Imatinib is a promising candidate for the treatment of fibrotic diseases. This retrospective study evaluated the use of imatinib for the treatment of refractory sclerotic chronic graft-versus-host disease in 14 patients with different hematologic malignancies. Imatinib was started at a median of 44 months after transplantation (range, 16-119 months after transplantation) and was administered for a median of 5.9 months from time of initiation (range, 2.1-74 months from time of initiation). With a median overall follow-up of 11.6 months from time of initiation (range, 4.1-74 months from time of initiation) of imatinib, 4 patients (29%) had to stop imatinib because of drug intolerance. All other adverse reactions were of mild-to-moderate grade and could be managed symptomatically. Overall, 7 patients responded to imatinib (50%; 95% confidence interval, 24%-76%) with 4 patients improving their Rodman score more than or equal to 90%. In addition, imatinib therapy allowed for a significant reduction of corticosteroid dosage. Despite its limited size, this cohort suggests some beneficial activity of imatinib in sclerotic chronic graft-versus-host disease, warranting further prospective investigations.

Chronic graft-versus-host disease (cGVHD) remains a major cause of morbidity and mortality after allogeneic stem cell transplantation (allo-SCT). Standard primary treatment of cGVHD is a combination of corticosteroids (CSs) and calcineurin inhibitors. There is no standard therapy for those who fail to respond to CS, and CS-resistant cGVHD is associated with high morbidity.1  From the clinical standpoint, many cGVHD patients present with features of autoimmune collagen vascular disease, with clinical manifestations similar to those of autoimmune scleroderma and systemic lupus erythematosus.2  Likewise, sclerotic cGVHD (ScGVHD) is one of the most severe forms of the disease and is frequently refractory to standard treatment approaches.3-5  Thus, therapeutic options are usually limited for those patients with severe ScGVHD. Imatinib mesylate (IM), a clinically well-tolerated tyrosine kinase inhibitor, has been shown to be effective in patients with chronic myeloid leukemia and those with stromal gastrointestinal tumors.6,7  IM exerts selective, dual inhibition of the transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) pathways.8  On the other hand, blockade of TGF-β or PDGF signaling has been shown to reduce the development of fibrosis in various experimental models.9-11  Therefore, IM is a promising candidate for the treatment of fibrotic diseases, such as ScGVHD. This retrospective analysis describes the outcome of 14 patients experiencing severe or refractory ScGVHD and who received oral IM as salvage therapy

Study design

This was a retrospective study performed in 2 allo-SCT centers in France (Lille, n = 12; and Nantes, n = 2), which examined the safety and efficacy of IM therapy for refractory ScGVHD. Informed consent was obtained according to institutional guidelines in accordance with the Declaration of Helsinki. The off-label use of imatinib in the setting of ScGVHD was authorized by the Institutional Review Board of Centre Hospitalier Regional Universitaire, according to French laws in a compassionate setting for those patients presenting with advanced and refractory ScGVHD clinical features.

Study evaluations and therapy

Patient and transplantation characteristics are summarized in Table 1. Acute and cGVHD grading was performed according to classic criteria. A detailed history and physical examination were performed and documented before IM initiation. Physical examination included skin score and measurement of range of motion. The modified Rodman skin score was used to assess cutaneous sclerosis changes.12  The Rodman skin score measured thickness, graded from 0 to 3 as follows: 0 indicates uninvolved skin; 1, skin involvement with ability to pinch; 2, inability to pinch; and 3, inability to move. The pinch was done in one movement of moderate intensity, and using only fingertips. All affected areas were assessed, and the results of all areas were added up to give a final overall score. Responses in cutaneous sclerosis were always measured in the areas with the most severe involvement. All 14 patients analyzed in this retrospective study (of whom 2 were previously reported13 ) had refractory cGHVD with significant cutaneous sclerosis manifestations, and who failed at least 2 lines of prior systemic immunosuppressive therapy. IM (Glivec/Gleevec, Novartis France) was started in oral doses, usually of 400 mg/day (patient 12, a child, received 100 mg/day). IM treatment duration was at the discretion of the attending physician. Patients who had received myeloablative or reduced-intensity conditioning were examined in this analysis, as were recipients of related and unrelated stem cell grafts. All recipients underwent transplantation at least 6 months before IM initiation, none had received donor lymphocyte infusions in the preceding 100 days, none had signs of late onset aGVHD, and none was currently undergoing extracorporeal phototherapy. All patients continued to receive standard prophylaxis against Pneumocystis carinii, Toxoplasma gondii, fungal, and herpesvirus infection during study therapy. First response to IM was assessed 2 months after the start of therapy, and then on a monthly basis. At each evaluation, the attending physician usually collected the subjective feelings of the patient and the macroscopically visible changes of the skin before treatment and during follow-up, and evaluated their respective organ system as well as their RS. All concomitant medications and adverse events were captured from the patients' medical source files. For the purpose of this analysis, complete response was defined as resolution of all manifestations in involved organs, whereas partial response was defined as an improvement more than 50% in at least one cGVHD manifestation, and minor response was defined as an improvement less than 50% without any new organ involvement or progression in a previously involved organ. Failure was defined as the absence of response after 2 months. Patients who progressed after an initial response to IM were recorded as in flair of their cGVHD.14  The benefit could be also evaluated in terms of CS taper. The CS dose received was assessed at last follow-up and compared with the previous CS dose received at time of initiation of IM therapy.

Table 1

Patient and transplantation characteristics

Case no.Age, ySex, R/DDiagnosisConditioning/stem cell sourceGVHD prophylaxisaGVHD gradecGVHDcGVHD sitesImmunosuppression before imatinib
34.7 F/M CML MAC/BM CSA Extensive Skin, joint contracture CS-CSA 
15.3 M/M CML RIC/BM CSA-MMF Extensive Skin, mouth, eyes CS-CSA-AZA-MMF 
53.6 M/F MDS MAC/BM CSA-MTX None Extensive Skin, bronchiolitis, joint contracture CS-CSA-MMF-RTX-ECP 
45.1 M/M HODG RIC/PBSC CSA-MMF None Extensive Skin, mouth CS-CSA-MMF 
59 M/M CML RIC/PBSC CSA-MMF Extensive Skin, mouth CS-TAC-MMF 
54.6 F/F MDS MAC/PBSC CSA-CS None Extensive Skin, eyes, mouth CS-CSA-MMF-RTX-ECP 
56.3 F/M MDS MAC/BM CSA-MTX Extensive Skin, mouth CS-ECP 
31 F/M AA MAC/PBSC CSA-CS-MMF None Extensive Skin, mouth, eyes, bronchiolitis, joint contracture CS-TAC-MMF 
25.6 M/F ALL MAC/BM TAC-CS-MMF None Extensive Skin, mouth, joint contracture CS-CSA-MMF-TAC-ECP 
10 22.8 M/F ALL MAC/BM CSA-CS Extensive Skin, mouth, liver CS-CSA-ECP 
11 35.2 F/F HODG RIC/PBSC SIR-MMF None Extensive Skin, mouth CS-CSA-MMF-ECP 
12 4.7 F/F AML MAC/BM CSA-MTX None Extensive Joint contracture without sclerotic lesions CS-CSA-MMF-ECP 
13 52 M/M MMM RIC/PBSC CSA None Extensive Skin, mouth, eyes, joint contracture CS-CSA-MMF-RTX 
14 56 M/F MMM MAC/BM CSA-MMF None Extensive Skin, mouth, eyes, liver, joint contracture CS-CSA-MMF-RTX 
Case no.Age, ySex, R/DDiagnosisConditioning/stem cell sourceGVHD prophylaxisaGVHD gradecGVHDcGVHD sitesImmunosuppression before imatinib
34.7 F/M CML MAC/BM CSA Extensive Skin, joint contracture CS-CSA 
15.3 M/M CML RIC/BM CSA-MMF Extensive Skin, mouth, eyes CS-CSA-AZA-MMF 
53.6 M/F MDS MAC/BM CSA-MTX None Extensive Skin, bronchiolitis, joint contracture CS-CSA-MMF-RTX-ECP 
45.1 M/M HODG RIC/PBSC CSA-MMF None Extensive Skin, mouth CS-CSA-MMF 
59 M/M CML RIC/PBSC CSA-MMF Extensive Skin, mouth CS-TAC-MMF 
54.6 F/F MDS MAC/PBSC CSA-CS None Extensive Skin, eyes, mouth CS-CSA-MMF-RTX-ECP 
56.3 F/M MDS MAC/BM CSA-MTX Extensive Skin, mouth CS-ECP 
31 F/M AA MAC/PBSC CSA-CS-MMF None Extensive Skin, mouth, eyes, bronchiolitis, joint contracture CS-TAC-MMF 
25.6 M/F ALL MAC/BM TAC-CS-MMF None Extensive Skin, mouth, joint contracture CS-CSA-MMF-TAC-ECP 
10 22.8 M/F ALL MAC/BM CSA-CS Extensive Skin, mouth, liver CS-CSA-ECP 
11 35.2 F/F HODG RIC/PBSC SIR-MMF None Extensive Skin, mouth CS-CSA-MMF-ECP 
12 4.7 F/F AML MAC/BM CSA-MTX None Extensive Joint contracture without sclerotic lesions CS-CSA-MMF-ECP 
13 52 M/M MMM RIC/PBSC CSA None Extensive Skin, mouth, eyes, joint contracture CS-CSA-MMF-RTX 
14 56 M/F MMM MAC/BM CSA-MMF None Extensive Skin, mouth, eyes, liver, joint contracture CS-CSA-MMF-RTX 

R/D indicates recipient/donor; aGVHD, acute graft-versus-host disease; cGVHD, chronic graft-versus-host disease; CML, chronic myeloid leukemia; MAC, myeloablative conditioning; BM, bone marrow; CSA, cyclosporine A; CS, corticosteroids; RIC, reduced intensity conditioning; MMF, mycophenolate mofetil; AZA, azathioprine; MDS, myelodysplastic syndrome; MTX, methotrexate; RTX, rituximab; ECP, extracorporeal photopheresis; HODG, Hodgkin disease; PBSC, peripheral blood stem cell; AA: aplastic anemia; TAC, tacrolimus; ALL, acute lymphoblastic leukemia; SIR, sirolimus; AML, acute myeloid leukemia; and MMM, myeloid metaplasia with myelofibrosis.

Statistical analysis

Descriptive statistical methodology was used for all analyses.

cGVHD features, responses to IM, and outcomes are summarized in Table 2. IM was started at a median time of 44 months (range, 16-119 months) after allo-SCT. Patients who did not develop drug-related intolerance received IM for a median of 5.9 months (range, 2.1-74 months) from time of initiation. With a median overall follow-up of 11.6 months (range, 4.1-74 months) from time of initiation of IM, 4 patients (29%) had to stop IM because of drug intolerance directly related to IM according to the investigator's assessment (patients 6, 8, 9, and 10). In the latter 4 patients, side effects (especially cramps) could not be managed using a lower dose of IM. Six other patients experienced some adverse reactions of mild to moderate grade that could be managed symptomatically and did not require IM discontinuation. IM dosage had to be reduced from 400 to 300 mg/day in one patient (patient 14) because of side effects. Overall, 7 patients responded to IM (50%; 95% confidence interval, 24%-76%) with 4 patients improving their Rodman score more than or equal to 90%. At last follow-up, 2 patients were in complete response and 5 were in partial responses of their cGVHD. In those responding patients, the patient felt improvement as soon as one month after the initiation of IM. In addition, IM therapy allowed for a significant reduction of CS dosage in those assessable patients. With an overall median follow-up of 56 months (range, 25-147 months) from transplantation, only one patient (patient 8) in this series of very advanced cGVHD patients died of refractory ScGVHD (infection).

Table 2

Outcomes after imatinib therapy

Case no.Maximal tolerated daily dose of IM, mgIM therapy duration at last follow-up, moSide effects/IM discontinuation at last follow-upRS before IM initiationRS after 2 mo of IM initiationcGVHD status at 2 mo of IMRS at last follow-upcGVHD response in other organs at last follow-up*cGVHD status at last follow-upPercentage CS reduction (last dosage mg/d)Overall follow-up, moFollow-up since IM initiation, moStatus at last follow-up
1 600 74.1+ None/no 10 PR Joints contracture CR 100% (0) 147.2 74.1 Alive 
400 14.0+ Diarrhea/no 22 PR Mouth, eyes CR 100% (0) 133.1 14.0 Alive 
400 5.8 Cough/yes 12 PR Bronchiolitis, joints contracture PR 100% (0) 41.5 11.7 Alive 
400 10.6 Nausea, diarrhea, lumbar bone pain/yes PR Mouth PR (>90%) 100% (0) 55.4 12.8 Alive 
400 2.1 Rash/yes 12 12 Failure NE — failure NE 65.4 14.5 Alive 
400 0.5 Paresthesia, muscle cramps, diarrhea/yes 10 NE NE NE — failure NE 95.5 11.7 Alive 
400 Muscle cramps, diarrhea/yes MR Mouth Relapse NE 25.1 8.9 Alive 
400 2.1 Headache, muscle cramps/yes 21 13 MR 21 Mouth, eyes, bronchiolitis, joints contractures Relapse NE 46.8 13.8 Died of infection 
400 2.0 Edema, muscle cramps, diarrhea/yes 11 MR Mouth, joints contracture Relapse NE 51.5 11.4 Alive 
10 400 1.5 Muscle cramps/yes 11 MR Mouth, liver Relapse NE 33.4 9.5 Alive 
11 400 11.4+ None/no 30 PR Mouth PR (>90%) 100% (0) 56.6 11.4 Alive 
12 100 4.1+ None/no 0 PR Joints contracture PR (>90%) 100% (0) 66.3 4.1 Alive 
13 400 6.0+ Edema/no 19 15 PR Eyes, joints contracture PR 100% (0) 30.1 6.0 Alive 
14 300 4.5+ Nausea, vomiting, diarrhea/no 12 11 MR 10 Eyes, joints contractures MR 0 (30) 67.2 4.5 Alive 
Case no.Maximal tolerated daily dose of IM, mgIM therapy duration at last follow-up, moSide effects/IM discontinuation at last follow-upRS before IM initiationRS after 2 mo of IM initiationcGVHD status at 2 mo of IMRS at last follow-upcGVHD response in other organs at last follow-up*cGVHD status at last follow-upPercentage CS reduction (last dosage mg/d)Overall follow-up, moFollow-up since IM initiation, moStatus at last follow-up
1 600 74.1+ None/no 10 PR Joints contracture CR 100% (0) 147.2 74.1 Alive 
400 14.0+ Diarrhea/no 22 PR Mouth, eyes CR 100% (0) 133.1 14.0 Alive 
400 5.8 Cough/yes 12 PR Bronchiolitis, joints contracture PR 100% (0) 41.5 11.7 Alive 
400 10.6 Nausea, diarrhea, lumbar bone pain/yes PR Mouth PR (>90%) 100% (0) 55.4 12.8 Alive 
400 2.1 Rash/yes 12 12 Failure NE — failure NE 65.4 14.5 Alive 
400 0.5 Paresthesia, muscle cramps, diarrhea/yes 10 NE NE NE — failure NE 95.5 11.7 Alive 
400 Muscle cramps, diarrhea/yes MR Mouth Relapse NE 25.1 8.9 Alive 
400 2.1 Headache, muscle cramps/yes 21 13 MR 21 Mouth, eyes, bronchiolitis, joints contractures Relapse NE 46.8 13.8 Died of infection 
400 2.0 Edema, muscle cramps, diarrhea/yes 11 MR Mouth, joints contracture Relapse NE 51.5 11.4 Alive 
10 400 1.5 Muscle cramps/yes 11 MR Mouth, liver Relapse NE 33.4 9.5 Alive 
11 400 11.4+ None/no 30 PR Mouth PR (>90%) 100% (0) 56.6 11.4 Alive 
12 100 4.1+ None/no 0 PR Joints contracture PR (>90%) 100% (0) 66.3 4.1 Alive 
13 400 6.0+ Edema/no 19 15 PR Eyes, joints contracture PR 100% (0) 30.1 6.0 Alive 
14 300 4.5+ Nausea, vomiting, diarrhea/no 12 11 MR 10 Eyes, joints contractures MR 0 (30) 67.2 4.5 Alive 

IM indicates imatinib; RS, Rodman score; cGVHD, chronic graft-versus-host disease; CS, corticosteroids; PR, partial response; CR, complete response; NE, not evaluable; and MR, minor response.

*

Only patients showing at least PR are mentioned.

Patient 1 had simultaneous disease recurrence (CML) at time of IM initiation.

Patient 12 did not have assessable sclerotic skin lesions.

Currently, there is no standard “second-line” therapy for CS-resistant cGVHD. Several candidate drugs were already tested with variable results, and the response of ScGVHD is usually disappointing.15  In our study, the global response rate to IM was high if considered in terms of salvage therapy and cutaneous sclerosis manifestation improvement. On the other hand, the incidence of IM-related adverse reactions reflected the usual rate observed in chronic myelogenous leukemia treatment.

In animal models, IM has been shown to prevent fibrosis through inhibition of PDGF signaling and fibroblast proliferation mediated by TGF-β.9,10  ScGVHD is a recalcitrant disease featuring multiorgan fibrosis and dysfunction. The ability of IM to abrogate the activation of the PDGF receptor entails its use in the treatment of ScGVHD. Indeed, IM at clinically relevant concentrations has potent antifibrotic effects in vitro and in vivo and can prevent the development of inflammation-driven experimental fibrosis when treatment was initiated before administration of the profibrotic stimulus.16  In addition, IM might be effective for the treatment of established fibrosis.17  At present, several trials aiming to examine the use of IM in the treatment of systemic sclerosis are currently underway.18  Despite its limited size, this cohort (and other case reports19,20 ) demonstrates evidence of beneficial activity of IM in ScGVHD. Hence, given its oral administration, efficacy, and safety profile, evaluation of the role of IM in refractory ScGVHD warrants further investigation in sufficiently powered, well-controlled multicenter prospective trials using the robust National Institutes of Health consensus staging and response criteria.21 

An Inside Blood analysis of this article appears at the front of this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank E. Gomez for his help in collecting data and the nursing staff for providing excellent care for our patients.

M.M. was supported by Région Pays de Loire, Association pour la Recherche sur le Cancer, Fondation de France, Fondation contre la Leucémie, Agence de Biomédecine, Association Cent pour Sang la Vie, and Association Laurette Fuguain, all of which have provided generous and continuous support for his clinical and basic research work.

Contribution: L.M. provided clinical care, recorded and collected clinical data, and commented on the manuscript; M.M provided clinical care, collected patient data, analyzed data, and wrote and revised the report; B.C., V.C., P.C., L.T., and J.-P.J. provided clinical care and recorded clinical data; and I.Y.-A conceived and designed the study, provided clinical care, collected patient data, analyzed data, and revised the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Ibrahim Yakoub-Agha, Service des Maladies du Sang, UAM allogreffes de CSH, Centre Hospitalier Regional Universitaire de Lille, F-59037 Lille Cedex, France; e-mail: i-yakoub-agha@chru-lille.fr.

1
Lee
 
SJ
Vogelsang
 
G
Flowers
 
ME
Chronic graft-versus-host disease.
Biol Blood Marrow Transplant
2003
, vol. 
9
 (pg. 
215
-
233
)
2
Baird
 
K
Pavletic
 
SZ
Chronic graft versus host disease.
Curr Opin Hematol
2006
, vol. 
13
 (pg. 
426
-
435
)
3
Marcellus
 
DC
Altomonte
 
VL
Farmer
 
ER
, et al. 
Etretinate therapy for refractory sclerodermatous chronic graft-versus-host disease.
Blood
1999
, vol. 
93
 (pg. 
66
-
70
)
4
Baudard
 
M
Vincent
 
A
Moreau
 
P
Kergueris
 
MF
Harousseau
 
JL
Milpied
 
N
Mycophenolate mofetil for the treatment of acute and chronic GVHD is effective and well tolerated but induces a high risk of infectious complications: a series of 21 BM or PBSC transplant patients.
Bone Marrow Transplant
2002
, vol. 
30
 (pg. 
287
-
295
)
5
Skert
 
C
Patriarca
 
F
Sperotto
 
A
, et al. 
Sclerodermatous chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: incidence, predictors and outcome.
Haematologica
2006
, vol. 
91
 (pg. 
258
-
261
)
6
Giralt
 
SA
Arora
 
M
Goldman
 
JM
, et al. 
Impact of imatinib therapy on the use of allogeneic haematopoietic progenitor cell transplantation for the treatment of chronic myeloid leukaemia.
Br J Haematol
2007
, vol. 
137
 (pg. 
461
-
467
)
7
Blanke
 
C
Current management of GIST.
Clin Adv Hematol Oncol
2004
, vol. 
2
 (pg. 
280
-
283
)
8
Vuorinen
 
K
Gao
 
F
Oury
 
TD
Kinnula
 
VL
Myllarniemi
 
M
Imatinib mesylate inhibits fibrogenesis in asbestos-induced interstitial pneumonia.
Exp Lung Res
2007
, vol. 
33
 (pg. 
357
-
373
)
9
Daniels
 
CE
Wilkes
 
MC
Edens
 
M
, et al. 
Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycin-mediated lung fibrosis.
J Clin Invest
2004
, vol. 
114
 (pg. 
1308
-
1316
)
10
Santiago
 
B
Gutierrez-Canas
 
I
Dotor
 
J
, et al. 
Topical application of a peptide inhibitor of transforming growth factor-beta1 ameliorates bleomycin-induced skin fibrosis.
J Invest Dermatol
2005
, vol. 
125
 (pg. 
450
-
455
)
11
Chaudhary
 
NI
Roth
 
GJ
Hilberg
 
F
, et al. 
Inhibition of PDGF, VEGF and FGF signalling attenuates fibrosis.
Eur Respir J
2007
, vol. 
29
 (pg. 
976
-
985
)
12
Clements
 
P
Lachenbruch
 
P
Siebold
 
J
, et al. 
Inter and intraobserver variability of total skin thickness score (modified Rodnan TSS) in systemic sclerosis.
J Rheumatol
1995
, vol. 
22
 (pg. 
1281
-
1285
)
13
Magro
 
L
Catteau
 
B
Coiteux
 
V
Bruno
 
B
Jouet
 
JP
Yakoub-Agha
 
I
Efficacy of imatinib mesylate in the treatment of refractory sclerodermatous chronic GVHD.
Bone Marrow Transplant
2008
, vol. 
42
 (pg. 
757
-
760
)
14
de Lavallade
 
H
Mohty
 
M
Faucher
 
C
Furst
 
S
El-Cheikh
 
J
Blaise
 
D
Low-dose methotrexate as salvage therapy for refractory graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation.
Haematologica
2006
, vol. 
91
 (pg. 
1438
-
1440
)
15
Fraser
 
CJ
Scott Baker
 
K
The management and outcome of chronic graft-versus-host disease.
Br J Haematol
2007
, vol. 
138
 (pg. 
131
-
145
)
16
Distler
 
JH
Jungel
 
A
Huber
 
LC
, et al. 
Imatinib mesylate reduces production of extracellular matrix and prevents development of experimental dermal fibrosis.
Arthritis Rheum
2007
, vol. 
56
 (pg. 
311
-
322
)
17
Akhmetshina
 
A
Venalis
 
P
Dees
 
C
, et al. 
Treatment with imatinib prevents fibrosis in different preclinical models of systemic sclerosis and induces regression of established fibrosis.
Arthritis Rheum
2009
, vol. 
60
 (pg. 
219
-
224
)
18
Bibi
 
Y
Gottlieb
 
AB
A potential role for imatinib and other small molecule tyrosine kinase inhibitors in the treatment of systemic and localized sclerosis.
J Am Acad Dermatol
2008
, vol. 
59
 (pg. 
654
-
658
)
19
Majhail
 
NS
Schiffer
 
CA
Weisdorf
 
DJ
Improvement of pulmonary function with imatinib mesylate in bronchiolitis obliterans following allogeneic hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2006
, vol. 
12
 (pg. 
789
-
791
)
20
Moreno-Romero
 
JA
Fernandez-Aviles
 
F
Carreras
 
E
Rovira
 
M
Martinez
 
C
Mascaro
 
JM
Imatinib as a potential treatment for sclerodermatous chronic graft-vs-host disease.
Arch Dermatol
2008
, vol. 
144
 (pg. 
1106
-
1109
)
21
Pavletic
 
SZ
Lee
 
SJ
Socie
 
G
Vogelsang
 
G
Chronic graft-versus-host disease: implications of the National Institutes of Health consensus development project on criteria for clinical trials.
Bone Marrow Transplant
2006
, vol. 
38
 (pg. 
645
-
651
)

Author notes

*L.M. and M.M. contributed equally to this study.

Sign in via your Institution