Interferon α (IFN-α) therapy improves prognosis in Philadelphia chromosome (Ph)–positive chronic myelogenous leukemia (CML). Polyethylene glycol (PEG) attached to IFN-α prolongs its half-life and may offer better therapy. The aims of this phase 1 study were to define the maximal tolerated dose (MTD), dose-limiting toxicities (DLTs), and response with PEG IFN-α-2b. Twenty-seven adults with Ph+ CML in chronic or accelerated phases, in whom IFN-α treatment had failed, were studied. Patients had hematologic (9 patients) or cytogenetic resistance (12 patients) or intolerance to IFN-α (6 patients). PEG IFN-α-2b was given as a weekly subcutaneous injection starting at 0.75 μg/kg weekly and escalating to 1.5, 3, 4.5, 6, 7.5, and 9.0 μg/kg. The MTD was defined at 7.5 to 9 μg/kg; DLT included severe fatigue, neurotoxicity, liver function abnormalities, and myelosuppression. Longer administration of PEG IFN-α-2b resulted in chronic side effects not observed earlier, which defined the MTD and DLT. The proposed phase 2 dose of PEG IFN-α-2b was 6 μg/kg weekly. Among 19 patients with active disease, 7 (37%) achieved complete hematologic response (CHR); 2 (11%) had a cytogenetic response (complete). Among 8 patients treated in CHR, 7 (87%) improved cytogenetic response to complete (4 patients) or partial (3 patients). All 6 patients intolerant to IFN-α tolerated PEG IFN-α-2b; 4 improved their cytogenetic response. The results show that PEG IFN-α-2b is easier to deliver (once weekly), better tolerated, and perhaps more effective than IFN-α.

Interferon α (IFN-α) therapy has been associated with improved prognosis in several cancers including chronic myelogenous leukemia (CML), lymphomas, multiple myeloma, renal cancer, and melanoma.1-7 In Philadelphia chromosome (Ph)-positive (Ph+) CML, single-arm and randomized studies have demonstrated the benefit of IFN-α alone or in combination with cytosine arabinoside (ara-C) in improving outcome of patients with CML.8-18 

Patient compliance to prolonged treatment with IFN-α in CML is an important factor for achieving clinical benefit.1-3,19Antitumor efficacy appears related to higher dose schedules of IFN-α daily therapy.1,20,21 Similarly, trials in CML suggest that an increased area under the curve (AUC), and associated prolonged tumor exposure to IFN-α, may be important in mediating the antileukemic effects. Therapy with IFN-α is cumbersome and associated with significant side effects requiring dose reductions and temporary or permanent treatment interruptions in 10% to 50% of patients.1-3 

Polyethylene glycol (PEG) is a linear, hydrophobic, uncharged, flexible polymer available in a variety of molecular weights.22,23A semisynthetic formulation (protein-polymer conjugate of IFN-α-2b) was developed by attaching a single PEG 12 000 molecule to the ε amino group of selected lysine residues in the IFN-α-2b molecule or the N-terminal amino acid.24 PEG modification of proteins has led to the development of several PEG-proteins of current or future potential importance in therapy, including PEG-asparaginase,25,26 PEG-erythropoietin,27PEG-granulocyte colony stimulating factor, recombinant PEG human megakaryocyte growth and development factor,28,29 and others.30,31 PEG modification of proteins prolongs their plasma half-life, reduces antigenicity and immunogenicity, and reduces sensitivity to proteolysis.32,33 The new PEG-proteins may also at times be more effective than the unmodified molecules.25,27,32-35 

We hypothesized that the new PEG IFN-α-2b, with its significantly prolonged plasma half-life from minutes to days, may allow easier treatment schedules (weekly instead of daily injections). By reducing immunogenicity and antigenicity, and by reducing access of the larger molecule to specific organs, it may also be associated with a lower incidence of side effects and improved patient tolerance and compliance. Finally, prolongation of plasma half-life and subsequent increase in AUC may enhance the therapeutic efficacy of PEG IFN-α-2b compared with IFN-α-2b. This report summarizes our phase 1 experience with PEG IFN-α-2b in patients with CML. In addition to defining the dose-limiting toxicity (DLT) and maximum tolerated dose (MTD), we present detailed plasma pharmacokinetic studies and analyze tolerance and response to PEG IFN-α-2b following exposure to IFN-α therapy.

Study group

Adults aged 18 years or older referred to or treated at our institution with a diagnosis of Ph+ CML were eligible. Entry criteria required (1) failure on prior IFN-α therapy; (2) adequate performance status (ECOG performance 0-2), adequate liver (bilirubin, aspartate aminotransferase [AST], alanine aminotrasnferase [ALT] ≤ 2 × normal) and renal functions (creatinine ≤ 2 mg/100 mL) and cardiac status; and (3) absence of serious neuropsychiatric complications, unless directly related to prior IFN-α therapy.

Failure on IFN-α could be because of (1) hematologic resistance defined by a failure to achieve a complete hematologic response (CHR) after 6 months of IFN-α therapy or rising white blood cell (WBC) count above 12 × 109/L despite optimal IFN-α–based therapy; (2) unsatisfactory cytogenetic response defined as lack of Ph suppression after 12 months or more of IFN-α therapy, Ph suppression to 35% Ph+ cells or more after 18 or more months of IFN-α therapy, or loss of cytogenetic response (> 30% increase in Ph+ metaphases); or (3) significant toxicity defined as grade 3 to 4 side effects.

Patients in blastic phase of CML were not eligible.36Patients in accelerated phase of CML, as previously defined,37 were not eligible to be treated, except if they had clonal evolution as the only accelerated phase criterion. One patient was registered and later re-evaluated as having more than 15% blasts (patient 26 in Table 5). He was analyzed as part of the study group. All patients signed the appropriate informed consent, as required by institutional guidelines.

Therapy

Treatment with PEG IFN-α-2b was given as a single subcutaneous (SQ) injection weekly. Patients were taught self-injections and were observed as outpatients for 1 month of treatment prior to continuing therapy under the care of their referring physicians. The starting dose schedule, based on 2 previous phase 1 studies in normal volunteers and in patients with compensated chronic hepatitis C, was 0.75 μg/kg SQ weekly.38,39 Subsequent dose escalations were to 1.5, 3, 4.5, 6, 7.5, and 9 μg/kg weekly.

The phase 1 study followed the classical “3 + 3 design” used in most phase 1 studies. Three patients were entered at one dose level and observed for at least 3 weeks. If none of the 3 experienced grade 3 or worse toxicity, subsequent patients were entered on the next dose level. If 1 of 3 patients experienced grade 3 or worse toxicity, 3 more patients were treated at the same dose level. If 1 of 6 patients experienced grade 3 or worse toxicity, subsequent patients were entered at the next dose level. If 3 or more patients (of 3-6 depending on the details of the level) experienced grade 3 or worse toxicity, the DLT was exceeded and subsequent patients were treated at the next lower dose level, or a dose in between depending on the experience at the previous level. If 2 of 6 patients experienced grade 3 toxicity at a particular dose level, it defined the DLT and MTD, and the recommended dose for phase 2 studies was then defined at a dose level below the MTD.

Pretreatment and follow-up studies

Pretreatment evaluation required history and physical examination, complete blood counts, differential and platelet counts, serum chemistries (SMA12) including liver and renal functions, bone marrow aspiration for morphology and cytogenetic analysis, and additional studies as indicated. All patients had received prior IFN-α therapy. Documentation of prior IFN-α therapy response, dose schedule, and toxicities was performed when such information was available.

Follow-up studies included complete blood count, platelet and differential at least weekly for 4 weeks, then every 2 to 4 weeks; SMA12 weekly for 4 weeks, then every 4 to 8 weeks; marrow aspiration and cytogenetic analysis at 6 weeks, 3 months, and then every 3 to 6 months on therapy.

Pharmacokinetic studies of PEG IFN-α-2b

The pharmacokinetics of PEG IFN-α-2b were assessed using sparsely sampled serum concentration values obtained at 0, 24, 48, 72, and 168 hours after dosing at weeks 1 and 4. Serum PEG IFN-α-2b concentrations were determined using a validated electrochemiluminescence (ECL) assay with a lower limit of quantitation (LOQ) of 50 pg/mL. The assay was linear and reproducible between 50 and 2000 pg/mL.40 

Individual serum PEG IFN-α-2b concentration-time data were used for pharmacokinetic analysis with model-independent methods.41Serum concentrations below the LOQ were reported as 0 pg/mL. The area under the serum concentration-time curve from time 0 to 168 hours (AUC[0-168 hours]) was calculated using the linear trapezoidal method. The average serum concentration (Cavg) was determined as follows: Cavg = [AUC(0-168 hr)] / 168 hr. The accumulation factor (R) was determined as follows: R = [AUC(0-168 hr), week 4] / [AUC(0-168 hr), week 1]. True Cmax, Tmax, and half-life values were not calculated due to the sparse sampling schedule.

Response and toxicity criteria

Response was defined as previously described.1,2 A CHR required normalization of blood counts (WBC count < 10 × 109/L, platelets < 450 × 109/L), no peripheral blasts, and disappearance of all signs and symptoms of CML, including palpable splenomegaly. CHR was further classified by the degree of cytogenetic response: complete if Ph+ cells were 0%; partial if Ph+ cells were 1% to 34%; minor if Ph+ cells were 35% to 90%.

Toxicity grade was based on the National Cancer Institute (NCI) common toxicity criteria.42 

Statistical considerations

Statistical methods were descriptive for response and side effects.

Study group

Twenty-seven patients with Ph+ CML were treated (Table1). Their median age was 47 years (range, 21-66 years); 5 (18%) were women. The median duration of disease was 50 months (range, 7-144 months). Eighteen patients had chronic phase CML (1 was in second chronic phase); 9 patients had accelerated phase CML based on the following: blasts 15% or more, 1 patient; cytogenetic clonal evolution only, 8 patients.

Table 1.

Characteristics of the 27 study group patients

CharacteristicNo.
Age at least 60 y [median] 3 [47]  
Splenomegaly 
Disease phase  
 Chronic 17  
 Second chronic 
 Accelerated 9  
Duration of disease (mo)  
 Fewer than 12 2  
 12-36 7  
 More than 36 18  
Hemoglobin (< 10 g/dL) 1  
WBC (> 20 × 109/L) 
Platelets (> 450 × 109/L) 6  
Reason for change from IFN-α  
 Hematologic resistance/partial hematologic response  4/5  
 Unsatisfactory cytogenetic response 12  
 Toxicities with IFN-α 6  
Philadelphia chromosome percent status before PEG IFN-α-2b  
 100% 20 
 35%-90% 4  
 15%-30% 3  
Disease status at start of therapy  
 Active 19  
 CHR 
CharacteristicNo.
Age at least 60 y [median] 3 [47]  
Splenomegaly 
Disease phase  
 Chronic 17  
 Second chronic 
 Accelerated 9  
Duration of disease (mo)  
 Fewer than 12 2  
 12-36 7  
 More than 36 18  
Hemoglobin (< 10 g/dL) 1  
WBC (> 20 × 109/L) 
Platelets (> 450 × 109/L) 6  
Reason for change from IFN-α  
 Hematologic resistance/partial hematologic response  4/5  
 Unsatisfactory cytogenetic response 12  
 Toxicities with IFN-α 6  
Philadelphia chromosome percent status before PEG IFN-α-2b  
 100% 20 
 35%-90% 4  
 15%-30% 3  
Disease status at start of therapy  
 Active 19  
 CHR 

Clonal evolution included double Ph, 2 patients; double Ph with trisomy 8, 1 patient; double Ph with trisomy 8 and isochromosome 17, 1 patient; trisomy 8, 1 patient; and other translocations [t(16;18), t(13;16), t(10;11)], 3 patients. The t(10;11) was also associated with additional changes.

Nineteen patients had active CML at the start of therapy; 8 patients were in CHR. Reasons for entering the study included hematologic resistance to IFN-α in 9 patients, unsatisfactory cytogenetic response (defined under “Patients and methods”) in 12 patients, and intolerance to IFN-α in 6 patients. Patient 13 (Table 5) who had a cytogenetic response and clonal evolution [t(9;22) plus t(16;18) in 15% of metaphases] is considered in the unsatisfactory cytogenetic response category. The median duration of IFN-α therapy in the 9 patients with hematologic resistance was 37 months (range, 8-118 months), and in the 12 patients with cytogenetic resistance 42 months (range, 12-92 months). Two patients in the group with intolerance to IFN-α still had a cytogenetic response at the start of PEG IFN-α-2b (patient 17, Ph 50%; patient 20, Ph 30%; Table 5), whereas the other 4 were 100% Ph+. One patient who was not hematologically resistant to IFN-α (patient 9 in Table 5) had a different Ph+ status between the last IFN-α evaluation and beginning PEG IFN-α-2b. In the evaluation for cytogenetic response, Ph status prior to beginning PEG IFN-α-2b was considered. The 6 patients intolerant to IFN-α (Table 5) were removed from the study for the following reasons: grade 3 toxicity with necrotizing skin reactions at the sites of injections (patient 1); grade 3 fatigue and difficult concentration, grade 2 joint aches, and shortness of breath (patient 9); grade 3 fatigue, aches, and depression, grade 2 nausea and stiffness (patient 14); grade 3 depression and grade 2 fatigue (patient 17); grade 3 pulmonary toxicity (patient 18); and grade 3 irritability and depression, and grade 2 fatigue and muscle and bone aches (patient 20).

Side effects

Toxicities at each dose level are detailed in Tables2 and 3. At the dose level ranges of PEG IFN-α-2b 0.75 to 4.5 μg/kg SQ weekly, side effects were mild to moderate and included flulike symptoms (fever, chills) most prominent with the first injection, lasting for 24 to 48 hours and subsiding, fatigue, and aches. Tolerance to fever and flulike symptoms developed after multiple injections, although fatigue tended to increase. Only one patient experienced grade 2 nausea at 4.5 μg/kg, and another had grade 2 skin rash at 4.5 μg/kg (not shown for simplification). No other patients experienced such toxicities at these levels or at dose levels of 6 to 9 μg/kg.

Table 2.

Nonhematologic side effects with PEG IFN-α-2b

Dose level
(μg/kg/wk)
No.Fatigue, achesDepression
2
NeurotoxicityFever, flu
2
Liver
abnormalities
Weight loss
1-2
23-423-423-4
0.75          
1.5         
     
4.5        
     
7.5  
 
Dose level
(μg/kg/wk)
No.Fatigue, achesDepression
2
NeurotoxicityFever, flu
2
Liver
abnormalities
Weight loss
1-2
23-423-423-4
0.75          
1.5         
     
4.5        
     
7.5  
 

Number of patients with toxicity grade is shown under each toxicity column.

Table 3.

Hematologic side effects with PEG IFN-α-2b

Dose level (μg/kg/wk)No.AnemiaThrombocytopeniaGranulocytopenia
23-423-423-4
0.75       
1.5     
     
4.5      
     
7.5 
  
Dose level (μg/kg/wk)No.AnemiaThrombocytopeniaGranulocytopenia
23-423-423-4
0.75       
1.5     
     
4.5      
     
7.5 
  

Number of patients with toxicity grade is shown under each toxicity column.

At PEG IFN-α-2b 6 μg/kg, one patient each had grade 2 fatigue, grade 2 depression, fever, and flulike symptoms. One patient developed grade 3 liver toxicity (AST 353 IU/L, ALT 343 IU/L) after 9 weeks of therapy, which reverted to normal 4 weeks after treatment interruption and remained normal with reinstitution of one dose level reduction of PEG-IFN-α-2b (4.5 μg/kg SQ weekly). One patient developed neutropenia of 0.9 × 109/L after 16 months of therapy.

At 7.5 to 9 μg/kg, grade 3 fatigue and aches were observed in 3 of 12 patients, grade 3 neurotoxicity in 3 of 12 patients, and grade 3 liver abnormalities in 2 of 12 patients. Neurotoxicity included apathy, difficulty in thinking and concentration, and memory problems. Thrombocytopenia less than 50 × 109/L was observed in 2 of 6 patients at 7.5 μg/kg, and in 2 of 6 patients at 9 μg/kg. Granulocytopenia less than 0.5 × 109/L was observed in 1 of 6 patients at 7.5 μg/kg and in 1 of 6 patients at 9 μg/kg. Although the toxicity evaluation to escalate to the next dose level was after 1 month of therapy, some of the grade 3 toxicities developed after longer periods of therapy. This explains the grade 3 to 4 myelosuppression at 1.5 μg/kg not accounted for in the dose escalation because it occurred later. Other grade 3 to 4 hepatic and neurotoxicities also occurred at times beyond the first month of study. Thus, the cumulative experience with longer therapy indicated that PEG IFN-α-2b dose schedules of 7.5 to 9 μg/kg weekly defined the MTD with reasonable certainty among the 12 patients treated at these 2 dose levels. The DLTs were defined by severe fatigue, severe neurotoxicity, liver dysfunction, and severe myelosuppression as defined by the NCI criteria. None of the marrow toxicities were associated with complications such as febrile infections or need for transfusions. The PEG IFN-α-2b dose schedule of 6 μg/kg weekly was the recommended dose as a single agent for phase 2 studies, one dose below the MTD, considering the small numbers of patients treated.

Response and tolerance

Response by disease status at start of PEG IFN-α-2b (active, CHR), and by prior last IFN-α response are shown in Table4. Among 19 patients with active disease at the start of therapy, 7 achieved CHR (37%), and 3 had a partial hematologic response (PHR), for an overall response rate of 53%. Two patients achieved a complete cytogenetic response (patients 9 and 14 in Table 5). One patient in accelerated phase with 15% or more blasts returned to a second chronic phase (patient 26 in Table 5). Among 8 patients entered into the study in CHR, 7 achieved or improved their cytogenetic response. Four patients with minor cytogenetic responses at the start of PEG IFN-α-2b therapy improved to partial cytogenetic response (2 patients: Ph from 75% to 25%, and Ph from 69% to 17%, respectively), and to complete cytogenetic response (2 patients; both from Ph 50% to 0%). The other 3 patients starting with a partial cytogenetic response either improved their response (1 patient: Ph 30% to 5%), or achieved a complete cytogenetic response (2 patients: Ph from 20% to 0% and from 5% to 0%, respectively). All 6 patients treated for IFN-α intolerance were able to tolerate PEG IFN-α-2b; 4 achieved a cytogenetic response, 1 partial and 3 complete.

Table 4.

Response to PEG IFN-α-2b by disease status and prior response to IFN-α

TreatedPHRCHRCytogenetic response
MinorPartialComplete
Disease status       
 Active 19 — — 
 CHR NA NA — 
Status prior to therapy       
 Hematologic resistance to IFN-α — — — — 
 Cytogenetic status prior to PEG IFN-α-2b       
  Ph 95%-100% 11 — NA — — 2  
  Ph 50%-75% — NA — 2  
  Ph 15%-35% — NA — 2  
Intolerance to IFN-α — — — 
TreatedPHRCHRCytogenetic response
MinorPartialComplete
Disease status       
 Active 19 — — 
 CHR NA NA — 
Status prior to therapy       
 Hematologic resistance to IFN-α — — — — 
 Cytogenetic status prior to PEG IFN-α-2b       
  Ph 95%-100% 11 — NA — — 2  
  Ph 50%-75% — NA — 2  
  Ph 15%-35% — NA — 2  
Intolerance to IFN-α — — — 

Entries are numbers of patients. Patient with accelerated phase CML and ≥ 15% blasts returned to second chronic phase, but is evaluated as resistant because he did not achieve PHR or CHR (patient 26 in Table 5).

NA indicates not applicable.

Table 5.

Details of patient and disease characteristics and response to PEG IFN-α-2b

Patient
no.
Age
(y)
CML duration
(mo)
PEG IFN-α-2b
dose (μg/kg/wk)
Last IFN-α
response
CML phase
(active/CHR)
WBC
pretreatment
(× 109/L)
Percent Ph+cellsOverall
response on
PEG IFN-α-2b
Pre-PEG
IFNα-2b
Lowest on
therapy
15-150 57 52 0.75 CHR Second CP-A 15.6 100 NA Resistant 
23 97 0.75 PHR Acc-A 25.4 100 NA Resistant 
60 43 0.75 PHR CP-A 14.6 100 NA Resistant 
55 58 1.5 CHR CP-A 13.3 100 100 CHR 
21 12 1.5 PHR Acc-A 21.8 100 NA Resistant 
55 137 1.5 Resistant CP-A 38.6 100 100 CHR 
46 36 CHR CP-A 48.2 100 NA Resistant 
66 21 Resistant CP-A 73.6 100 100 CHR 
95-150 59 22 Minor CG CP-A 18.2 100 Complete CG 
10 40 38 4.5 CHR CP-A 12.3 95 NA Resistant 
11 31 79 4.5 Resistant CP-A 14.7 100 100 CHR 
12 35 109 4.5 CHR Acc-CHR 6.4 100 NA Resistant 
13 59 Partial CG Acc-CHR 4.4 15 Complete CG 
145-150 56 CHR CP-A 11.9 100 Complete CG  
15 34 99 Minor CG CP-CHR 8.0 50 Complete CG 
16 61 138 7.5 Resistant Acc-A 53.2 100 100 Resistant 
175-150 41 60 7.5 Minor CG CP-CHR 7.9 50 Complete CG 
185-150 33 78 7.5 CHR CP-A 9.3 100 NA Resistant 
19 47 50 7.5 CHR Acc-A 19.0 100 100 PHR 
205-150 50 25 7.5 Partial CG CP-CHR 4.6 30 Partial CG 
21 50 144 7.5 PHR CP-A 52.0 100 100 CHR 
22 45 45 CHR Acc-A 13.6 100 100 PHR 
23 33 90 Partial CG CP-CHR 6.5 20 Complete CG 
24 41 106 CHR CP-A 10.5 100 100 PHR 
25 47 30 Minor CG Acc-CHR 5.4 69 17 Partial CG 
265-151 56 50 PHR Acc-A5-151 71.1 100 100 Second CP  
27 54 14 Minor CG CP-CHR 7.2 75 25 Partial CG 
Patient
no.
Age
(y)
CML duration
(mo)
PEG IFN-α-2b
dose (μg/kg/wk)
Last IFN-α
response
CML phase
(active/CHR)
WBC
pretreatment
(× 109/L)
Percent Ph+cellsOverall
response on
PEG IFN-α-2b
Pre-PEG
IFNα-2b
Lowest on
therapy
15-150 57 52 0.75 CHR Second CP-A 15.6 100 NA Resistant 
23 97 0.75 PHR Acc-A 25.4 100 NA Resistant 
60 43 0.75 PHR CP-A 14.6 100 NA Resistant 
55 58 1.5 CHR CP-A 13.3 100 100 CHR 
21 12 1.5 PHR Acc-A 21.8 100 NA Resistant 
55 137 1.5 Resistant CP-A 38.6 100 100 CHR 
46 36 CHR CP-A 48.2 100 NA Resistant 
66 21 Resistant CP-A 73.6 100 100 CHR 
95-150 59 22 Minor CG CP-A 18.2 100 Complete CG 
10 40 38 4.5 CHR CP-A 12.3 95 NA Resistant 
11 31 79 4.5 Resistant CP-A 14.7 100 100 CHR 
12 35 109 4.5 CHR Acc-CHR 6.4 100 NA Resistant 
13 59 Partial CG Acc-CHR 4.4 15 Complete CG 
145-150 56 CHR CP-A 11.9 100 Complete CG  
15 34 99 Minor CG CP-CHR 8.0 50 Complete CG 
16 61 138 7.5 Resistant Acc-A 53.2 100 100 Resistant 
175-150 41 60 7.5 Minor CG CP-CHR 7.9 50 Complete CG 
185-150 33 78 7.5 CHR CP-A 9.3 100 NA Resistant 
19 47 50 7.5 CHR Acc-A 19.0 100 100 PHR 
205-150 50 25 7.5 Partial CG CP-CHR 4.6 30 Partial CG 
21 50 144 7.5 PHR CP-A 52.0 100 100 CHR 
22 45 45 CHR Acc-A 13.6 100 100 PHR 
23 33 90 Partial CG CP-CHR 6.5 20 Complete CG 
24 41 106 CHR CP-A 10.5 100 100 PHR 
25 47 30 Minor CG Acc-CHR 5.4 69 17 Partial CG 
265-151 56 50 PHR Acc-A5-151 71.1 100 100 Second CP  
27 54 14 Minor CG CP-CHR 7.2 75 25 Partial CG 

CP indicates chronic phase; Acc, accelerated phase; NA, not applicable; A, active disease; CG, cytogenetic; Minor CG, Ph 35%-90%; partial CG, Ph 1%-34%; complete CG, Ph 0%.

F5-150

Entered for IFN-α intolerance.

F5-151

Accelerated blasts ≥ 15%.

Table 5 lists the individual patient and disease characteristics including prior response to IFN-α, disease status at the start of PEG IFN-α-2b, and response evaluation.

Table 6 details the follow-up cytogenetic studies among patients who achieved improved cytogenetic response with PEG IFN-α-2b. The median follow-up of patients is 25 months (range, 2-34 months). Currently, 24 of the 27 patients are alive, and 10 continue on PEG IFN-α-2b.

Table 6.

Follow-up cytogenetic studies among patients with improved cytogenetic response on PEG IFN-α-2b

PatientPercent Ph+ metaphases on PEG IFN-α-2b therapy
PretreatmentBest (overall)At 3 monthsAt 6 monthsAt 9 monthsAt 12 monthsAfter 12 months
[month of therapy]
100 — 95 95 100 65 [18], 40 [26], 0 [30] 
13 15 10 10 15 0 [14], 30 [17], 10 [20], 65 [24], 25 [26] 
14 100 100 95 95 75 55 [15], 0 [25] 
15 50 60 40 30 13 0 [16], 25 [25] 
17 50 0 [16], 0 [24] 
20 30 14 35 Insufficient  10 [13], 30 [17], 45 [24] 
23 20 15 5 [19] 
25 69 17 (1 mo) 90 90 100 — — 
27 75 25 75 47 25 — 25 [21] 
PatientPercent Ph+ metaphases on PEG IFN-α-2b therapy
PretreatmentBest (overall)At 3 monthsAt 6 monthsAt 9 monthsAt 12 monthsAfter 12 months
[month of therapy]
100 — 95 95 100 65 [18], 40 [26], 0 [30] 
13 15 10 10 15 0 [14], 30 [17], 10 [20], 65 [24], 25 [26] 
14 100 100 95 95 75 55 [15], 0 [25] 
15 50 60 40 30 13 0 [16], 25 [25] 
17 50 0 [16], 0 [24] 
20 30 14 35 Insufficient  10 [13], 30 [17], 45 [24] 
23 20 15 5 [19] 
25 69 17 (1 mo) 90 90 100 — — 
27 75 25 75 47 25 — 25 [21] 

Pharmacokinetic studies

PEG IFN-α-2b was well absorbed following SQ administration to patients with CML (Table 7). Serum concentrations increased in a dose-related manner at week 1, but not at week 4, which may be due to the high interpatient variability and the small number of patients (Figure 1). There appeared to be no dose-related changes in the accumulation of PEG IFN-α-2b (Table 7).

Table 7.

Mean pharmacokinetic parameters

ParameterPEG IFN-α-2b weekly dose (μg/kg)
0.751.53.04.56.07.59.0
Week 1        
 n    3    3    3    3    3    6    6  
 Cavg (pg/mL)    184    513    1 013    2 907    3 243    3 062    4 393  
 AUC(0-168 h) (pg/h/mL)    30 984    86 244    170 196    488 440    544 752    514 416    738 072  
Week 4                             
 Cavg (pg/mL)    288    725    1 338    6 931    5 548    3 006    4 592  
 AUC(0-168 h) (pg/h/mL)    48 384    121 868    224 756    1 164 476    932 124    505 022    771 410  
 R    1.54 1.59 1.36 2.997-150 1.65 1.26 1.19 
ParameterPEG IFN-α-2b weekly dose (μg/kg)
0.751.53.04.56.07.59.0
Week 1        
 n    3    3    3    3    3    6    6  
 Cavg (pg/mL)    184    513    1 013    2 907    3 243    3 062    4 393  
 AUC(0-168 h) (pg/h/mL)    30 984    86 244    170 196    488 440    544 752    514 416    738 072  
Week 4                             
 Cavg (pg/mL)    288    725    1 338    6 931    5 548    3 006    4 592  
 AUC(0-168 h) (pg/h/mL)    48 384    121 868    224 756    1 164 476    932 124    505 022    771 410  
 R    1.54 1.59 1.36 2.997-150 1.65 1.26 1.19 
F7-150

Including subject 11 (R = 1.49, excluding subject 11).

Fig. 1.

Concentration-time profiles.

The mean concentration-time profiles of PEG IFN-α-2b are shown for different dose levels (0.75-9.0 μg/kg) as measured on week 1 (dose 1) and week 4 (dose 4).

Fig. 1.

Concentration-time profiles.

The mean concentration-time profiles of PEG IFN-α-2b are shown for different dose levels (0.75-9.0 μg/kg) as measured on week 1 (dose 1) and week 4 (dose 4).

Close modal

Two previous phase 1 studies of PEG IFN-α-2b had been conducted that defined our current starting dose.38,39 In normal volunteers randomized to receive a single SQ injection of PEG IFN-α-2b (up to 0.5 μg/kg) or IFN-α-2b, side effects included flulike symptoms, redness at the injection site, headache, fatigue and malaise, and myelosuppression. Among patients with chronic active hepatitis C, PEG IFN-α-2b doses up to 1.5 μg/kg weekly were well tolerated without DLT, and a similar spectrum of side effects observed. Side effects were less severe at PEG IFN-α-2b doses up to 1 μg/kg relative to IFN-α-2b 3 MU (million units) 3 times weekly. Pharmacokinetics studies showed better profiles for PEG IFN-α-2b compared with IFN-α-2b. The mean absorption half-lives were similar at 4.6 versus 2.3 hours; however, the elimination half-life was 8- to 10-fold greater for PEG IFN-α-2b compared with IFN-α-2b (44 versus 4-5 hours). Mean apparent clearance was 10-fold less for PEG IFN-α-2b and the volume of distributions similar for both compounds.

In our phase 1 study of PEG IFN-α-2b in CML, the findings were encouraging. The DLT was defined by significant thrombocytopenia, but side effects including fatigue, weight loss, and neurotoxicity were observed. The MTD was defined at PEG IFN-α-2b dose of 7.5 to 9 μg/kg SQ weekly. The proposed dose of PEG IFN-α-2b for phase 2 studies is 6 μg/kg weekly. However, as with many other phase 1 to 2 studies this dose has to be evaluated according to the cumulative experience with longer durations of therapy, toxicities with combinations (eg, with ara-C), and with different characteristics of study group (eg, no prior IFN-α exposure, older patients).

The predicted AUC comparative dose to PEG IFN-α-2b 0.3 μg/kg weekly is IFN-α 9 MU weekly. Thus the PEG IFN-α-2b dose of 6 μ/kg weekly chosen for phase 2 studies is equivalent to IFN-α-2b 180 MU weekly, that is, 27 MU daily (about 15 MU/m2daily). This dose is at least 3 times the IFN-α doses used in previous CML studies, without increased toxicities. This may explain the superior anti-CML efficacy of PEG IFN-α-2b compared with IFN-α.

Of interest were the favorable results observed in particular study groups. For example, among 9 patients with definite hematologic resistance to prior optimal IFN-α therapy, 4 patients (44%) demonstrated response to PEG IFN-α-2b. Among patients starting with 100% Ph+ disease, 2 of 11 had improved cytogenetic response (Table 4). Cytogenetic responses improved in 9 of 27 patients treated (33%). Finally, all 6 patients treated because of intolerance to IFN-α were able to tolerate PEG IFN-α-2b, and 4 of them had a favorable response (Table 4). This indicates the better general tolerance of patients to PEG IFN-α-2b compared with IFN-α. Thus, of a total of 27 patients in whom IFN-α therapy had failed, 13 (48%) had a favorable response to PEG IFN-α-2b (either CHR or improved cytogenetic response).

PEG IFN-α-2b was well absorbed following SQ administration in CML. PEG IFN-α-2b serum concentrations increased in a dose-related manner at week 1 but not at week 4 (Table 7). This may have been due to high interpatient variability and the small number of patients assessed in this study.

In summary, PEG IFN-α-2b appears to provide easier schedule delivery, better tolerance, and perhaps improved efficacy compared with regular IFN-α. Future studies will evaluate PEG IFN-α-2b in combinations with other active anti-CML agents such as cytarabine, homoharringtonine, and the BCR-ABL tyrosine kinase inhibitor STI571 (Gleevec, Novartis, East Hanover, New Jersey), as well as in front-line CML therapy.

Supported in part by research funding from Schering to M.T. and H.K.

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
Kantarjian
HM
Deisseroth
A
Kurzrock
LR
Estrov
A
Talpaz
M
Chronic myelogenous leukemia: a concise update.
Blood.
82
1993
691
703
2
Kantarjian
HM
O'Brien
S
Anderlini
P
Talpaz
M
Treatment of chronic myelogenous leukemia: current status and investigational options.
Blood.
87
1996
3069
3081
3
Faderl
S
Talpaz
M
Estrov
Z
Kantarjian
HM
Chronic myelogenous leukemia: biology and therapy.
Ann Intern Med.
31
1999
207
219
4
Savage
PD
Muss
HB
Renal cell cancer.
Biologic Therapy of Cancer.
DeVita
VT
Hellman
S
Rosenberg
SA
1995
373
387
Lippincott
Philadelphia, PA
5
Solal-Celigny
P
Lepage
E
Brousse
N
et al
Recombinant interferon alfa-2b combined with a regimen containing doxorubicin in patients with advanced follicular lymphoma.
N Engl J Med.
329
1993
1608
1614
6
Ludwig
H
Cohen
AM
Polliack
A
et al
Interferon-alpha for induction and maintenance in multiple myeloma: results of two multi-center randomized trials and summary of other studies.
Ann Oncol.
6
1995
467
476
7
Kirkwood
JM
Strawderman
MH
Ernstoff
MS
Smith
TJ
Borden
EC
Blum
RH
Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group trial EST 1684.
J Clin Oncol.
14
1996
7
17
8
Talpaz
M
Kantarjian
H
Kurzrock
R
et al
Interferon-alpha produces sustained cytogenetic responses in chronic myelogenous leukemia.
Ann Intern Med.
114
1991
532
538
9
Kantarjian
HM
Smith
TL
O'Brien
S
et al
Prolonged survival in chronic myelogenous leukemia after cytogenetic response to interferon-α therapy.
Ann Intern Med.
122
1995
254
261
10
Italian Cooperative Study Group on Chronic Myeloid Leukemia
Interferon alfa-2a compared with conventional chemotherapy for the treatment of chronic myeloid leukemia.
N Engl J Med.
330
1994
820
825
11
Allan
NC
Richards
SM
Shepherd
PCA
et al
UK Medical Research Council randomized, multi-center trial of interferon-α n1 for chronic myeloid leukaemia: improved survival irrespective of cytogenetic response.
Lancet.
345
1995
1392
1397
12
Chronic Myeloid Leukemia Trialists' Collaborative Group
Interferon alfa versus chemotherapy for chronic myeloid leukemia: a meta-analysis of seven randomized trials.
J Natl Cancer Inst.
89
1997
1616
1620
13
Kantarjian
HM
O'Brien
S
Smith
TL
et al
Treatment of Philadelphia chromosome-positive early chronic phase chronic myelogenous leukemia with daily doses of interferon alpha and low-dose cytarabine.
J Clin Oncol.
17
1999
284
292
14
Kantarjian
H
Keating
M
Estey
E
et al
Treatment of advanced Philadelphia chromosome-positive chronic myelogenous leukemia with interferon-α and low-dose cytarabine.
J Clin Oncol.
10
1992
772
778
15
Arthur
CK
Ma
DFF
Combined interferon apha-2a and cytosine arabinoside as first-line treatment for chronic myeloid leukemia.
Acta Haematol.
89(suppl 1)
1993
15
21
16
Thaler
J
Hilbe
W
Apfelbeck
U
et al
Interferon-alpha-2c and low-dose ara-C for the treatment of patients with CML: results of the Austrian multi-center phase study.
Leuk Res.
21
1997
75
80
17
Guilhot
F
Chastang
C
Michallet
M
et al
Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia.
N Engl M Med.
337
1997
223
229
18
Rosti
G
Bonifazi
F
De Vivo
A
et al
Cytarabine increases karyotypic response and survival in αIFN treated chronic myelogenous leukemia patients: results of a national prospective randomized trial of the Italian Cooperative Study Group on CML [abstract].
Blood.
94
1999
600a
19
Ansari
H
Hasford
J
Hehlmann
R
Fallacies of the intention-to-treat analysis. The German CML Study Group [abstract].
J Mol Med.
75
1997
8243
20
Talpaz
M
Kantarjian
H
Low-dose interferon-a in chronic myelogenous leukemia.
Ann Intern Med.
122
1995
728
729
21
Alimena
G
Morra
E
Lazzarino
M
et al
Interferon alpha-2b as therapy for Ph1-positive chronic myelogenous leukemia: a study of 82 patients treated with intermittent or daily administration.
Blood.
72
1988
642
647
22
Francis
GE
Fisher
D
Delgado
C
Malik
F
Gardiner
A
Neale
D
PEGylation of cytokines and other therapeutic proteins and peptides: the importance of biological optimisation of coupling techniques.
Int J Hematol.
68
1998
1
18
23
Francis
GE
Delgado
C
Fisher
D
Malik
F
Agrawal
AK
Polyethylene glycol modification: relevance of improved methodology to tumour targeting.
J Drug Target.
3
1996
321
340
24
Wang
YS
Youngster
S
Bausch
J
Zhang
R
McNemar
C
Wyss
DF
Identification of the major positional isomer of pegylated interferon alpha-2b.
Biochemistry.
39
2000
10634
10640
25
Asselin
BL
Whitin
JC
Coppola
DJ
Rupp
IP
Sallan
SE
Cohen
HJ
Comparative pharmacokinetic studies of three asparaginase preparations.
J Clin Oncol.
11
1993
1780
1786
26
Ettinger
LJ
Asselin
B
Poplack
DG
Kurtzberg
J
Toxicity profile of PEG-L-asparaginase in native-L-asparaginase hypersensitive and non-hypersensitive patients with acute lymphoblastic leukemia (ALL).
Med Pediatr Oncol.
21
1993
556
560
27
Kotasek
D
The ARANESP 980291 Study Group
Berg
R
Poulsen
E
Colowick
A
Randomized, double-blind, placebo controlled, phase I/II dose finding study of ARANESP administered once every three weeks in solid tumor patients [abstract].
Blood.
96
2000
294a
28
Komatsu
N
Okamoto
T
Yoshida
T
et al
Pegylated recombinant human megakaryocyte growth and development factor increased platelet counts in patients with aplastic anemia and myelodysplastic syndrome [abstract].
Blood.
96
2000
296a
29
Bolwell
B
Vredenburg
J
Overmoyer
B
et al
Phase I study of pegylated recombinant human megakaryocyte growth and development factor in breast cancer patients after autologous peripheral blood progenitor cell transplantation.
Bone Marrow Transplant.
26
2000
141
145
30
Tsutsumi
Y
Onda
M
Nagata
S
Lee
B
Kreitman
RJ
Pastan
I
Site-specific chemical modification with polyethylene glycol of recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) improves antitumor activity and reduces animal toxicity and immunogenicity.
Proc Natl Acad Sci U S A.
97
2000
8548
8553
31
Safra
T
Groshen
S
Jeffers
S
et al
Treatment of patients with ovarian carcinoma with pegylated liposomal doxorubicin.
Cancer.
91
2001
90
100
32
Chaffee
S
Mary
A
Stiehm
ER
Girault
D
Fischer
A
Hershfield
MS
IgG antibody response to polyethylene glycol-modified adenosine deaminase in patients with adenosine deaminase deficiency.
J Clin Invest.
89
1992
1643
1651
33
Kitamura
K
Takahashi
T
Takashina
K
et al
Polyethylene glycol modification of the monoclonal antibody A7 enhances its tumor localization.
Biochem Biophys Res Commun.
171
1990
1387
1394
34
Zeuzem
S
Feinman
SV
Rasenack
J
et al
Peginterferon alpha-2 in patients with chronic hepatitis C.
N Engl J Med.
343
2000
1666
1672
35
Heathcote
EJ
Shiffman
ML
Cooksley
WGE
et al
Peginterferon alfa-2a in patients with chronic hepatitis C and cirrhosis.
N Engl J Med.
343
2000
1673
1680
36
Kantarjian
HM
Keating
MJ
Talpaz
M
et al
Chronic myelogenous leukemia in blast crisis: an analysis of 242 patients.
Am J Med.
83
1987
445
454
37
Kantarjian
HM
Dixon
D
Keating
MJ
et al
Characteristics of accelerated disease in chronic myelogenous leukemia.
Cancer.
61
1988
1441
1446
38
195-010 Study Report Data on file, Schering-Plough Research Institute; Kenilworth, NJ.
39
195-060. Data on file, Schering-Plough Research Institute; Kenilworth, NJ.
40
Obenauer-Kutner
LJ
Jacobs
SJ
Kolz
K
Tobias
LM
Bordens
RW
A highly sensitive electrochemiluminescence immunoassay for interferon alfa-2b in human serum.
J Immunol Methods.
206
1997
25
33
41
Gibaldi
M
Perrier
D
Pharmacokinetics.
2nd ed.
1982
409
417
Marcel Dekker
New York, NY
42
National Cancer Institute Common Toxicity Criteria, version 2.0 (1.30.1998). http://ctep.info.nih.gov Accessed January 2001.

Author notes

Hagop M. Kantarjian, Department of Leukemia, M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Box 428, Houston, TX 77030.

Sign in via your Institution