Intensive chemotherapy given in early chronic phase of chronic myelogenous leukemia (CML) has resulted in high numbers of circulating Philadelphia (Ph) chromosome–negative hematopoietic progenitor cells (HPC). We have autografted 30 consecutive patients with CML in chronic phase with HPC collected in this way to facilitate restoration of Ph-negative hematopoiesis in bone marrow after high-dose therapy. Hematopoietic recovery to greater than 0.5 ×109/L neutrophils and to greater than 25 × 109/L platelets occurred in all patients, a median of 13 (range, 9 to 32) days and 16 (range, 6 to 106) days postautograft, respectively. Regenerating marrow cells were Ph-negative in 16 (53%) patients and greater than 66% Ph-negative in 10 (33%) patients. Twenty-eight patients are alive 6 to 76 months (median, 24 months) after autografting. Three patients have developed blast crisis from which 2 have died. Eight patients are in complete cytogenetic remission at a median of 20 (range, 6 to 44) months with a median ratio BCR-ABL/ABL of 0.002 (range, <0.001 to 0.01). Eight patients are in major cytogenetic remission at a median of 22 (range, 6 to 48) months. No patient died as a consequence of the treatment. All patients had some degree of stomatitis that was severe in 15 (50%) patients. Gastrointestinal and hepatic toxicities were observed in about one fourth of patients. Thus, autografting with Ph-negative mobilized HPC can result in prolonged restoration of Ph-negative hematopoiesis for some patients with CML; moreover, most autograft recipients report normal or near normal activity levels, suggesting that this procedure need not to be associated either with prolonged convalescence or with chronic debility.

CHRONIC MYELOGENOUS leukemia (CML) is a malignant disease of hematopoietic stem cells. Conventional treatment with chemotherapy controls signs of disease but neither postpones onset of acute phase nor prolongs survival. Except for MD Anderson,1 interferon-α can offer significant cytogenetic remission in 9% to 19% rates.2-4 Transplantation with stem cells obtained from either related or unrelated donors can result in prolonged disease-free survival, and it is curative in many patients.5-7 Unfortunately, donor stem-cell transplants may be offered to only a minority of CML patients. In these last years, several investigators have observed that diploid hematopoietic progenitor cells (HPC) coexist with their malignant counterparts in the marrow of some patients with CML,8-13 observations that led to clinical trials testing autografting after high-dose chemoradiotherapy regimens.14-20 

Beginning in July 1989, our group initiated a pilot study with intensive chemotherapy to induce aplasia followed by leukaphereses of peripheral blood HPC, in an attempt to verify whether residual diploid cells have, in the repopulating phase, a proliferative advantage over Philadelphia (Ph) chromosome–positive cells and if they could be collected in quantities suitable for autografting applications. These studies showed that it was possible to collect diploid cells also in patients with advanced phases of disease cytogenetically refractory to interferon therapy.21 In a subsequent study, we confirmed that patients treated early in chronic phase with chemotherapy had a higher number of diploid cells than patients in the more advanced phase of disease.22 

Here we present the results from 30 consecutive patients autografted with Ph-negative or prevalently Ph-negative (≤34% Ph-positive) HPC during their chronic phase of CML.

Patients with Ph-positive CML who were less than 65 years old, ineligible for allogeneic stem-cell transplantation, and in morphologic chronic phase entered this study. All patients had previously received only hydroxyurea to reduce white blood cells less than or equal to 20 × 109/L. The first cohort of patients (n = 11) were mobilized with the ICE protocol consisting of idarubicin 8 mg/m2/d intravenously for 5 days; cytarabine (ARA-C) 800 mg/m2 by 2-hour infusion for 5 days; and etoposide 150 mg/m2/d by 2-hour infusion for 3 days. Subsequently, 19 patients were given a mini-ICE protocol (the same three drugs at the same doses but idarubicin and ARA-C were given for 3 days only). Beginning 8 days after the end of the chemotherapy, recombinant human granulocyte colony-stimulating factor (rhG-CSF) was administered daily at a dosage of 5 μg/kg until the total neutrophil count was more than 1.0 × 109/L for 3 consecutive days. Leukaphereses were performed using CS 3000 PLUS machine (Baxter, Deerfield, IL) or Dideco Excel (Milan, Italy), generally starting on the first day that the white blood cell count exceeded 0.8 × 109/L and the concentration of CD34+ cells was at least 5/μL on peripheral blood. The next leukapheresis was usually performed when the white blood cell count had surpassed 109/L and the circulating CD34+ cells were more than 10/μL. The collections were continued until the total number of CD34+ cells collected was at least 2 × 106/kg. Each leukapheresis product was analyzed cytogenetically and for CD34+ and colony-forming unit–granulocyte-macrophage (CFU-GM) numbers. The total leukapheresis product for each patient was frozen using a Planner R-203 (SACI, Milan, Italy). Between June 1992 and April 1998, 40 candidates had their peripheral blood HPC treated with mobilization procedure, but 10 of them could not be autografted because of high level of Ph-positive cells in the leukapheretic product (6 patients), very low level of CD34+ cells (<1 × 106/kg; 3 patients), and matched unrelated donor (MUD) transplant in the last 1 patient.

Patients.

The 30 patients were aged 22 to 62 years (median 46 years) at the time of autografting (Table 1). All patients were in first chronic phase. The treatment was approved by the local ethical committee, and patients gave informed consent before entry into the study.

Table 1.

Details of Patients at Autografting, Outcome, and Survival

Patients Age/ Sex Sokal Index Interval Diagnosis to Autografting (mo) Mobilizing TherapyCytogenetics on HPC Preautografting (Ph-positive/metas)HighDose Therapy Marrow Cytogenetics Ph-Positive (%) Outcome BCR-ABL/ABL RatioFollow-up From ASCT (mo)
TP  59/F  1.05  ICE  0/126  IVT  0  A/Ph+ 100%   76 
BG  46/M  1.45  17  ICE  0/119  IVT  Died of BC disease   29  
BF1  44/F  0.75  30 ICE  0/119  BU  0  A/Ph+ 46%   42 
BC  53/F  0.71  10  ICE  0/87  IVT  A/Ph+ 10%   48  
BP  37/M  0.77  ICE  0/75  IVT  0  A/Ph+ 100%   48 
MC1  48/F  0.85  6  ICE  0/30  IVT  15 A/Ph+ 26%   48  
SE1  40/M  0.79  ICE  0/178  BU  0  Alive, in CCR  <0.001  44 
GR  47/M  0.91  9  ICE  0/114  IVT  30 Died of BC disease   18  
VG1  39/M  0.87  ICE  0/137  BU  0  Alive, in CCR  0.003  41 
GO  46/M  0.66  9  ICE  0/128  BU  A/Ph+ 50%   39  
SS  60/M  0.84  mini-ICE  17/285  BU  0  A/Ph+ 65%  31  
UK  49/F  0.65  5  ICE  14/203  BU 0  A/Ph+ 85%   36  
PN  30/M  1.43 14  mini-ICE  0/178  BU  A/Ph+ 100%/BC   25  
PG  56/M  3.03 4  mini-ICE  0/59  BU  0  A/Ph+ 100%  26  
BS  22/F  0.51  8  mini-ICE  12/124 BU  100  A/Ph+ 100%   23  
NF  57/M 1  3  mini-ICE  13/106  BU  25 A/Ph+ 29%   26  
DG  41/M  0.68  mini-ICE  0/42  BU  0  Alive, in CCR  0.002  24 
CE  62/M  0.92  6  mini-ICE  0/40  BU  20 A/Ph+ 20%   23  
GG  27/M  1.03  mini-ICE  0/100  BU  0  Alive, in CCR  0.002 21  
PL  37/F  1.35  6  mini-ICE  0/88  BU 5  A/Ph+ 20%   19  
MA  42/M  0.59 4  mini-ICE  7/51  BU  30  A/Ph+ 12%  21  
BM  34/M  1.69  4  mini-ICE  65/176 BU  95  A/Ph+ 58%   18  
PM  46/M 0.614  3  mini-ICE  0/248  BU  0  Alive, in CCR 0.001  19  
CR  48/M  0.63  7  mini-ICE  0/69 BU  0  Alive, in CCR  0.01  15  
SP  32/M 1.0  11  mini-ICE  0/35  BU  0  Alive, in CCR 0.01  7  
MM  52/M  0.71  6  mini-ICE  0/40 BU  6  A/Ph+ 6%   13  
MG  45/M 1.06  4  mini-ICE  1/30  BU  90 A/Ph+ 90%   13  
FV  34/M  0.6  mini-ICE  0/30  BU  0  Alive, in CCR  0.01  
GA  49/M  0.72  14  mini-ICE  0/130  BU  40 A/Ph+ 46%   6  
MA  47/M  0.61  mini-ICE  2/2  BU  10  A/Ph+ 10%  
Patients Age/ Sex Sokal Index Interval Diagnosis to Autografting (mo) Mobilizing TherapyCytogenetics on HPC Preautografting (Ph-positive/metas)HighDose Therapy Marrow Cytogenetics Ph-Positive (%) Outcome BCR-ABL/ABL RatioFollow-up From ASCT (mo)
TP  59/F  1.05  ICE  0/126  IVT  0  A/Ph+ 100%   76 
BG  46/M  1.45  17  ICE  0/119  IVT  Died of BC disease   29  
BF1  44/F  0.75  30 ICE  0/119  BU  0  A/Ph+ 46%   42 
BC  53/F  0.71  10  ICE  0/87  IVT  A/Ph+ 10%   48  
BP  37/M  0.77  ICE  0/75  IVT  0  A/Ph+ 100%   48 
MC1  48/F  0.85  6  ICE  0/30  IVT  15 A/Ph+ 26%   48  
SE1  40/M  0.79  ICE  0/178  BU  0  Alive, in CCR  <0.001  44 
GR  47/M  0.91  9  ICE  0/114  IVT  30 Died of BC disease   18  
VG1  39/M  0.87  ICE  0/137  BU  0  Alive, in CCR  0.003  41 
GO  46/M  0.66  9  ICE  0/128  BU  A/Ph+ 50%   39  
SS  60/M  0.84  mini-ICE  17/285  BU  0  A/Ph+ 65%  31  
UK  49/F  0.65  5  ICE  14/203  BU 0  A/Ph+ 85%   36  
PN  30/M  1.43 14  mini-ICE  0/178  BU  A/Ph+ 100%/BC   25  
PG  56/M  3.03 4  mini-ICE  0/59  BU  0  A/Ph+ 100%  26  
BS  22/F  0.51  8  mini-ICE  12/124 BU  100  A/Ph+ 100%   23  
NF  57/M 1  3  mini-ICE  13/106  BU  25 A/Ph+ 29%   26  
DG  41/M  0.68  mini-ICE  0/42  BU  0  Alive, in CCR  0.002  24 
CE  62/M  0.92  6  mini-ICE  0/40  BU  20 A/Ph+ 20%   23  
GG  27/M  1.03  mini-ICE  0/100  BU  0  Alive, in CCR  0.002 21  
PL  37/F  1.35  6  mini-ICE  0/88  BU 5  A/Ph+ 20%   19  
MA  42/M  0.59 4  mini-ICE  7/51  BU  30  A/Ph+ 12%  21  
BM  34/M  1.69  4  mini-ICE  65/176 BU  95  A/Ph+ 58%   18  
PM  46/M 0.614  3  mini-ICE  0/248  BU  0  Alive, in CCR 0.001  19  
CR  48/M  0.63  7  mini-ICE  0/69 BU  0  Alive, in CCR  0.01  15  
SP  32/M 1.0  11  mini-ICE  0/35  BU  0  Alive, in CCR 0.01  7  
MM  52/M  0.71  6  mini-ICE  0/40 BU  6  A/Ph+ 6%   13  
MG  45/M 1.06  4  mini-ICE  1/30  BU  90 A/Ph+ 90%   13  
FV  34/M  0.6  mini-ICE  0/30  BU  0  Alive, in CCR  0.01  
GA  49/M  0.72  14  mini-ICE  0/130  BU  40 A/Ph+ 46%   6  
MA  47/M  0.61  mini-ICE  2/2  BU  10  A/Ph+ 10%  

Abbreviations: IVT, idarubicin, etoposide, TBI; BU, busulfan; ASCT, autografting; CCR, complete cytogenetic remission; BC, blastic crisis.

High-dose therapy.

Patients were treated with one of the two intensive therapy regimens. The median time from diagnosis and autograft was 6 months (range, 3 to 30 months). Four patients received autograft over 11 months after diagnosis. Two of them decided to wait for research of MUD, and in the meantime they were treated with hydroxyurea. MUD research was negative; therefore, they were autografted at 14 months from diagnosis. The other 2 patients did not agree to receive autograft so early, and they were given only hydroxyurea. After 17 and 30 months they underwent autografting.

Six patients received idarubicin (50 mg intravenously in single dose over 3 hours) on day −11; etoposide (800 mg/m2 as a 2-hour infusion) on days −8/−7; and total body irradiation (850 cGy in single dose) on day −2. Peripheral HPC were reinfused on day 0. The subsequent 24 patients received busulfan alone at 4 mg/kg/d orally for 4 consecutive days (total 16 mg/kg). A loading dose of 600 mg oral phenytoin was administered 1 day before the start of chemotherapy, and phenytoin was continued at a dose of 100 mg/d for 5 days to prevent epileptic seizures. All patients received hydration with urine alkalinization, allopurinol, and ranitidine. Peripheral HPC were reinfused 5 days after the end of chemotherapy.

Supportive care.

Patients were managed in conventional rooms (without high-efficiency particulate air filtration) and given intravenous antibiotics and amphotericin B, irradiated blood products, and intravenous nutrition as indicated.

Assessment for autografting.

Marrow samples were taken at regular intervals after autografting for morphologic and cytogenetic analyses. BCR-ABL/ABL ratio was performed after autografting and during the follow-up only on patients resulting Ph-negative.

Definition of response.

Hematologic remission was defined as the absence of morphologic evidence of CML in the blood and marrow. Cytogenetic remission was defined as the presence of 100% (complete) or 66% to 99% (major) Ph-negative metaphases in Giemsa-banded preparations from 24- to 48-hour marrow cultures. BCR-ABL/ABL ratio was evaluated for assessing the BCR-ABL transcript and BCR-ABL/ABL ratio on marrow cells during the follow-up as described.23 

Treatment after autografting.

Therapy with interferon-α 2a or 2b was begun earlier after engraftment and given at 3 M/U daily for 5 days per week for 8 weeks together with interleukin-2 (IL-2) at 2 × 106 daily for 5 days every 9 weeks. This course was repeated for 3 times, and after that the patient received interferon therapy alone.

Statistical methods.

Actuarial curves were estimated according to the Kaplan and Meier method.24 Surviving patients were censored on the last day of follow-up.

Composition of the autograft.

The number of CD34+ cells infused ranged from 1.06 to 74.5 × 106/kg (median, 4.04) and the number of CFU-GM from 0.11 to 108 × 104/kg (median, 19.2). Twenty-two patients were reinfused with Ph-negative cells only and 14 (64%) achieved marrow cytogenetic remission postgraft. Six patients were infused with less than 34% Ph-positive cells and 2 (33%) of them achieved marrow cytogenetic remission postautograft. The last 2 patients were reinfused with greater than 34% Ph-positive cells, and they achieved 10% and 95% Ph-positive cells postautograft. No correlation is possible considering the very few patients reinfused with Ph-positive greater than 34%.

Hematopoietic recovery.

Recovery of neutrophils to greater than 0.5 × 109/L occurred at a median of 13 (range, 9 to 32) days and recovery of platelets to greater than or equal to 25 × 109/L at a median of 16 (range, 6 to 106) days. During the first 2 months following hematopoietic recovery after autografting, cytogenetic analysis showed complete remission in marrow cells in 16 (53%) of the 30 patients and major remission in 10 (33%) patients (Table 1). Four patients showed Ph-positive cells greater than 34% of marrow cells. In 8 patients, platelets recovery greater than 50 × 109/L did not occur during the first 8 weeks after autografting. In 1 patient this recovery occurred after 15 weeks. After this time, in all cases engraftment was achieved and there were no cases of late cytopenia.

Toxicity.

All patients had some degree of stomatitis, which was severe in 15 patients (Table 2). Gastrointestinal and hepatic toxicities were observed in about one fourth of patients. No other major toxicities were encountered.25 Specifically, no patient had busulfan-related pulmonary toxicity, hepatic veno-occlusive disease, or epileptic seizures.

Table 2.

Toxicity After High-Dose Therapy in 30 Patients Autografted for CML

Cardiac Bladder Renal Respiratory HepaticCNS Stomatitis Gastrointestinal
Grade III or less  0  0   0  0   0  15 (50%)  3 (10%) 
Grade II or less  0  0  1 (3%)  0  4 (13%)  6 (20%)  3 (10%)  
Grade I only  0  0  1 (3%) 0  3 (10%)  0  3 (10%)  1 (3%)  
Total with any degree of toxicity  0  0  2 (6%)  0  7 (23%)  24 (80%)  7 (23%) 
Cardiac Bladder Renal Respiratory HepaticCNS Stomatitis Gastrointestinal
Grade III or less  0  0   0  0   0  15 (50%)  3 (10%) 
Grade II or less  0  0  1 (3%)  0  4 (13%)  6 (20%)  3 (10%)  
Grade I only  0  0  1 (3%) 0  3 (10%)  0  3 (10%)  1 (3%)  
Total with any degree of toxicity  0  0  2 (6%)  0  7 (23%)  24 (80%)  7 (23%) 

Abbreviation: CNS, central nervous system.

Two patients died of myeloid blastic transformation at 18 and 29 months after autografting. Twenty-eight patients are alive 6 to 76 months (median, 24 months) after autografting, with an actuarial survival of 87% (95% confidential limit [CL]: 54 to 97). Eight of these maintain complete cytogenetic remission at a median of 20 (range, 6 to 44) months after autografting. The median ratio BCR-ABL/ABL was of 0.002 (range, <0.001 to 0.01). Between 3 and 17 months (median 5 months), Ph-positive cells became detectable in 8 of the 16 patients who achieved complete cytogenetic remission (Fig2). These 8 patients are now in major cytogenetic remission at a median of 22 (range, 6 to 48) months. All patients (Ph-negative and Ph-positive) remain on interferon-α therapy. The status of each patient on the last day of follow-up is shown in Table 1. Survival plot after autografting is shown in Fig1.

Fig. 1.

Actuarial survival after autografting. Patients who remain in complete (▴) or in major (▵) cytogenetic remission are indicated.

Fig. 1.

Actuarial survival after autografting. Patients who remain in complete (▴) or in major (▵) cytogenetic remission are indicated.

Close modal
Fig. 2.

Time to recurrence of greater than 1% Ph-positive cells after infusion of mobilized Ph-negative cells in 16 patients in whom complete cytogenetic remission was achieved.

Fig. 2.

Time to recurrence of greater than 1% Ph-positive cells after infusion of mobilized Ph-negative cells in 16 patients in whom complete cytogenetic remission was achieved.

Close modal

The general rationale for autografting in first chronic phase is that a reduction in the target population of leukemic stem cells available for secondary mutational events will delay emergence of a blast-phase subclone and thereby prolong survival. The fact that Ph-positive cells subsequently became detectable is not surprising in view of experience with syngeneic transplants and evidence that immune effectors cells in allografting can make an important contribution to the prevention of disease recurrence after allografting. Most of the published experience in autografting for CML in chronic phase has involved the infusion of Ph-positive cells obtained from untreated patients at diagnosis.14,16,20,26 Although significant decreases in the percentage of Ph-positive marrow metaphases posttransplant have been noted in many patients, only a minority of patients have shown less than 25% Ph-positive cells, and only exceptional cases were completely Ph-negative early or at greater than or equal to 1 year posttransplantation.14,16 Several investigators suggest that total survival may be extended in autografted patients compared with historic controls, particularly in those who show some Ph-negative cells after grafting.14,16,27,28 However, it has been noted that a significant percentage of such patients may require repeated autografts because of early or late graft failure.14 

There is limited experience to date using Ph-negative cells in an attempt to increase the curative potential of autografting in CML. Approaches to obtaining such cells have involved the use of repeated courses of intensive chemotherapy,29 in vitro purging,12,15,17,31-37 or long-term culture8,10,18 and in vivo purging.21,22,30,31,46-48 

A total of 40 patients were treated with intensive chemotherapy and G-CSF. Of these, 10 patients could not be autografted because 1 patient underwent MUD, 6 patients collected highly contaminated Ph-positive cells, and the last 3 patients achieved a very low level of CD34+ cells (<1 × 106/kg).

In this report we have analyzed the results achieved in 30 consecutive patients autografted with Ph-negative or prevalently Ph-negative cells combined with adequate number of HPC. It was shown that the restoration of normal hematopoiesis was achieved in the majority of patients, suggesting that the chance of graft failure is minimized with cells mobilized with this procedure. The speed of hematopoietic recovery was consistent with an autograft-derived origin of predominantly diploid cells. No patient died as a consequence of the treatment. The majority of patients had stomatitis, which was severe in 50% of them, and only a minority of patients encountered some degree of hepatic and gastrointestinal toxicities. Specifically, no patient had busulfan-related pulmonary toxicity, hepatic veno-occlusive disease, or epileptic seizures. Thus, we can reasonably agree with the London data that the use of busulfan produces results at least as good as those achieved with total body irradiation (TBI)-containing regimens.38 There has been predominantly Ph-negative hematopoiesis in the majority of our patients for months postgrafting, and with the exception of 3 patients evolved in blastic crisis, there have been no hematologic relapses to date. The median follow-up postautograft was 24 (range, 6 to 76) months, and the actuarial survival at 3.5 years postautograft was 87% (95% CL: 54 to 97). Finally, most autograft recipients report normal or near-normal activity levels, suggesting that this procedure need not be associated either with prolonged convalescence or with chronic debility.

All autografted patients received postgraft immunobiological therapy with rhIL-2 and interferon-α in the attempt to delay the blastic evolution. Combination therapies of rhIL-2 and interferon-α have been shown to function synergistically in augmenting cytolytic activity in mice, both in vitro and in vivo.39-45 Both rhIL-2 and interferon-α were able to increase the suppressed natural killer cytolytic activity of lymphocytes isolated from CML patients.43,44 Recently, the efficacy of this combination has been confirmed in chronic and advanced phases of CML.45 

In conclusion, our data suggest that early transplantation with mobilized Ph-negative or prevalently Ph-negative is a technique worth developing further. Because it is well tolerated, it can increase the age for investigative treatment for CML; 23% of the 30 patients we have autografted were between 52 and 62 years of age. Taken together, the results achieved by our group suggest that this approach, when applied in early chronic phase, is able to restore and to maintain major or complete cytogenetic response in greater than 50% of the initial population. This percentage seems to be superior to that obtained with interferon therapy alone; however, such a small series cannot be compared with the results of large-scale trials. Randomized trials are needed to evaluate our procedure versus interferon-α ± low-dose ARA-C. This approach is being investigated in three randomized trials under way in Germany, the United States/United Kingdom (ECOG-MRC CML trial), and European Group for Blood and Marrow Transplantation.

We thank Mara Capurro for manuscript preparation.

Supported by Associazione Italiana Ricerca contro il Cancro (AIRC), 1998.

Address reprint request to Angelo M. Carella, MD, Hematology/ABMT Unit, Via Acerbi, 10/22, 16148 Genoa, Italy; e-mail:amcarella@smartino.ge.it.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Talpaz
M
Kantarjian
H
Kurzrock
R
Trujillo
JM
Gutterman
JU
Interferon-alpha produces sustained cytogenetic responses in chronic myelogenous leukemia.
Ann Intern Med
114
1991
532
2
Allan
NC
Richards
SM
Shepherd
PCA
UK Medical Research Council randomised, multicentre trial of interferon-alpha n1 for chronic myeloid leukemia: Improved survival irrespective of cytogenetic response.
Lancet
345
1995
1392
3
Hehlmann
R
Heimpel
H
Hasford
H
Kolb
HJ
Pralle
H
Hossefeld
DK
Queisser
W
Loffler
H
Hochhaus
A
Heinze
B
Georgii
A
Bartram
CR
Greisshammer
M
Bergmann
L
Essers
U
Falge
C
Queisser
U
Meyer
P
Schmitz
N
Eimermacher
H
Walther
F
Fett
W
Kleeberg
UR
Kabisch
A
Nerl
C
Zimmermann
R
Meuret
G
Tichelli
A
Kanz
L
Tigges
FJ
Schmid
L
Brockhaus
W
Tobler
A
Reiter
A
Perker
M
Emmerich
B
Verpoort
K
Zankovich
R
Wossow
PV
Prummer
O
Thiele
J
Buhr
T
Carbonell
F
Ansari
H
the German CML Study Group
Randomized comparison of interferon-alpha with busulfan and hydroxyurea in chronic myelogenous leukemia (CML).
Blood
84
1994
4064
4
The Italian Cooperative Study Group on Chronic Myeloid Leukemia
Interferon alfa-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia.
N Engl J Med
330
1994
820
5
Thomas
ED
Clift
RA
Fefer
A
Appelbaum
FR
Beutty
P
Bensinger
WI
Buckner
CD
Cheever
MA
Deeg
HJ
Doney
K
Fluornoy
N
Greenberg
P
Hansen
JA
Martin
P
McGuffin
R
Romberg
R
Sanders
JE
Singer
J
Stewart
P
Storb
R
Sullivan
K
Weiden
PL
Whiterspoon
R
Marrow transplantation for the treatment of chronic myelogenous leukemia.
Ann Intern Med
104
1986
155
6
Goldman
JM
Apperley
JF
Jones
L
Marcus
R
Goolden
AW
Batchelor
R
Hale
G
Waldmann
H
Reid
CD
Hows
J
Bone marrow transplantation for patients with chronic myeloid leukemia.
N Engl J Med
314
1986
202
7
Hansen
JA
Gooley
TA
Martin
PJ
Appelbaum
F
Channcey
TR
Clift
RA
Petersdort
EW
Radich
J
Sanders
JE
Storb
R
Sullivan
K
Anasetti
C
Bone marrow transplantation from unrelated donors for patients with chronic myelogenous leukemia.
N Engl J Med
328
1998
962
8
Coulombel
L
Kalousek
DK
Eaves
CJ
Gupta
CM
Eaves
CA
Long-term marrow culture reveals chromosomally normal hematopoietic progenitor cells in patients with Philadelphia chromosome-positive chronic myelogenous leukemia.
N Engl J Med
308
1983
1493
9
Udomsakdi
C
Eaves
CJ
Lansdorp
PM
Eaves
AC
Phenotypic heterogeneity of primitive leukemic hematopoietic cells in patients with chronic myeloid leukemia.
Blood
80
1992
2522
10
Eaves
C
Udomsakdi
C
Cashman
J
Barnett
M
Eaves
A
The biology of normal and neoplastic stem cells in CML.
Leuk Lymphoma
11
1993
245
11
Verfaillie
CM
Miller
WJ
Boylan
K
McGlave
PB
Selection of benign primitive hematopoietic progenitors in chronic myelogenous leukemia on the basis of HLA-DR antigen expression.
Blood
79
1992
1003
12
Petzer
AL
Eaves
CJ
Lansdorp
PM
Ponchio
L
Barnett
MJ
Eaves
AC
Characterization of primitive subpopulations of normal and leukemic cells present in the blood of patients with newly diagnosed as well as established chronic myeloid leukemia.
Blood
88
1996
2162
13
McGlave
P
Mamus
S
Vilen
B
Dewald
G
Effect of recombinant gamma interferon on chronic myelogenous leukemia bone marrow progenitors.
Exp Hematol
15
1987
331
14
Hoyle
C
Gray
R
Goldman
J
Autografting for patients with CML in chronic phase: An update.
Br J Haematol
86
1994
76
15
Carlo Stella
C
Mangoni
L
Piovani
G
In vitro marrow purging in chronic myelogenous leukaemia: Effect of mafosfamide and recombinant granulocyte-macrophage colony-stimulating factor.
Bone Marrow Transplant
8
1991
265
16
Reiffers
J
Trouette
R
Marit
G
Marit
G
Montastruc
M
Faberes
C
Cony-Mokhoul
P
David
B
Bourdeau
J
Bilhou-Nabera
C
Lacombe
F
Feuillatre-Fabre
F
Vezon
G
Bernard
PH
Broustet
A
Autologous blood stem cell transplantation for chronic granulocytic leukaemia in transformation: A report of 47 case.
Br J Haematol
77
1991
339
17
De Fabritiis
P
Petti
MC
Montefusco
E
De Propris
MS
Sala
R
Bellucci
R
Mancini
M
Lisci
A
Bonetto
F
Geiser
T
Calabretta
B
Mandelli
F
BCR-ABL antisense oligodoxynucleotide in vitro purging and autologous bone marrow transplantation for patients with chronic myelogenous leukemia in advanced phase.
Blood
91
1998
3156
18
Barnett
MJ
Eaves
CJ
Phillips
GL
Gascoyne
RD
Hogge
DE
Horsman
DE
Humphries
RK
Klingemann
HG
Lansdorp
PM
Nantel
SH
Reece
DE
Shepherd
JD
Spinelli
JJ
Sutherland
HJ
Eaves
AC
Autografting with cultured marrow in chronic myelogenous leukemia: Results of a pilot study.
Blood
84
1994
724
19
McGlave
PB
Arthur
D
Miller
WJ
Lasky
L
Kersey
J
Autologous transplantation for CML using marrow treated ex vivo with recombinant human interferon gamma.
Bone Marrow Transplant
6
1990
115
20
McGlave
PB
De Fabritiis
P
Deisseroth
A
Goldman
J
Barnett
M
Reiffers
J
Simonsson
B
Carella
A
Aeppli
D
Autologous transplants for chronic myelogenous leukemia: Results from eight transplant groups.
Lancet
343
1994
1486
21
Carella
AM
Podestà
M
Frassoni
F
Raffo
MR
Pollicardo
N
Pungolino
E
Vimercati
R
Sessarego
M
Parodi
C
Rabitti
C
Ferrero
R
Benvenuto
F
Figari
O
Carlier
P
Levcasis
G
Valbonesi
M
Vitale
V
Giordano
D
Pierluigi
D
Nati
S
Guerraccio
A
Rosso
C
Saglio
G
Collection of “normal” blood repopulating cells during early hemopoietic recovery after intensive conventional chemotherapy in chronic myelogenous leukemia.
Bone Marrow Transplant
12
1993
267
22
Carella
AM
Cunningham
I
Lerma
E
Dejana
A
Benvenuto
F
Podestà
M
Celesti
L
Chimirri
F
Abate
M
Vassallo
F
Figari
O
Parodi
C
Sessarego
M
Valbonesi
M
Carlier
P
Prencipe
E
Gatti
AM
Van Den Berg
D
Hoffman
R
Frassoni
F
Mobilization and transplantation of Philadelphia-negative peripheral blood progenitor cells early in chronic myelogenous leukemia.
J Clin Oncol
15
1997
1575
23
Corsetti
MT
Podestà
M
Lerma
E
Li Pira
G
Manca
F
Carella
AM
Peripheral blood progenitor cells mobilized early at diagnosis in patients with chronic myelogenous leukemia contain very low amount of BCR-ABL transcripts.
Leukemia
12
1998
998
24
Kaplan
EL
Meier
P
Non parametric estimation from incomplete observations.
J Am Stat Assoc (B)
53
1958
457
25
Bearman
SI
Appelbaum
FR
Buckner
CD
Petersen
FB
Fisher
LD
Clift
RA
Thomas
ED
Regimen-related toxicity in patients undergoing bone marrow transplantation.
J Clin Oncol
6
1988
1562
26
Meloni
G
Vignetti
M
Cardillo
AMF
Ablative chemotherapy followed by peripheral blood stem cells reinfusion for chronic myelogenous leukemia in chronic phase.
Bone Marrow Transplant
14
1994
103
(abstr, suppl 3)
27
Simonsson
B
Oberg
G
Killander
A
Bjoreman
M
Bjorkholm
M
Gahrton
G
Stenke
L
Turesson
I
Udén
AM
Malm
C
Vilén
L
Wahlin
A
Lofvenber
E
Carneskog
J
Westin
J
for the Swedish CML-group
Intensive treatment in order to minimize the Ph-positive clone in chronic myelogenic leukemia (CML).
Bone Marrow Transplant
14
1994
555
(suppl 3)
28
McGlave
P
Miller
J
Miller
W
Perry
E
Fautsch
S
Ramsay
NKC
Verfaillie
CM
Weisdorf
D
Autologous marrow transplant therapy for chronic myelogenous leukemia using marrow treated ex vivo with human recombinant interferon gamma.
Blood
84
1994
537a
(abstr, suppl 1)
29
Vogler
WR
Winton
EF
James
S
O’Neill
S
Granade
S
Mallard
G
Autologous marrow transplantation after karyotipic conversion to normal in blastic phase of chronic myelocytic leukemia.
Am J Med
75
1983
1080
30
Fischer
T
Neubamer
A
Mohm
J
Huhn
D
Dusemann
C
Link
H
Arseniev
L
Bussing
B
Novotny
J
Ganser
A
Duyster
J
Bunjes
D
Kreiter
S
Aulitzky
W
Hehlmann
R
Huber
C
Chemotherapy-induced mobilization of karyotypically normal PBSC for autografting in CML.
Bone Marrow Transplant
21
1998
1029
31
Heinzinger
M
Waller
CF
Rosenstiel
A
Scheid
S
Burger
KJ
Lange
W
Quality of IL-3 and G-CSF mobilized peripheral blood stem cells in patients with early chronic phase CML.
Leukemia
12
1998
333
32
Becker
M
Fabrega
S
Belloc
F
Rice
A
Barbu
V
Reiffers
J
Interferon gamma is effective for BM purging in a patient with CML.
Bone Marrow Transplant
12
1993
155
33
Gewirtz
A
Treatment of CML with c-myb antisense oligodeoxynucleotides.
Bone Marrow Transplant
14
1994
S57
S61
(suppl 3)
34
Luger
SM
Ratajczak
MZ
Stadtmauer
EA
Mangan
P
Magee
D
Silberstein
L
Edelstein
M
Nowell
P
Gewirtz
AM
Autografting for chronic myeloid leukemia (CML) with C-MYB antisense oligodeoxynucleotides purged bone marrow: A preliminary report.
Blood
84
1994
151a
(abstr, suppl 1)
35
O’Brien
SG
Rule
SA
Ratajczak
MZ
Luger
SM
Goldman
JM
Gewirtz
AM
Autografting for CML using bone marrow purged with MYB antisense oligonucleotide.
Br J Haematol
89
1995
12
(abstr, suppl 1)
36
Kiehntopf
M
Esquivel
EL
Brach
MA
Herrmann
F
Clinical applications of ribozymes.
Lancet
345
1995
1027
37
Pachuk
CJ
Yoon
K
Moelling
K
Coney
LR
Selective cleavage of bcr-abl chimeric RNAs by a ribozyme targeted to non-contiguous sequences.
Nucleic Acids Res
22
1994
301
38
O’Brien
S
Goldman
JM
Busulfan alone as cytoreduction before autografting for chronic myelogenous leukemia.
Blood
91
1998
1091
39
Brunda
MJ
Tarnowsld
D
Davatelis
V
Interaction of recombinant interferons with recombinant interleukin-2. Differential effects on natural killer cell activity and interleukin-2 activated killer cells.
Int J Cancer
37
1986
787
40
Brunda
MJ
Bellantoni
D
Sutich
V
In vivo anti-tumor activity of combinations of interferon alpha and interleukin-2 in a murine model. Correlation of efficacy with the induction of cytotoxic cells resembling natural killer cells.
Int J Cancer
40
1987
365
41
Igio
M
Sakurai
M
Tamura
T
Saijo
N
Hoshi
A
In vivo anti-tumor activity of multiple injections of recombinant interleukin-2, alone and in combination with three different types of recombinant interferon, on various syngeneic murine tumors.
Cancer Res
48
1988
260
42
Kuribayashi
K
Gillis
S
Kern
DE
Henney
CS
Murine NK cell cultures: Effects of interleukin-2 and interferon on cell growth and cytotoxic activity.
J Immunol
126
1981
2321
43
Verfaillie
C
Kay
N
Miller
W
McGlave
P
Diminished A-LAK cytotoxicity and proliferation accompany disease progression in chronic myelogenous leukemia.
Blood
76
1990
401
44
Hauch
M
Gazzola
MV
Small
T
Bordignon
C
Barnett
L
Cunningham
I
Castro-Malaspinia
H
O’Reilly
RJ
Keever
CA
Anti-leukemia potential of interleukin-2 activated natural killer cells after bone marrow transplantation for chronic myelogenous leukemia.
Blood
75
1990
2250
45
Nagler
A
Ackerstein
A
Barak
V
Slavin
S
Treatment of chronic myelogenous leukemia with rhIL-2 and interferon-α-2a.
J Hematotherapy
11
1994
75
46
Talpaz
M
Kantarjian
H
Liang
J
Calvert
L
Hamer
J
Tibbits
P
Durett
A
Claxton
D
Giralt
S
Khouri
I
Przpiorka
D
Van Besien
K
Andersson
B
Mehra
R
Gajewski
J
Seong
D
Hester
J
Estey
E
Korbling
M
Pollicardo
N
Berenson
R
Heimfeld
S
Champlin
R
Deisseroth
AB
Percentage of Philadelphia-chromosome (Ph)-negative and Ph-positive cells found after autologous transplantation for CML depends on percentage of diploid cells induced by conventional-dose chemotherapy before collection of autologous cells.
Blood
85
1995
3257
47
Deisseroth
AB
Zu
Z
Claxton
D
Hanania
EG
Fu
S
Ellerson
D
Goldberg
L
Thomas
M
Janicek
K
Anderson
WF
Hester
J
Korbling
M
Durett
A
Moen
R
Berenson
R
Heimfeld
S
Hamer
J
Calvert
L
Tibbits
P
Talpaz
M
Kantarjian
H
Champlin
R
Reading
C
Genetic marking shows that Ph+ cells present in autologous transplant of chronic myelogenous leukemia (CML) contribute to relapse after autologous bone marrow in CML.
Blood
83
1994
3068
48
Chalmers
EA
Franklin
IM
Kelsey
SM
Newland
AC
Clark
RE
Sproul
AM
Crotty
G
McCann
SR
Fielding
A
Goldstone
AH
Hepplestone
A
Watson
W
Sharp
RA
Tansey
P
Treatment of chronic myeloid leukaemia in first chronic phase with idarubicin and cytarabine: Mobilization of Philadelphia-negative peripheral blood stem cells.
Br J Haematol
96
1997
627
Sign in via your Institution