Review Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 14, 2014; 20(46): 17368-17375
Published online Dec 14, 2014. doi: 10.3748/wjg.v20.i46.17368
Hematopoietic stem cell transplantation for non-malignant gastrointestinal diseases
Abdulbaqi Al-toma, Department of Internal Medicine and Gastroenterology, 3430 EM Nieuwegein, The Netherlands
Petula Nijeboer, Gerd Bouma, Chris JJ Mulder, Department of Gastroenterology, VU University Medical Centre, 1005 MB Amsterdam, The Netherlands
Otto Visser, Department of Hematology, VU University Medical Centre, 1005 MB Amsterdam, The Netherlands
Author contributions: Al-toma A and Nijeboer P have performed literature search and wrote the manuscript; Bouma G, Visser O and Mulder CJJ have critically revised the manuscript, provided scientific input and feedback.
Correspondence to: Abdulbaqi Al-toma, MD, PhD, Gastroenterologist, Department of internal medicine and gastroenterology, St. Antonius hospital, Koekoeslaan 1, 3430 EM Nieuwegein, The Netherlands. a.altoma@antoniusziekenhuis.nl
Telephone: +31-30-6099111 Fax: +31-30-6056357
Received: March 7, 2014
Revised: April 30, 2014
Accepted: September 29, 2014
Published online: December 14, 2014

Abstract

Both, autologous and allogeneic hematopoietic stem cell transplantation (HSCT) can be used to cure or ameliorate a variety of malignant and non-malignant diseases. The rationale behind this strategy is based on the concept of immunoablation using high-dose chemotherapy, with subsequent regeneration of naive T-lymphocytes derived from reinfused hematopoietic progenitor cells. In addition, the use of HSCT allows for the administration of high-dose chemotherapy (whether or not combined with immunomodulating agents such as antithymocyte globulin) resulting in a prompt remission in therapy-refractory patients. This review gives an update of the major areas of successful uses of HSCT in non-malignant gastrointestinal disorders. A Medline search has been conducted and all relevant published data were analyzed. HSCT has been proved successful in treating refractory Crohn’s disease (CD). Patients with refractory celiac disease type II and a high risk of developing enteropathy associated T-cell lymphoma have shown promising improvement. Data concerning HSCT and mesenchymal SCT in end-stage chronic liver diseases are encouraging. In refractory autoimmune gastrointestinal diseases high-dose chemotherapy followed by HSCT seems feasible and safe and might result in long-term improvement of disease activity. Mesenchymal SCT for a selected group of CD is promising and may represent a significant therapeutic alternative in treating fistulas in CD.

Key Words: Hematopoietic stem cell transplantation, Mesenchymal stem cells, Non-malignant gastrointestinal diseases, Celiac disease, Refractory celiac disease, Lymphoma, Crohn’s, Ulcerative colitis, Cirrhosis

Core tip: Hematopoietic stem cell transplantation (HSCT) can be used to treat malignant and non-malignant diseases. This therapeutic modality is based on using immunoablation followed by reinfusion of hematopoietic progenitor cells to regenerate naive T-lymphocytes. HSCT and mesenchymal SCT have been proved successful in treating refractory inflammatory conditions such as Crohn’s disease and refractory celiac disease type II. The ultimate target of aggressively treating this type of celiac disease is to prevent development of lymphoma. Data in end-stage liver diseases are also encouraging.



INTRODUCTION

In cancer patients, myeloablative doses of radiation or high dose chemotherapy (HDC) or both have been used followed by infusion of autologous hematopoietic cells to restore bone marrow function[1-3].

Recently, HDC followed by hematopoietic stem cell transplantation (HSCT) has been applied increasingly thanks to the availability of supportive measures, such as availability of antibiotics to prevent or treat infections during marrow aplasia, improvement in transfusion support and the use of hematopoietic growth factors to “mobilize” HSCs to shorten the recovery time of marrow function[1]. In addition, an important progress has been achieved in manipulating, mobilizing bone marrow stem cells and recruitment of hematopoietic stem cells (HSCs) from the peripheral blood[3,4].

The peripheral blood contains HSCs[4-6]; these cells can be easily harvested and then utilized to hasten hematological recovery after ablative therapy[7]. Specific chemotherapeutic regimens, granulocyte colony stimulating factor and/or new agents such as Plerixafor effectively increase the number of HSCs in the peripheral blood providing adequate autologous stem cell harvest[8,9].

In the last two decades, HSCT is gaining wider acceptance in the management of difficult to treat autoimmune diseases[10-16].

HSCT has been conducted in treating refractory Crohn’s disease (CD). More recently, mesenchymal SCT (MSCT) has been explored in the management of CD complicated by fistulas. Encouraging results have been reported. In a group of refractory celiac disease, we have reported impressive results using autologous SCT (auto-SCT).

The efficacy and safety of HSCT and MSCT are being evaluated in patients with end-stage chronic liver diseases, both viral and autoimmune-induced, and the preliminary results seem to be promising. So far, limited data have been published about auto-SCT in autoimmune liver diseases.

We tried here to provide an overview of the experience gained thus far in treating non-malignant gastrointestinal diseases.

BACKGROUND

HSCs are capable of regenerating immune cells; this characteristic provides the theoretical possibility of resetting the immune system without autoimmunity. Meanwhile, MSCs have immunosuppressive effect, which might be beneficial in different inflammatory disorders[17,18].

The possible mechanism for using stem cell therapy for inflammatory disorders is to induce a state of immunoablation using HDC followed by reinfusing hematopoietic progenitor cells to achieve regeneration of naive T-lymphocytes derived. This procedure could reset the immune system after first eliminating the patient’s own system by preparative chemo[-immuno] therapy[1].

Animal studies and the use of HSCT in experimental forms of autoimmune diseases have contributed significantly to the knowledge and subsequently the application of auto-HSCT in autoimmune diseases[19-21].

Clinical observations in patients with autoimmune diseases such as severe systemic sclerosis, who received HSCT to treat concomitant hematological diseases, paralleled the experience obtained from animal studies[22,23].

The limited therapeutic options for severe, uncontrolled autoimmune diseases made it necessary to explore HSCT as a treatment modality. To be acceptable for non-oncological indications this modality should have an acceptable mortality and morbidity rate. Auto-HSCT has gained priority over allo-HSCT because it is associated with low morbidity and mortality rates both in oncology[24,25] and in immune disorders (7%)[10].

For non-malignant conditions auto-HSCT may be applied using either myeloablative or non-myeloablative regimens[13]. Myeloablation results in an irreversible aplasia of the bone marrow; therefore HSCs should be provided to restore the marrow function. On the other hand, non-myeloablation is designed for autoimmune diseases; following such a regimen, hematopoietic recovery will occur without infusion of HSCs[13].

MSCs are distinct lineage of stem cells, having no unique phenotypic marker, representing 1 in 10000 nucleated cells in the bone marrow[26]. Different tissues can provide the source of these cells, e.g., bone marrow, skeletal muscle, adipose tissue, synovial membranes, umbilical cord blood and placenta[27].

MSCs give rise to many cell lineages, thus promoting regeneration of damaged tissue in vivo[28]. Also these cells exert important immunomodulatory functions[29], can regenerate clonally[30] and exhibit anti-proliferative and anti-inflammatory properties, making them candidates for treatment of immune-mediated inflammatory diseases[31].

APPLICATION IN INFLAMMATORY BOWEL DISEASE

CD is a relapsing-remitting disorder with enhanced T-helper cell reaction[32-34]. HSCT is considered as a valuable option in the treatment of CD because it has been shown to be effective in treating autoimmune disorders sharing a similar pathogenic background.

The mechanism of positive effect of HSCT in not entirely clear; however allo-HSCT may change the genetic constitution that predisposes to CD. On the other hand, auto-HSCT helps to eliminate committed lymphocytes and also facilitates relatively safe use of immunosuppression[35].

The first evidence for using stem cell therapy in refractory CD came from reports showing that CD improves after stem cell therapy for other concomitant disorders[36].

In 1993 Drakos et al[37] reported an improvement of a patient with CD who received allo-HSCT for a malignant lymphoma. Five years later, allo-HSCT was performed in six non-Hodgkin lymphoma (NHL) patients who also had concomitant CD. Three patients remained in remission after withdrawal of immunosuppressives[38].

In a report by Ditschkowski et al[39] Eleven inflammatory bowel disease (IBD) patients (seven CD and four ulcerative colitis) underwent allo-HSCT in connection with various hematology disorders. Ten transplant recipients stayed in remission, follow up 34 mo.

Numerous case reports and case series have been published dealing with the clinical response of CD who received auto-SCT to treat concomitant conditions[40-43]. One such example is a young patient diagnosed with CD and needed intensive treatment with anti-inflammatory and immunosuppressive agents[40]. He remained in remission after auto-HSCT for NHL.

Table 1 summarizes the literature on indirect evidence for effectiveness of HSCT in IBD. These data indicate that HSCT might benefit CD but this benefit is not universal.

Table 1 Indirect evidence for efficacy of hematopoietic stem cell transplantation in inflammatory bowel disease.
Ref.SCTOriginal indicationNo./type IBDConditioningResults/remarks
Drakos et al[37]Allo-SCTNHL1/fistulizing CDBCNU/Etoposide, Cy, ara-C and MelphalanRemission/6 mo follow up
Kashyap et al[40]Auto-SCTNHL1/Perianal CDCy,VP16+TBIRemission 7 yr/Crohn’s at age 13 yr ASCT at 20 yr
Lopez-Cubero et al[38]Allo-SCTLeukemia5/CDCy/TBI3 pts has lasting remission
Söderholm et al[41]Auto-SCTAML1/Fistulizing CDCy/TBIRemission 5 yr
Ditschkowski et al[39]Auto-SCTAML/CML/MDS11/7 CD +4 UCVariousRemission in 10/Follow up 34 mo
Anumakonda et al[42]Auto-SCTNHL1/CDCHOPRemission 8 yr/Relapse after 8 yr

In 2003, Burt et al[44] and Craig et al[45] provided the first direct evidence for the efficacy of HSCT for CD. In 2 patients diarrhea resolved following transplantation. Crohn’s disease activity index (CDAI) is normalized. Simultaneously, another group reported a complete remission in one patient using the same conditioning regimen[46].

Another report by Oyama et al[47] has provided the results of using HSCT refractory CD. A total of 12 patients have been treated, eleven of them remained in remission [follow-up of 18.5 mo (range, 7-37 mo)].

In 2010 Burt et al[48] have presented their experience in using auto-HSCT for IBD. Twenty-four patients with CD were treated. 91% stayed in remission for 1 year post transplantation. A similar outcome was reported by another group who treated 10 CD patients (remission rates of 80% after 1 year)[49].

Hasselblatt et al[50] have reported the outcome of 12 patients with refractory CD treated with auto-HSCT. Five patients achieved a clinical and endoscopic remission within 6 mo after auto-HSCT. However, relapses occurred in 7 patients, but disease activity was mild and could be controlled by low-dose corticosteroids and immunosuppressive therapy.

Allo-HSCT for CD has also been performed; in 2009 a report on a successful use of allo-HSCT in treating a 9-year-old child was published[51]. The investigators have hypothesized that a nonsense mutation in the IL10RB gene is probably the genetic cause of IBD in the affected patient and given the severity of the disease, they considered allo-HSCT as treatment. The patient underwent conditioning with the use of alemtuzumab (1 mg/kg), fludarabine (180 mg/m2), treosulfan (42 mg/m2), and thiotepa (10 mg/kg). Anal fistulas resolved and the patient has remained in remission more than a year after HSCT.

MSCs have been tested for fistulizing and also luminal CD[52]. In fistulizing disease, the differentiation potential of these cells is thought to be necessary to achieve closure of fistulas, while their immunosuppressive effect is the rationale for their use in luminal disease.

Rectovaginal fistulas are difficult to treat. García-Olmo et al[52] used lipoaspirate to provide stem cells in a patient with rectovaginal fistula. Subsequently, the patient had no complains related to the fistula. The same group has tested the usefulness and safety of MSCT in treating fistulas[53]. Nine fistulas were treated. Six fistulas were considered healed (75%), (follow-up 22 mo; range 12-30).

Forbes et al[54] published the result of a phase II study using allo-MSCs for refractory luminal CD. An open-label, multicenter study included 16 patients (21-55 years old; 6 men) with infliximab-or adalimumab-refractory, endoscopically confirmed, active luminal CD (CDAI > 250). Subjects were given intravenous infusions of allogeneic MSCs (2 × 106 cells/kg body weight) weekly for 4 wk. The primary end point was clinical response (decrease in CDAI > 100 points) 42 d after the first MSC administration; secondary end points were clinical remission (CDAI, < 150), endoscopic improvement [a CD endoscopic index of severity (CDEIS) value, < 3 or a decrease by > 5], quality of life, level of C-reactive protein, and safety. Among the 15 patients who completed the study, the mean CDAI score was reduced from 370 (median, 327; range, 256-603) to 203 (median, 129) at day 42 (P < 0.0001). Twelve patients had a clinical response (80%; 95%CI: 72%-88%; mean reduction in CDAI, 211; range 102-367), 8 had clinical remission (53%; range, 43%-64%; mean CDAI at day 42, 94; range, 44-130). Seven patients had endoscopic improvement (47%), for whom the mean CDEIS scores decreased from 21.5 (range, 3.3-33) to 11.0 (range, 0.3-18.5).

Table 2 summarizes the literatures on direct evidence for effectiveness of HSCT and MSCT in IBD. We may conclude that auto-HSCT is feasible, safe and also effective in achieving and maintaining remission in CD. Furthermore, MSCT may represent a significant therapeutic alternative in treating CD. Still larger randomized or observational multicenter trials are required to confirm the efficacy of this therapy.

Table 2 Direct evidence for efficacy of hematopoietic stem cell transplantation and mesenchymal stem cell transplantation in inflammatory bowel disease.
Ref.SCT/MSCTNo. treated/type IBDConditioningResults
Burt et al[44]Auto-HSCT2/Crohn’s, fistulas, CDAI > 250Cy + ATGCDAI,CRP, albumin normalized; remission > 1 yr
Kreisel et al[46]Auto-HSCT1/ileocolic Crohn’sCy + ATG9 mo complete remission
García-Olmo et al[52]Auto-HSCT Adipose tissue1/recurrent rectovaginal fistulasNone necessary3 mo asymptomatic
García-Olmo et al[53]MSCT5/fistulasNone necessaryFollow up 22 mo; 75% healing
Oyama et al[47]Auto-HSCT12/fistula 6, stenosis 3, perianal 4; most have surgeryCy + ATG11 sustained remission (CDAI < 150) after median 18.5 mo; 1 relapse > 15 mo
Cassinotti et al[49]Auto-HSCT4/stenosis, fistulas, bleedingCy + ATGAll clinical remission at 3 m, 1 relapse after 4 mo; Sustained remission in 3 after 16.5 m o
Glocker et al[51]Allo-HSCT1/perianal fistulaAlemtuzumab, fludarabine, treosulfan, thiotepaRemission 2 yr; 9-year-old child
Burt et al[48]Auto-HSCT24/Crohn’sCy + ATG91% remission 1 yr, 57% for 3 yr, 19% for 5 yr
Cassinotti et al[49]Auto-HSCT10/Crohn’sCy + ATG80% remission > 1 yr, 40% > 3 yr and 30% > 5 yr
Hasselblatt et al[50]Auto-HSCT12/Crohn’sCy + G-CSF5 complete remission within 6 m, relapses occurred in 7. Mean follow-up 3.1 yr (range 0.5-10.3)
Forbes et al[54]MSCT, weekly for 4 wk16/Crohn’s disease (CDAI > 250)None necessaryCDAI declined from median 327 to 129 (day 42). Clinical response 12 pts; Clinical remission 8 pts. Endoscopic improvement in 7 pts

Concerning ulcerative colitis, improvement of disease activity after stem cell transplantation for other indications has been reported. Four patients remained in remission after receiving auto-HSCT for leukemia[39]. Two patients with ulcerative colitis remained in remission after they underwent an auto-HSCT in connection with leukemia[55].

HSCT IN REFRACTORY CELIAC DISEASE

In celiac disease, gluten-derived peptides bind to HLA-DQ2 and HLA-DQ8 molecules on antigen-presenting cells[56]. These molecules then in turn present these peptides and induce a CD4+ T cell response which results in an inflammatory response[57-59]. In a small minority (0.5%-1%) of adult celiac patients symptoms persist despite strict adherence to a gluten free diet[60]. After excluding other causes of villous atrophy, these patients are diagnosed with refractory celiac disease (RCD).

These refractory patients are further subdivided into 2 types depending on immunophenotyping by flow cytometry[61-65] of the intraepithelial lymphocytes (IEL): patients with normal intraepithelial lymphocytes are classified as (RCD-I), while those those with more than 20% aberrant intraepithelial lymphocytes are classified as (RCD II)[59]. Aberrant IEL are characterized by T-cell specific CD3 in their cytoplasm, yet lack surface expression of CD3 and CD8. RCD-II is usually resistant to any known therapy[66-71]. This entity frequently transforms into an aggressive enteropathy-type-associated T cell lymphoma (EATL)[71-73].

The safety and efficacy of auto-SCT in RCD type II has been tested[74,75]. Between 2004 and 2010, 18 RCD-II patients were evaluated for auto-HSCT as a consequence of unresponsiveness or partial response to cladribine therapy[74]. Thirteen patients underwent conditioning and transplantation. One patient died due to transplantation-related complications. In the majority of patients, clinical improvement was observed after auto-HSCT. Complete histological remission was achieved in 5 patients. One patient developed EATL despite auto-HSCT after a follow-up of four years. Recent, yet unpublished, data show excellent overall long-term survival of 87 mo.

Auto-HSCT seems safe and might result in a long-term clinical remission with a better quality of life in RCD-II patients. Moreover, this treatment strategy seems to prevent or delay the development of EATL. These observations argue for an aggressive therapeutic approach for those RCD-II patients eligible for auto-HSCT.

HSCT IN CHRONIC LIVER DISEASES

Liver transplantation is the standard treatment in advanced liver cirrhosis, however it has significant shortcomings, e.g., a long waiting list, expensive and numerous complications[76-78]. In addition, the vastly increasing prevalence of end-stage liver disease without a parallel increase in donor livers has precipitated a search for alternative therapies.

Circulating stem cells can differentiate into mature hepatocytes or cholangiocytes in vivo[79]. MSCs differentiate into functional hepatocyte-like cells in animal experiments[79]. MSCs can protect against experimental liver fibrosis in rats[80] and also result in regression of fibrosis in mice[81]. Therefore, stem cell therapy might be useful in treating chronic liver disease such as liver cirrhosis.

Patients with liver cirrhosis who had been treated with MSCs had a good clinical outcome with improvement in de Model End Stage Liver Disease score and quality of life[82-84].

The largest series thus far reported comes from Egypt[85] where auto-HSCT was conducted in 48 patients, 36 with chronic end-stage hepatitis C-induced liver disease and 12 with end-stage autoimmune liver disease. Treatment was generally well tolerated; 10 patients (20.8%) died during 12 mo of follow-up and two had developed portal hypertension. There was clinical and biochemical improvement. Patients had a statistically significant decrease in ascites and marked improvement in albumin, bilirubin, clotting status, and aminotransferase levels. However, the authors raise concern about the development of portal hypertension after HSCT. They further postulated that the incidence of this serious complication can be minimized by employing hepatic artery infusion as the sole approach to transplantation.

SAFETY ISSUES

The age limit of stem cell recipients has changed over time due to the availability of good supportive care. For RCD-II, we use an age limit of seventy years[74,75].

Regimen-related toxicity can be serious; this is particularly true for recipients of allo-HSCT. Opportunistic infections and pancytopenia occur much less frequently after auto-HSCT than allo-HSCT recipients.

Intensive therapies including HSCT aim to reintegrate patients expeditiously in their work and social activities. Usually a good quality of life can be gained within few months after HSCT[86].

For MSCs, clinical risk data largely stem from studies in steroid-refractory graft versus host disease. Other than the possibility of an increased short-term risk of pneumonia in these highly immunocompromised patients, MSCs have been considered safe[87]. There are no reports of increased risk from other infections or tumors, despite concerns based on theoretical mechanistic grounds[88]. No significant adverse effects have been reported from CD studies of locally administered allo-MSCs[89], intravenous auto-MSCs[90], or intravenous allo-MSCs[91].

FUTURE PERSPECTIVES

Rigorous safety evaluations of new therapies are essential. Future prospective observational multicenter studies with standardized inclusion criteria, conditioning regimens, and if possible supportive care, are still necessary to substantiate the already published data. Still long term follow up data concerning response are lacking. Criteria for the selection and the timing of providing stem cell transplantation in patients with chronic inflammatory diseases and premalignant conditions are strongly needed. Standardization of immunosuppression, optimizing conditioning regimens and also integration of immunotherapy in treatment protocols are areas for future research. There is still much to learn and understand about MSC function, immunopathogenic mechanisms in treating different disease conditions and standardization of preparation methods.

In gastroenterology, it seems that HSCT and MSCT gain steadily, and deservedly, more grounds in the arsenal of treatment of inflammatory disease and premalignant conditions, like IBD, refractory celiac disease and liver cirrhosis.

Footnotes

P- Reviewer: Huang YH S- Editor: Qi Y L- Editor: A E- Editor: Ma S

References
1.  Storb RF, Champlin R, Riddell SR, Murata M, Bryant S, Warren EH. Non-myeloablative transplants for malignant disease. Hematology Am Soc Hematol Educ Program. 2001;375-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 73]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
2.  Thomas ED, Ferrebee JW. Prolonged storage of marrow and its use in the treatment of radiation injury. Transfusion. 1962;2:115-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
3.  Zhang M, Huang B. The multi-differentiation potential of peripheral blood mononuclear cells. Stem Cell Res Ther. 2012;3:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
4.  Barr RD, Whang-Peng J, Perry S. Hemopoietic stem cells in human peripheral blood. Science. 1975;190:284-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 106]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
5.  Abrams RA, McCormack K, Bowles C, Deisseroth AB. Cyclophosphamide treatment expands the circulating hematopoietic stem cell pool in dogs. J Clin Invest. 1981;67:1392-1399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
6.  de Haan G, Van Zant G. Stem cells from birth to death: The history and the future. J Am Aging Assoc. 2002;25:79-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
7.  Pelus LM, Farag SS. Increased mobilization and yield of stem cells using plerixafor in combination with granulocyte-colony stimulating factor for the treatment of non-Hodgkin’s lymphoma and multiple myeloma. Stem Cells Cloning. 2011;4:11-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
8.  Gabús R, Borelli G, Ferrando M, Bódega E, Citrín E, Jiménez CO, Alvarez R. Mobilization of hematopoietic progenitor cells with granulocyte colony stimulating factors for autologous transplant in hematologic malignancies: a single center experience. Rev Bras Hematol Hemoter. 2011;33:410-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
9.  Sheridan WP, Begley CG, Juttner CA, Szer J, To LB, Maher D, McGrath KM, Morstyn G, Fox RM. Effect of peripheral-blood progenitor cells mobilised by filgrastim (G-CSF) on platelet recovery after high-dose chemotherapy. Lancet. 1992;339:640-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 610]  [Cited by in F6Publishing: 620]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
10.  Gratwohl A, Passweg J, Bocelli-Tyndall C, Fassas A, van Laar JM, Farge D, Andolina M, Arnold R, Carreras E, Finke J. Autologous hematopoietic stem cell transplantation for autoimmune diseases. Bone Marrow Transplant. 2005;35:869-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 141]  [Article Influence: 7.4]  [Reference Citation Analysis (1)]
11.  Kapoor S, Wilson AG, Sharrack B, Lobo A, Akil M, Sun L, Dalley CD, Snowden JA. Haemopoietic stem cell transplantation--an evolving treatment for severe autoimmune and inflammatory diseases in rheumatology, neurology and gastroenterology. Hematology. 2007;12:179-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
12.  Nash RA, McSweeney PA, Crofford LJ, Abidi M, Chen CS, Godwin JD, Gooley TA, Holmberg L, Henstorf G, LeMaistre CF. High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: long-term follow-up of the US multicenter pilot study. Blood. 2007;110:1388-1396.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Burt RK, Marmont A, Oyama Y, Slavin S, Arnold R, Hiepe F, Fassas A, Snowden J, Schuening F, Myint H. Randomized controlled trials of autologous hematopoietic stem cell transplantation for autoimmune diseases: the evolution from myeloablative to lymphoablative transplant regimens. Arthritis Rheum. 2006;54:3750-3760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 86]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
14.  Oyama Y, Barr WG, Statkute L, Corbridge T, Gonda EA, Jovanovic B, Testori A, Burt RK. Autologous non-myeloablative hematopoietic stem cell transplantation in patients with systemic sclerosis. Bone Marrow Transplant. 2007;40:549-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 57]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
15.  Jayne D, Passweg J, Marmont A, Farge D, Zhao X, Arnold R, Hiepe F, Lisukov I, Musso M, Ou-Yang J. Autologous stem cell transplantation for systemic lupus erythematosus. Lupus. 2004;13:168-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 162]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
16.  Snowden JA, Saccardi R, Allez M, Ardizzone S, Arnold R, Cervera R, Denton C, Hawkey C, Labopin M, Mancardi G. Haematopoietic SCT in severe autoimmune diseases: updated guidelines of the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2012;47:770-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 210]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
17.  Gregorini M, Maccario R, Avanzini MA, Corradetti V, Moretta A, Libetta C, Esposito P, Bosio F, Dal Canton A, Rampino T. Antineutrophil cytoplasmic antibody-associated renal vasculitis treated with autologous mesenchymal stromal cells: evaluation of the contribution of immune-mediated mechanisms. Mayo Clin Proc. 2013;88:1174-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
18.  Pak J. Regeneration of human bones in hip osteonecrosis and human cartilage in knee osteoarthritis with autologous adipose-tissue-derived stem cells: a case series. J Med Case Rep. 2011;5:296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 167]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
19.  van Bekkum DW. Stem cell transplantation in experimental models of autoimmune disease. J Clin Immunol. 2000;20:10-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
20.  van Gelder M, Mulder AH, van Bekkum DW. Treatment of relapsing experimental autoimmune encephalomyelitis with largely MHC-matched allogeneic bone marrow transplantation. Transplantation. 1996;62:810-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
21.  van Bekkum DW. Conditioning regimens for the treatment of experimental arthritis with autologous bone marrow transplantation. Bone Marrow Transplant. 2000;25:357-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 43]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
22.  Machold KP, Smolen JS. Stem cell transplantation: limits and hopes. Ann Rheum Dis. 2001;60:548-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
23.  Verburg RJ, Flierman R, Sont JK, Ponchel F, van Dreunen L, Levarht EW, Welling MM, Toes RE, Isaacs JD, van Laar JM. Outcome of intensive immunosuppression and autologous stem cell transplantation in patients with severe rheumatoid arthritis is associated with the composition of synovial T cell infiltration. Ann Rheum Dis. 2005;64:1397-1405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
24.  Mijovic A, Pamphilon D. Harvesting, processing and inventory management of peripheral blood stem cells. Asian J Transfus Sci. 2007;1:16-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
25.  Pelus LM. Peripheral blood stem cell mobilization: new regimens, new cells, where do we stand. Curr Opin Hematol. 2008;15:285-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 68]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25:2739-2749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1697]  [Cited by in F6Publishing: 1616]  [Article Influence: 95.1]  [Reference Citation Analysis (0)]
27.  Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11055]  [Cited by in F6Publishing: 11799]  [Article Influence: 694.1]  [Reference Citation Analysis (1)]
28.  Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2360]  [Cited by in F6Publishing: 2515]  [Article Influence: 167.7]  [Reference Citation Analysis (0)]
29.  Gonzalez-Rey E, Anderson P, González MA, Rico L, Büscher D, Delgado M. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut. 2009;58:929-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 482]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
30.  Dazzi F, van Laar JM, Cope A, Tyndall A. Cell therapy for autoimmune diseases. Arthritis Res Ther. 2007;9:206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 60]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
31.  Panés J, Salas A. Mechanisms underlying the beneficial effects of stem cell therapies for inflammatory bowel diseases. Gut. 2009;58:898-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
32.  Timmer A. Environmental influences on inflammatory bowel disease manifestations. Lessons from epidemiology. Dig Dis. 2003;21:91-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
33.  Hughes AL. Consistent across-tissue signatures of differential gene expression in Crohn’s disease. Immunogenetics. 2005;57:709-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
34.  Matricon J, Barnich N, Ardid D. Immunopathogenesis of inflammatory bowel disease. Self Nonself. 2010;1:299-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 150]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
35.  Brittan M, Hunt T, Jeffery R, Poulsom R, Forbes SJ, Hodivala-Dilke K, Goldman J, Alison MR, Wright NA. Bone marrow derivation of pericryptal myofibroblasts in the mouse and human small intestine and colon. Gut. 2002;50:752-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 205]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
36.  Hawkey CJ, Snowden JA, Lobo A, Beglinger C, Tyndall A. Stem cell transplantation for inflammatory bowel disease: practical and ethical issues. Gut. 2000;46:869-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
37.  Drakos PE, Nagler A, Or R. Case of Crohn’s disease in bone marrow transplantation. Am J Hematol. 1993;43:157-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
38.  Lopez-Cubero SO, Sullivan KM, McDonald GB. Course of Crohn’s disease after allogeneic marrow transplantation. Gastroenterology. 1998;114:433-440.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Ditschkowski M, Einsele H, Schwerdtfeger R, Bunjes D, Trenschel R, Beelen DW, Elmaagacli AH. Improvement of inflammatory bowel disease after allogeneic stem-cell transplantation. Transplantation. 2003;75:1745-1747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
40.  Kashyap A, Forman SJ. Autologous bone marrow transplantation for non-Hodgkin’s lymphoma resulting in long-term remission of coincidental Crohn’s disease. Br J Haematol. 1998;103:651-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 81]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
41.  Söderholm JD, Malm C, Juliusson G, Sjödahl R. Long-term endoscopic remission of crohn disease after autologous stem cell transplantation for acute myeloid leukaemia. Scand J Gastroenterol. 2002;37:613-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 35]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Anumakonda V, Hayee B, Chung-Faye G. Remission and relapse of Crohn’s disease following autologous haematopoietic stem cell transplantation for non-Hodgkin’s lymphoma. Gut. 2007;56:1325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
43.  Sonwalkar SA, James RM, Ahmad T, Zhang L, Verbeke CS, Barnard DL, Jewell DP, Hull MA. Fulminant Crohn’s colitis after allogeneic stem cell transplantation. Gut. 2003;52:1518-1521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
44.  Burt RK, Traynor A, Oyama Y, Craig R. High-dose immune suppression and autologous hematopoietic stem cell transplantation in refractory Crohn disease. Blood. 2003;101:2064-2066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 79]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
45.  Craig RM, Traynor A, Oyama Y, Burt RK. Hematopoietic stem cell transplantation for severe Crohn’s disease. Bone Marrow Transplant. 2003;32 Suppl 1:S57-S59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
46.  Kreisel W, Potthoff K, Bertz H, Schmitt-Graeff A, Ruf G, Rasenack J, Finke J. Complete remission of Crohn’s disease after high-dose cyclophosphamide and autologous stem cell transplantation. Bone Marrow Transplant. 2003;32:337-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
47.  Oyama Y, Craig RM, Traynor AE, Quigley K, Statkute L, Halverson A, Brush M, Verda L, Kowalska B, Krosnjar N. Autologous hematopoietic stem cell transplantation in patients with refractory Crohn’s disease. Gastroenterology. 2005;128:552-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 190]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
48.  Burt RK, Craig RM, Milanetti F, Quigley K, Gozdziak P, Bucha J, Testori A, Halverson A, Verda L, de Villiers WJ. Autologous nonmyeloablative hematopoietic stem cell transplantation in patients with severe anti-TNF refractory Crohn disease: long-term follow-up. Blood. 2010;116:6123-6132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 139]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
49.  Cassinotti A, Annaloro C, Ardizzone S, Onida F, Della Volpe A, Clerici M, Usardi P, Greco S, Maconi G, Porro GB. Autologous haematopoietic stem cell transplantation without CD34+ cell selection in refractory Crohn’s disease. Gut. 2008;57:211-217.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Hasselblatt P, Drognitz K, Potthoff K, Bertz H, Kruis W, Schmidt C, Stallmach A, Schmitt-Graeff A, Finke J, Kreisel W. Remission of refractory Crohn’s disease by high-dose cyclophosphamide and autologous peripheral blood stem cell transplantation. Aliment Pharmacol Ther. 2012;36:725-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
51.  Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schäffer AA, Noyan F, Perro M, Diestelhorst J, Allroth A, Murugan D. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033-2045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1121]  [Cited by in F6Publishing: 1015]  [Article Influence: 67.7]  [Reference Citation Analysis (0)]
52.  García-Olmo D, García-Arranz M, García LG, Cuellar ES, Blanco IF, Prianes LA, Montes JA, Pinto FL, Marcos DH, García-Sancho L. Autologous stem cell transplantation for treatment of rectovaginal fistula in perianal Crohn’s disease: a new cell-based therapy. Int J Colorectal Dis. 2003;18:451-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 216]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
53.  García-Olmo D, García-Arranz M, Herreros D, Pascual I, Peiro C, Rodríguez-Montes JA. A phase I clinical trial of the treatment of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48:1416-1423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 604]  [Cited by in F6Publishing: 556]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
54.  Forbes GM, Sturm MJ, Leong RW, Sparrow MP, Segarajasingam D, Cummins AG, Phillips M, Herrmann RP. A phase 2 study of allogeneic mesenchymal stromal cells for luminal Crohn’s disease refractory to biologic therapy. Clin Gastroenterol Hepatol. 2014;12:64-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 238]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
55.  Tyndall A, Gratwohl A. Blood and marrow stem cell transplants in autoimmune disease. A consensus report written on behalf of the European League Against Rheumatism (EULAR) and the European Group for Blood and Marrow Transplantation (EBMT). Br J Rheumatol. 1997;36:390-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 76]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
56.  Tjon JM, van Bergen J, Koning F. Celiac disease: how complicated can it get? Immunogenetics. 2010;62:641-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 110]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
57.  Sollid LM. Coeliac disease: dissecting a complex inflammatory disorder. Nat Rev Immunol. 2002;2:647-655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 686]  [Cited by in F6Publishing: 640]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
58.  Vader W, Stepniak D, Kooy Y, Mearin L, Thompson A, van Rood JJ, Spaenij L, Koning F. The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses. Proc Natl Acad Sci USA. 2003;100:12390-12395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 267]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
59.  Nijeboer P, van Wanrooij RL, Tack GJ, Mulder CJ, Bouma G. Update on the diagnosis and management of refractory coeliac disease. Gastroenterol Res Pract. 2013;2013:518483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 56]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
60.  Daum S, Cellier C, Mulder CJ. Refractory coeliac disease. Best Pract Res Clin Gastroenterol. 2005;19:413-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 199]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
61.  van Wanrooij RL, Schreurs MW, Bouma G, von Blomberg BM, Tack GJ, Verbeek WH, Mulder CJ. Accurate classification of RCD requires flow cytometry. Gut. 2010;59:1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
62.  Cellier C, Patey N, Mauvieux L, Jabri B, Delabesse E, Cervoni JP, Burtin ML, Guy-Grand D, Bouhnik Y, Modigliani R. Abnormal intestinal intraepithelial lymphocytes in refractory sprue. Gastroenterology. 1998;114:471-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 238]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
63.  Eiras P, Roldán E, Camarero C, Olivares F, Bootello A, Roy G. Flow cytometry description of a novel CD3-/CD7+ intraepithelial lymphocyte subset in human duodenal biopsies: potential diagnostic value in coeliac disease. Cytometry. 1998;34:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Patey-Mariaud De Serre N, Cellier C, Jabri B, Delabesse E, Verkarre V, Roche B, Lavergne A, Brière J, Mauvieux L, Leborgne M. Distinction between coeliac disease and refractory sprue: a simple immunohistochemical method. Histopathology. 2000;37:70-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 121]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
65.  Verbeek WH, Goerres MS, von Blomberg BM, Oudejans JJ, Scholten PE, Hadithi M, Al-Toma A, Schreurs MW, Mulder CJ. Flow cytometric determination of aberrant intra-epithelial lymphocytes predicts T-cell lymphoma development more accurately than T-cell clonality analysis in Refractory Celiac Disease. Clin Immunol. 2008;126:48-56.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Goerres MS, Meijer JW, Wahab PJ, Kerckhaert JA, Groenen PJ, Van Krieken JH, Mulder CJ. Azathioprine and prednisone combination therapy in refractory coeliac disease. Aliment Pharmacol Ther. 2003;18:487-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 100]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
67.  Wahab PJ, Crusius JB, Meijer JW, Uil JJ, Mulder CJ. Cyclosporin in the treatment of adults with refractory coeliac disease--an open pilot study. Aliment Pharmacol Ther. 2000;14:767-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 66]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
68.  Mulder CJ, Wahab PJ, Meijer JW, Metselaar E. A pilot study of recombinant human interleukin-10 in adults with refractory coeliac disease. Eur J Gastroenterol Hepatol. 2001;13:1183-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 59]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
69.  Cellier C, Delabesse E, Helmer C, Patey N, Matuchansky C, Jabri B, Macintyre E, Cerf-Bensussan N, Brousse N. Refractory sprue, coeliac disease, and enteropathy-associated T-cell lymphoma. French Coeliac Disease Study Group. Lancet. 2000;356:203-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 444]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
70.  Mauriño E, Niveloni S, Cherñavsky A, Pedreira S, Mazure R, Vazquez H, Reyes H, Fiorini A, Smecuol E, Cabanne A. Azathioprine in refractory sprue: results from a prospective, open-label study. Am J Gastroenterol. 2002;97:2595-2602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
71.  Al-Toma A, Goerres MS, Meijer JW, von Blomberg BM, Wahab PJ, Kerckhaert JA, Mulder CJ. Cladribine therapy in refractory celiac disease with aberrant T cells. Clin Gastroenterol Hepatol. 2006;4:1322-1327; quiz 1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 98]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
72.  Delabie J, Holte H, Vose JM, Ullrich F, Jaffe ES, Savage KJ, Connors JM, Rimsza L, Harris NL, Müller-Hermelink K. Enteropathy-associated T-cell lymphoma: clinical and histological findings from the international peripheral T-cell lymphoma project. Blood. 2011;118:148-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 209]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
73.  Tack GJ, Verbeek WH, Al-Toma A, Kuik DJ, Schreurs MW, Visser O, Mulder CJ. Evaluation of Cladribine treatment in refractory celiac disease type II. World J Gastroenterol. 2011;17:506-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 52]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
74.  Al-toma A, Visser OJ, van Roessel HM, von Blomberg BM, Verbeek WH, Scholten PE, Ossenkoppele GJ, Huijgens PC, Mulder CJ. Autologous hematopoietic stem cell transplantation in refractory celiac disease with aberrant T cells. Blood. 2007;109:2243-2249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 112]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
75.  Tack GJ, Wondergem MJ, Al-Toma A, Verbeek WH, Schmittel A, Machado MV, Perri F, Ossenkoppele GJ, Huijgens PC, Schreurs MW. Auto-SCT in refractory celiac disease type II patients unresponsive to cladribine therapy. Bone Marrow Transplant. 2011;46:840-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
76.  Said A, Lucey MR. Liver transplantation: an update. Curr Opin Gastroenterol. 2006;22:272-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
77.  Karakayali H, Boyvat F, Coskun M, Isiklar I, Sözen H, Filik L, Yilmaz U, Gür G, Haberal M. Venous complications after orthotopic liver transplantation. Transplant Proc. 2006;38:604-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
78.  Dondero F, Farges O, Belghiti J, Francoz C, Sommacale D, Durand F, Sauvanet A, Janny S, Varma D, Vilgrain V. A prospective analysis of living-liver donation shows a high rate of adverse events. J Hepatobiliary Pancreat Surg. 2006;13:117-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
79.  Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, Wang X, Finegold M, Weissman IL, Grompe M. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med. 2000;6:1229-1234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1764]  [Cited by in F6Publishing: 1845]  [Article Influence: 76.9]  [Reference Citation Analysis (0)]
80.  Zhao DC, Lei JX, Chen R, Yu WH, Zhang XM, Li SN, Xiang P. Bone marrow-derived mesenchymal stem cells protect against experimental liver fibrosis in rats. World J Gastroenterol. 2005;11:3431-3440.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Fang B, Shi M, Liao L, Yang S, Liu Y, Zhao RC. Systemic infusion of FLK1(+) mesenchymal stem cells ameliorate carbon tetrachloride-induced liver fibrosis in mice. Transplantation. 2004;78:83-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 227]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
82.  Gordon MY, Levicar N, Pai M, Bachellier P, Dimarakis I, Al-Allaf F, M’Hamdi H, Thalji T, Welsh JP, Marley SB. Characterization and clinical application of human CD34+ stem/progenitor cell populations mobilized into the blood by granulocyte colony-stimulating factor. Stem Cells. 2006;24:1822-1830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 232]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
83.  Mohamadnejad M, Alimoghaddam K, Mohyeddin-Bonab M, Bagheri M, Bashtar M, Ghanaati H, Baharvand H, Ghavamzadeh A, Malekzadeh R. Phase 1 trial of autologous bone marrow mesenchymal stem cell transplantation in patients with decompensated liver cirrhosis. Arch Iran Med. 2007;10:459-466.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Kharaziha P, Hellström PM, Noorinayer B, Farzaneh F, Aghajani K, Jafari F, Telkabadi M, Atashi A, Honardoost M, Zali MR. Improvement of liver function in liver cirrhosis patients after autologous mesenchymal stem cell injection: a phase I-II clinical trial. Eur J Gastroenterol Hepatol. 2009;21:1199-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 307]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
85.  Salama H, Zekri AR, Zern M, Bahnassy A, Loutfy S, Shalaby S, Vigen C, Burke W, Mostafa M, Medhat E. Autologous hematopoietic stem cell transplantation in 48 patients with end-stage chronic liver diseases. Cell Transplant. 2010;19:1475-1486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 82]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
86.  Pidala J, Anasetti C, Jim H. Health-related quality of life following haematopoietic cell transplantation: patient education, evaluation and intervention. Br J Haematol. 2010;148:373-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 83]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
87.  Forslöw U, Blennow O, LeBlanc K, Ringdén O, Gustafsson B, Mattsson J, Remberger M. Treatment with mesenchymal stromal cells is a risk factor for pneumonia-related death after allogeneic hematopoietic stem cell transplantation. Eur J Haematol. 2012;89:220-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
88.  Mishra PJ, Mishra PJ, Glod JW, Banerjee D. Mesenchymal stem cells: flip side of the coin. Cancer Res. 2009;69:1255-1258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 135]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
89.  Garcia-Olmo D, Herreros D, Pascual I, Pascual JA, Del-Valle E, Zorrilla J, De-La-Quintana P, Garcia-Arranz M, Pascual M. Expanded adipose-derived stem cells for the treatment of complex perianal fistula: a phase II clinical trial. Dis Colon Rectum. 2009;52:79-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 598]  [Cited by in F6Publishing: 541]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
90.  Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, Kooy-Winkelaar EM, Koning F, Zwaginga JJ, Fidder HH. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study. Gut. 2010;59:1662-1669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 471]  [Cited by in F6Publishing: 448]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
91.  Onken J, Jaffe T, Custer L. Long-term safety of prochymal adult mesenchymal stem cells in Crohn’s disease. Gastroenterology. 2008;134:A661.  [PubMed]  [DOI]  [Cited in This Article: ]