of AHSCT on progressive MS remains to be determined. Both guidelines were limited in terms of rigour of development, as the details of systematic searches for evidence, criteria for selecting evidence, and methods of formulating the recommendations were not reported. However, the guidelines were the updated versions of their previous ones and were developed in Europe, the US, and Canada. Despite these limitations, all included studies showed profound effects of AHSCT in halting disease activity and preventing disability in RRMS patients. The findings of the clinical studies and the recommendations of the guidelines are likely to be applicable to the Canadian context.

Conclusions and Implications for Decision- or Policy-Making

Two RCTs16,17 and 4 retrospective cohort studies18-21 were included to address the clinical effectiveness of AHSCT in patients with MS. Two evidence-based guidelines12,22 on the use of AHSCT in MS were identified. No studies regarding the cost-effectiveness of AHSCT for MS were identified.

Compared to ALZ or other DMTs, patients in the AHSCT group had significant improvement in clinical outcomes including disease progression, clinical relapse, NRS, MSFC score, and NEDA. Patients treated with AHSCT also experienced improvement in quality of life and a decrease in MRI activity such as new T2 lesions and Gd-enhancing lesions. For safety, there was no treatment-related mortality or life-threatening complications including PML after treatment with AHSCT. The most common AEs in the first 100 days after AHSCT treatment were febrile neutropenia, organ infections, sepsis and viral reactivations (e.g., Epstein-Barr virus, cytomegalovirus, varicella-zoster virus). Long-term AEs included the development of new autoimmune diseases, mainly thyroid disease. Studies comparing different AHSCT conditioning regimens such as myeloablative and non-myeloablative were not identified. Thus, AHSCT appears to be efficacious with expected adverse events. Treatment with DMTs including ALZs and MTZ were also associated with high rates of infection. Thyroid disease occurred more frequent in the patients treated with ALZ compared to AHSCT patients. The identified guidelines recommend the use of AHSCT as standard of care for the treatment of highly active RRMS patients refractory to DMTs and suggest that the treatment may be appropriate for progressive forms of MS with an active inflammatory component. As the follow-up period was 3 years in most included studies, longer-term efficacy and safety data of AHSCT remain to be determined. The findings in this report should be interpreted with caution because of some limitations of the included studies (e.g., small sample sizes, and limited in study design). Future large and well-designed RCTs with inclusion of more recent and potent DMTs are warranted to further evaluate the role of AHSCT in MS with longer-term efficacy and safety data. Moreover, trials aiming at refining treatment procedure by comparing different conditioning regimens are also needed.

References

1.MS Society of Canada. Prevalence and incidence of MS in Canada and around the world. 2020; https://mssociety.ca/research-news/article/prevalence-and-incidence-of-ms-in-canada-and-around-the-world#:~:text=Canada%20continues%20to%20have%20one,MS%20%E2%80%93%201%20in%20every%20400. Accessed 2021 Apr 22.

2.National Multiple Sclerosis Society. Types of MS. 2020; https://www.nationalmssociety.org/What-is-MS/Types-of-MS. Accessed 2021 Apr 22.

3.Thompson AJ, Banwell BL, Barkhof F, et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018;17(2):162-173. PubMed

4.Barnett Y, Garber JY, Barnett MH. MRI biomarkers of disease progression in multiple sclerosis: old dog, new tricks? Quant Imaging Med Surg. 2020;10(2):527-532. PubMed

5.Multiple Sclerosis Society of Canada. Relapses and relapse management. 2017; https://mssociety.ca/library/document/XtQHJqhSi1CGjrzKMgeNUuAc5kBPv3Wy/original.pdf. Accessed 2021 Apr 22.

6.Manouchehrinia A, Beiki O, Hillert J. Clinical course of multiple sclerosis: a nationwide cohort study. Mult Scler. 2017;23(11):1488-1495. PubMed

7.Fambiatos A, Jokubaitis V, Horakova D, et al. Risk of secondary progressive multiple sclerosis: a longitudinal study. Mult Scler. 2020;26(1):79-90. PubMed

8.MS Society of Canada. Disease-modifying therapies. 2021; https://mssociety.ca/managing-ms/treatments/medications/disease-modifying-therapies-dmts. Accessed 2021 Apr 22.

9.Vargas DL, Tyor WR. Update on disease-modifying therapies for multiple sclerosis. J Investig Med. 2017;65(5):883-891. PubMed

10.Mariottini A, De Matteis E, Muraro PA. Haematopoietic stem cell transplantation for multiple sclerosis: current status. BioDrugs. 2020;34(3):307-325. PubMed

11.Jessop H, Farge D, Saccardi R, et al. General information for patients and carers considering haematopoietic stem cell transplantation (HSCT) for severe autoimmune diseases (ADs): a position statement from the EBMT Autoimmune Diseases Working Party (ADWP), the EBMT Nurses Group, the EBMT Patient, Family and Donor Committee and the Joint Accreditation Committee of ISCT and EBMT (JACIE). Bone Marrow Transplant. 2019;54(7):933-942. PubMed

12.Sharrack B, Saccardi R, Alexander T, et al. Autologous haematopoietic stem cell transplantation and other cellular therapy in multiple sclerosis and immune-mediated neurological diseases: updated guidelines and recommendations from the EBMT Autoimmune Diseases Working Party (ADWP) and the Joint Accreditation Committee of EBMT and ISCT (JACIE). Bone Marrow Transplant. 2020;55(2):283-306. PubMed

13.Downs SH, Black N. The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J Epidemiol Community Health. 1998;52(6):377-384. PubMed

14.Agree Next Steps Consortium. The AGREE II Instrument. Hamilton (ON): AGREE Enterprise; 2017: https://www.agreetrust.org/wp-content/uploads/2017/12/AGREE-II-Users-Manual-and-23-item-Instrument-2009-Update-2017.pdf. Accessed 2021 Apr 22.

15.Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62(10):e1-e34. PubMed

16.Burt RK, Balabanov R, Burman J, et al. 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;321(2):165-174. PubMed

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

18.Alping P, Burman J, Lycke J, Frisell T, Piehl F. Safety of alemtuzumab and autologous hematopoietic stem cell transplantation compared to noninduction therapies for multiple sclerosis. Neurology. 2021;96(11):e1574-e1584. PubMed

19.Zhukovsky C, Sandgren S, Silfverberg T, et al. Autologous haematopoietic stem cell transplantation compared with alemtuzumab for relapsing-remitting multiple sclerosis: an observational study. J Neurol Neurosurg Psychiatry. 2021;92(2):189-194. PubMed

20.Boffa G, Lapucci C, Sbragia E, et al. Aggressive multiple sclerosis: a single-centre, real-world treatment experience with autologous haematopoietic stem cell transplantation and alemtuzumab. Eur J Neurol. 2047;27(10):2047-2055.

21.Mariottini A, Innocenti C, Forci B, et al. Safety and efficacy of autologous hematopoietic stem-cell transplantation following natalizumab discontinuation in aggressive multiple sclerosis. Eur J Neurol. 2019;26(4):624-630. PubMed

22.Cohen JA, Baldassari LE, Atkins HL, et al. Autologous hematopoietic cell transplantation for treatment-refractory relapsing multiple sclerosis: position statement from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2019;25(5):845-854. PubMed

Appendix 1: Selection of Included Studies

Figure 1: Selection of Included Studies

Four hundred and 64 citations were identified, of which 423 were excluded. Forty-one electronic literature and no grey literature potentially relevant full-text reports were retrieved for scrutiny. In total, 8 reports are included in the review.

Appendix 2: Characteristics of Included Publications

Table 2: Characteristics of Included Primary Clinical Studies

Study citation, country, funding source

Study design

Population characteristics

Intervention and comparator(s)

Clinical outcomes, length of follow-up

Burt et al., 201916

US

Funding: No industry support or pharmaceutical support

Phase III, open-label, multicenter, parallel, 1:1 RCT

Sample size determination: Yes

ITT analysis: No

Adjustment for multiple comparisons: No

Patients with RRMS (N = 110)

Mean age, years:

  • HSCT: 35.6

  • DMT: 35.6

% female:

  • HSCT: 62

  • DMT: 66

Mean duration of disease, months:

  • HSCT: 63.1

  • DMT: 84.8

Mean EDSS:

  • HSCT: 3.4

  • DMT: 3.3

Mean number of Gd-enhancing lesions on MRI:

  • HSCT: 4.5

  • DMT: 4.9

Mean MRI T2-weighted lesion volume, cm3

  • HSCT: 16.4

  • DMT: 12.5

Non-myeloablative HSCT (n = 55): DMT was discontinued and variable washout periods were observed before admission for HSCT.

DMT (n = 55): Received an FDA-approved DMT of higher efficacy or a different class than the therapy taken at the time of randomization. Could also receive immune-modulating or immunosuppressive drugs. Ocrelizumab and alemtuzumab were excluded. Patients experiencing progression of disability after at least 1 year of treatment were allowed to cross over to receive HSCT.

Primary Outcome: Disease progression (an increase in EDSS score of at least 1 point on 2 evaluations 6 months apart after at least 1 year of treatment)

Secondary Outcomes:

  • Survival

  • Relapses (a neurologic symptom lasting > 24 hours; not associated with infection, fever, or heat intolerance)

  • NRS (range 0 to 100 in 1-point increments from worst [0] to no [100] disability; MCID, 10)

  • T2-weighted lesion volume (% change from baseline)

  • SF-36 QoL (range 1 to 100; higher scores = more favourable health state)

  • MSFC score (incorporates a timed 25-ft walk test, the 9-Hole Peg test [a measure of arm function], and the PASAT [scored as the total number correct out of 60; data shown are % of correct answers])

Post-hoc Outcomes:

  • Time to first relapse

  • NEDA (i.e., no progression, no relapses, and no new or enlarging lesions on MRI)

  • Outcomes of patients in DMT group who crossed over to receive HSCT

Adverse events: Transplantation-related; hospitalizations; ED visits; infections; new medical problems

Follow-up: 5 years

Mancardi et al., 210517

Italy

Funding: Italian Multiple Sclerosis Foundation

phase II, open-label, multicenter, parallel, 1:1 RCT

Sample size determinations: Yes

ITT analysis: Yes

Adjustment for multiple comparison: No

Patients with a defined MS (secondary-progressive MS or RRMS) (N = 21)

Mean age, years:

  • AHSCT: 36

  • MTX: 35

% female:

  • AHSCT: 56

  • MTX: 75

Median EDSS (range)

  • AHSCT: 6.5 (5.5 to 6.5)

  • MTX: 6 (5.5 to 6.5)

Mean disease duration (range), years:

  • AHSCT: 10.5 (5 to 20)

  • MTX: 9.8 (2 to 23)

Myeloablative AHSCT (n = 9)

MTX (n = 12): 20 mg every month for 6 months

Primary Outcomes:

  • Number of new T2 MRI lesions

Secondary Outcomes:

  • Gd-enhancing lesions on T1-weighted MRI

  • Relapses

  • Time to disability progression

  • Time of appearance of the first new T2 MRI lesions

Safety:

  • AHSCT-related mortality

  • Early AEs

  • Serious AEs

Follow-up: 4 years

Alping et al., 202118

Sweden

Funding: Patient-Centered Outcomes Research Institute award.

Retrospective cohort study

Sample size determination: No

Adjustment for covariates: No

Patients with MS (mainly RRMS) (N = 271)

Mean age, years: 34.0

% female: 65.0

Mean MS duration, years: 6.7

MS type, %:

  • RRMS: 92.8

  • PPMS: 1.2

  • SPMS: 6.1

Mean EDSS score: 3.1

Both non-myeloablative and myeloablative AHSCT (n = 139)

ALZ (n = 132)

Matched non-induction therapies (Natalizumab, dimethyl fumarate, rituximab, fingolimod; n = 2,486)

Outcomes:

  • Death

  • Thyroid disease

  • Non-thyroid autoimmune disease

  • Infection

Follow-up: 3 years

Zhukovsky et al., 202119

Sweden

Funding: No industry support or pharmaceutical support

Retrospective cohort study

Sample size determination: No

Adjustment for covariates: No

Patients with RRMS treated with AHSCT or alemtuzumab from 1 January 2011 to 31 December 2018 (N = 144)

Median age (IQR):

  • AHSCT: 30 (26 to 37)

  • ALZ: 35 (30 to 41); P = 0.005

% female:

  • AHSCT: 40.8

  • ALZ: 78.6

Mean duration of disease, years:

  • AHSCT: 6.4

  • ALZ: 7.0

ARR 1 year before treatment:

  • AHSCT: 1.4

  • ALZ: 0.54; P < 0.0001

Median EDSS (IQR):

  • AHSCT: 3 (2 to 4)

  • ALZ: 2 (1 to 2.5); P < 0.0001

Median ARMSS (IQR):

  • AHSCT: 6.1 (4.2 to 7.3)

  • ALZ: 78.6 (2.0 to 5.5); P < 0.0001

Non-myeloablative AHSCT (n = 69)

ALZ (n = 75): 60 mg over 5 days and a repeated dose of 36 mg over 3 days after 1 year. New courses of 36 mg were administered if clinical relapses and/or new MRI lesions occurred.

Primary Outcomes:

  • NEDA at 3 years (defined as absence of clinical relapses, CDW and MRI events)

Secondary Outcomes:

  • Freedom from MRI events

  • Clinical relapses (a period of acute worsening of neurologic function lasting ≥ 24 hour not attributable to an external cause such as increased body temperature or acute infection)

  • Freedom from CDW (an increase in EDSS score with at least 1 point from baseline sustained between 2 follow-up visits separated in time by no less than 6 months)

  • ARR (number of relapses occurring during a time period divided by the number of years in that time period)

  • Proportion of patients (EDSS ≥ 2) with CDI/stability/CDW (CDI, a decrease in EDSS score with at least 1 point from baseline sustained between 2 follow-up visits separated in time by no less than 6 months)

  • AEs of grade 3 or higher according to CTCAE v5.0 within the first 100 days after treatment

  • Late AEs after treatment (autoimmune or infectious AEs grade 2 or higher, or any AEs grade 3 or higher present at 100 days from treatment or occurring thereafter.

Follow-up: 3 years

Boffa et al., 202020

Italy

Funding: Did not receive any funding support

Retrospective cohort study

Sample size determination: No

Adjustment for covariates: Yes

Patients with aggressive RRMS (multiple [≥ 2] relapses with incomplete resolution in the past year; > 2 MRI scans showing new or enlarging T2 lesions or Gd-enhancing lesions despite active treatment; or EDSS score ≥ 4 within 5 years of onset; no response to therapy with 1 or more DMTs for up to 1 year) (N = 57)

Mean age, years:

  • AHSCT: 32.1, ALZ: 35.1

% female:

  • AHSCT: 76, ALZ: 75

Median EDSS (IQR):

  • AHSCT: 6 (4.5 to 7), ALZ: 3 (1 to 4)

Mean disease duration, years:

  • AHSCT: 9.5, ALZ: 7.2

Mean ARR:

  • AHSCT: 3.2, ALZ: 1.7

MRI activity, %:

  • AHSCT: 88, ALZ: 44

Mean number of Gd-enhancing lesions:

  • AHSCT: 15.5, ALZ: 1.6

Mean follow-up, months:

  • AHSCT: 50.9, ALZ: 26.3

Myeloablative AHSCT (n = 25)

ALZ (n = 32): 12 mg per day on 5 consecutive days and the second of 12 mg per day on 3 consecutive days, 12 months apart.

Outcomes:

Primary:

  • Time to first relapse

  • Time to confirm disability worsening

  • Time to first evidence of MRI activity

  • Time to first evidence of disease activity (according to NEDA definition)

Secondary:

  • ARR at 12, 24 and 36 months

  • 6-month confirmed EDSS changes at 12 and 24 months

  • Safety

Follow-up: 3 years

Mariottini et al., 201921

Italy

Funding: Not reported

Retrospective cohort study

Patients with RRMS who discontinued from natalizumab after at least 6 administrations and with at least 6 months of follow-up (N = 55)

Median age (IQR), years:

  • AHSCT: 35 (31 to 43)

  • DMT: 39 (35 to 47)

% female:

  • AHSCT: 81.8

  • DMT: 75.6

Median disease duration (IQR), years:

  • AHSCT: 13.0 (5.0 to 16.0)

  • DMT: 9.0 (5.0 to 13.0)

Median EDSS score (IQR):

  • AHSCT: 3.0 (1.5 to 4.5)

  • DMT: 2.5 (2.4 to 4.0)

Myeloablative AHSCT (n = 11)

DMT (n = 41): First-line (interferons, glatiramer acetate, dimethyl fumarate) or second-line (fingolimod, alemtuzumab, rituximab, cyclophosphamide, mitoxantrone)

Outcomes:

  • NEDA

  • Clinical relapses

  • Disability worsening

  • Safety (AEs)

Follow-up: 3 years

AE = adverse event; AHSCT = autologous hematopoietic stem cell transplantation; ALZ = alemtuzumab; ARMSS = age-related multiple sclerosis severity score; ARR = annualized relapse rate; CDI = confirmed disability improvement; CDW = confirmed disability worsening; DMT = disease-modifying therapy; EDSS = Expanded Disability Status Scale; FDA = FDA; Gd = gadolinium; HSCT = hematopoietic stem cell transplantation; IQR = interquartile range; IQR = interquartile range; ITT = intention-to-treat; MCID = minimal clinically important difference; MRI = MRI; MSFC = Multiple Sclerosis Functional Composite; MTX = mitoxantrone; MS = multiple sclerosis; NEDA = no evidence of disease activity; NRS = Neurologic Rating Scale; PASAT = Paced Auditory Serial Addition Test; PPMS = primary-progressive MS; QoL = quality of life; RCT = randomized controlled trial; RRMS = relapse-remitting multiple sclerosis; SF-36 = Short Form 36; SPMS = secondary-progressive MS.

Table 3: Characteristics of Included Guidelines

Intended users, target population

Intervention and practice considered

Major outcomes considered

Evidence collection, selection, and synthesis

Evidence quality assessment

Recommendations development and evaluation

Guideline validation

EBMT, Sharrack et al., 202012

Intended users: Transplant physicians, nurses, neurologists

Target population: Not primarily targeted at patients, families, and non-specialist health professional carers

HSCT in MS and immune-mediated neurologic diseases

Efficacy and safety of AHSCT

Evidence was found from PubMed searches and recent EBMT congress presentations.

Evidence selection and synthesis: Not reported

Evidence levels (I, II, III)a based on consideration of health benefits, side effects and risks, and balanced against non-HSCT options.

Recommendationsb were classified as S, CO, D or GNR

Updated of its previous guidelines

Recommendations were developed by clinicians, nurses, statisticians and data management persons, all with experience in HSCT.

Each recommendation provides potential for auditing clinical practice. The guideline considers resource implications and other issues relevant to implementations of HSCT.

Published in peer-reviewed journal

ASBMT, Cohen et al., 201922

Intended Users: Neurologists in treating MS and transplantation physicians with experience in AHSCT

Target Population: Patients with MS

AHSCT in MS

Efficacy and safety of AHSCT in MS

Evidence identified from the Embase and Ovid MEDLINE databases. One author reviewed all search results.

Evidence selection and synthesis: Not reported

Evidence quality assessment: None, just by consensus from a panel of experts (expert opinion)

Updated of its previous guidelines

Recommendations were developed by a panel of experts in AHSCT and MS.

Published in peer-reviewed journal

AHSCT = autologous hematopoietic stem cell transplantation; ASBMT = American Society for Blood and Marrow Transplantation; HSCT = Hematopoietic stem cell transplantation; EBMT = European Group for Blood and Marrow Transplantation; MS = multiple sclerosis; RCT = randomized controlled trial.

Note that this table has been formatted for accessibility but has not been copy-edited.

aLevels of evidence:

I: from at least on well-executed RCT.

II: from at least 1 well-designed clinical trial without randomization; cohort or case-controlled analytic studies (preferably from more than 1 centre); multiple time-series studies; or dramatic results from uncontrolled experiments.

III: from opinions of respected authorities based on clinical experience, descriptive studies, or reports from expert committees.

bRecommendations:

S: Standard of care – Indications categorized as S are reasonably well defined and results compare favourably (or are superior) to those of non-transplant treatment approaches.

CO: clinical option – The CO category applies to indications for which the results of small patient cohorts show efficacy and acceptable toxicity of the HSCT procedure, but confirmatory randomized studies are missing, often as a result of low patient numbers.

D: Developmental – Indications have been classified as D when the experience is limited, and additional research is needed to define the role of HSCT.

GNR = Generally not recommended – The GNR category comprises a variety of clinical scenarios in which the use of HSCT cannot be recommended to provide a clinical benefit to the patient, including early disease stages when results of conventional treatment do not normally justify the additional risk of a HSCT, very advanced forms of a disease in which the chance of success is so small that does not justify the risks for patient and donor, and indications in which the transplant modality may not be adequate for the characteristics of the disease.

Appendix 3: Critical Appraisal of Included Publications

Note that this appendix has not been copy-edited.

Table 4: Strengths and Limitations of Clinical Studies Using the Downs and Black Checklist13

Item

Burt et al., 201916

Mancardi et al., 201517

Alping et al., 202118

Zhukovsky et al., 202119

Boffa et al., 202020

Mariottini et al., 201921

Reporting

1. Is the hypothesis/aim/objective of the study clearly described?

Yes

Yes

Yes

Yes

Yes

Yes

2. Are the main outcomes to be measured clearly described in the Introduction or Methods section?

Yes

Yes

Yes

Yes

Yes

Yes

3. Are the characteristics of the patients included in the study clearly described?

Yes

Yes

Yes

Yes

Yes

Yes

4. Are the interventions of interest clearly described?

Yes

Yes

Yes

Yes

Yes

Yes

5. Are the distributions of principal confounders in each group of subjects to be compared clearly described?

NA – RCT, characteristic balanced between groups

NA – RCT, characteristic balanced between groups

Yes

Yes

Yes

NA –characteristic balanced between groups

6. Are the main findings of the study clearly described?

Yes

Yes

Yes

Yes

Yes

Yes

7. Does the study provide estimates of the random variability in the data for the main outcomes?

Yes – 95% CI provided

Yes – 95% CI provided

Yes – 95% CI provided

Yes – 95% CI provided

Yes – 95% CI provided

Yes – interquartile range provided

8. Have all important adverse events that may be a consequence of the intervention being reported?

Yes

Yes

Yes – mainly reported on safety data

Yes

Yes

Yes

9. Have the characteristics of patients lost to follow-up been described?

No

NA – no patients lost to follow-up

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

10. Have actual P values been reported (e.g., 0.035 rather than < 0.05) for the main outcomes except where the P value is less than 0.001?

Yes

Yes

Yes

Yes

Yes

Yes

External validity

11. Were the subjects asked to participate in the study representative of the entire population from which they were recruited?

Probably no – from 4 centres

No – small sample size

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

12. Were the subjects who were prepared to participate representative of the entire population from which they were recruited?

Probably no – only 12% included

No

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

13. Were the staff, places, and facilities where the patients were treated, representative of the treatment the majority of the patients receive?

Yes

Yes

Yes

Yes

Yes

Yes

Internal validity – bias

14. Was an attempt made to blind study subjects to the intervention they have received?

No – not possible

No – not possible

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

15. Was an attempt made to blind those measuring the main outcomes of the intervention?

No – not possible

No – not possible

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

16. If any of the results of the study were based on “data dredging”, was this made clear?

NA

NA

NA

NA

NA

NA

17. In trials and cohort studies, so the analyses adjust for different lengths of follow-up of patients, or in case-control studies, is the time period between the intervention and outcome the same for cases and controls?

Yes

Yes

Yes

Yes

Yes

Yes

18. Were the statistical tests used to assess the main outcomes appropriate?

Yes

Yes

Yes

Yes

Yes

Yes

19. Was compliance with the intervention/s reliable?

NA

NA

NA

NA

NA

NA

20. Were the main outcome measures used accurate (valid and reliable)?

Yes

Yes

Yes

Yes

Yes

Yes

Internal validity – confounding (selection bias)

21. Were the patients in different intervention groups (trials and cohort studies) or were the cases and controls (case-control studies) recruited from the same population?

Yes

Unclear

Yes – From the Swedish Multiple Sclerosis Register

Yes

Yes

Yes

22. Were study subjects in different intervention groups (trial and cohort studies) or were the cases and controls (case-controls studies) recruited over the same period of time?

Yes – between 2005 to 2016

Yes

Yes

Yes – between 1 January 2011 to 31 December 2018

Unclear

Yes – between January 2010 to June 2016

23. Were study subjects randomized to intervention groups?

Yes

Yes

No

No

No

No

24. Was the randomized intervention assignment concealed from both patients and health care staff until recruitment was complete and irrevocable?

Unclear

Unclear

NA

NA

NA

NA

25. Was the adequate adjustment for confounding in the analyses from which the main findings were drawn?

NA – RCT

NA – RCT

No

No

Yes – multivariate analysis

No

26. Were losses of patients to follow-up taken into account?

No – no ITT analysis

NA – all patients were follow-up

NA – retrospective study

NA – retrospective study

NA – retrospective study

NA – retrospective study

27. Did the study have sufficient power to detect a clinically important effect where the P value for a difference being due to chance is less than 5%?

Yes – sample size determination

Yes – sample size determination

No – no sample size determination

No – no sample size determination

No – no sample size determination

No – no sample size determination

ITT = intention-to-treat; NA = not applicable; RCT = randomized controlled trial.

Table 5: Strengths and Limitations of Guidelines Using AGREE II14

Item

EBMT, Sharrack et al., 202012

ASBMT, Cohen et al., 201922

Domain 1: Scope and Purpose

1. The overall objective(s) of the guideline is (are) specifically described.

Yes

Yes

2. The health question(s) covered by the guideline is (are) specifically described.

Yes

Yes

3. The population (patients, public, etc.) to whom the guideline is meant to apply is specifically described.

Yes

Yes

Domain 2: Stakeholder Involvement

4. The guideline development group includes individuals from all relevant professional groups.

Yes

Yes

5. The views and preferences of the target population (patients, public, etc.) have been sought.

Unclear

Unclear

6. The target users of the guideline are clearly defined.

Yes

Yes

Domain 3: Rigour of Development

7. Systematic methods were used to search for evidence.

NR

NR

8. The criteria for selecting the evidence are clearly described.

NR

Yes

9. The strengths and limitations of the body of evidence are clearly described.

Yes

No

10. The methods for formulating the recommendations are clearly described.

NR

NR

11. The health benefits, side effects, and risks have been considered in formulating the recommendations.

Yes

Yes

12. There is an explicit link between the recommendations and the supporting evidence.

Probably yes

Probably yes

13. The guideline has been externally reviewed by experts before its publication.

Yes

Yes

14. A procedure for updating the guideline is provided.

Yes

Yes

Domain 4: Clarity of Presentation

15. The recommendations are specific and unambiguous.

Yes

Yes

16. The different options for management of the condition or health issue are clearly presented.

Yes

Yes

17. Key recommendations are easily identifiable.

Yes

Yes

Domain 5: Applicability

18. The guideline describes facilitators and barriers to its application.

Yes

No

19. The guideline provides advice and/or tools on how the recommendations can be put into practice.

Yes

No

20. The potential resource implications of applying the recommendations have been considered.

Yes

No

21. The guideline presents monitoring and/or auditing criteria.

Yes

No

Domain 6: Editorial Independence

22. The views of the funding body have not influenced the content of the guideline.

Unclear

Unclear

23. Competing interests of guideline development group members have been recorded and addressed.

Yes

Yes

AGREE II = Appraisal of Guidelines for Research and Evaluation II; ASBMT = American Society for Blood and Marrow Transplantation; EBMT = European Group for Blood and Marrow Transplantation; NR = not reported; NA = not applicable.

Appendix 4: Main Study Findings and Authors’ Conclusions

Note that this appendix has not been copy-edited.

Summary of Findings of Included Primary Clinical Studies

Main study findings

Burt et al., 201916

Non-myeloablative hematopoietic stem cell therapy (HSCT) (n = 55) versus DMT (n = 55)

Primary end point

Disease progression (EDSS score increase of ≥ 1):

Median time to progression:

Proportion of patients with disease progression:

Secondary end points

Relapse in the first year:

Proportion of patients with relapse:

Mean change in EDSS score from baseline to 1 year:

Mean change in NRS score from baseline to 1 year:

Mean change in MRI T2-weighted lesion volume from baseline to 1 year:

Mean change in timed 25-feet walk from baseline to 1 year:

Mean change in 9-Hole Peg Test from baseline to 1 year:

Mean change in Paced Auditory Serial Addition Test from baseline to 1 year:

Mean change in MSFC score from baseline to 1 year:

Mean change in SF-36 quality of life score from baseline to 1 year:

Post-hoc analysis

Median time to first relapse:

Proportion with NEDA:

- DMT:

Outcomes of patients in the DMT groups who crossed over to HSCT:

Adverse events:

- HSCT:

- DMT:

Rate of infection per patient per year:

Author’s conclusion

“In this preliminary study of patients with relapsing-remitting MS, nonmyeloablative HSCT, compared to DMT, results in prolonged time to disease progression. Further research is needed to replicate these findings and to assess long-term outcomes and safety.”16 (p. 165)

Main study findings

Mancardi et al., 201517

Myeloablative AHSCT (n = 9) versus MTX (n = 12)

MRI outcomes for 17 patients (AHSCT [n = 8] versus MTX [n = 9]; lost 4 patients [1 in HSCT, 3 in MTX])

- New T2 lesions over 4 years:

Gd + lesions during 4 years:

Clinical outcome, disability, and relapses for 20 out of 21 randomized patients (AHSCT [n = 9] versus MTX [n = 11])

ARR:

Progression at end of follow-up:

EDSS change: No differences between groups at year 1, 2, 3 and 4

Safety (no statistical comparison)

Author’s conclusion

“Intense immunosuppression followed by AHSCT is significantly superior to MTX in reducing MRI activity in severe cases of MS. These results strongly support further phase III studies with primary clinical endpoints.”17 (p. 981)

Main study findings

Alping et al., 202118

Myeloablative AHSCT (n = 139) versus ALZ (n = 132) versus matched patients treated with non-induction therapies (Natalizumab, dimethyl fumarate, rituximab, fingolimod; n = 2,486)

Safety

Mortality:

Thyroid disease:

Non-thyroid autoimmune disease:

Any infection:

Infection diagnosed ≥ 6 months from therapy initiation:

Types of infections:

Author’s conclusion

“We confirmed a higher incidence of thyroid disease in alemtuzumab- and, to a smaller extent, AHSCT-treated patients and found a higher incidence of infection for AHSCT compared to both alemtuzumab and noninduction therapies. The incidence of non-thyroid autoimmune disease was low for both therapies.”18 (p. e1574)

Main study findings

Zhukovsky et al., 202119

Non-myeloablative AHSCT (n = 69) versus ALZ (n = 75)

Primary end point

Secondary end points

Author’s conclusion

“In this observational cohort study, treatment with AHSCT was associated with a higher likelihood of maintaining ‘no evidence of disease activity’. Adverse events were more frequent with AHSCT in the first 100 days, but thereafter more common in patients treated with ALZ.”19 (p. 189)

Main study findings

Boffa et al., 202020

Myeloablative AHSCT (n = 25) versus ALZ (n = 32)

Primary end point

Secondary outcomes

Safety

Author’s conclusion

“Alemtuzumab and AHSCT are effective treatment choices for aggressive multiple sclerosis. AHSCT seems to be superior to alemtuzumab in inducing complete disease control and on promoting short-term disability improvement.”20 (p. 2047)

Main study findings

Mariottini et al., 201921

Myeloablative AHSCT (n = 11) versus DMT (n = 41) in patients withdrew from natalizumab (NTZ) treatment

Safety/AEs after AHSCT

NEDA at 3 years of follow-up:

ARR (clinical relapses):

Disability worsening:

Author’s conclusion

“These data suggest that an aggressive therapy should be established after NTZ with the shortest possible washout period. AHSCT after 6 months from NTZ withdrawal appears to be safe.”21 (p. 624)

Table 6: Summary of Recommendations in Included Guidelines

Recommendations and supporting evidence

Quality of evidence and strength of recommendations

EBMT, Sharrack et al., 202012

Recommendations of AHSCT for MS:

  • “AHSCT should be offered to patients with RRMS with high clinical and MRI inflammatory disease activity (at least 2 clinical relapses, or one clinical relapse with Gd-enhancing or new T2 MRI lesions at a separate time point, in the previous 12 months) despite the use of one or more lines of approved DMTs. Evidence best supports treatment in patients who are able to ambulate independently (EDSS 5.5 or less), who are younger than 45 years and have disease duration less than 10 years.”12 (p. 289)

Level of evidence: I

Strength of recommendation: S

  • “Patients with ‘aggressive’ MS, who develop severe disability in the previous 12 months, are suitable candidates for AHSCT. Given the potential for irreversible disability, such patients may be considered even before failing a full course of DMT.”12 (p. 289)

Level of evidence: II

Strength of recommendation: CO

  • “Patients with SPMS should be considered for AHSCT, preferably in a prospective clinical trial, only when inflammatory activity is still evident (clinical relapses and Gd-enhancing or new T2 MRI lesions) with documented disability progression in the previous 12 months.”12 (p. 289)

Level of evidence: II

Strength of recommendation: CO

  • “Patients with PPMS should be considered for AHSCT, preferably in a prospective clinical trial, only when inflammatory activity is still evident (Gd-enhancing or new T2 MRI lesions) with documented disability progression in the previous 12 months.”12 (p. 289)

Level of evidence: II

Strength of recommendation: CO

  • “Paediatric patients with MS who have breakthrough inflammatory disease with less toxic treatments may be considered for AHSCT.”12 (p. 289)

Level of evidence: II

Strength of recommendation: CO

ASBMT, Cohen et al., 201922

Position Statement:

The ASBMT Task Force recommends revising the recommended indication for AHCT in MS to ‘standard of care, clinical evidence available’, for patients with relapsing forms of MS (RRMS or progressive MS with superimposed activity) who have prognostic factors that indicate a high risk of future disability, including ongoing clinical relapse or MRI lesion activity despite treatment with available DMTs, especially if disease activity continues despite treatment with high-efficacy DMTs and/or worsening disability.”22 (p. 851)

None

AHCT = autologous hematopoietic cell transplantation; AHSCT = autologous hematopoietic stem cell transplantation; ASBMT = American Society for Bone Marrow Transplantation; DMT = disease-modifying therapy; EDSS = Expanded Disability Status Scale; Gd = gadolinium; MRI = MRI; MS = multiple sclerosis; PPMS = primary-progressive MS; RRMS = relapsing-remitting MS; SPMS = secondary-progressive MS.

Appendix 5: References of Potential Interest

Note that this appendix has not been copy-edited.

Previous CADTH Reports

1.Stem cell transplant for multiple sclerosis: clinical effectiveness, cost-effectiveness, and guidelines (CADTH Rapid response report: reference list). Ottawa (ON): CADTH; 2020: https://www.cadth.ca/sites/default/files/pdf/htis/2020/RA1150%20Stem%20Cell%20Transplant%20for%20MS%20Final.pdf. Accessed 2021 Apr 22.

Additional References

2.Burt RK, Tappenden P, Han X, et al. Health economics and patient outcomes of hematopoietic stem cell transplantation versus disease-modifying therapies for relapsing remitting multiple sclerosis in the United States of America. Mult Scler Relat. 2020;45:102404. PubMed

3.Miller AE, Chitnis T, Cohen BA, et al. autologous hematopoietic stem cell transplant in multiple sclerosis: recommendations of the National Multiple Sclerosis Society. JAMA Neurol. 2021;78(2):241-246. PubMed

4.Laureys G, Willekens B, Vanopdenbosch L, et al. A Belgian consensus protocol for autologous hematopoietic stem cell transplantation in multiple sclerosis. Acta Neurol Belg. 2018;118(2):161-168. PubMed

5.Ge F, Lin H, Li Z, Chang T. Efficacy and safety of autologous hematopoietic stem-cell transplantation in multiple sclerosis: a systematic review and meta-analysis. Neurol Sci. 2019;40(3):479-487. PubMed