Journal Information
Vol. 43. Issue 1.
Pages 65-86 (January - March 2021)
Share
Share
Download PDF
More article options
Visits
6302
Vol. 43. Issue 1.
Pages 65-86 (January - March 2021)
Review article
Open Access
A review of hematopoietic stem cell transplantation for autoimmune diseases: multiple sclerosis, systemic sclerosis and Crohn's disease. Position paper of the Brazilian Society of Bone Marrow Transplantation
Visits
6302
Maria Carolina Oliveiraa, Juliana Bernardes Eliasa, Daniela Aparecida de Moraesa, Belinda Pinto Simõesa, Morgani Rodriguesb, Andreza Alice Feitosa Ribeirob, Lilian Piron-Ruizc, Milton Arthur Ruizc, Nelson Hamerschlakb,
Corresponding author
hamer@einstein.br

Corresponding author at: Unidade de Transplante de Medula Óssea do Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/520, São Paulo, SP CEP: 05652-900, Brazil.
a Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
b Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
c Associação Portuguesa de Beneficência de São José do Rio Preto, São José do Rio Preto, SP, Brazil
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (6)
Table 1. Hematopoietic stem cell transplants for autoimmune diseases according to the European Society for Blood and Marrow Transplantation (EBMT).1
Table 2. Results of studies on multiple sclerosis disease.
Table 3. Results of studies on systemic sclerosis disease.
Table 4. Results of Crohn's disease study outcomes.
Show moreShow less
Abstract

Autoimmune diseases are an important field for the development of bone marrow transplantation, or hematopoietic stem cell transplantation. In Europe alone, almost 3000 procedures have been registered so far. The Brazilian Society for Bone Marrow Transplantation (Sociedade Brasileira de Transplantes de Medula Óssea) organized consensus meetings for the Autoimmune Diseases Group, to review the available literature on hematopoietic stem cell transplantation for autoimmune diseases, aiming to gather data that support the procedure for these patients. Three autoimmune diseases for which there are evidence-based indications for hematopoietic stem cell transplantation are multiple sclerosis, systemic sclerosis and Crohn's disease. The professional stem cell transplant societies in America, Europe and Brazil (Sociedade Brasileira de Transplantes de Medula Óssea) currently consider hematopoietic stem cell transplantation as a therapeutic modality for these three autoimmune diseases. This article reviews the evidence available.

Keywords:
Multiple sclerosis
Crohn disease
Systemic scleroderma
Diffuse scleroderma
Bone marrow transplantation
Full Text
Introduction

Autoimmune diseases have become an important field for the development of hematopoietic stem cell transplantation (HSCT). The European Society for Blood and Marrow Transplantation (EBMT) recently published an update of transplant indications for autoimmune diseases, with a total of 2809 cases registered at that point. The cases are summarized in Table 1.1

Table 1.

Hematopoietic stem cell transplants for autoimmune diseases according to the European Society for Blood and Marrow Transplantation (EBMT).1

Autoimmune diseases  n
Multiple sclerosis  Total:  1415 
Connective tissue diseases  Total:  771 
Systemic sclerosis  609   
Systemic lupus erythematosus  110   
Polymyositis-dermatomyositis  17   
Sjogren's syndrome   
Antiphospholipid syndrome   
Other connective tissue diseases  25   
Rheumatoid arthritis (inflammatory arthritis)  Total:  190 
Inflammatory bowel diseases  Total:  200 
Crohn's disease  179   
Celiac disease  16   
Ulcerative colitis   
Other inflammatory bowel diseases   
Hematologic diseases  Total:  56 
Immune thrombocytopenia  28   
Autoimmune hemolytic anemia  13   
Evans’ syndrome  10   
Other hematological diseases   
Vasculitis  Total:  48 
Granulomatous and polyangiitis  10   
Behcet's disease   
Takayasu's arteritis   
Polyarteritis   
Churg Strauss syndrome   
Other vasculitis  21   
Other neurological diseases  Total:  109 
Insulin dependent diabetes  Total:  20 
Other autoimmune diseases  Total:  23 
Total    2809 

Although autologous HSCT (AHSCT) has been used worldwide to treat autoimmune diseases, with slightly different approaches among centers, the main strategy remains similar. Briefly, the procedure consists of a first phase, when autologous hematopoietic stem cells are harvested and cryopreserved, and a second phase, including conditioning regimen and stem cell infusion. Patients undergo mobilization of hematopoietic stem cells from the bone marrow to peripheral blood through a combination of drugs including filgrastim (a granulocyte colony stimulating factor, G-CSF), followed by cell harvesting by leukapheresis and cryopreservation. Most transplant centers use 2–4g/m2 cyclophosphamide and granulocyte colony-stimulating factor (G-CSF) for subsequent collection of hematopoietic progenitor cells.2–13 The scheme allows for collection of more cells than the use of G-CSF as a single agent, and very few events of mobilization failure are described. Additionally, the administered cyclophosphamide promotes initial control of disease activity, contributing to the therapeutic effect of the transplant procedure.10,11,13

At the second phase of HSCT, patients receive immunoablative doses of a lymphoablative conditioning regimen that may include chemotherapeutic agents, immunotherapeutic agents and/or irradiation, followed by autologous cell infusion. A period of aplasia follows, after which there is engraftment and reconstitution of a tolerant and no longer self-reactive immune system.10,14 The purpose of HSCT in autoimmune diseases is to promote immune depletion, eliminate autoreactive lymphocytes, and to reprogramme the immune system, leading to the reestablishment of immune tolerance and, thereby, prolonged disease control.15

The Brazilian Society for Bone Marrow Transplantation (Sociedade Brasileira de Transplantes de Medula Óssea, SBMTO) organized consensus meetings for the Autoimmune Diseases Group, to review the available literature on HSCT for autoimmune diseases, aiming to gather evidence that support the procedure for these patients. Three of the most important indications for HSCT, sufficiently evidence-based, are multiple sclerosis (MS), systemic sclerosis (SSc) and Crohn's disease. As the following sections show, the American Society for Transplantation and Cellular Therapy (ASTCT), the European Society (EBMT), and the Brazilian SBMTO currently consider HSCT as part of the already consolidated treatment procedures for these three autoimmune diseases, outside research trial settings.

Multiple sclerosis

Multiple sclerosis (MS) is a demyelinating chronic inflammatory disease of the central nervous system that mainly affects young adults, and has heterogeneous clinical and pathological characteristics resulting from different pathways of tissue injury.16 Inflammation, demyelination, and axonal degeneration are the main pathological mechanisms that cause clinical manifestations.17 However, the cause of MS is unknown18,19 and it is considered an incurable disease. The most widely accepted theory is that MS begins as an autoimmune inflammatory reaction mediated by autoreactive lymphocytes.16,20 Microglia activation and chronic neurodegeneration occur later. Regardless of the immune pattern of early demyelination, analyzes of active lesions over time suggest that there is a single immuno-effector mechanism in each person.21

MS begins at the median age of 23.5 years, and mean age of 30 years.22 Mortality among patients with MS is not much different from the general population, but neurological deficits progress in all patients. It is expected that in 15 years from diagnosis, 50% of patients will need help walking, and after 25 years, most cannot walk anymore.23,24

MS symptoms are non-specific and may include paresthesias and paresis due to transverse myelitis, ataxia due to cerebellar injury, tremor, monocular visual loss due to optic neuritis, double vision due to brainstem dysfunction, fatigue and sphincter changes. If there is a relapse episode, the functional disability scale score used in neurological examination increases, indicating worsening. During remission, the patient remains stable within his or her disability score. Numerous scales have been described to evaluate MS, the most used being the Expanded Disability Status Scale (EDSS), described by Kurtzke.25

MS is classified in three clinical types, which receive different treatment approaches. The first is the relapsing remitting type (RRMS), resulting from inflammatory multifocal lesions that cause clinical recurrence and activity of detectable lesions on magnetic resonance imaging (increase or appearance of new lesions on hyperintense T2 or increase of gadolinium on T1 lesions). It manifests with episodic relapses followed by partial or total recovery from dysfunction, interspersed with remission periods of at least 30 days.23,24

After an average time of 10–15 years, the episodes become less frequent, followed by less evident recovery, with accumulation of sequelae and gradual worsening of the neurological picture that is typical of the secondary progressive form (SPMS). This is characterized by neurodegeneration, but may still have superimposed inflammatory activity.23,24

The third clinical type of MS, the primary progressive MS (PPMS), occur in about 10–15% of patients, at the mean age onset of approximately 40 years, and occurs equally in males and females. It is characterized by progressive accumulation of disability from disease onset. Can occur occasional plateaus, minor and temporary improvements but after all continues progressing. The diagnosis is made based only on patient's history. This “malignant form” or “aggressive form” of MS, is characterized by rapidly progressive course, leading to significant disability, or even death, in multiple neurologic systems in a relatively short time after disease onset.23,24

The worsening of symptoms is highly heterogeneous among patients with MS. The extent of inflammatory activity affects MS prognosis substantially. Because of that, the goal of treatment approaches is the absence of disease activity clinically or at magnetic resonance imaging and the improvement of disability or of the EDSS score. This is called “no evidence of disease activity” (NEDA).26

Conventional therapeutic approaches for multiple sclerosis

As of December 2017, the US Food and Drug Administration (FDA) has approved 15 drugs considered as capable of modifying MS course: 5 interferon-beta preparations; two glatiramer acetate preparations; the monoclonal antibodies natalizumab, alemtuzumab, daclizumab and ocrelizumab (the first B-cell targeted therapy); the chemotherapeutic agent mitoxantrone; and small molecule oral agents fingolimod, dimethyl fumarate and teriflunomide. Dalfampridine (aminoperidine) has been approved as a therapy to treat and improve gait speed.21 However, none of these are curative therapies. They are partially effective in reducing relapse rates and disease progression. Furthermore, all disease-modifying therapies present safety concerns, with the risk increasing over time. Better treatment strategies are required for patients with relapsing MS, for those who present inflammatory activity despite the treatment used and for those with treatment side effects.27

Transplant in multiple sclerosis

Since the mid-1990s, with the first studies on animal models with subsequent clinical application, autologous transplantation (AHSCT) has been an important tool in inducing an “immunotolerant” immune reconstitution.2,3 Thousands of transplants for treating MS have been performed worldwide, with more than 700 evaluated in studies.6,28–46 Some studies showed progression-free survival of more than five years,39,43 with superior neurological improvement in patients with relapsing-remission type and in those with inflammatory activity observed on magnetic resonance imaging.43,47,48Table 2 shows the results of the main studies focused on the use of HSCT to treat MS: there are 8 retrospective studies and 6 single-arm clinical trials on AHSCT for MS, and only 2 randomized clinical trials focusing on the comparison of AHSCT versus disease-modifying therapies for MS. There are also 3 meta-analysis and systematic reviews and one position paper.6,9,10,39,49–57

Table 2.

Results of studies on multiple sclerosis disease.

Site/year of publication(ref)  Study design  Comparisons  Endpoints  Inclusion criteria/  N  Transplant regimen  PFS/outcomes  Relapse/progression rate  Non-relapse mortality  OS  Follow-up 
Retrospective studies
Italy 201249  Retrospective case series  None  EDSS progression free-survival  MS treated previously with DMT with severe clinical course past year (EDSS worsening1.0)  74(33 RRMS and 41 SPMS)  BEAM+rabbit ATG  EDSS PFS at 5y 66% for all patientsRRMS EDSS PFS 5y 71%  30% for RRMS vs 10% for SPMS  2.7%  95.9%  Median 4 years (8 mo–10.5 yrs) 
Swedish 201456  Retrospective case series  None  Relapse free survival, MRI event free survival, EDSS PFS and DFS  MS treated previously with DMT  52 but analyzed 41(40 RRMS, 5 SPMS, 2 PPMS,)  BEAM+ATG in 41 and Cy (200mg/kg)+ATG in 7  At 5y: EDSS PFS 77%; clinical relapse free survival 87%; MRI event-free survival 85% and DFS: 68%  At 5 years, 4 patients had clinical relapse, 5 had MRI activity, 8 had EDSS progression  Zero TRM  100%  Mean 4.0 years (1–9 yrs) 
Northwestern/Chicago 201555  Retrospective case series  None  Time to EDSS improvement and time to EDSS worsening both at least 1.0 point  RRMS failed DMT, with 2 or more treated relapses or 1 treated relapse with Gd lesion at a separate time/RRMS 123, SPMS 28  151 but 145 analyzed  CY 200mg/kg+alentuzumab 20mg or rabbit ATG 6mg/kg  EDSS improved 50% at 2 years, 64% at 4 yearsRelapse-free survival was 89% at 2 years and 80% at 4 years;PFS was 92% at 2 years and 87% at 4 years and disease activity–free survival was 80% and 68% respectively  The proportion of patients with a 1.0 or greater change in EDSS score was 10% in 19 of 112 patients with an indication of progression at 1 year and11% in 9 of 82 for progression at 2 years; and 11% in 7 of 64 for progression at 3 years;and 8% in 3 of 36 for progression at 4 years; and5% in 4 of 27 for progression at 5 years.  Zero TRM  99.3% (1 death due hypertensive cardiovascular disease)  2,5 years (6 month–5 yrs) 
CIBMTR/EBMT Review53  Retrospective registry review  None  PFS  Data base of AHSCT for MS/46 RRMS, 186 SPMS, 32 PPMS, 17 PRMS  RRMS failed DMT, with 2 or more treated relapses or 1 treated relapse with Gd lesion at a separate time/RRMS 123, SPMS 28  High intensity in 53/281 (18.9%); intermediate intensity in 49/281 (17.4%); low intensity in 49/281 (17.4%).  Overall PFS was 46%; among patients with RRMs/PRMS PFS was 82% at 3 years andEDSS PFS 73% at 5 years    2.8%  93% at 5 years  Median 6.6 yrs 
Czech Republic 201039  Retrospective case series  None  Reported efficacy of AHSCT experience in MS  All patients treated with AHSCT 10 years11 RRMS, 15 SPMS  Data base of AHSCT for MS/46 RRMS, 186 SPMS, 32 PPMS, 17 PRMS  BEAM in 26 and BEAM+ATG in 16  PFS at 3 years 70.8% and 29.2% at 6 years  Median annual relapse rate within the year before ASCT was 2, while the median annual relapse rate was 0 within the first 2 years after ASCT  Zero  96.1% (1 death due to glioblastoma multiforme at 60 months of follow-up)  Median 66 months (11–132) 
Spain 201757  Retrospective case series  None  Reported toxicity and the long-term efficacy of AHSCT  RRMS/SPMS under treatment with one of the MS-approved drugs for more than 1 year, who experienced one or more relapses in the previous year and worsening of at least 1 point in disability (EDSS)22 RRMS, 9 SPMS  31  BEAM+rabbit ATG  100% of RRMs were free of disability at 6 months; and 60% achieving sustained disability recovery at 6 months;22% of SPMS were free of disability at 6 months; and 10% achieving sustained disability recovery at 6 months  32.3% (10) had at least one relapse post-AHSCT; 6 in the RRMS group (27.2%) and 4 in the SPMS group (44.4%). 7 (22.6%) experienced progression of disability, all within SP form.  Zero  96.7% (1 death 13 years after transplant from aspiration pneumonia after progressing and reaching an EDSS of 9.5.)  Median 8.4 years (2–16) 
Single-arm prospective study
Russia 201558  Single arm clinical trial  None  Safety and efficacy (treatment response)  All patients treated with AHSCT 6 years: EDSS 1.5–8.0, ±Gd lesions, and no treatment with interferons or immunosuppressive agents within 3 months before enrollment  99(43 RRMS, 56 SPMS, 18 PPMS, 3 PRMS)  Reduced intensity conditioning regimen based on BEAM  EFS for the whole group was 80%. In the group with RRMS, EFS was 83.3% and in the group with progressive course 75.5%92% EDSS PFS 5 years  16.7% at 8 years.  Zero  100%  Median 48.9 months 
Northwestern/Chicago 200959  Single arm clinical trial  None  EDSS PFS and reversal of neurologic disability  RRMS with 2 steroid treated relapses in previous 12 month.  21(21 RRMS)  Cychophosphamide+alemtuzumab/rabbit ATG  100% EDSS PFS 3 years  5 patients (24%) relapsed but achieved remission after further immunosuppressionAfter a mean of 37 months (range 24–48 months), all patients were free from progression (no deterioration in EDSS score), and 16 were free of relapses  Zero  100%  Mean 3.1 years (1.5–10) 
Canada 201662  Single arm clinical trial  None  MS activity-free survival (absence of clinical relapse, new MRI lesion or progression of EDSS)  Disease activity despite 1 year of DMT.  24(12 RRMS, 12 SPMS)  Cyclophosphamide/busulfan  70% PFS 3 years  No clinical relapse  4.2%  95%  Median 6.1 year (±2.5) 
HALT-MS 2015, 201760,61  Single arm clinical trial  None  Time to treatment failure (death or MS activity)  RRMS with failure of DMTs during the prior 18 months  24(24 RRMS)  BEAM+rabbit ATG  70% NEDA 5 years, 91% EDSS PFS  Two participants had disease progression and died at .2.5 years and .3.5 years after AHSCT; a third participant also had disease progression at 15 months and died at 4.5 years post-HCTOf 24 patients 7 did not maintain EFS by close of follow-up by an increase in EDSS 0.5 (n=2), clinical relapse (n=3) or development of new MRI lesions (n=2)  Zero  87.5%  Median 4.9 year (6–12) 
Australia 201863  Single arm clinical trial  None  EFS (NEDA)  RRMS with at least 1 relapse or one new MRI lesion in the past year despite DMT/SPMS worsening with at least 1 MRI lesion in the past year.  35(20 RRMS, 15 SPMS)  BEAM+horse ATG  60% NEDA 3 year  Clinical relapses occurred in 3 patients at 12, 13 and 14 months, respectively after AHSCT  Zero  100%  Median 6.9 year (0.7–21.6) 
Randomized study
ASTIMS 201552  Phase II, AHSCT vs mitoxantrone  AHSCT vs mitoxantrone  Cumulative number of new T2 lesion MRI 4 years after randomization  Worsening in EDSS and one or more MRI lesion last year despite DMTs  21 (9 to AHSCT)7 RRMS, 13 SPMS, 1 PPMS  BEAM+ATG  New T2 lesion MRI: 2.5 AHSCT vs 8 mitoxantrone 4 years  EDSS progression was 57% in AHSCT vs 48% in the mitoxantrone (p=0.5)  Zero  100%  4 years 
MIST 201855  Phase3 AHSCT vs conventional DMT  AHSCT vs DMT  6-Month worsening EDSS1.0 after one year's post-AHSCT or DMT treatment  At least 2 clinical relapse, or one relapse in MRI lesion at different time in the last year despite DMT  110 (55 AHSCT)110 RRMS  Cyclophosphamide+rabbit ATG  At 1 year, mean EDSS improved from 3.38% to 2.36% with AHSCT and worsened from 3.31 to 3.98 with DMTs. At 1 year, mean T2 lesion volume on MRI decreased in the AHSCT, but increased in The DMT group.  15.4% AHSCT vs 85.3% of relapse at 5 years. Disability worsening occur in 5.8% AHSCT vs 66.7% DMT group; median time to pression 24 months in the DMT.  Zero  100%  Up to 5 years 

N: number of enrolled subjects; PFS: progression-free survival; OS: overall survival; EDSS: Expanded Disability Status Scale; TRM: transplant-related mortality; FU: follow-up; AHSCT: autologous hematopoietic stem cell transplantation; SPMS: secondary progressive MS; PPMS: primary progressive MS; PRMS: progressive relapsing; DMT: disease modifying treatment; NEDA: no evidence of disease activity (absence of relapse, disability worsening on the EDSS or MRI lesion activity); MRI: magnetic resonance imaging; Gd: gadolinium-enhanced lesion on MRI.

These studies together support the efficacy of AHSCT in patients with relapsing form of MS. Although they differed in design, population, conditioning protocol and only two were randomized with a control group, many patients experience disease activity control for a long time. The largest retrospective study was a review from the CIBMTR/EBMT register in 2017 that analyzed 281 patients.53 Overall progression-free survival rate was 46%; among patients with RRMS or PRMS, progression-free survival rates were 82% at 3 years and 73% at 5 years; OS at 5 years was 93%. Factors associated with neurologic progression post AHCT was older age, progressive versus relapsing form of MS and more than two previous disease modifying therapies (DMTs).

All the single-arm35,58–63 clinical trials demonstrated high efficacy of AHSCT for RRMS or PRMS. The first reported was by Burt et al., with 21 patients with RRMS conditioned with cyclophosphamide plus rabbit ATG/alemtuzumab, showing a EDSS progression free survival at 3 years of 100% and EDSS improvements ≥1 in 81% and no change in 9.5%, OS of 100%.59 Recently Moore at al. reported 35 patients with RRMS and SPMS in a phase 2 study conditioned with BEAM and horse ATG showing a MS activity-free survival of 82% at one year 65% at 2 years and 60% at 3 years; looking at RRMS only the MS activity-free survival was 90% at one year and 70% at 3 years.63

Only two randomized controlled trials are available: in the ASTIMS study, Mancardy et al. compared AHSCT versus mitoxantrone; 9 out 21 were randomized to AHSCT.52 They were conditioned with BEAM plus rabbit ATG. Over 4 years, the median number of new T2-weighted MRI lesions was 2.5 in the AHSCT versus 8 in the mitoxantrone group (p=0.00016) and none of those who received AHSCT had new gadolinium-enhanced MRI lesions, but 56% of those on mitoxantrone had at least 1 MRI lesion (p=0.029); EDSS progression was 57% in AHSCT vs 48% in the mitoxantrone group (p=0.50). The phase 3 MIST trial55 compared the efficacy of AHSCT versus DMTs. The conditioning was non-myeloablative, with cyclophosphamide plus rabbit ATG and 110 patients were randomized 1:1. With a follow-up of up to 5 years, disability worsening occurred in 5.8% AHSCT versus 66.7% of DMT group. At 1 year, relapse occurred in 2% in the AHSCT and 68.2% in the DMT group 9; with p<0.001 and at 5 years, relapse occurred in 15.4% AHSCT vs 85.2% DMT group; no deaths or grade 4 toxicities related to transplantation were reported.

A meta-analysis of AHSCT for MS evaluated the safety and efficacy of transplant. Led by Somani at al., it evaluated 15 studies including more than 764 transplanted patients. A proportion of NEDA (no evidence of disease activity) was found in 2 years in 83% and in 5 years 67%; the transplant-related mortality was estimated in 2.3%, the rate of EDDS progression was 17.1% at 2 years and 23.3% at 5 years.54 Another interesting study done by Sormani et al.64 evaluated a comparison of NEDA status in studies of AHSCT (n=66) with various conventional e high-efficacy MS DMTs (n=216). The use of AHSCT led to NEDA rates of 78–83% at 2 years and 60–68% at 5 years. On the other hand, DMTs lead to reported NEDA rates of 13–46% at 2 years.

Transplant-related mortality (TRM) has improved over the past 20 years.14,54 The initial reports described TRM as high as 9.5% (37, revised in 65), which later decreased to 1.3%, according to EBMT data.6,9 The incorporation of less myeloablative regimens, center experience and center accreditation have contributed to the improved outcomes.8,59,65–68

Recently the American Society of Bone Marrow Transplantation50 has conducted a comprehensive literature review, including many of the papers already discussed here, and recommended autologous HSCT as “standard of care, clinical evidence available” for treatment-refractory relapsing MS.

Guidelines for transplantation in multiple sclerosis

Considering the results presented above, HSCT can be considered for patients with relapsing-remitting form of MS, with EDSS less than 6, who have not responded to conventional treatment or progressive MS with superimposed activity. Especially if disease activity continues despite treatment with high-efficacy DMTs and worsening disability. The treatment for other MS types should follow the guidelines proposed by SBMTO previously,69 shown in Box 1 and Box 2, and using the same reduced-intensity conditioning with cyclophosphamide (200mg/kg) and rabbit anti-thymocyte (4.5mg/kg). Intensive vigilance should be maintained for short and long-term post-transplant complications, especially fever, deep vein thrombosis, pulmonary embolism, urinary tract infections, falls, metabolic syndrome and coronary disease.65,70

Box 1.

Indications for autologousa hematopoietic stem cell transplantation (HSCT) in multiple sclerosis (MS).

Patients under 60 years old who are not responsive to the current first line standard therapy and who present EDDS between 3 and 6 
 
Patients with inflammatory activity in the forms:- relapsing-remitting: IB;- secondary progressive with inflammatory activity (clinical and imaging): IIB;- primary progressive with inflammatory activity (clinical and imaging): IIB 
 
Patients with the “malignant” form of multiple sclerosis who developed severe disability in the previous year: IIB 

EDSS: Expanded Disability Status Scale.

a Allogeneic HSCT is not recommended for any category of MS.

Box 2.

Contraindications to hematopoietic stem cell transplantation in multiple sclerosis.

• Advanced disease and no inflammatory activity 
• EDSS>6.0 
• Renal impairment: serum creatinine>2mg/dL (177μmol/L) or creatinine clearance (CrCL)<50. 
• Liver dysfunction: Frank cirrhosis. Other condition related to liver dysfunction (hepatitis, alcohol abuse, hepatic steatosis and iron overload) need consultation with a hepatologist to contraindication 
• Cardiac disease: left ventricular ejection fraction (LVEF) ≤40% for BEAM condition and LVEF ≤50% using chemotherapy regimens with known cardiac toxicity (e.g., cyclophosphamide) or uncontrolled coronary artery disease or uncontrolled arrhythmias. 
• Pulmonary dysfunction: corrected DLCO<50% 
• Poorly controlled chronic diseases (diabetes, hypertension) 
• Pregnancy 
• HIV positivity 
• History of previous malignancy 
• Psychiatric disorders 

EDSS: Expanded Disability Status Scale.

Systemic sclerosis

Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by fibrosis of the skin and internal organs, and vasculopathy.71,72 The disease usually begins with vascular hyperreactivity and endothelial activation, which, associated with inflammatory phenomena, promotes progressive tissue damage and fibrosis.73 The etiology, still not completely understood, includes a susceptible genetic background associated with environmental stimuli, which promote immune dysregulation and, as a consequence, injury to tissues.71,73

Patients with severe and progressive forms of SSc have reduced quality of life and mortality rates that can reach 50% in five years and 70% in 10 years.74 Cardiopulmonary dysfunction is the leading cause of death, especially in patients with interstitial lung disease and pulmonary hypertension.71 A meta-analysis has shown that the mortality of patients with SSc has not decreased in the past 40 years, despite new available treatment options.75

Clinical presentation

Skin involvement is a hallmark of SSc, and its extent enables clinical classification into three subtypes: limited, diffuse and sine scleroderma (without cutaneous involvement). The extent of skin involvement correlates with disease presentation and severity. Patients with the diffuse form of SSc suffer from interstitial lung disease, cardiac and renal involvement more frequently.72 This subtype tends to progress faster and with greater severity, morbidity and mortality than other forms of the disease.72,76,77 On the other hand, limited systemic sclerosis patients tend to present pulmonary hypertension as a life-threatening manifestation.

Conventional therapeutic approaches

The management of patients with systemic sclerosis (SSc) includes pharmacological and non-pharmacological approaches.78 Hand and extremity warming and protection against trauma, dietary and postural measures to reduce gastroesophageal reflux and treatment of skin and digital ulcers are recommended for most patients, given the high frequency of these manifestations.78

Conventional drug treatment is quite limited and ineffective in controlling the progression of the disease. There are no drugs that treat SSc globally, and therapeutic strategies are often organ or manifestation-directed. Currently, international rheumatology societies have developed recommendations for the treatment of patients with SSc, always according to individual manifestations.78,79 Randomized clinical studies have investigated synthetic and biological immunomodulatory agents, showing modest benefits and no repercussions on disease-related mortality, with exception of angiotensin-converting enzyme (ACE) inhibitors, which in the past significantly decreased the mortality of patients with scleroderma renal crises.59,78

Interstitial lung disease is a severe manifestation of SSc and the leading cause of death in this disease. The available treatments have modest and short-lived success rates on the control of interstitial lung disease.71,78–81 Best outcomes are described for cyclophosphamide and mycophenolate mofetil, with short-term benefit when compared with placebo.82,83 More recently, rituximab, tocilizumab, nintedanib and pirfenidone, among other therapeutic agents, have been investigated with promising results, yet to be confirmed.84–87 Isolated diffuse skin involvement, when severe and progressive, is also associated with high mortality rates and poor quality of life, with outcomes comparable to those of patients with visceral involvement.86 Thus, systemic immunosuppression is recommended for selected cases.87

Hematopoietic stem cell transplantation

In the past years 20 years, AHSCT has been indicated as treatment for patients with severe and rapidly progressive SSc.10 According to data from international transplant registries, the number of transplanted patients per year has progressively increased, reflecting the good outcomes of this therapeutic approach.14

The most recently adopted indication criteria for transplantation are the following10,88,89:

  • An established diagnosis of systemic sclerosis, according to the 2013 ACR/EULAR classification criteria90.

  • Patients with diffuse form of the disease, with a minimum modified Rodnan's score of 16, and worsening by at least 25% in the last 6 months, under immunosuppressive treatment.

  • Patients with interstitial lung involvement with a decline in predicted forced vital capacity (FVC) or carbon monoxide (CO) diffusion percentages greater than 10% in the preceding 6 months, while under immunosuppressive treatment.

The exclusion criteria for HSCT to consider based on available studies are the following10,88,89:

  • Age over 60 years;

  • Current pregnancy;

  • Inability to practice effective contraception;

  • Inability to accept infertility as a possible consequence after transplantation;

  • Severe psychiatric disorder;

  • Current acute or chronic infection;

  • Previous malignancy, except localized and treated cervical and thyroid cancer;

  • Major organ dysfunction, as follows:

    • Liver – increased alanine aminotransferase (ALT) or aspartate aminotransferase (AST), over three times the normal range; or increased serum bilirubin levels, over three times the normal range, except when Gilbert's syndrome present;

    • Kidneys – creatinine clearance below 40ml/min; or creatinine levels above 2mg/dL;

    • Lungs – forced vital capacity or hemoglobin-adjusted CO diffusion below 40% of predicted;

    • Heart – left ventricular ejection fraction below 50%, constrictive pericarditis, ventricular arrhythmias, extensive myocardial fibrosis, systolic pulmonary artery pressure greater than 40mmHg or mean pulmonary artery pressure greater than 25mmHg, evidence of ventricular diastolic dysfunction, septal dyssynergia.

Historically, the initial studies were important to establish recommendations and guide currently adopted procedures. Phase I/II studies showed that autologous HSCT (AHSCT) improves skin involvement and at least stabilizes the pulmonary condition (Table 3).11,12,91–96 These studies also evidenced the importance of a proper heart assessment as a strategy to decrease transplant-related mortality. Cardiac dysfunctions that occur during transplantation accounted for many of the transplant-related deaths described in the initial studies.97 Currently, there are detailed cardiac assessment recommendations,98 aimed at identifying patients with previous cardiac lesions and, therefore, at a higher risk of cardiotoxicity.

Table 3.

Results of studies on systemic sclerosis disease.

Site/year of publication(ref)  Study design  Comparisons  Endpoints  Inclusion criteria  N  Transplant regimen  PFS  Relapse/progression rate  Non-relapse mortality  OS 
Single-arm studies
Europe+USA multicenter/200191  Phase I/II  None, single arm  Feasibility, mortality and preliminary response to treatment  Diffuse SSc or limited SSC with interstitial lung disease or pulmonary hypertension  41 (37 transplanted)  Multiple, most CY 150–200mg/kg±ATG/alemtuzumab and/or TBI  46% at 4y  23% (7/30) at 4y  17% (7/41)  73% at 4y 
France multicenter/200293  Phase I/II  None, single arm  Feasibility, tolerance and efficacy  Early (<4y) diffuse SSc plus visceral involvement  12 (11 transplanted)  CY or MelphalanCD34+ selected graft  45% at mean 18 months  50% (5/10) at mean 18 months FU  9% (1/11)  64% at mean 18 months FU 
USA multicenter/2002104  Pilot study  None, single arm  Safety and potential efficacy  Early (<3y) diffuse SSc plus visceral involvement or progressive interstitial lung disease  19  TBI 800cGy+CY 120mg/kg+90mg/kg eATGCD34+ selected graft  63% at mean 15 months  25% (4/16) at mean 15 months  16% (3/19)  79% at mean 15 months 
Europe multicenter/200494  Retrospective analysis of phase I and II studies  All studies were non-comparative, single arm  Safety and efficacy  Early (<3y) diffuse SSc or progressive interstitial lung disease  57  Multiple, most CY 150–200mg/kg±ATG/alemtuzumab and/or TBI  58% at 5y  48% at 5y  8.7% (5/57)  72% at 5y 
USA multicenter/200795  Phase II  None, single arm  Safety and efficacy  Early (<4y) diffuse SSc plus visceral involvement or progressive interstitial lung disease  34  TBI 800cGy+CY 120mg/kg+90mg/kg eATGCD34+ selected graft  64% at 5y  15% (4/26) at 5y  23% (8/34)  64% at 5y 
USA (Chicago)/200796  Phase I  None, single arm  Safety (toxicity and transplant-related mortality)  Diffuse SSc plus visceral involvement or pulmonary disease regardless of skin involvement  10  CY 200mg/kg+7.5mg/kg rATGUnselected graft  70% at mean 25.5 months  22% (2/9) at mean 25.5 months  Zero  90% at mean 25.5 months 
Germany (Berlin)/201212  Retrospective analysis  None, single arm  Safety and efficacy  CY inefficacy or diffuse SSc with bad prognosis  26  CY 200mg/kg+40mg/kg rATG*CD34+ selected graft  53% at 3y  39% (9/23) at 3y  4% (1/26)  74% at 3y 
EUA (Chicago) and Brazil (Ribeirão Preto)/201389  Retrospective analysis  None, single arm  Efficacy and use of cardiac screening  Diffuse SSc plus visceral involvement or pulmonary disease regardless of skin involvement  90  CY 200mg/kg+4.5-6.5mg/kg rATGNo graft selection  70% at 5y  15% (13/85) at 5y  6% (5/90)  78% at 5y 
Italy (Milan)/201711  Phase II  HSCT vs historical cohort  Safety and efficacy  Diffuse SSc with progression of skin or of visceral disease  18 HSCT+36 SSc controls  CY 200mg/kg+7.5mg/kg rATGCD34+ selected graft  Not available.Significantly higher PFS in HSCT than in SSc control group  Not available.Significantly lower disease progression in HSCT than in SSc control group  5.6% (1/18) in HSCT group.Not available for SSc control group  89% in HSCT group and39% in SSc controls, at 5y 
Europe (multicenter) and Brazil (Ribeirão Preto)/2020102  Prospective observational  None, single group  Safety and efficacy  Transplanted patients with progressive SSc with data registered to the EBMT database  80  In 76 patients: median CY 200mg/kg+rATGIn 4 patients: Thiotepa 10mg/kg+CY 100mg/kg+rATGCD34+ selection in 44%  82% at 2y  13% (11/85) in 2y  6.25% (5/80)  90% in 2y 
Randomized studies
ASSIST (Chicago, USA)/201199  Phase II, open label, randomized 1:1Patient cross-over from CY to HSCT arm allowed at 12 months of FU.  HSCT versus 6 monthly IV CY pulses  Improvement in skin score or lung function in 12 months of follow-up  Diffuse SSc plus visceral involvement or progressive interstitial lung disease  19 (10 HSCT arm and 9 CY arm)  CY 200mg/kg+6.5mg/kg rATGUnselected graft  100% in HSCT arm (10 patients) and 11% in CY arm, at 12 months FU.7 patients from CY arm crossed over to HSCT at 12 months FU and all improved.At 2.6y of mean FU after HSCT, PFS was 88%  Zero in HSCT armand 89% (8/9) in CY arm at 12 months of FU.12% (2/17) in all transplanted patients (original and crossed-over) at mean 2.6y of FU  Zero in HSCT and CY arms, at mean 2.6y of FU  100% at mean 2.6y of FU 
ASTIS (Europe multicenter)/201488  Phase III, open label, randomized 1:1  HSCT versus12 monthly CY pulses  Progression/relapse-free survival, toxicity, efficacy.  Early (<4y) diffuse SSc plus major organ involvement or progressive diffuse skin disease (<2y) with no visceral involvement  156 (79 HSCT arm and 77 CY arm)  CY 200mg/kg+7.5mg/kg rATGCD34+ selected graft  77% in HSCT arm and 65% in CY arm, at 4y FY  11% (7/66) in HSCT arm and 35% (20/57) in CY arm at 4y FU  10% (8/79) in HSCT arm and zero in CY arm at 4y FU  80% in HSCT arm and 65% in CY arm at 4y FU 
SCOT (USA multicenter)/2018100  Phase II, open label, randomized 1:1  HSCT versus12 monthly CY pulses  Global rank composite score at 54 months of FU  Any form of early SSc (<5y) plus interstitial lung disease or previous scleroderma renal crisis  55 (33 HSCT arm and 32 CY rm)  TBI 800cGy+CY 120mg/kg+90mg/kg eATGCD34+ selected graft  74% in HSCT arm and 47% in CY arm, at 72 months FU  18% (6/33) in HSCT arm and 41% (14/34) in CY arm, at 54 months FU  6% (2/33) in HSCT arm.Zero in the CY group  86% in HSCT arm and 51% in CY arm, at 72 months FU 

PFS: progression-free survival; OS: overall survival; N: number of enrolled subjects; USA: United States of America; y: years; SSc: systemic sclerosis; TBI: total body irradiation; IV CY: intravenous cyclophosphamide; eATG: equine anti-thymoglobulin; rATG: rabbit anti-thymoglobulin; FU: follow-up; HSCT: hematopoietic stem cell transplantation; rATG*: this center used ATG-Fresenius (Neovii-Biotech, Germany).

The three randomized studies more recently published show that HSCT surpasses conventional treatment in SSc patients, promoting longer overall survival, longer disease-free survival and higher quality of life (Table 3).88,99,100 These results are essential to convince the community about the efficacy of HSCT. In fact, since 2017, AHSCT has been recommended by the European League Against Rheumatism (EULAR) guidelines, for patients with SSc at risk of organ failure.78

Treatment protocols have been refined and incorporated into the routine of several transplant centers. However, a few points of debate still remain. There is still no consensus among centers whether to adopt graft selection or to use non-selected grafts. While CD34+ graft selection may prevent reinfusion of autoreactive cells during transplant and potential disease reactivation, it adds costs to the procedure and increases the risk of contamination and cell death. A retrospective study from the European Blood and Marrow Transplant Group (EBMT) compared SSc patients transplanted with or without graft selection, showing similar clinical outcomes.101 Although not statistically significant, patients who received unselected grafts tended to have better overall and disease-free survival than patients treated with selected grafts. More recently, a second prospective observational study from the EBMT reported, conversely, higher disease improvement rates after HSCT with CD34+ graft selection, although there was no impact on the progression-free survival.102 A third, smaller, recent study has also shown better outcomes in patients transplanted with selected versus non-selected grafts (Table 3).103 In summary, the question shall not be settled without a prospective randomized trial.

Most transplant centers adopt non-myeloablative conditioning regimens, however a group of North American multicenter investigators, from 26 transplant sites, has consistently reported their experience with myeloablative protocols including full body irradiation.95,104 In this scenario, the most recently published randomized trial (SCOT trial) has shown good results in the myeloablative transplant group (Table 3).100

The experience with allogeneic transplantation for systemic sclerosis is limited to isolated case reports.14,105,106 Therefore, clinical evidence is insufficient to recommend this treatment modality for patients with SSc.

Guidelines for transplantation in systemic sclerosis

The three randomized studies already published provide solid evidence to recommend autologous hematopoietic stem cell transplantation for severe and progressive cases of systemic sclerosis. According to the Oxford Center for Evidence-Based Medicine's evidence table,107 we consider the evidence to have Grade A and Level 1B. Patients should be thoroughly evaluated for heart dysfunction and fibrosis before transplantation and the procedure should be performed in centers with experience in managing patients with systemic sclerosis during transplantation.

Crohn's disease

Crohn's disease is a chronic relapse-remitting inflammatory bowel disorder that can affect any site of the digestive tract.108 Currently, Crohn's is a global disease, with increasing worldwide incidence and prevalence.109 In Brazil, the prevalence of Crohn's disease varies according to the region of the country, from 12.8/100,000 in the Northeastern area, to 14.1 and 24.3/100,000 in the states of Espírito Santo and São Paulo, respectively.110–112

The disease is immunologically mediated and heterogeneous among the affected patients. The imbalanced and dysregulated immunity of the intestinal mucosa provides an inappropriate response against original intestinal flora or luminal antigens. These responses are responsible for the symptoms and the intestinal lesions observed in Crohn's disease. Disturbances in the intestinal mucosa occur in patients genetically predisposed and exposed to environmental triggers.113

The mucosal disturbance may be linked to abnormalities in the toll-like recognition receptors (TLRs), which play an essential role in the pathogenesis of numerous autoimmune diseases. The perturbation of the TLR signaling pathway in intestinal macrophages has been associated with tolerance breakdown in autoimmune diseases.114 Patients with Crohn's disease present defects in the innate immune pathway, and inadequate T cell responses to pathogenic mutations in the nucleotide-binding oligomerization domain 2 (NOD2) and autophagy-related protein 16-1 (ATG16L1) genes that impair bacterial sensing and clearing.108 Imbalances between natural (nTreg) and induced regulatory T-cells (iTreg), and effector T-helper (Th) cells (Th1/Th17) that defend the mucosa from bacteria, fungi and viruses are associated with Crohn's disease.108,115 The Genoma Wide Association Study Project identified 71 susceptibility loci on 17 chromosomes and regions related to inflammatory bowel disease, renamed IBD1 to IBD9.116

The age at diagnosis, disease location and behavior along the gastrointestinal tract are defined according to Montreal classification.117 The cardinal symptoms of Crohn's disease include abdominal pain, diarrhea, hematochezia, bloody stools, fatigue, weight loss, fever, recurrent fistulas, and extraintestinal manifestations.118 Extraintestinal manifestations are common, such as arthropathy (both axial and peripheral), ocular involvement (uveitis, scleritis, and episcleritis), dermatological (including pyoderma gangrenosum and erythema nodosum), nephrolithiasis, hepatobiliary involvement (primary sclerosing cholangitis), cholelithiasis, venous or arterial thromboembolism, and other associated immune-mediated diseases.119

The majority of patients (80%) present small bowel involvement, 1/3 only ileitis, 1/3 ileocolonic involvement and in 1/3, Crohn's disease is restricted to the colon and rectum. In a North American cohort, 30% of patients presented perianal disease, while in Europe this complication was detected in only 9% of patients.120 Oral and gastroduodenal involvement is estimated in approximately 10% of Crohn's disease patients.120 Fistulas are common: enteric, cutaneous, vesical, vaginal, anal, or rectal. Complications in the involved organs, such as abdominal masses and abscesses, are frequent, with perianal fistulizing disease in up to one-quarter of the patients.119 At diagnosis, most (71%) patients present a non-stricturing and non-penetrating profile of disease, but 21% have a stricturing and 8% a penetrating behavior.120

Surgery due to intestinal inflammation or related complications, such as stenosis, fistulas or perforations, occurs in 13% of patients in the first year of diagnosis, and 22% in five years of follow-up. The 10-year cumulative risk of major abdominal surgery is of approximately 55%.108,109,119,120

Diagnosis of Crohn's disease is based on the combination of a history of chronic intestinal inflammation symptoms and colonoscopy, magnetic resonance enterography and histopathological findings.119 Routine laboratory studies include complete blood count, basic metabolic panel, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), liver function tests, iron distribution studies, vitamin B12 and D levels, β2 microglobulin and an immunologic panel study.121

Ileocolonoscopy and biopsy are considered gold standard tools for diagnosis of Crohn's disease. When visualizing the ileum, right, transverse and left colon, abnormalities in the rectum mucosa and lesions are observed as cobblestones, with nodularity, edema, superficial or deep ulcerations, friability, or stenosis. The Crohn's Disease Endoscopy Severity Index (CDEIS) and the Simple Endoscopy Score for Crohn's disease (SES-CD) were developed and validated to evaluate the compromised intestinal surface, and to characterize the severity of the involvement.122,123 All lesions should be biopsied for histological study. Upper endoscopy should only be performed in patients with upper gastrointestinal signs and symptoms and wireless capsule endoscopy may be an option for patients without intestinal strictures.119 Magnetic resonance enterography is a suitable method for the diagnosis, as the location, extension, disease activity, presence of obstructions, fistulas, and severity of inflammatory lesions of Crohn's disease need to be determined.124

Disease activity is measured by scores, such as the Crohn's Disease Activity Index (CDAI), and the Harvey Bradshaw index (HBi).125,126 A CDAI below 150 is defined as disease remission. A HBi below five defines remission, 5–7 mildly active disease, 8–16 moderately active disease and higher than 16, severe disease.

Treatment aims to stabilize Crohn's disease, reduce symptoms, and heal intestinal lesions. Anti-inflammatory drugs, immunosuppressive agents, corticosteroids, and biological agents are prescribed alone or in combination. Drugs are usually administered in a stepwise sequence, named “up and down” treatment. In more severe cases, early indication of biological agents associated with immunosuppressants remain controversial and debated.108

Hematopoietic stem cell therapy

Hematopoietic stem cell transplantation (HSCT) has emerged as a potential treatment for Crohn's disease due to the chronicity of the disease and lack of further therapeutic options in refractory patients. Additionally, since 1993, there are several case reports in the literature of Crohn's disease patients with concomitant leukemia or lymphoma who improved from the former when transplanted for the latter.127–132

In 2003, investigators from the Northwestern University (Chicago, USA) published their successful experience with the first two patients treated with AHSCT for Crohn's disease as primary indication.133,134 A subsequent update from the same investigators described dramatic clinical remissions in 11 from a total of 12 transplanted patients, in 18 months of follow-up.135 In 2010, a last update described the long-term follow-up of 24 Crohn's disease patients transplanted in Chicago, showing high rates of disease progression over a five-year follow-up (Table 4).4

Table 4.

Results of Crohn's disease study outcomes.

Site/year of publication(ref)  Study design  Comparisons  Endpoints  Inclusion criteria  N  Transplant regimen  PFS  Relapse/progression rate  Non-relapse mortality  OS 
Single-arm studies
Italy (Milan) 2007136  Phase I/IIProspective  None, single arm  Safety and clinical/endoscopic response  CDAI>250Active intestinal lesionsFailed>2 ISC-reactive protein>1mg/dL  CY 200mg/kg+7.5mg/kg rATGUnselected graft  75% at 16.5 months mean FU  25% (1/4) at 16.5 months mean FU  Zero at 16.5 months  100% at 16.5 months 
EUA (Chicago) 20104  Phase I/IIProspective  None, single arm  Safety and clinical outcomes  CDAI>250Crohn's severity index>16Failed anti-TNF  24  CY 200mg/kg+6mg/kg rATG or 90mg/kg eATGCD34+ selected graft  19% at 5y  15/23 at mean FU of 4.4y  Zero TRM1 accidental death at 3y  95% at 5y 
Netherlands 2011137  Case series  None, single arm  None described  Active diseaseFailed anti-TNFRisk of resection surgery  3(only 2 transplanted)  CY 200mg/kg+90mg/kg eATGCD34+ selected graft  Zero at 5y (both patients relapsed at 3 and 12m post-AHSCT  100% (2/2) at 5y  Zero  100% at 5y 
Germany (Freiburg) 2012138  Phase I/IIProspective  None, single arm  Safety, feasibility and efficacy  Active diseaseFailed conventional therapy  12 (only 9 transplanted)  CY 200mg/kgCD34+ selected graft  17% (2/12) at mean follow-up of 3.1y  78% (7/9) at 3.1y  Zero at mean 3.1y  100% at mean 3.1y 
United Kingdom (3 centers) 2014139  Retrospective case series  None, single arm  Long-term clinical outcomes  Active disease failed conventional therapy  CY 200mg/kg+7.5mg/kg rATGCD34+ selected graft in 2 patients  Zero (6/6 relapses)  100% at median 10 months after AHSCT  Zero  100% at median 87 months FU 
Brazil (S. José do Rio Preto) 2017142  Phase I/IIProspective  None, single arm  Safety and early clinical outcomes (first 30 days after AHSCT)  CDAI>150Active intestinal lesionsRisk of resection surgeryFailed anti-TNF  14  CY 200mg/kg+6.5mg/kg rATGUnselected graft  93% at 30 days  1/14 at 30 days  Zero at 30 days  100% at 30 days 
Europe multicenter (EBMT) 2018143  Retrospective survey  None, single arm  Safety and efficacy  Patients transplanted from 1997 to 2015Excludes ASTIC patients  82  86% CY 200mg/kg+ATG11% CY 200mg/kg+CD34 selection  54% at 1y27% at 41 months  73% (60/81) at median 10 months after AHSCT  1.2% (1/82)  97% at 5y 
Spain (Madrid) 2019144  Retrospective case series  None, single arm  Safety and efficacy  Patients with active Crohn's disease transplanted from 2011 to 2017.  CY 200mg/kg+rATG (dose not described)Unselected graft  29% at 48 months  71% (5/7) at 48 months  Zero  100% 
Randomized study
ASTIC (EBMT, multicenter) 2016140  Phase II, open label, randomized 1:1  Patients randomized after mobilization to immediate AHSCT or control treatment with AHSCT deferred for 1y  Sustained clinical and endoscopic/radiologic remission at 1 y  Active diseaseImpaired function/quality of lifeFailed ≥3 IS/biologicals  45 (23 AHSCT and 22 control treatment)  CY 200mg/kg+7.5mg/kg rATGUnselected graft  8.7% (2/23) in the AHSCT group4.5% (1/22) in the control group  95% (20/21) in the AHSCT group at 1y94% (16/17) in the control group at 1y  4.3% (1/23) at 1y in the transplant groupZero in the control group  96% at 1y in the transplant group100% in the control group 

PFS: progression-free survival; OS: overall survival; N: number of enrolled subjects; USA: United States of America; y: years; CDAI: Crohn's disease activity index; TRM: transplant-related mortality; IV CY: intravenous cyclophosphamide; eATG: equine anti-thymoglobulin; rATG: rabbit anti-thymoglobulin; FU: follow-up; HSCT: hematopoietic stem cell transplantation; anti-TNF: treatment with anti-tumor necrosis factor monoclonal antibody; IS: immunosuppressors; biologicals: biological agents.

The European centers also reproduced the North-American protocol, with similar outcomes of low toxicity and high rates of short-term disease remission.136 Longer follow-up, however, confirmed the high relapse/progression rates.137–144 A European multicenter randomized study (Autologous Stem Cell Transplantation in Refractory Crohn's Disease, ASTIC) had a very strict study design and ambitious endpoints, and thus failed to show superiority of AHSCT versus mobilization only (Table 4).140 A later reassessment of the same results, with more traditional endpoints, enabled more optimistic conclusions; that AHSCT promotes clinical and endoscopic benefits, despite a high burden of adverse events.145 To date, the EBMT registry reports Crohn's disease as the third most frequent autoimmune disease indication for AHSCT.141

In Brazil, the first report of AHSCT for Crohn's disease was published in 2013.142 To date, at least 57 procedures have been performed in four Brazilian centers (unpublished data). A single institution Brazilian trial enrolled 14 Crohn's disease patients for AHSCT with high rate of disease remissions and improved quality of life at 30 days post-transplantation (Table 4).146 Longer follow-up of the quality of life in these patients, evaluated by the IBDQ (Inflammatory Bowel Disease Questionnaire) and SF-36 (Short Form-36) questionnaires show sustained benefit at four years post-AHSCT (data not published).

The field of AHSCT for Crohn's disease has advanced over time, in parallel to the learning curve in other autoimmune disease indications. Patient selection and disease stratification are important steps that precede AHSCT, and aim to improve safety and post-transplantation outcomes.46,143 Perianal disease, fistulas and intra-abdominal abscesses, as well as presence of ostomies, are not considered absolute exclusion criteria, but increase risks associated to the procedure and should be carefully considered before patient enrollment. Moreover, hematopoietic progenitor cells may be successfully and safely mobilized from the peripheral blood with low (2g/m2) doses of cyclophosphamide.147 Disease reactivation over time is still high (Table 4) and whether CD34+ graft selection has any effect on long-term control remains to be defined.4,136–140,142,143 Finally, there is evidence to suggest that after AHSCT, patients become more responsive to conventional therapy than before the procedure.140,141 These aspects should be explored in future studies.

The new ongoing protocols, ASTIC-Lite (ASTIC-low intensity therapy evaluation, EBMT), and the AutoCrohn2 (São José do Rio Preto, Brazil), include a more refined patient assessment and stratification with accurate clinical, immunological, magnetic resonance and microbiota studies. These studies aim for a better understanding of the therapeutic potential of AHSCT for patients with Crohn's disease.147–149

Guidelines for transplantation in Crohn's disease

Autologous HSCT has the potential to induce clinical remission and improve quality of life in patients with Crohn's disease with poor prognosis or that are refractory to immunosuppressants or biologic agents. AHSCT is considered safe, but associated with a high number of adverse events, mainly infectious. Therefore, AHSCT for Crohn's disease should be performed by experienced centers, with specialized teams to manage gastrointestinal and infectious complications. Early referral is recommended to optimize clinical outcomes and minimize risks.

Conclusions

Scientific evidence supports the clinical use of autologous hematopoietic stem cell transplantation (AHSCT) for multiple sclerosis (MS), systemic sclerosis and Crohn's disease. In systemic sclerosis, AHSCT is indicated due to the severity of the disease and lack of therapeutic options, with effectiveness confirmed by phase III randomized trials. In MS, treatment with AHSCT is supported by 25 years of research, including recent phase III trials comparing transplant to new drugs. In Crohn's disease refractory to treatment with immunosuppressor and biological agents, long-term benefits have been shown after AHSCT, despite low rates of sustained disease remission.

The role of HSCT in these and in other autoimmune diseases is likely to evolve further with increased clinical experience, especially with regards to optimal timing of transplant. It is expected that improvements in design of clinical trials and experimental studies further expand the impact of HSCT in this field.

Conflicts of interest

The authors declare no conflicts of interest.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References
[1]
J.A. Snowden, M. Badoglio, T. Alexander.
The rise of autologous HCT for autoimmune diseases: what is behind it and what does it mean for the future of treatment? An update on behalf of the EBMT Autoimmune Diseases Working Party.
Expert Rev Clin Immunol, 15 (2018), pp. 981-985
[2]
R.A. Nash, J.D. Bowen, P.A. McSweeney, S.Z. Pavletic, K.R. Maravilla, M. Park, et al.
High-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation for severe multiple sclerosis.
Blood, 102 (2003), pp. 2364-2372
[3]
T. Kozák, E. Havrdová, J. Pit’ha, E. Gregora, R. Pytlík, J. Maaloufová, et al.
High-dose immunosuppressive therapy with PBPC support in the treatment of poor risk multiple sclerosis.
Bone Marrow Transplant, 25 (2000), pp. 525-531
[4]
R.K. Burt, R.M. Craig, F. Milanetti, K. Quigley, P. Gozdziak, J. Bucha, et al.
Autologous nonmyeloablative hematopoietic stem cell transplantation in patients with severe anti-TNF refractory Crohn disease: long-term follow-up.
Blood, 116 (2010), pp. 6123-6132
[5]
C.J. Hawkey, J. Lindsay, J. Gribben.
Stem cell transplantation for refractory Crohn disease – reply.
[6]
G. Mancardi, R. Saccardi.
Autologous haematopoietic stem-cell transplantation in multiple sclerosis.
Lancet Neurol, 7 (2008), pp. 626-636
[7]
L. Su, J. Xu, B.-X. Ji, S.-G. Wan, C.-Y. Lu, H.-Q. Dong, et al.
Autologous peripheral blood stem cell transplantation for severe multiple sclerosis.
Int J Hematol, 84 (2006), pp. 276-281
[8]
H. Openshaw, B.T. Lund, A. Kashyap, R. Atkinson, I. Sniecinski, L.P. Weiner, et al.
Peripheral blood stem cell transplantation in multiple sclerosis with busulfan and cyclophosphamide conditioning: report of toxicity and immunological monitoring.
Biol Blood Marrow Transplant, 6 (2000), pp. 563-575
[9]
D. Farge, M. Labopin, A. Tyndall, A. Fassas, G.L. Mancardi, J. Van Laar, et al.
Autologous hematopoietic stem cell transplantation for autoimmune diseases: an observational study on 12 years’ experience from the European Group for Blood and Marrow Transplantation Working Party on Autoimmune Diseases.
Haematologica, 95 (2010), pp. 284-292
[10]
J.A. Snowden, R. Saccardi, M. Allez, S. Ardizzone, R. Arnold, R. Cervera, et al.
Haematopoietic SCT in severe autoimmune diseases: updated guidelines of the European Group for Blood and Marrow Transplantation.
Bone Marrow Transplant, 47 (2012), pp. 770-790
[11]
N. Del Papa, F. Onida, E. Zaccara, G. Saporiti, W. Maglione, E. Tagliaferri, et al.
Autologous hematopoietic stem cell transplantation has better outcomes than conventional therapies in patients with rapidly progressive systemic sclerosis.
Bone Marrow Transplant, 52 (2017), pp. 53-58
[12]
J.C. Henes, M. Schmalzing, W. Vogel, G. Riemekasten, F. Fend, L. Kanz, et al.
Optimization of autologous stem cell transplantation for systemic sclerosis – a single-center longterm experience in 26 patients with severe organ manifestations.
J Rheumatol, 39 (2012), pp. 269-275
[13]
G.C. De Santis, B. de Pina Almeida Prado, K. de Lima Prata, D.M. Brunetta, M.D. Orellana, P.V.B. Palma, et al.
Mobilization and harvesting of PBPC in newly diagnosed type 1 diabetes mellitus.
Bone Marrow Transplant, 47 (2012), pp. 993-994
[14]
J.A. Snowden, M. Badoglio, M. Labopin, S. Giebel, E. McGrath, Z. Marjanovic, et al.
Evolution, trends, outcomes, and economics of hematopoietic stem cell transplantation in severe autoimmune diseases.
Blood Adv, 1 (2017), pp. 2742-2755
[15]
T. Alexander, A. Thiel, O. Rosen, G. Massenkeil, A. Sattler, S. Kohler, et al.
Depletion of autoreactive immunologic memory followed by autologous hematopoietic stem cell transplantation in patients with refractory SLE induces long-term remission through de novo generation of a juvenile and tolerant immune system.
[16]
H.L. Weiner.
Multiple sclerosis is an inflammatory T-cell-mediated autoimmune disease.
Arch Neurol, 61 (2004), pp. 1613
[17]
A. Compston, A. Coles.
Multiple sclerosis.
Lancet, 372 (2008), pp. 1502-1517
[18]
D.S. Goodin.
The epidemiology of multiple sclerosis.
Handbook of clinical neurology [Internet], (2014), pp. 231-266
[19]
A. Nylander, D.A. Hafler.
Multiple sclerosis.
J Clin Invest, 122 (2012), pp. 1180-1188
[20]
E.S. Roach.
Is multiple sclerosis an autoimmune disorder?.
Arch Neurol, 61 (2004), pp. 1615
[21]
D.S. Reich, C.F. Lucchinetti, P.A. Calabresi.
Multiple sclerosis.
N Engl J Med, 378 (2018), pp. 169-180
[22]
S.V. Ramagopalan, A.D. Sadovnick.
Epidemiology of multiple sclerosis.
Neurol Clin, 29 (2011), pp. 207-217
[23]
Atlas multiple sclerosis resources in the world [Internet].
(2008),
[24]
MS movement problems | multiple sclerosis.
Multiple Sclerosis International Federation, (2016),
[25]
J.F. Kurtzke.
Clinical definition for multiple sclerosis treatment trials.
Ann Neurol, 36 (1994), pp. S73-S79
[26]
G. Giovannoni, B. Turner, S. Gnanapavan, C. Offiah, K. Schmierer, M. Marta.
Is it time to target no evident disease activity (NEDA) in multiple sclerosis?.
Mult Scler Relat Disord, 4 (2015), pp. 329-333
[27]
T. Garry, S. Krieger, M. Fabian.
MS research update [Internet].
(2018),
[28]
R.K. Burt, B.A. Cohen, E. Russell, K. Spero, A. Joshi, Y. Oyama, et al.
Hematopoietic stem cell transplantation for progressive multiple sclerosis: failure of a total body irradiation-based conditioning regimen to prevent disease progression in patients with high disability scores.
Blood, 102 (2003), pp. 2373-2378
[29]
E. Carreras, A. Saiz, P. Marín, C. Martínez, M. Rovira, N. Villamor, et al.
CD34+ selected autologous peripheral blood stem cell transplantation for multiple sclerosis: report of toxicity and treatment results at one year of follow-up in 15 patients.
Haematologica, 88 (2003), pp. 306-314
[30]
J.P.A. Samijn, P.A.W. te Boekhorst, T. Mondria, P.A. van Doorn, H.Z. Flach, F.G.A. van der Meché, et al.
Intense T cell depletion followed by autologous bone marrow transplantation for severe multiple sclerosis.
J Neurol Neurosurg Psychiatry, 77 (2006), pp. 46-50
[31]
H. Atkins, M. Freedman.
Immune ablation followed by autologous hematopoietic stem cell transplantation for the treatment of poor prognosis multiple sclerosis.
Methods Mol Biology, (2009), pp. 231-246
[32]
A. Fassas, V.K. Kimiskidis, I. Sakellari, K. Kapinas, A. Anagnostopoulos, V. Tsimourtou, et al.
Long-term results of stem cell transplantation for MS: a single-center experience.
Neurology, 76 (2011), pp. 1066-1070
[33]
A. Saiz, Y. Blanco, J. Berenguer, M. Gómez-Choco, E. Carreras, T. Arbizu, et al.
Clinical outcome 6 years after autologous hematopoietic stem cell transplantation in multiple sclerosis.
Neurologia, 23 (2008), pp. 405-407
[34]
T. Kozák, E. Havrdová, J. Pit’ha, E. Gregora, R. Pytlík, J. Maaloufová, et al.
Immunoablative therapy with autologous stem cell transplantation in the treatment of poor risk multiple sclerosis.
Transplant Proc, 33 (2001), pp. 2179-2181
[35]
Y.L. Shevchenko, A.A. Novik, A.N. Kuznetsov, B.V. Afanasiev, I.A. Lisukov, V.A. Kozlov, et al.
High-dose immunosuppressive therapy with autologous hematopoietic stem cell transplantation as a treatment option in multiple sclerosis.
Exp Hematol, 36 (2008), pp. 922-928
[36]
J. Xu, B. Ji, L. Su, H. Dong, X. Sun, C. Liu.
Clinical outcomes after autologous haematopoietic stem cell transplantation in patients with progressive multiple sclerosis.
Chin Med J (Engl), 119 (2006), pp. 1851-1855
[37]
X.-S. Ni, J. Ouyang, W.-H. Zhu, C. Wang, B. Chen.
Autologous hematopoietic stem cell transplantation for progressive multiple sclerosis: report of efficacy and safety at three yr of follow up in 21 patients.
Clin Transplant, 20 (2006), pp. 485-489
[38]
R. Saccardi, G.L. Mancardi, A. Solari, A. Bosi, P. Bruzzi, P. Di Bartolomeo, et al.
Autologous HSCT for severe progressive multiple sclerosis in a multicenter trial: impact on disease activity and quality of life.
Blood, 105 (2005), pp. 2601-2607
[39]
E. Krasulová, M. Trneny, T. Kozák, B. Vacková, D. Pohlreich, D. Kemlink, et al.
High-dose immunoablation with autologous haematopoietic stem cell transplantation in aggressive multiple sclerosis: a single centre 10-year experience.
Mult Scler, 16 (2010), pp. 685-693
[40]
M. Breban, R.E. Hammer, J.A. Richardson, J.D. Taurog.
Transfer of the inflammatory disease of HLA-B27 transgenic rats by bone marrow engraftment.
J Exp Med, 178 (1993), pp. 1607-1616
[41]
I. de Kleer, B. Vastert, M. Klein, G. Teklenburg, G. Arkesteijn, G.P. Yung, et al.
Autologous stem cell transplantation for autoimmunity induces immunologic self-tolerance by reprogramming autoreactive T cells and restoring the CD4+CD25+ immune regulatory network.
Blood, 107 (2006), pp. 1696-1702
[42]
T. Takayama, Y. Nishioka, L. Lu, M.T. Lotze, H. Tahara, A.W. Thomson.
Retroviral delivery of viral interleukin-10 into myeloid dendritic cells markedly inhibits their allostimulatory activity and promotes the induction of T-cell hyporesponsiveness.
Transplantation, 66 (1998), pp. 1567-1574
[43]
M.M. Herrmann, S. Gaertner, C. Stadelmann, J. van den Brandt, R. Böscke, W. Budach, et al.
Tolerance induction by bone marrow transplantation in a multiple sclerosis model.
Blood, 106 (2005), pp. 1875-1883
[44]
R.K. Burt, B. Cohen, J. Rose, F. Petersen, Y. Oyama, D. Stefoski, et al.
Hematopoietic stem cell transplantation for multiple sclerosis.
Arch Neurol, 62 (2005), pp. 860-864
[45]
R.A. Good, T. Verjee.
Historical and current perspectives on bone marrow transplantation for prevention and treatment of immunodeficiencies and autoimmunities.
Biol Blood Marrow Transplant, 7 (2001), pp. 123-135
[46]
A. Tyndall, A. Fassas, J. Passweg, C. Ruiz de Elvira, M. Attal, P. Brooks, et al.
Autologous haematopoietic stem cell transplants for autoimmune disease--feasibility and transplant-related mortality. Autoimmune Disease and Lymphoma Working Parties of the European Group for Blood and Marrow Transplantation, the European League Against Rheumatism and the International Stem Cell Project for Autoimmune Disease.
Bone Marrow Transplant, 24 (1999), pp. 729-734
[47]
G.L. Mancardi, A. Murialdo, P. Rossi, F. Gualandi, G. Martino, A. Marmont, et al.
Autologous stem cell transplantation as rescue therapy in malignant forms of multiple sclerosis.
Mult Scler J, 11 (2005), pp. 367-371
[48]
J. Fagius, J. Lundgren, G. Oberg.
Early highly aggressive MS successfully treated by hematopoietic stem cell transplantation.
Mult Scler, 15 (2009), pp. 229-237
[49]
G.L. Mancardi, M.P. Sormani, M. Di Gioia, L. Vuolo, F. Gualandi, M.P. Amato, et al.
Autologous haematopoietic stem cell transplantation with an intermediate intensity conditioning regimen in multiple sclerosis: the Italian multi-centre experience.
Mult Scler, 18 (2012), pp. 835-842
[50]
J.A. Cohen, L.E. Baldassari, H.L. Atkins, J.D. Bowen, C. Bredeson, P.A. Carpenter, 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, 25 (2019), pp. 845-854
[51]
Mancardi GL. ASTIMS: High dose immunoablation and autologous haematopoietic stem cell transplantation versus mitoxantrone therapy in severe multiple sclerosis | EBMT [Internet]. Available from: https://www.ebmt.org/research/studies/astims-high-dose-immunoablation-and-autologous-haematopoietic-stem-cell [cited 16.11.19].
[52]
G.L. Mancardi, M.P. Sormani, F. Gualandi, A. Saiz, E. Carreras, E. Merelli, et al.
Autologous hematopoietic stem cell transplantation in multiple sclerosis: a phase II trial.
Neurology, 84 (2015), pp. 981-988
[53]
P.A. Muraro, M. Pasquini, H.L. Atkins, J.D. Bowen, D. Farge, A. Fassas, et al.
Long-term outcomes after autologous hematopoietic stem cell transplantation for multiple sclerosis.
[54]
M.P. Sormani, P.A. Muraro, I. Schiavetti, A. Signori, A. Laroni, R. Saccardi, et al.
Autologous hematopoietic stem cell transplantation in multiple sclerosis: a meta-analysis.
Neurology, 88 (2017), pp. 2115-2122
[55]
R.K. Burt, R. Balabanov, J. Burman, B. Sharrack, J.A. Snowden, M.C. Oliveira, et al.
Effect of nonmyeloablative hematopoietic stem cell transplantation vs continued disease-modifying therapy on disease progression in patients with relapsing-remitting multiple sclerosis.
[56]
J. Burman, E. Iacobaeus, A. Svenningsson, J. Lycke, M. Gunnarsson, P. Nilsson, et al.
Autologous haematopoietic stem cell transplantation for aggressive multiple sclerosis: the Swedish experience.
J Neurol Neurosurg Psychiatry, 85 (2014), pp. 1116-1121
[57]
B. Casanova, I. Jarque, F. Gascón, J.C. Hernández-Boluda, F. Pérez-Miralles, J. de la Rubia, et al.
Autologous hematopoietic stem cell transplantation in relapsing-remitting multiple sclerosis: comparison with secondary progressive multiple sclerosis.
Neurol Sci, 38 (2017), pp. 1213-1221
[58]
J.L. Shevchenko, A.N. Kuznetsov, T.I. Ionova, V.Y. Melnichenko, D.A. Fedorenko, A.V. Kartashov, et al.
Autologous hematopoietic stem cell transplantation with reduced-intensity conditioning in multiple sclerosis.
Exp Hematol, 40 (2012), pp. 892-898
[59]
R.K. Burt, Y. Loh, B. Cohen, D. Stefoski, D. Stefosky, R. Balabanov, et al.
Autologous non-myeloablative haemopoietic stem cell transplantation in relapsing-remitting multiple sclerosis: a phase I/II study.
Lancet Neurol, 8 (2009), pp. 244-253
[60]
R.A. Nash, G.J. Hutton, M.K. Racke, U. Popat, S.M. Devine, L.M. Griffith, et al.
High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for relapsing-remitting multiple sclerosis (HALT-MS).
[61]
R.A. Nash, G.J. Hutton, M.K. Racke, U. Popat, S.M. Devine, K.C. Steinmiller, et al.
High-dose immunosuppressive therapy and autologous HCT for relapsing-remitting MS.
Neurology, 88 (2017), pp. 842-852
[62]
H.L. Atkins, M. Bowman, D. Allan, G. Anstee, D.L. Arnold, A. Bar-Or, et al.
Immunoablation and autologous haemopoietic stem-cell transplantation for aggressive multiple sclerosis: a multicentre single-group phase 2 trial.
[63]
J.J. Moore, J.C. Massey, C.D. Ford, M.L. Khoo, J.J. Zaunders, K. Hendrawan, et al.
Prospective phase II clinical trial of autologous haematopoietic stem cell transplant for treatment refractory multiple sclerosis.
J Neurol Neurosurg Psychiatry, 90 (2019), pp. 514-521
[64]
M.P. Sormani, P.A. Muraro, R. Saccardi, G. Mancardi.
NEDA status in highly active MS can be more easily obtained with autologous hematopoietic stem cell transplantation than other drugs.
Mult Scler, 23 (2017), pp. 201-204
[65]
N. Hamerschlak, M. Rodrigues, D.A. Moraes, M.C. Oliveira, A.B.P.L. Stracieri, F. Pieroni, et al.
Brazilian experience with two conditioning regimens in patients with multiple sclerosis: BEAM/horse ATG and CY/rabbit ATG.
Bone Marrow Transplant, 45 (2010), pp. 239-248
[66]
R. Saccardi, T. Kozak, C. Bocelli-Tyndall, A. Fassas, A. Kazis, E. Havrdova, et al.
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, 12 (2006), pp. 814-823
[67]
M.C. Pasquini, J. Voltarelli, H.L. Atkins, N. Hamerschlak, X. Zhong, K.W. Ahn, et al.
Transplantation for autoimmune diseases in north and South America: a report of the Center for International Blood and Marrow Transplant Research.
Biol Blood Marrow Transplant, 18 (2012), pp. 1471-1478
[68]
R.K. Burt, R. Balabanov, X. Han, B. Sharrack, A. Morgan, K. Quigley, et al.
Association of nonmyeloablative hematopoietic stem cell transplantation with neurological disability in patients with relapsing-remitting multiple sclerosis.
JAMA, 313 (2015), pp. 275-284
[69]
M.C. Rodrigues, O. de, N. Hamerschlak, D.A. de Moraes, B.P. Simões, M. Rodrigues, et al.
Guidelines of the Brazilian society of bone Marrow transplantation on hematopoietic stem cell transplantation as a treatment for the autoimmune diseases systemic sclerosis and multiple sclerosis.
Rev Bras Hematol Hemoter, 35 (2013), pp. 134-143
[70]
Z. DeFilipp, R.F. Duarte, J.A. Snowden, N.S. Majhail, D.M. Greenfield, J.L. Miranda, et al.
Metabolic syndrome and cardiovascular disease after hematopoietic cell transplantation: screening and preventive practice recommendations from the CIBMTR and EBMT.
Biol Blood Marrow Transplant, 22 (2016), pp. 1493-1503
[71]
C.P. Denton, A.U. Wells, J.G. Coghlan.
Major lung complications of systemic sclerosis.
Nat Rev Rheumatol, 14 (2018), pp. 511-527
[72]
C.P. Denton, D. Khanna.
Systemic sclerosis.
Lancet, 390 (2017), pp. 1685-1699
[73]
J. Varga, D. Abraham.
Systemic sclerosis: a prototypic multisystem fibrotic disorder.
J Clin Invest, 117 (2007), pp. 557-567
[74]
R.T. Domsic, S.I. Nihtyanova, S.R. Wisniewski, M.J. Fine, M. Lucas, C.K. Kwoh, et al.
Derivation and validation of a prediction rule for two-year mortality in early diffuse cutaneous systemic sclerosis.
Arthritis Rheumatol, 66 (2014), pp. 1616-1624
[75]
M. Elhai, C. Meune, J. Avouac, A. Kahan, Y. Allanore.
Trends in mortality in patients with systemic sclerosis over 40 years: a systematic review and meta-analysis of cohort studies.
Rheumatology (Oxford), 51 (2012), pp. 1017-1026
[76]
E.C. LeRoy, T.A. Medsger.
Criteria for the classification of early systemic sclerosis.
J Rheumatol, 28 (2001), pp. 1573-1576
[77]
F.A. Wollheim.
Classification of systemic sclerosis. Visions and reality.
Rheumatology, 44 (2005), pp. 1212-1216
[78]
O. Kowal-Bielecka, J. Fransen, J. Avouac, M. Becker, A. Kulak, Y. Allanore, et al.
Update of EULAR recommendations for the treatment of systemic sclerosis.
Ann Rheum Dis, 76 (2017), pp. 1327-1339
[79]
Sampaio-Barros PD, Zimmermann AF, Müller CdS, Borges CTL, Freire EAM, Maretti GB.
Recommendations for the management and treatment of systemic sclerosis.
Rev Bras Reumatol, 53 (2013), pp. 258-275
[80]
J.A. Lopez-Ovejero, S.D. Saal, W.A. D’Angelo, J.S. Cheigh, K.H. Stenzel, J.H. Laragh.
Reversal of vascular and renal crises of scleroderma by oral angiotensin-converting-enzyme blockade.
N Engl J Med, 300 (1979), pp. 1417-1419
[81]
V.D. Steen, J.P. Costantino, A.P. Shapiro, T.A. Medsger.
Outcome of renal crisis in systemic sclerosis: relation to availability of angiotensin converting enzyme (ACE) inhibitors.
Ann Intern Med, 11 (1990), pp. 352-357
[82]
D.P. Tashkin, R. Elashoff, P.J. Clements, M.D. Roth, D.E. Furst, R.M. Silver, et al.
Effects of 1-year treatment with cyclophosphamide on outcomes at 2 years in scleroderma lung disease.
Am J Respir Crit Care Med, 176 (2007), pp. 1026-1034
[83]
E.R. Volkmann, D.P. Tashkin, N. Li, M.D. Roth, D. Khanna, A.-M. Hoffmann-Vold, et al.
Mycophenolate mofetil versus placebo for systemic sclerosis-related interstitial lung disease: an analysis of scleroderma lung studies I and II.
Arthritis Rheumatol, 69 (2017), pp. 1451-1460
[84]
D. Khanna, C.P. Denton, A. Jahreis, J.M. van Laar, T.M. Frech, M.E. Anderson, et al.
Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial.
Lancet, 387 (2016), pp. 2630-2640
[85]
D. Khanna, C.P. Denton, C.J.F. Lin, J.M. van Laar, T.M. Frech, M.E. Anderson, et al.
Safety and efficacy of subcutaneous tocilizumab in systemic sclerosis: results from the open-label period of a phase II randomised controlled trial (faSScinate).
Ann Rheum Dis, 77 (2018), pp. 212-220
[86]
L. Shand, M. Lunt, S. Nihtyanova, M. Hoseini, A. Silman, C.M. Black, et al.
Relationship between change in skin score and disease outcome in diffuse cutaneous systemic sclerosis: application of a latent linear trajectory model.
Arthritis Rheum, 56 (2007), pp. 2422-2431
[87]
M. Elhai, M. Boubaya, O. Distler, V. Smith, M. Matucci-Cerinic, J.J. Alegre Sancho, et al.
Outcomes of patients with systemic sclerosis treated with rituximab in contemporary practice: a prospective cohort study.
Ann Rheum Dis, 78 (2019), pp. 979-987
[88]
J.M. van Laar, D. Farge, J.K. Sont, K. Naraghi, Z. Marjanovic, J. Larghero, et al.
Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial.
JAMA, 311 (2014), pp. 2490-2498
[89]
R.K. Burt, M.C. Oliveira, S.J. Shah, D.A. Moraes, B. Simoes, M. Gheorghiade, et al.
Cardiac involvement and treatment-related mortality after non-myeloablative haemopoietic stem-cell transplantation with unselected autologous peripheral blood for patients with systemic sclerosis: a retrospective analysis.
Lancet, 381 (2013), pp. 1116-1124
[90]
F. van den Hoogen, D. Khanna, J. Fransen, S.R. Johnson, M. Baron, A. Tyndall, et al.
Classification criteria for systemic sclerosis: an American College of Rheumatology/European League Against Rheumatism Collaborative Initiative.
Arthritis Rheum, 65 (2013), pp. 2737-2747
[91]
M. Binks, J.R. Passweg, D. Furst, P. McSweeney, K. Sullivan, C. Besenthal, et al.
Phase I/II trial of autologous stem cell transplantation in systemic sclerosis: procedure related mortality and impact on skin disease.
Ann Rheum Dis, 60 (2001), pp. 577-584
[92]
R.K. Burt, Y. Oyama, A. Traynor, K. Quigley, M. Brush, J. Rodriguez, et al.
Hematopoietic stem cell transplantation for systemic sclerosis with rapid improvement in skin scores: is neoangiogenesis occurring?.
Bone Marrow Transplant, 32 (2003), pp. S65-S67
[93]
D. Farge, J.P. Marolleau, S. Zohar, Z. Marjanovic, J. Cabane, N. Mounier, et al.
Autologous bone marrow transplantation in the treatment of refractory systemic sclerosis: early results from a French multicentre phase I–II study.
Br J Haematol, 119 (2002), pp. 726-739
[94]
D. Farge, J. Passweg, J.M. van Laar, Z. Marjanovic, C. Besenthal, J. Finke, et al.
Autologous stem cell transplantation in the treatment of systemic sclerosis: report from the EBMT/EULAR Registry.
Ann Rheum Dis, 63 (2004), pp. 974-981
[95]
R.A. Nash, P.A. McSweeney, L.J. Crofford, M. Abidi, C.-S. Chen, J.D. Godwin, et al.
High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: long-term follow-up of the US multicenter pilot study.
Blood, 110 (2007), pp. 1388-1396
[96]
Y. Oyama, W.G. Barr, L. Statkute, T. Corbridge, E.A. Gonda, B. Jovanovic, et al.
Autologous non-myeloablative hematopoietic stem cell transplantation in patients with systemic sclerosis.
Bone Marrow Transplant, 40 (2007), pp. 549-555
[97]
R.K. Burt, M.C. Oliveira, S.J. Shah.
Cardiac assessment before stem cell transplantation for systemic sclerosis.
[98]
D. Farge, R.K. Burt, M.-C. Oliveira, E. Mousseaux, M. Rovira, Z. Marjanovic, et al.
Cardiopulmonary assessment of patients with systemic sclerosis for hematopoietic stem cell transplantation: recommendations from the European Society for Blood and Marrow Transplantation Autoimmune Diseases Working Party and collaborating partners.
Bone Marrow Transplant, 52 (2017), pp. 1495-1503
[99]
R.K. Burt, S.J. Shah, K. Dill, T. Grant, M. Gheorghiade, J. Schroeder, et al.
Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an open-label, randomised phase 2 trial.
[100]
K.M. Sullivan, E.A. Goldmuntz, L. Keyes-Elstein, P.A. McSweeney, A. Pinckney, B. Welch, et al.
Myeloablative autologous stem-cell transplantation for severe scleroderma.
N Engl J Med, 378 (2018), pp. 35-47
[101]
M.C. Oliveira, M. Labopin, J. Henes, J. Moore, N.D. Papa, A. Cras, et al.
Does ex vivo CD34+ positive selection influence outcome after autologous hematopoietic stem cell transplantation in systemic sclerosis patients?.
Bone Marrow Transplant, 51 (2016), pp. 501-505
[102]
J. Henes, M.C. Oliveira, M. Labopin, M. Badoglio, H.U. Scherer, N. Del Papa, et al.
Autologous stem cell transplantation for progressive systemic sclerosis: a prospective non-interventional study from the European Society for Blood and Marrow Transplantation Autoimmune Disease Working Party.
[103]
M. Ayano, H. Tsukamoto, H. Mitoma, Y. Kimoto, M. Akahoshi, Y. Arinobu, et al.
CD34-selected versus unmanipulated autologous haematopoietic stem cell transplantation in the treatment of severe systemic sclerosis: a post hoc analysis of a phase I/II clinical trial conducted in Japan.
Arthritis Res Ther, 21 (2019), pp. 30
[104]
P.A. McSweeney, R.A. Nash, K.M. Sullivan, J. Storek, L.J. Crofford, R. Dansey, et al.
High-dose immunosuppressive therapy for severe systemic sclerosis: initial outcomes.
Blood, 100 (2002), pp. 1602-1610
[105]
Y. Loh, Y. Oyama, L. Statkute, L. Verda, K. Quigley, K. Yaung, et al.
Non-myeloablative allogeneic hematopoietic stem cell transplantation for severe systemic sclerosis: graft-versus-autoimmunity without graft-versus-host disease?.
Bone Marrow Transplant, 39 (2007), pp. 435-437
[106]
M. Shiratsuchi, S. Motomura, Y. Abe, S. Shiokawa, J. Nishimura.
Long-term follow-up after nonmyeloablative allogeneic hematopoietic stem cell transplantation for systemic sclerosis.
Clin Rheumatol, 27 (2008), pp. 1207-1209
[107]
Oxford Centre for Evidence-based Medicine – Levels of Evidence (March 2009) – CEBM [Internet].
(2009),
[108]
D.C. Baumgart, W.J. Sandborn.
Crohn's disease.
Lancet, 380 (2012), pp. 1590-1605
[109]
D. Baumgart.
The natural history of inflammatory bowel disease.
Crohn's disease and ulcerative colitis, Springer, (2012),
[110]
J.M.L. Parente, C.S.R. Coy, V. Campelo, M.P.P.D. Parente, L.A. Costa, R.M. da Silva, et al.
Inflammatory bowel disease in an underdeveloped region of Northeastern Brazil.
World J Gastroenterol, 21 (2015), pp. 1197-1206
[111]
A. Lima Martins, R.A. Volpato, M.d.P. Zago-Gomes.
The prevalence and phenotype in Brazilian patients with inflammatory bowel disease.
BMC Gastroenterol, 18 (2018), pp. 87
[112]
R.G. Gasparini, L.Y. Sassaki, R. Saad-Hossne.
Inflammatory bowel disease epidemiology in São Paulo State, Brazil.
Clin Exp Gastroenterol, 11 (2018), pp. 423-429
[113]
J.D. Feuerstein, A.S. Cheifetz.
Crohn disease: epidemiology, diagnosis, and management.
Mayo Clin Proc, 92 (2017), pp. 1088-1103
[114]
Z. Zhou, M. Ding, L. Huang, G. Gilkeson, R. Lang, W. Jiang.
Toll-like receptor-mediated immune responses in intestinal macrophages; implications for mucosal immunity and autoimmune diseases.
Clin Immunol, 173 (2016), pp. 81-86
[115]
A.G. Pockley, J.O. Lindsay, G.A. Foulds, S. Rutella, J.G. Gribben, T. Alexander, et al.
Immune reconstitution after autologous hematopoietic stem cell transplantation in Crohn's disease: current status and future directions. A review on behalf of the EBMT Autoimmune Diseases Working Party and the autologous stem cell transplantation in refra.
Front Immunol, 9 (2018), pp. 646
[116]
B. Verstockt, K.G. Smith, J.C. Lee.
Genome-wide association studies in Crohn's disease: past, present and future.
Clin Transl Immunol, 7 (2018), pp. e1001
[117]
M.S. Silverberg, J. Satsangi, T. Ahmad, I.D.R. Arnott, C.N. Bernstein, S.R. Brant, et al.
Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology.
Can J Gastroenterol, 19 (2005), pp. 5A-36A
[118]
G.R. Lichtenstein, S.B. Hanauer, W.J. Sandborn, Practice Parameters Committee of American College of Gastroenterology.
Management of Crohn's disease in adults.
Am J Gastroenterol, 104 (2009), pp. 465-483
[119]
G.R. Lichtenstein, E.V. Loftus, K.L. Isaacs, M.D. Regueiro, L.B. Gerson, B.E. Sands.
ACG Clinical Guideline: management of Crohnʼs disease in adults.
Am J Gastroenterol, 113 (2018), pp. 481-517
[120]
J. Burisch, G. Kiudelis, L. Kupcinskas, H.A.L. Kievit, K.W. Andersen, V. Andersen, et al.
Natural disease course of Crohn's disease during the first 5 years after diagnosis in a European population-based inception cohort: an Epi-IBD study.
[121]
M. Gajendran, P. Loganathan, A.P. Catinella, J.G. Hashash.
A comprehensive review and update on Crohn's disease.
Disease-a-Month, 64 (2018), pp. 20-57
[122]
M. Daperno, G. D’Haens, G. Van Assche, F. Baert, P. Bulois, V. Maunoury, et al.
Development and validation of a new, simplified endoscopic activity score for Crohn's disease: the SES-CD.
Gastrointest Endosc, 60 (2004), pp. 505-512
[123]
R. Khanna, S.A. Nelson, B.G. Feagan, G. D’Haens, W.J. Sandborn, G. Zou, et al.
Endoscopic scoring indices for evaluation of disease activity in Crohn's disease.
Cochrane Database Syst Rev, (2016), pp. CD010642
[124]
A.K. Lunder, L.T. Bakstad, J. Jahnsen, A. Borthne, J.R. Hov, M. Vatn, et al.
Assessment of bowel inflammation and strictures by magnetic resonance enterography in long-term Crohn's disease.
J Crohns Colitis, 13 (2019), pp. 607-614
[125]
W.R. Best, J.M. Becktel, J.W. Singleton, F. Kern.
Development of a Crohn's disease activity index. National Cooperative Crohn's Disease Study.
Gastroenterology, 70 (1976), pp. 439-444
[126]
R.F. Harvey, J.M. Bradshaw.
A simple index of Crohn's-disease activity.
[127]
P.E. Drakos, A. Nagler, R. Or.
Case of Crohn's disease in bone marrow transplantation.
Am J Hematol, 43 (1993), pp. 157-158
[128]
A. Kashyap, S.J. Forman.
Autologous bone marrow transplantation for non-Hodgkin's lymphoma resulting in long-term remission of coincidental Crohn's disease.
Br J Haematol, 103 (1998), pp. 651-652
[129]
S.O. Lopez-Cubero, K.M. Sullivan, G.B. McDonald.
Course of Crohn's disease after allogeneic marrow transplantation.
Gastroenterology, 114 (1998), pp. 433-440
[130]
M. Musso, F. Porretto, A. Crescimanno, F. Bondì, V. Polizzi, R. Scalone.
Crohn's disease complicated by relapsed extranodal Hodgkin's lymphoma: prolonged complete remission after unmanipulated PBPC autotransplant.
Bone Marrow Transplant, 26 (2000), pp. 921-923
[131]
J.D. Söderholm, C. Malm, G. Juliusson, R. Sjödahl.
Long-term endoscopic remission of crohn disease after autologous stem cell transplantation for acute myeloid leukaemia.
Scand J Gastroenterol, 37 (2002), pp. 613-616
[132]
M. Ditschkowski, H. Einsele, R. Schwerdtfeger, D. Bunjes, R. Trenschel, D.W. Beelen, et al.
Improvement of inflammatory bowel disease after allogeneic stem-cell transplantation.
Transplantation, 75 (2003), pp. 1745-1747
[133]
R.M. Craig, A. Traynor, Y. Oyama, R.K. Burt.
Hematopoietic stem cell transplantation for severe Crohn's disease.
Bone Marrow Transplant, 32 (2003), pp. S57-S59
[134]
R.K. Burt, A. Traynor, Y. Oyama, R. Craig.
High-dose immune suppression and autologous hematopoietic stem cell transplantation in refractory Crohn disease.
Blood], 101 (2003), pp. 2064-2066
[135]
R.K. Burt, A. Marmont, Y. Oyama, S. Slavin, R. Arnold, F. Hiepe, et al.
Randomized controlled trials of autologous hematopoietic stem cell transplantation for autoimmune diseases: the evolution from myeloablative to lymphoablative transplant regimens.
Arthritis Rheum, 54 (2006), pp. 3750-3760
[136]
A. Cassinotti, C. Annaloro, S. Ardizzone, F. Onida, A.D. Volpe, M. Clerici, et al.
Autologous haematopoietic stem cell transplantation without CD34+ cell selection in refractory Crohn's disease.
[137]
D.W. Hommes, M. Duijvestein, Z. Zelinkova, P.C.F. Stokkers, M.H. Ley, J. Stoker, et al.
Long-term follow-up of autologous hematopoietic stem cell transplantation for severe refractory Crohn's disease.
J Crohn's Colitis, 5 (2011), pp. 543-549
[138]
P. Hasselblatt, K. Drognitz, K. Potthoff, H. Bertz, W. Kruis, C. Schmidt, et al.
Remission of refractory Crohn's disease by high-dose cyclophosphamide and autologous peripheral blood stem cell transplantation.
Aliment Pharmacol Ther, 36 (2012), pp. 725-735
[139]
J.A. Snowden, A. Ansari, S. Sachchithanantham, G. Jackson, N. Thompson, A. Lobo, et al.
Autologous stem cell transplantation in severe treatment-resistant Crohn's disease: long-term follow-up of UK patients treated on compassionate basis.
QJM., 107 (2014), pp. 871-877
[140]
C.J. Hawkey, M. Allez, M.M. Clark, M. Labopin, J.O. Lindsay, E. Ricart, et al.
Autologous hematopoetic stem cell transplantation for refractory Crohn disease a randomized clinical trial.
JAMA, 314 (2015), pp. 2524-2534
[141]
J.A. Snowden, J. Panés, T. Alexander, M. Allez, S. Ardizzone, D. Dierickx, et al.
Autologous haematopoietic stem cell transplantation (AHSCT) in severe Crohn's disease: a review on behalf of ECCO and EBMT.
J Crohns Colitis, 12 (2018), pp. 476-488
[142]
M.A. Ruiz, R.L. Kaiser Junior, M.A. Gouvêa Faria, L.G. de Quadros.
Remission of refractory Crohn's disease after autologous hematopoietic stem cell transplantation.
Rev Bras Hematol Hemoter, 37 (2015), pp. 136-139
[143]
C.K. Brierley, C. Castilla-Llorente, M. Labopin, M. Badoglio, M. Rovira, E. Ricart, et al.
Autologous haematopoietic stem cell transplantation for Crohn's disease: a retrospective survey of long-term outcomes from the European Society for Blood and Marrow Transplantation.
J Crohns Colitis, 12 (2018), pp. 1097-1103
[144]
N. Hernanz, M. Sierra, N. Volpato, L. Núñez-Gómez, F. Mesonero, P. Herrera-Puente, et al.
Autologous haematopoietic stem cell transplantation in refractory Crohn's disease: experience in our centre.
Gastroenterol Hepatol, 42 (2019), pp. 16-22
[145]
J.O. Lindsay, M. Allez, M. Clark, M. Labopin, E. Ricart, G. Rogler, et al.
Autologous stem-cell transplantation in treatment-refractory Crohn's disease: an analysis of pooled data from the ASTIC trial.
Lancet Gastroenterol Hepatol, 2 (2017), pp. 399-406
[146]
M.A. Ruiz, R.L. Kaiser, L.G. de Quadros, L. Piron-Ruiz, T. Peña-Arciniegas, M.A.G. Faria, et al.
Low toxicity and favorable clinical and quality of life impact after non-myeloablative autologous hematopoietic stem cell transplant in Crohn's disease.
BMC Res Notes, 10 (2017), pp. 495
[147]
M.A. Ruiz, R.L. Kaiser, L.P. Ruiz, T. Peña-Arciniegas, L. Castiglioni, P.S. Saran, et al.
Crohn's disease patients effectively mobilize peripheral blood stem cells to perform autologous haematopoietic stem cell transplantation.
bioRxiv, (2018), pp. 348763
[148]
N. Pereira, M. Torres, M. Moraes, I. Alencar, L. Bouzas.
Seleção de doador de medula óssea ou de sangue periférico para o transplante de células-tronco hematopoiéticas.
Diretrizes da Sociedade Brasileira de Transplante de Medula Óssea [Internet], Sociedade Brasileira de Transplante de Medula Óssea (SBTMO), (2012), pp. 15-18
[149]
J.A. Snowden, C. Hawkey, D. Hind, L. Swaby, K. Mellor, R. Emsley, et al.
Autologous stem cell transplantation in refractory Crohn's disease – low intensity therapy evaluation (ASTIClite): study protocols for a multicentre, randomised controlled trial and observational follow up study.
BMC Gastroenterol, 19 (2019), pp. 82
Copyright © 2021. Associação Brasileira de Hematologia, Hemoterapia e Terapia Celular
Idiomas
Hematology, Transfusion and Cell Therapy
Article options
Tools