Systemic sclerosis (SSc) is a multisystem disease of presumed autoimmune pathogenesis for which no proven effective treatment exists. High-dose immunosuppressive therapy (HDIT) has been proposed as an investigational treatment for severe autoimmune diseases. Nineteen patients with poor-prognosis SSc underwent HDIT. The median age was 40 years (range, 23-61 years), the median modified Rodnan skin score (a measure of dermal sclerosis) was 31, and the median DLCO was 57%. Conditioning therapy involved 800 cGy total body irradiation (TBI) (± lung shielding to approximately 200 cGy), 120 mg/kg cyclophosphamide, and 90 mg/kg equine antithymocyte globulin. CD34-selected granulocyte–colony-stimulating factor–mobilized autologous blood stem cells provided hematopoietic rescue. With median follow-up at 14.7 months, the Kaplan-Meier estimated 2-year survival rate was 79%. Three patients died of treatment complications and one of disease progression. Two of the first 8 patients had fatal regimen-related pulmonary injury, a complication not found among 11 subsequent patients who received lung shielding for TBI. Overall, internal organ functions were stable to slightly worse after HDIT, and 4 patients had progressive or nonresponsive disease. As measured by modified Rodnan skin scores and modified health assessment questionnaire disability index (mHAQ-DI) scores, significant disease responses occurred in 12 of 12 patients evaluated at 1 year after HDIT. In conclusion, though important treatment-related toxicities occurred after HDIT for SSc, modifications of initial approaches appear to reduce treatment risks. Responses in skin and mHAQ-DI scores exceed those reported with other therapies, suggesting that HDIT is a promising new therapy for SSc that should be evaluated in prospective randomized studies.

Systemic sclerosis (SSc) is an uncommon multisystem disease characterized by diffuse inflammation and subsequent fibrosis of skin together with varying degrees of internal organ involvement. Internal organ involvement entails poor prognosis, and deaths result primarily from pulmonary failure and cardiac events. Although the pathogenesis of SSc remains unclear, early disease is thought to be primarily immunologic in nature with secondary involvement of tissue fibroblasts.1 Because of this immunosuppressive agents have been used to treat SSc. Although some reports have suggested that immunosuppressive agents are effective in SSc, controlled trials have not confirmed this.2 Given the lack of effective treatment together with known prognostic factors for early mortality,3 patients with SSc were considered appropriate candidates for investigation of high-dose immunosuppressive therapy (HDIT).4,5 

Previous experience with allogeneic and autologous stem cell transplantation together with experimental rodent studies formed the basis for our initial HDIT protocols.6,7 An underlying hypothesis for this study was that HDIT followed by the infusion of lymphocyte-depleted (CD34-selected) autologous peripheral blood stem cells (PBSCs) would allow near or complete immune ablation leading to disease control. This would be followed by regeneration of a self-tolerant immune system from multipotential hematopoietic progenitors. Results of this study and similar studies in other autoimmune diseases also could potentially provide important insights into the pathogenesis and mechanisms of autoimmune diseases. We have previously reviewed issues pertaining to protocol development6 and the selection of patients with SSc8 for trials of HDIT followed by autologous hematopoietic cell transplantation. Here we report initial results investigating HDIT for severe SSc.

Study design and eligibility

This pilot study was designed to evaluate the safety and potential efficacy of HDIT using total body irradiation (TBI), cyclophosphamide (Cy), and equine antithymocyte globulin for severe SSc. Primary end points were grades 3-4 regimen-related toxicity and engraftment. Secondary end points included disease response, safety of granulocyte–colony-stimulating factor (G-CSF) mobilization, and immunologic recovery. Limited pooled data from 8 patients in this study have previously been reported.9 

This report describes 19 consecutive patients enrolled between January 1997 and August 1980 at the Fred Hutchinson Cancer Research Center (FCHRC) and Virginia Mason Medical Center in Seattle (n = 13), Wayne State University/Karmanos Cancer Institute (n = 3), University of Michigan (n = 2), and Loma Linda University (n = 1). Written institutional review board–approved consent for treatment was obtained from all patients. Figure 1 shows patient eligibility criteria that predict 5-year mortality risks of approximately 50% with conventional treatment.8 

Fig. 1.

Major eligibility criteria for study.

Fig. 1.

Major eligibility criteria for study.

Close modal

Treatment and supportive care

Patients underwent PBSC mobilization as outpatients using G-CSF at 16 μg/kg per day subcutaneously, with the first apheresis on day 4. G-CSF–mobilized PBSC products were CD34-selected10using an Isolex 300I (Nexell, Irvine, CA) with targets greater than 3.5 × 106 CD34+ cells/kg, though lower doses were used in 2 patients. Unmodified PBSC (more than 3.0 × 106 CD34+ cells/kg) were also stored for treating engraftment failure or severe immunodeficiency after HDIT. Cells were cryopreserved using standard techniques. Grafts were evaluated for content of CD34+ cells and T cells (CD3+) by flow cytometry. HDIT consisted of fractionated total body irradiation (TBI) delivered as 200 cGy per fraction, 2 fractions per day, on day −5 and day −4, for a total dose of 800 cGy TBI, 60 mg/kg Cy intravenously on each of days −3 and −2, and 15 mg/kg equine antithymocyte globulin (ATGAM; Pharmacia, Peapack, NJ) intravenously on days −5, −3, −1, +1, +3, and +5. Methylprednisolone (1 mg/kg) was given intravenously with each dose of ATG. Individual TBI treatments were delivered with a minimum of 5 hours between fractions. TBI was administered from dual-opposed cobalt Co 60 sources or linear accelerators at dose rates of 7 cGy to 15 cGy per minute. TBI without lung shielding was used for the first 8 patients, and the subsequent 11 patients were treated with TBI modified to shield the lungs with 5 half-value layers of lead (97% shielding) after the first 200 cGy fraction. As protection against lung toxicity and to standardize steroid therapy prednisone, 0.5 mg/kg prednisone per day was given from the start of conditioning until day +30 and was tapered over 1 month in patients 5 to 19. This also had the potential benefit of protecting against ATG-induced serum sickness. CD34-selected PBSCs were infused on day 0. G-CSF (5 μg/kg per day) was given intravenously from day 0 until the absolute neutrophil count was greater than 500/μL for 3 days. Patients were hospitalized from the start of conditioning therapy until neutrophil engraftment and resolution of major toxicities. Infection prophylaxis included trimethoprim-sulfamethoxazole for Pneumocystis carinii,11 fluconazole forCandida,12 and ganciclovir for cytomegalovirus (CMV) reactivation.13 

Evaluation of outcomes

Neutrophil engraftment was defined as occurring on the first of 3 consecutive postnadir days with more than 500 neutrophils per microliter, and platelet engraftment was defined as occurring on the first of 3 consecutive days with more than 20 000 platelets per microliter without transfusion. Regimen-related toxicities were defined according to the Bearman scale as those occurring within 28 days of transplantation.14 

Formal disease status evaluations were performed before growth factor mobilization and at 3, 12, 24, and 36 months after HDIT. These included a modified Rodnan skin score (mRSS),15 pulmonary function testing, echocardiogram, serum creatinine, 24-hour urinary creatinine clearance and protein, autoantibodies, and modified Health Assessment Questionnaire Disability Index (mHAQ-DI).16 Briefly, the mRSS uses physical examination to measure dermal thickening. Seventeen anatomic sites are scored 0 to 3 (0 indicates normal) resulting in a composite score of 0 to 51. The mHAQ-DI is a validated tool for assessing activities of daily living in SSc.3,17-19 It is a functional index that involves a self-assessment questionnaire of daily living activities and is reported as a composite score on a scale of 0 to 3, with 0 indicating normal.

Immune recovery

Peripheral blood (PB) T cells, including CD4+ and CD8+ subsets, natural killer (NK) cells, and B cells, were measured in a single laboratory before PBSC mobilization, then at approximately 7, 30, 90, 180, 365, and 730 days after transplantation using 3-color flow cytometry.20-22 B cells were defined as mononuclear cells (MNCs) expressing CD19 or CD20 and not expressing T, NK, monocyte, or progenitor cell markers CD10 and CD34. CD4+ cells were CD3+ CD4+ CD8-MNC. CD8+ cells were CD3+ CD8+ CD4-MNC. NK cells were CD16+ or CD56+ MNC not expressing CD3 or CD14.21,22 

Statistics

Outcomes are reported as of April 2001. Medians and ranges are reported unless otherwise specified. Survival was estimated using the method of Kaplan and Meier,23 and associated confidence intervals were estimated using the Greenwood formula. Post-HDIT values of parameters were compared with pre-HDIT values using the signed-rank test. All reported P values are 2-sided without adjustments for multiple comparisons.

Patient characteristics

Table 1 summarizes patient characteristics before transplantation. The median age was 40 years (range, 23-61 years), and median disease duration was 20 months (range, 4-41 months). Sixteen of 19 patients were female. The median pretransplantation DLCO was 57%, and the median mRSS was 31.

Table 1.

Pretransplantation characteristics of patients

Pt.Study UPNSexAge, yGroup*Primary organDLCO/FVCSerum creatininemRSSDisease duration, moScl-70 +vePrior therapy
FH11956 47 76/74 1.4 47 38 No Hcq, St, D-pen, CSA 
FH12946 38 48/72 29 30 Yes Hcq, CSA, St 
FH13316 23 57/62 0.7 30 28 Yes Mtx, D-pen, Ctx, St 
FH13483 40 47/75 0.5 32 22 Yes IVIG, St 
FH13833 39 46/77 0.5 30 35 Yes CSA, D-pen 
FH14767 61 67/83 0.8 35 No St, Cy 
FH14709 56 52/76 0.5 50 No D-pen 
UM 1 40 62/69 0.8 39 15 Yes Mtx 
FH15570 49 45/75 0.8 14 13 Yes St, Aza, Mtx 
10 KCI-1 27 66/78 0.9 18 No Mtx, St 
11 FH16089 37 57/87 0.6 40 11 No St, Mtx, Rl 
12 FH15782 55 57/64 0.4 27 29 No St, Cy 
13 KCI-2 48 47/65 0.9 24 20 Yes Cy, St 
14 UM-2 23 66/103 18 Yes None 
15 FH15735 23 84/65 0.6 31 34 ND D-pen, St, Cy 
16 KCI-3 42 56/67 1.3 36 41 No Mtx 
17 FH16891 28 47/54 0.6 43 19 Yes St, D-pen 
18 FH16855 40 69/66 0.6 19 28 Yes Enb 
19 LLU-1 34 72/61 0.4 32 16 No Hcq, Mtx 
 Median  40   57/72 0.7 31 20   
Pt.Study UPNSexAge, yGroup*Primary organDLCO/FVCSerum creatininemRSSDisease duration, moScl-70 +vePrior therapy
FH11956 47 76/74 1.4 47 38 No Hcq, St, D-pen, CSA 
FH12946 38 48/72 29 30 Yes Hcq, CSA, St 
FH13316 23 57/62 0.7 30 28 Yes Mtx, D-pen, Ctx, St 
FH13483 40 47/75 0.5 32 22 Yes IVIG, St 
FH13833 39 46/77 0.5 30 35 Yes CSA, D-pen 
FH14767 61 67/83 0.8 35 No St, Cy 
FH14709 56 52/76 0.5 50 No D-pen 
UM 1 40 62/69 0.8 39 15 Yes Mtx 
FH15570 49 45/75 0.8 14 13 Yes St, Aza, Mtx 
10 KCI-1 27 66/78 0.9 18 No Mtx, St 
11 FH16089 37 57/87 0.6 40 11 No St, Mtx, Rl 
12 FH15782 55 57/64 0.4 27 29 No St, Cy 
13 KCI-2 48 47/65 0.9 24 20 Yes Cy, St 
14 UM-2 23 66/103 18 Yes None 
15 FH15735 23 84/65 0.6 31 34 ND D-pen, St, Cy 
16 KCI-3 42 56/67 1.3 36 41 No Mtx 
17 FH16891 28 47/54 0.6 43 19 Yes St, D-pen 
18 FH16855 40 69/66 0.6 19 28 Yes Enb 
19 LLU-1 34 72/61 0.4 32 16 No Hcq, Mtx 
 Median  40   57/72 0.7 31 20   
*

Eligibility group as per Table 1.

Primary organ dysfunction for which HDIT was performed. P indicates pulmonary; R, renal.

DLCO corrected for hemoglobin expressed as %; FVC, forced vital capacity expressed as %. FH indicates Fred Hutchinson Cancer Research Center; KCI, Karmanos Cancer Institute; UM, University of Michigan; LLU, Loma Linda University Medical Center; Aza, azathioprine; CSA, cyclosporine; Cy, cyclophosphamide; D-Pen, D-penicillamine; Enb, Enbrel; F, female; Hcq, Hydroxychloroquine; IVIG, intravenous immunoglobulin; M, male; mRSS, modified Rodnan skin score; Mtx, methotrexate; Rl, relaxin; St, corticosteroids.

PBSC collection and engraftment

G-CSF was tolerated without major complications, and all grafts were collected with a single course of G-CSF. Four patients had what appeared to be low-grade disease flares. These were manifested by mild skin inflammation including edema (n = 2), skin tightness (n = 1), erythema (n = 1), and arthralgias (n = 2) that resolved without specific therapy after G-CSF was stopped. Data from PBSC mobilizations and the CD34 selections of the PBSC products are summarized in Table2. Engraftment occurred promptly (Table2) with full platelet and neutrophil recovery in all patients. Patient 1 had longstanding erythropoietin-responsive anemia because of scleroderma-induced renal insufficiency that persisted after HDIT.

Table 2.

PBSC mobilization, grafts, and engraftment

MedianRangeNo. patients
Mobilization and grafts    
 Number of aphereses* 2 -7 19 
 CD34+ cells × 106/kg 4.2 2.7 -7.4 19  
 Purity (%) 91.5 55 -96 19  
 T cells × 104/kg 3.6 0 -25.2 17 
Engraftment    
 Days to ANC > 500/μL 7 -13 19  
 Days to platelets > 20 000/μL 10 9 -25 19 
MedianRangeNo. patients
Mobilization and grafts    
 Number of aphereses* 2 -7 19 
 CD34+ cells × 106/kg 4.2 2.7 -7.4 19  
 Purity (%) 91.5 55 -96 19  
 T cells × 104/kg 3.6 0 -25.2 17 
Engraftment    
 Days to ANC > 500/μL 7 -13 19  
 Days to platelets > 20 000/μL 10 9 -25 19 
*

To collect CD34-selected autografts and backup.

T-cell data were incomplete for 2 patients.

Day 1 of 3 consecutive days.

Infections

Bacteremia developed in 3 patients, venous catheter infection in 2 patients, urinary tract infection in 1 patient, and thumb cellulitis in 1 patient (7 patients overall). These conditions were readily managed with antibacterial agents. No fungal infections developed. CMV reactivations (antigenemia) occurred in 3 of 10 CMV-seropositive patients and were treated effectively with ganciclovir. Epstein-Barr virus (EBV)–related posttransplantation lymphoproliferative disorder (PTLD) and recurrent herpes simplex virus (HSV) infections occurred in one patient.

Regimen-related toxicities

Conditioning therapy was well tolerated by most patients. Mucositis was mild (grades 0 to 2) and other early regimen toxicities were grade 2 or less except for grade 4 pulmonary toxicity in patient 4. Three patients had fatal treatment-related complications. Patient 4 (baseline DLCO of 46%) acquired pneumonitis at 14 days. Patient 7 (baseline DLCO of 52%) had pneumonitis at 2 months. Both had gradual progressive respiratory failure and died after transient stabilization with high-dose corticosteroids. Bronchoalveolar fluid analyses from both patients showed no evidence of bacterial, fungal, or viral infection. Histologic analysis of an open lung biopsy specimen taken from patient 4 at the time of presentation showed acute bronchiolitis and alveolitis together with background fibrosis related to the underlying SSc. No evidence of infection was found on histopathologic examination. Biopsy was not performed on patient 7 for safety reasons, and autopsy permission was denied. Because both of these pulmonary toxicities appeared to be regimen-related, lung shielding was adopted for subsequent patients.

Patient 10 had fatal PTLD. During conditioning therapy, rabbit ATG (2.5 mg/kg 6 times) was substituted for horse ATG because of a hypersensitivity reaction. Recurrent acyclovir-resistant HSV infections occurred early after HDIT. In contrast to all but one other patient, PB T cells were undetectable at 1 month after HDIT. Two months after HDIT, cardiorespiratory arrest occurred requiring mechanical ventilation. Elevated serum troponin levels suggested possible myocardial injury. After the patient remained stable for 2 weeks, rapidly increasing PB lymphocytosis developed, and liver, spleen, and lymph nodes became enlarged. EBV-related PTLD was diagnosed on lymph node biopsy. Death from multiorgan failure occurred shortly after rituximab treatment was initiated.24 Autopsy tissues from lungs, liver, and bone marrow showed extensive invasion with lymphocytes.

Survival

At a median follow-up of 15 months (range, 8-45 months), 15 of 19 (79%) patients were alive with a projected 2-year survival rate of 78.9% (95% CI, 60.6%-97.3%; Figure2). Table 3summarizes survival and status.

Fig. 2.

Survival after HDIT.

Kaplan-Meier survival estimate as of April 1, 2001.

Fig. 2.

Survival after HDIT.

Kaplan-Meier survival estimate as of April 1, 2001.

Close modal
Table 3.

Summary of outcomes

Pt.Baseline mRSSBaseline DLCOSurvival, moStatusLast disease evaluation
Months after HDITmRSSDLCO, %mHAQ-DI
47 76 44.8 Alive, Cy for PD (lung disease) 36 10 55 1.0 
29 48 39.6 Alive, FK-506 for PD (lung disease) 36 12 42 0  
30 57 36.1 Alive, no therapy for SSc 36 69 
32 47 3-150 Died on + 58 pulmonary toxicity — — — — 
30 46 31.9 Alive, no therapy for SSc 24 20 42 0.375  
35 67 24.8 Alive, no therapy for SSc 24 60 0.125 
50 52 3-150 Died on + 79 pulmonary toxicity — — — — 
39 62 22.9 Alive, no therapy for SSc 12 26 57 0.2  
14 45 17.1 Alive, no therapy for SSc 12 52 
10 66 3-150 Died on + 64 EBV-PTLD — — — — 
11 40 57 14.7 Alive, no therapy for SSc 12 24 60 0.75  
12 27 57 14.5 Alive, no therapy for SSc 12 18 41 0.25 
13 24 47 12.8 Alive, no therapy for SSc 12 54 0.5  
14 18 66 12.8 Alive, no therapy for SSc 12 10 66 
15 31 84 12.8 Alive, prednisone 8 mg/d 12 21 60 1.25 
16 36 56 3-150 Died + 123 of PD (renal failure) — — — — 
17 43 47 10.8 Alive, no therapy for SSc 46 28 2.25 (3) 
18 19 69 9.6 Alive, no therapy for SSc 15 49 2.125  
19 32 72 7.5 Alive, no therapy for SSc 52 0.450 
Pt.Baseline mRSSBaseline DLCOSurvival, moStatusLast disease evaluation
Months after HDITmRSSDLCO, %mHAQ-DI
47 76 44.8 Alive, Cy for PD (lung disease) 36 10 55 1.0 
29 48 39.6 Alive, FK-506 for PD (lung disease) 36 12 42 0  
30 57 36.1 Alive, no therapy for SSc 36 69 
32 47 3-150 Died on + 58 pulmonary toxicity — — — — 
30 46 31.9 Alive, no therapy for SSc 24 20 42 0.375  
35 67 24.8 Alive, no therapy for SSc 24 60 0.125 
50 52 3-150 Died on + 79 pulmonary toxicity — — — — 
39 62 22.9 Alive, no therapy for SSc 12 26 57 0.2  
14 45 17.1 Alive, no therapy for SSc 12 52 
10 66 3-150 Died on + 64 EBV-PTLD — — — — 
11 40 57 14.7 Alive, no therapy for SSc 12 24 60 0.75  
12 27 57 14.5 Alive, no therapy for SSc 12 18 41 0.25 
13 24 47 12.8 Alive, no therapy for SSc 12 54 0.5  
14 18 66 12.8 Alive, no therapy for SSc 12 10 66 
15 31 84 12.8 Alive, prednisone 8 mg/d 12 21 60 1.25 
16 36 56 3-150 Died + 123 of PD (renal failure) — — — — 
17 43 47 10.8 Alive, no therapy for SSc 46 28 2.25 (3) 
18 19 69 9.6 Alive, no therapy for SSc 15 49 2.125  
19 32 72 7.5 Alive, no therapy for SSc 52 0.450 

PD indicates progressive disease; mRSS, modified Rodnan skin score, mHAQ-DI, modified Health Assessment Questionnaire Disability Index.

F3-150

Patient died.

Disease evaluations

Table 4 summarizes various parameters at baseline and at 3, 12, 24, and 36 months after HDIT. Thirteen of 15 (86.7%) patients at 3-month follow-up and 12 of 12 (100%) patients at 12-month follow-up had disease response as determined by mRSS (more than 25% improvement) or mHAQ-DI (more than 0.4-point improvement). No patient had progression of mRSS (more than 25% increase) or mHAQ-DI (more than 0.4-point increase). Three patients had disease progression including 2 with pulmonary and 1 with renal disease.

Table 4.

Changes in organ function, mRSS, and mHAQ-DI after HDIT

ParameterMonths after HDITNo. patientsMedian difference from baselineP
mRSS 15 − 7  (1 to − 25) .002 
 12 12 − 13  (− 8 to − 19) .0005 
 24 − 19  (− 10 to − 32) .06 
 36 − 21  (− 17 to − 37) .25 
mHAQ-DI 14 − 1.0  (0 to − 2) .001 
 12 11 − 1.675  (0 to − 2.26) .002 
 24 − 1.875  (− .125 to − 2.01) .06 
 36 − 2.01  (− 2 to − 2.125) .25  
Serum creatinine 15 0.1  (− 0.1 to 0.6) .02 
 12 11 0.1  (− 0.1 to 0.8) .02 
 24 0.3  (0.2 to 0.7) .06  
 36 0.5  (0.2 to 0.8) .50  
DLCO 15 − 11  (− 32 to 3) .001  
 12 12 − 5  (− 26 to 7) .11  
 24 − 4  (− 22 to 10) .88  
 36 4  (− 28 to 12) 1.0 
FVC 16 1  (− 16 to 7) .87 
 12 12 4  (− 12 to 13) .10  
 24 3  (− 2 to 16) .19  
 36 − 2  (− 17 to 24) 1.0  
LVEF 12 − 2  (− 17 to 5) .02  
 12 − 0.5  (− 12 to 5) .81  
 24 − 2.5  (− 6 to 1) 1.0  
 36 0  (− 3 to 3) 1.0 
ParameterMonths after HDITNo. patientsMedian difference from baselineP
mRSS 15 − 7  (1 to − 25) .002 
 12 12 − 13  (− 8 to − 19) .0005 
 24 − 19  (− 10 to − 32) .06 
 36 − 21  (− 17 to − 37) .25 
mHAQ-DI 14 − 1.0  (0 to − 2) .001 
 12 11 − 1.675  (0 to − 2.26) .002 
 24 − 1.875  (− .125 to − 2.01) .06 
 36 − 2.01  (− 2 to − 2.125) .25  
Serum creatinine 15 0.1  (− 0.1 to 0.6) .02 
 12 11 0.1  (− 0.1 to 0.8) .02 
 24 0.3  (0.2 to 0.7) .06  
 36 0.5  (0.2 to 0.8) .50  
DLCO 15 − 11  (− 32 to 3) .001  
 12 12 − 5  (− 26 to 7) .11  
 24 − 4  (− 22 to 10) .88  
 36 4  (− 28 to 12) 1.0 
FVC 16 1  (− 16 to 7) .87 
 12 12 4  (− 12 to 13) .10  
 24 3  (− 2 to 16) .19  
 36 − 2  (− 17 to 24) 1.0  
LVEF 12 − 2  (− 17 to 5) .02  
 12 − 0.5  (− 12 to 5) .81  
 24 − 2.5  (− 6 to 1) 1.0  
 36 0  (− 3 to 3) 1.0 

mRSS indicated modified Rodnan skin score expressed as points on a scale of 0-51. mHAQ-DI expressed as points on a scale of 0-3. A baseline mHAQ-DI score was not obtained on patient 13. Serum creatinine was expressed in μg/dL. Carbon monoxide diffusing capacity was corrected for hemoglobin; percentage predicted. Forced vital capacity, percentage of predicted. Left ventricular ejection fraction, percentage.

Compared with baseline, the median decrease in mRSS values was 21% (range, −3%-86%) at 3 months (P = .002) and 39% (range, 32%-93%) at 12 months (P = .0005). These reductions began early after HDIT and continued for 3 years (Figure3A).

Fig. 3.

Disease evaluation after HDIT.

(A) mRSS after HDIT. (B) DLCO percentage of predicted values corrected for hemoglobin. (C) mHAQ-DI values after HDIT.

Fig. 3.

Disease evaluation after HDIT.

(A) mRSS after HDIT. (B) DLCO percentage of predicted values corrected for hemoglobin. (C) mHAQ-DI values after HDIT.

Close modal

Among survivors the median decrease in DLCO values was statistically significant at 3 months (P = .001) but not at 12 months (P = .11) (Table 4; Figure 3B). Among 6 patients with lung shielding, the median DLCO change was −3% (−24%-7%), not significantly different than −5% (−26%-4%) among 6 patients without lung shielding. Progressive pulmonary disease requiring therapy developed in 2 patients. Patient 1 acquired pneumonitis at 3 years and commenced treatment with oral Cy. Patient 2 had progressive pneumonitis 12 months after HDIT that responded to FK-506 therapy. Neither patient had progression of skin disease.

Overall renal function deteriorated after HDIT, but the magnitude of the changes (median creatinine increase, 0.1) was not clinically meaningful. Patient 1, who had a prior scleroderma renal crisis, contracted mild renal failure possibly related to the discontinuation of ACE inhibitors during transplantation. Patient 16 had rapidly progressive renal failure from SSc at 3 months and died suddenly of multiorgan failure 123 days after transplantation. Left ventricular ejection fractions were overall slightly reduced at 3 months but were no different from baseline at 12 months after HDIT, and no patient had evidence of progressive cardiac disease. Patient 15 had severe gastrointestinal disease that remained stable with 12-month follow-up.

Quality-of-life assessments using mHAQ-DI showed marked improvements early after HDIT (Table 4; Figure 3C). Median reductions were 1.0 (0-2.0) at 3 months (P = .001) and 1.675 (0-2.26) at 12 months (P = .002). At 3 months 9 of 13 (69%) and at 12 months 10 of 10 (100%) evaluable patients with baseline scores of 0.5 or greater had mHAQ-DI reductions of more than 0.4 points.

Ten of 19 patients had detectable autoantibodies to topoisomerase I (anti-Scl-70) before transplantation. Of 9 evaluable patients, one (patient 9) became anti–Scl-70 negative for 12 months after HDIT, whereas the others remained persistently positive.

Immune reconstitution

HDIT induced profound lymphocytopenia. Recovery to normal values was the fastest for NK cells, intermediate for B cells and CD8 T cells, and slowest for CD4 T cells (Figure 4). Whereas the tempos of recoveries of NK cells and B cells were similar to those reported after autografting using unmodified PBSCs, recoveries of CD4 and CD8 T cells appeared to be slower.25-27 Most T cells were of memory/effector phenotype (data not shown).

Fig. 4.

Immune recovery after HDIT.

Median lymphocyte subset counts in the transplant recipients before transplantation (before G-CSF mobilization, arbitrarily displayed as day −30) and approximately on days 7, 30, 80, 180, 365, and 730. Error bars denote the 10th to 90th percentiles. Solid thick horizontal lines that represent the 10th to 90th percentiles and the broken thin horizontal lines that represent the medians of 104 healthy adults indicate reference ranges. Numbers of patients studied were 13 before transplantation, 11 on day 7, 17 on day 30, 11 on day 80, 11 on day 180, 9 on day 365, and 3 on day 730. Lymphocyte subset counts were determined using 3-color flow cytometry as previously described.20 

Fig. 4.

Immune recovery after HDIT.

Median lymphocyte subset counts in the transplant recipients before transplantation (before G-CSF mobilization, arbitrarily displayed as day −30) and approximately on days 7, 30, 80, 180, 365, and 730. Error bars denote the 10th to 90th percentiles. Solid thick horizontal lines that represent the 10th to 90th percentiles and the broken thin horizontal lines that represent the medians of 104 healthy adults indicate reference ranges. Numbers of patients studied were 13 before transplantation, 11 on day 7, 17 on day 30, 11 on day 80, 11 on day 180, 9 on day 365, and 3 on day 730. Lymphocyte subset counts were determined using 3-color flow cytometry as previously described.20 

Close modal

Although there is an overall 15% 8-year mortality rate for patients with SSc, subsets of patients with much poorer prognoses can be identified by skin scores and involvement of lungs, heart, or kidneys.8,28,29 High skin scores and significant pulmonary involvement predict an approximately 50% mortality rate within 5 years.30 The lack of effective therapies and likely autoimmune pathogenesis for SSc, together with preclinical and clinical transplantation data in autoimmunity, prompted our investigation of HDIT. Several case reports31,32 and retrospective registry reports9,33 concerning HDIT for SSc have been published. Limitations of the SSc-specific registry report9 included the wide heterogeneity of patient entry criteria of multiple small studies, the multiple different transplantation regimens and graft manipulation approaches that were used, the limited data that were reported, and the lack of standardized measures of disease response. Here we provide detailed outcome descriptions from the largest single study of HDIT for SSc.

We hypothesized that intensive immunosuppression would ablate immune responses driving disease activity, thus leading to disease stabilization or disease responses. The infused CD34-selected hematopoietic progenitors, depleted in vitro of disease-causing mature lymphoid elements, might then be able to generate a new nonautoreactive immune system. To deliver intensive and potentially immunoablative conditioning therapy, we used agents (TBI, Cy, and ATG) with potent immunosuppressive properties. In preclinical rodent studies, TBI was more immunosuppressive than chemotherapy conditioning.34TBI has been an integral part of conditioning regimens aimed at suppressing immunity before allogeneic hematopoietic cell transplantation to prevent graft rejection. Although we selected an attenuated TBI dose two thirds of that usually used in transplantation for malignancy (1200 cGy or more), 800 cGy TBI is myeloablative35 and requires progenitor cell support.

PBSCs were mobilized with G-CSF but without Cy to avert unnecessary neutropenia and cardiotoxicity,9 shorten treatment, and minimize costs. Adequate progenitor cell grafts were collected without serious disease activation. Limited prior exposure to marrow toxic agents among these patients might have accounted for the efficacy of G-CSF mobilization. Several patients had what appeared to be mild disease exacerbation that occurred during G-CSF treatment and resolved when drug treatment stopped. Disease flares, in some instances severe, have been reported with G-CSF mobilization in patients with other autoimmune diseases,36-38 and this has encouraged the use of Cy-based PBSC mobilization by some investigators. In multiple sclerosis, severe and even life-threatening neurologic changes were reported when G-CSF alone was used to mobilize PBSCs.36 In patients with rheumatoid arthritis G-CSF, induced flares of RA; however, as in this study, they were not life threatening and generally resolved with discontinuation of the drug.37 Although potentially useful suppression of autoimmunity may occur with Cy-based PBSC mobilization,38 fatal toxicities have been reported in SSc patients.9,38 Results of this study showed that G-CSF mobilization without Cy was safe and effective and that it may be the preferred approach for SSc patients.

Because severe immunosuppression was anticipated, unmodified PBSCs were stored as a precaution against immunodeficiency, and infection prophylaxis was based on current recommendations for allogeneic transplantation. HDIT produced profound reductions of PB T cells, NK cells, and B cells, but it did not produce complete immune ablation because T cells were regenerated presumably from residual T cells. CD4+ cell recovery appeared delayed compared to that previously reported after unmodified autografts.25-27Because adults have reduced thymic function,39-41 slow recovery of naive T cells from hematopoietic progenitors was expected in these patients. Infection risks may therefore be prolonged compared with the use of unmodified autografts that contain much larger numbers of mature lymphocytes. Further follow-up is needed to determine the long-term effects of HDIT on immunity in these patients.

Fatal PTLD in patient 16 most likely resulted from profound T-cell suppression leading to uncontrolled growth of EBV-infected B cells. PTLD is rare after autologous transplantation, typically occurring after allogeneic hematopoietic cell transplantation using T-cell depletion techniques or T-cell–directed antibodies.42,43In this case, rabbit ATG (used because of demonstrated hypersensitivity to horse ATG), which is likely a more potent immunosuppressive than horse ATG, might have contributed to PTLD when a highly immunosuppressive TBI-based regimen was used.44-46Observations from this case indicate that there are limits to the degree of immunosuppression that can be safely achieved after HDIT. Possible future approaches to dealing with the problem of PTLD in these studies could include the use of rituximab or autologous expanded EBV-specific T cells47,48 given either preemptively or in response to increasing levels of plasma EBV DNA levels.

Two patients died of what appeared to be regimen-induced pulmonary toxicity. Both had high mRSS and compromised pulmonary function that otherwise predicted poor survival. There was no firm histologic, cytologic, or clinical evidence that progressive SSc after HDIT contributed to these deaths. However, though BAL findings excluded infection as a cause for the pulmonary changes, BAL results and biopsy findings in patient 4 were not specific for regimen-related lung injury. Our conclusions that regimen-related effects were primarily responsible for lung injury were also based on the timing of these events in relation to HDIT, their rapid onset and progression, the lack of other manifestations of progressive SSc, and the subsequent finding that lung shielding appeared to protect against pulmonary toxicity in later patients. As seen after transplantation for malignancy, DLCO reductions occurred early after conditioning therapy. These usually recovered to approximately baseline over the first year. Although our results may suggest an increased sensitivity of scleroderma lungs to high-dose conditioning therapy, pre-existing pulmonary compromise is known to predispose to transplantation complications.49Because of de-escalation of the regimen through the introduction of lung shielding, none of 11 subsequent patients had similar serious lung injury.

In other studies of SSc, baseline mRSS and mHAQ-DI have been surrogates for survival.8,15,50 In retrospective analyses of prospective treatment trials in SSc, the skin scores were stable over 12 months and were improved at 2 and 3 years after enrollment,50,51 whereas mHAQ-DI scores were stable at 12 months and minimally changed at 24 months.52 In this study, at 1 year after HDIT, 12 of 12 patients had major improvements in mRSS or mHAQ-DI. Initial responses, particularly mHAQ-DI, frequently occurred by 3 months after HDIT. Although it is possible that the prednisone given after transplantation might have contributed to initial responses, the short duration of this therapy, together with the fact that 12 of 19 patients had received steroids before HDIT, argues against an important role for corticosteroids in producing the sustained skin responses observed in this study. These impressive and rapid responses are important findings in that they appear to substantially exceed responses observed in controlled studies of other therapies.51,52 For example, a 25% or greater improvement in the mRSS is significant and outside the range of intraobserver error.15,50 The mHAQ-DI changes are less variable, and results here were even more impressive. No previously reported therapy has produced similar improvements in the mRSS and mHAQ-DI in patients with severe SSc. However, pulmonary disease did not appear to respond to HDIT, possibly because irreversible damage might have already occurred. It is possible that the stable internal organ disease observed after HDIT in these patients represents a positive effect given the poor underlying prognosis for SSc and the likelihood for further progressive pulmonary compromise. Any benefit in this regard, together with the significance of responses observed in the mRSS and mHAQ-DI, cannot be adequately assessed without a concurrent control group.

Autoantibodies to Scl-70 (anti–topoisomerase I) were present in 10 patients before HDIT. Nine of these patients were survivors who could be evaluated after HDIT, and 8 remained positive for antibodies to topoisomerase I. Thus, with limited follow-up, anti–Scl-70 was not eliminated, and it remains to be determined whether autoantibody levels will be useful for evaluating disease responses after HDIT for SSc.

Of concern for this study were the potential toxicities of the preparative regimen. High-dose, involved-field radiation (eg, 5000 cGy) given for the treatment of malignancy in SSc patients may cause accelerated fibrosis within the radiation fields.53,54 In our study, however, rapid skin improvements were typical, suggesting that TBI doses were too low to elicit fibrosis or that control of systemic autoimmunity contributed to these responses. Although secondary cancers may result from TBI, lower dose-fractionated TBI delivered at low dose rates, as used here, is less oncogenic.55 An International Bone Marrow Transplant Registry report on 19 229 patients showed that with less than 1000 cGy TBI, late cancers were not increased compared with non-TBI–containing regimens.55 Because TBI-based regimens are more lymphoablative than chemotherapy-only regimens and given the promising results observed with respect to skin and mHAQ-DI scores, we have continued to investigate the use of TBI/Cy/ATG conditioning. Investigators in Europe are testing chemotherapy-only regimens. Ultimately, short-term safety and long-term outcomes may determine which regimens are superior.

Based on this and other reports,9,33,56-58 HDIT for SSc may be associated with increased risk for severe complications compared with HDIT for other autoimmune diseases, most likely because of the nature of pre-existing advanced disease-related organ damage. Modifications introduced into our protocol after initial unfavorable outcomes, including lung shielding for TBI, have resulted in no further episodes of pulmonary toxicity. Although the disease responses reported here are encouraging with respect to the potential for prolonging survival and improving function, disease progression in some initial patients treated suggests that the potential for HDIT producing definitive disease cures could be limited. However, this cannot be adequately evaluated from this study. Importantly, our early experience and the subsequent protocol modifications have set the stage for further studies to formally evaluate the efficacy of HDIT for SSc. The role of HDIT as a therapy for SSc will be more clearly defined by planned prospective randomized studies comparing this approach against conventional dose treatment using Cy, which in uncontrolled studies has shown some evidence of efficacy.59-65 

We thank the medical, nursing, and support staff at the listed institutions for their important contributions to this study through their careful and dedicated care of the patients.

Supported in part by the National Institute of Allergy and Infectious Diseases (grant N01-A1-05410), the National Institutes of Health (grants HL36444, AI46108, HL03701, CA15704, and M01-RR00042), the Department of Health and Human Services, the University of Michigan Venture Investment Fund (grant VIF.097), and Amgen, Inc.

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 U.S.C. section 1734.

1
Furst
DE
Clements
PJ
Hypothesis for the pathogenesis of systemic sclerosis.
J Rheumatol.
48(suppl)
1997
53
57
2
Furst
DE
Clements
PJ
Hillis
S
et al
Immunosuppression with chlorambucil, versus placebo, for scleroderma: results of a three-year, parallel, randomized, double-blind study.
Arthritis Rheum.
32
1989
584
593
3
Furst
DE
Clements
PJ
Steen
VD
et al
The modified Rodnan skin score is an accurate reflection of skin biopsy thickness in systemic sclerosis.
J Rheumatol.
25
1998
84
88
4
Sullivan
KM
Furst
DE
The evolving role of blood and marrow transplantation for the treatment of autoimmune diseases.
J Rheumatol.
48(suppl)
1997
1
4
5
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.
36
1997
390
392
6
McSweeney
PA
Nash
RA
Storb
R
Furst
DE
Gauthier
J
Sullivan
KM
Autologous stem cell transplantation for autoimmune diseases: issues in protocol development.
J Rheumatol.
48(suppl)
1997
79
84
7
Nash
RA
McSweeney
PA
Storb
R
et al
Development of a protocol for allogeneic marrow transplantation for severe systemic sclerosis: paradigm for autoimmune disease.
J Rheumatol.
48(suppl)
1997
72
78
8
Clements
PJ
Furst
DE
Choosing appropriate patients with systemic sclerosis for treatment by autologous stem cell transplantation.
J Rheumatol.
48(suppl)
1997
85
88
9
Binks
M
Passweg
JR
Furst
D
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
577
584
10
Rowley
SD
Loken
M
Radich
J
et al
Isolation of CD34+ cells from blood stem cell components using the Baxter Isolex system.
Bone Marrow Transplant.
21
1998
1253
1262
11
Sullivan
KM
Meyers
JD
Flournoy
N
Storb
R
Thomas
ED
Early and late interstitial pneumonia following human bone marrow transplantation.
Int J Cell Cloning.
4(suppl 1)
1986
107
121
12
Slavin
MA
Osborne
B
Adams
R
et al
Efficacy and safety of fluconazole prophylaxis for fungal infections after marrow transplantation: a prospective, randomized, double-blind study.
J Infect Dis.
171
1995
1545
1552
13
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood.
88
1996
4063
4071
14
Bearman
SI
Appelbaum
FR
Buckner
CD
et al
Regimen-related toxicity in patients undergoing bone marrow transplantation.
J Clin Oncol.
6
1988
1562
1568
15
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.
22
1995
1281
1285
16
Clements
PJ
Wong
WK
Hurwitz
EL
et al
The Disability Index of the Health Assessment Questionnaire is a predictor and correlate of outcome in the high-dose versus low-dose penicillamine in systemic sclerosis trial.
Arthritis Rheum.
44
2001
653
661
17
Poole
JL
Steen
VD
The use of the Health Assessment Questionnaire (HAQ) to determine physical disability in systemic sclerosis.
Arthritis Care Res.
4
1991
27
31
18
Steen
VD
Medsger
TA
Jr
The value of the Health Assessment Questionnaire and special patient-generated scales to demonstrate change in systemic sclerosis patients over time.
Arthritis Rheum.
40
1997
1984
1991
19
Clements
PJ
Wong
WK
Hurwitz
EL
et al
Correlates of the disability index of the health assessment questionnaire: a measure of functional impairment in systemic sclerosis.
Arthritis Rheum.
42
1999
2372
2380
20
Storek
J
Dawson
MA
Storer
B
et al
Immune reconstitution after allogeneic marrow transplantation compared with blood stem cell transplantation.
Blood.
97
2001
3380
3389
21
Storek
J
Dawson
MA
Maloney
DG
Comparison of two flow cytometric methods enumerating CD4 T cells and CD8 T cells.
Cytometry.
33
1998
76
82
22
Storek
J
Ferrara
S
Ku
N
Giorgi
JV
Champlin
RE
Saxon
A
B cell reconstitution after human bone marrow transplantation: recapitulation of ontogeny?
Bone Marrow Transplant.
12
1993
387
398
23
Kaplan
EL
Meier
P
Nonparametric estimations from incomplete observations.
J Am Stat Assoc.
53
2001
457
481
24
Milpied
N
Vasseur
B
Parquet
N
et al
Humanized anti-CD20 monoclonal antibody (Rituximab) in post transplant B-lymphoproliferative disorder: a retrospective analysis on 32 patients.
Ann Oncol.
11(suppl 1)
2000
113
116
25
Roberts
MM
To
LB
Gillis
D
et al
Immune reconstitution following peripheral blood stem cell transplantation, autologous bone marrow transplantation and allogeneic bone marrow transplantation.
Bone Marrow Transplant.
12
1993
469
475
26
Ashihara
E
Shimazaki
C
Yamagata
N
et al
Reconstitution of lymphocyte subsets after peripheral blood stem cell transplantation: two-color flow cytometric analysis.
Bone Marrow Transplant.
13
1994
377
381
27
Rosillo
MC
Ortuno
F
Moraleda
JM
et al
Immune recovery after autologous or rhG-CSF primed PBSC transplantation.
Eur J Haematol.
56
1996
301
307
28
Steen
VD
Medsger
TA
Jr
Severe organ involvement in systemic sclerosis with diffuse scleroderma.
Arthritis Rheum.
43
2000
2437
2444
29
Bryan
C
Knight
C
Black
CM
Silman
AJ
Prediction of five-year survival following presentation with scleroderma: development of a simple model using three disease factors at first visit.
Arthritis Rheum.
42
1999
2660
2665
30
Clements
PJ
Lachenbruch
PA
Ng
SC
Simmons
M
Sterz
M
Furst
DE
Skin score: a semiquantitative measure of cutaneous involvement that improves prediction of prognosis in systemic sclerosis.
Arthritis Rheum.
33
1990
1256
1263
31
Tyndall
A
Black
C
Finke
J
et al
Treatment of systemic sclerosis with autologous haemopoietic stem cell transplantation [abstract].
Lancet.
349
1997
254
32
Martini
A
Maccario
R
Ravelli
A
et al
Marked and sustained improvement 2 years after autologous stem cell transplantation in a girl with systemic sclerosis [abstract].
Rheumatology (Oxford).
38
1999
773
33
Tyndall
A
Fassas
A
Passweg
J
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
729
734
34
van Bekkum
DW
Conditioning regimens for the treatment of experimental arthritis with autologous bone marrow transplantation.
Bone Marrow Transplant.
25
2000
357
364
35
Storb
R
Raff
RF
Graham
T
et al
Marrow toxicity of fractionated vs. single dose total body irradiation is identical in a canine model.
Int J Radiat Oncol Biol Phys.
26
1993
275
283
36
Openshaw
H
Stuve
O
Antel
JP
et al
Multiple sclerosis flares associated with recombinant granulocyte colony-stimulating factor.
Neurology.
54
2000
2147
2150
37
Snowden
JA
Biggs
JC
Milliken
ST
et al
A randomised, blinded, placebo-controlled, dose escalation study of the tolerability and efficacy of filgrastim for haemopoietic stem cell mobilisation in patients with severe active rheumatoid arthritis.
Bone Marrow Transplant.
22
1998
1035
1041
38
Burt
RK
Fassas
A
Snowden
J
et al
Collection of hematopoietic stem cells from patients with autoimmune diseases.
Bone Marrow Transplant.
28
2001
1
12
39
Storek
J
Witherspoon
RP
Storb
R
T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life.
Bone Marrow Transplant.
16
1995
413
425
40
Weinberg
K
Annett
G
Kashyap
A
Lenarsky
C
Forman
SJ
Parkman
R
The effect of thymic function on immunocompetence following bone marrow transplantation.
Biol Blood Marrow Transplant.
1
1995
18
23
41
Storek
J
Witherspoon
RP
Immunologic reconstitution after hematopoietic stem cell transplantation.
Clinical Bone Marrow and Blood Stem Cell Transplantation. Cambridge
Atkinson
K
2000
111
146
Cambridge University Press
United Kingdom
42
Zutter
MM
Martin
PJ
Sale
GE
et al
Epstein-Barr virus lymphoproliferation after bone marrow transplantation.
Blood.
72
1988
520
529
43
Curtis
RE
Travis
LB
Rowlings
PA
et al
Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study.
Blood.
94
1999
2208
2216
44
Nash
RA
Dansey
R
Storek
J
et al
Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disorder (PTLD) after high-dose immunosuppressive therapy (HDIT) and autologous CD34-selected stem cell transplantation (SCT) for severe autoimmune diseases [abstract].
Blood.
96
2000
406
45
Brennan
DC
Flavin
K
Lowell
JA
et al
A randomized, double-blinded comparison of Thymoglobulin versus Atgam for induction immunosuppressive therapy in adult renal transplant recipients.
Transplantation.
67
1999
1011
1018
46
McCaul
KG
Nevill
TJ
Barnett
MJ
et al
Treatment of steroid-resistant acute graft-versus-host disease with rabbit antithymocyte globulin.
J Hematother Stem Cell Res.
9
2000
367
374
47
Rooney
CM
Smith
CA
Ng
CY
et al
Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation.
Lancet.
345
1995
9
13
48
Heslop
HE
Ng
CY
Li
C
et al
Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes.
Nat Med.
2
1996
551
555
49
Crawford
SW
Fisher
L
Predictive value of pulmonary function tests before marrow transplantation.
Chest.
101
1992
1257
1264
50
Clements
PJ
Lachenbruch
PA
Seibold
JR
et al
Skin thickness score in systemic sclerosis: an assessment of interobserver variability in 3 independent studies.
J Rheumatol.
20
1993
1892
1896
51
Clements
P
Lachenbruch
P
Furst
D
Paulus
H
The course of skin involvement in systemic sclerosis over three years in a trial of chlorambucil versus placebo.
Arthritis Rheum.
36
1993
1575
1579
52
Clements
PJ
Furst
DE
Wong
WK
et al
High-dose versus low-dose D-penicillamine in early diffuse systemic sclerosis: analysis of a two-year, double-blind, randomized, controlled clinical trial.
Arthritis Rheum.
42
1999
1194
1203
53
Aref
I
Cross
P
Nair
B
Case report: severe fibrosis in a patient with scleroderma and previous radiotherapy—a case report and literature review.
Br J Radiol.
69
1996
1055
1056
54
Abu-Shakra
M
Lee
P
Exaggerated fibrosis in patients with systemic sclerosis (scleroderma) following radiation therapy.
J Rheumatol.
20
1993
1601
1603
55
Curtis
RE
Rowlings
PA
Deeg
HJ
et al
Solid cancers after bone marrow transplantation.
N Engl J Med.
336
1997
897
904
56
Fassas
A
Anagnostopoulos
A
Kazis
A
et al
Peripheral blood stem cell transplantation in the treatment of progressive multiple sclerosis: first results of a pilot study.
Bone Marrow Transplant.
20
1997
631
638
57
Snowden
JA
Biggs
JC
Milliken
ST
Fuller
A
Brooks
PM
A phase I/II dose escalation study of intensified cyclophosphamide and autologous blood stem cell rescue in severe, active rheumatoid arthritis.
Arthritis Rheum.
42
1999
2286
2292
58
Traynor
AE
Schroeder
J
Rosa
RM
et al
Treatment of severe systemic lupus erythematosus with high-dose chemotherapy and haemopoietic stem-cell transplantation: a phase I study.
Lancet.
356
2000
701
707
59
Silver
RM
Warrick
JH
Kinsella
MB
Staudt
LS
Baumann
MH
Strange
C
Cyclophosphamide and low-dose prednisone therapy in patients with systemic sclerosis (scleroderma) with interstitial lung disease.
J Rheumatol.
20
1993
838
844
60
Akesson
A
Scheja
A
Lundin
A
Wollheim
FA
Improved pulmonary function in systemic sclerosis after treatment with cyclophosphamide.
Arthritis Rheum.
37
1994
729
735
61
Behr
J
Vogelmeier
C
Beinert
T
et al
Bronchoalveolar lavage for evaluation and management of scleroderma disease of the lung.
Am J Respir Crit Care Med.
154
1996
400
406
62
Schnabel
A
Reuter
M
Gross
WL
Intravenous pulse cyclophosphamide in the treatment of interstitial lung disease due to collagen vascular diseases.
Arthritis Rheum.
41
1998
1215
1220
63
Steen
VD
Lanz
JK
Jr
Conte
C
Owens
GR
Medsger
TA
Jr
Therapy for severe interstitial lung disease in systemic sclerosis: a retrospective study.
Arthritis Rheum.
37
1994
1290
1296
64
Davas
EM
Peppas
C
Maragou
M
Alvanou
E
Hondros
D
Dantis
PC
Intravenous cyclophosphamide pulse therapy for the treatment of lung disease associated with scleroderma.
Clin Rheumatol.
18
1999
455
461
65
White
B
Moore
WC
Wigley
FM
Xiao
HQ
Wise
RA
Cyclophosphamide is associated with pulmonary function and survival benefit in patients with scleroderma and alveolitis.
Ann Intern Med.
132
2000
947
954
66
Subcommittee for scleroderma criteria of the American Rheumatism Association Diagnostic and Therapeutic Criteria Committee
Preliminary criteria for the classification of systemic sclerosis (scleroderma).
Arthritis Rheum.
23
1980
581
590

Author notes

Peter A. McSweeney, Bone Marrow Transplant Program, University of Colorado Health Sciences Center, B-190, 4200 East 9th Ave, Denver, CO 80262; e-mail: peter.mcsweeney@uchsc.edu.

Sign in via your Institution