Immune-mediated effects appear to play a major role in controlling minimal residual disease (MRD). We, therefore, investigated the role of recombinant human interleukin-2 (rIL-2) given concomitantly with interferon-α (IFN-α) in malignant lymphoma (ML) patients with responding disease following autologous bone marrow or blood stem cell transplantation (ABSCT). Fifty-six patients were included in this investigation. Thirty-two patients had non-Hodgkin's lymphoma (NHL) and 24 patients had Hodgkin's disease (HD). Sixty-one patients (NHL 36, HD 25) served as historical controls. Patients from both groups had similar demographic characteristics, the same stage of disease at presentation, status of disease at transplantation, conditioning regimens, and type of transplant. rIL-2 and IFN-α were selfadministered in two cycles beginning 2.5 to 10.5 months (median, 4 months) posttransplant and separated by a 4-week interval. Each cycle consisted of IFN-α subcutaneously (SC) 3 × 106 U/d × 5 d/wk combined with rIL-2 SC 3 to 6 IU/m2/d × 5 d/wk for 4 weeks. The incidence of survival and disease-free survival (DFS) was significantly higher in the group under investigation than in the historical controls (P < .01). Of 56 patients with ML treated with IFN-α + rIL-2, 45 patients are DFS (80.4%) after a follow-up of 7 to 78 months (median, 34 months), whereas in the historical controls, 32 of 61 (52.5%) patients are disease free, in a follow-up of 4 to 84 months (median, 23 months) posttransplant (P < .01). Our preliminary results are encouraging and suggest that home administered immunotherapy with IFN-α and rIL-2 is relatively well tolerated and may intensify remission in ML patients with MRD following ABSCT.

HIGH-DOSE CHEMOTHERAPY in conjunction with autologous bone marrow or blood stem cell transplantation (ABSCT) is increasingly being used for the treatment of patients with non-Hodgkin's lymphoma (NHL) and Hodgkin's disease (HD).1-12 There are data indicating that this treatment regimen may improve long-term disease-free survival (DFS) in several categories of patients with malignant lymphoma (ML).3,6,7,12 However, the relapse rate still remains high.1-12 Residual tumor cells resistant to high-dose chemoradiotherapy may be responsible for the still relatively high rate of disease recurrence following this procedure. It is believed that the high relapse rate of disease following ABSCT is due to the lack of immune response of immunocompetent cells against residual tumor cells.13-17 

Recently it has been demonstrated that immune-mediated graft versus lymphoma similarly to graft versus leukemia effect may be induced in certain categories of NHL recipients of allogeneic marrow grafts.6,18-20 This suggests that one of the ways to reduce relapse rates post-ABSCT may be to intensify immune-mediated effector mechanisms against residual tumor cells.21,22 A number of cytokines have demonstrated antitumor activity in lymphoma patients. One of these is interferon-α (IFN-α), which when administered by itself or in conjunction with chemotherapy, has been shown to be effective in evoking antitumor response in 40% to 50% of the patients with low-grade NHL studied, including complete response (CR) in 5% to 10% of these patients.23-25 The results of IFN-α given together with chemotherapy have proven significantly better than chemotherapy alone.26 

Recombinant interleukin (rIL-2) is another novel activator of natural killer (NK) cells and T lymphocytes with cytotoxic capacities27,28 that has been shown to have an antitumor effect in several animal models25-30 including recipients of syngeneic bone marrow transplant29,30 and in pilot clinical trials on patients with NHL and HD.31-37 This cytokine has achieved encouraging preliminary results including some complete CRs over a follow-up period of up to 26 months.35 Combination therapy models of rIL-2 and IFN have been demonstrated to augment NK cytolytic activity both in vitro and in vivo in mice and humans.36-44 In view of the above and to reduce relapse rates following autologous transplant, rIL-2 has been administrated in pilot clinical trials to lymphoma patients post-ABSCT with encouraging results.45-49 We have conducted a phase IIb clinical trial on 56 ML patients with minimal residual disease (MRD) post-ABSCT, treated with a combination of rIL-2 and IFN-α subcutaneously (SC), in an outpatient setting and compared the results with 61 matched historical controls.

Patients.Fifty-six patients (36 men, 20 women), median age, 35 (10 to 53) years were enrolled in the study. Thirty-two patients had NHL and 24 patients had HD. Sixty-one ML patients (NHL 36, HD 25) served as historical controls (Table 1). Disease stage, sex, and age were statistically similar in the study group and the historical controls: 44 patients (79%) in the study group and 43 patients (70%) in the historical controls were stage III-IV at diagnosis, while 12 patients (21%) in the study group and 18 patients (30%) in the historical controls were stage I-II (Table 2). Thirty-three patients (58%) in the study group and 37 patients (60%) in the historical control group had B symptoms (Table 2). The frequency of NHL high-grade histology was 36% in the controls in comparison with 22% in the study group. Most of the patients in the study group and historical controls were transplanted in an advanced stage of disease, as 63% of the study group patients and 69% of the historical controls were transplanted after first relapse or more (Table 2). Conditioning regimen (TECAM) included thiotepa (40 mg/m2 × 4 days), etoposide (200 mg/m2 × 4 days), cytosar (200 mg/m2 × 4 days), cyclophosphamide (60 mg/kg × 1 day), and melphalan (60 mg/m2 × 2 days). There was no difference in the conditioning regimen between the immunotherapy-treated patients and the historical control group. Twenty-four hours after the last chemotherapy treatment, the patients received nonpurged cryopreserved autologous marrow or peripheral blood stem cells collected with a Cs 3000plus (Baxter Healthcare Corp, Deerfield, IL) following 5 days of treatment with 10 μg/kg granulocyte colony-stimulating factor (neupogen).

Table 1.

CMI Following ABSCT in ML Patients Versus Historical Controls: Demographic Characteristics

CMIMaleFemalePAge (yr)PNo. of Patients
HD 18 NS 28 (10-45) NS 24 (48%) 
 − 16  30 (10-37)  25 (52%) 
NHL 18 14 NS 41 (23-53) .02 32 (47%) 
 − 25 11  36 (5-62)  36 (53%) 
CMIMaleFemalePAge (yr)PNo. of Patients
HD 18 NS 28 (10-45) NS 24 (48%) 
 − 16  30 (10-37)  25 (52%) 
NHL 18 14 NS 41 (23-53) .02 32 (47%) 
 − 25 11  36 (5-62)  36 (53%) 

Abbreviation: NS, not significant.

Table 2.

CMI Following ABSCT in ML Patients Versus Historical Controls: Disease Characteristics

CMINodular SclerosisHistologyLymphocyte DepletedPSystemic SymptomsP
Mixed CellularityCMIAB
HD 19 NS HD+ 19 NS 
 − 18  − 16  
  Low Grade 
  Intermediate 
  Grades 
  High Grade 
   
NHL 16 NS NHL+ 18 14 NS 
  20 13  − 15 21 
CMINodular SclerosisHistologyLymphocyte DepletedPSystemic SymptomsP
Mixed CellularityCMIAB
HD 19 NS HD+ 19 NS 
 − 18  − 16  
  Low Grade 
  Intermediate 
  Grades 
  High Grade 
   
NHL 16 NS NHL+ 18 14 NS 
  20 13  − 15 21 
 CMI Disease Stage P CMI 1st CR 2nd CR Disease Status at BMT 
   II III IV     >2nd CR PR PD P 
HD 12 NS HD+ NS 
 − 10  − 
NHL 17 NS NHL+ 13 NS 
 − 22  − 10 11 
 CMI Disease Stage P CMI 1st CR 2nd CR Disease Status at BMT 
   II III IV     >2nd CR PR PD P 
HD 12 NS HD+ NS 
 − 10  − 
NHL 17 NS NHL+ 13 NS 
 − 22  − 10 11 

Abbreviations: 1st CR, first complete remission; 2nd CR, second complete remission; PR, partial response; PD, progressive disease.

The median time interval between ABCST and cell-mediated immunotherapy (CMI) was fixed at 4 (2.5 to 10) months, in view of the need for adequate hematopoietic reconstitution before the initiation of cytokine therapy. Only patients without disease progression for 4 months posttransplant were considered eligible for the CMI protocol. Hence, patients in the historical control group with disease progression during the 4 months post-ABSCT were also excluded from further analysis. None of the patients were intentionally excluded from the CMI protocol. Refusal to enter an experimental protocol, technical difficulties in obtaining cytokines, mainly rIL-2, due to inadequate supply, were among the reasons for noninclusion in the study.

Study parameters.Before the initiation of immunotherapy, all patients had their complete medical history taken and underwent physical examination, chest x-ray, electrocardiogram, prothrombin time (PT), partial thromboplastin time (PTT), complete blood count, and biochemistry tests. Patients were examined once weekly. Hematologic tests (complete blood counts including the differential, as well as biochemistry tests) were performed once weekly. Patients were monitored for adverse effects and drug toxicity. Survival, DFS, and relapse rates were assessed.

Treatment schedule.After stabilization of peripheral blood counts (white blood cell >2.5 × 109/L and platelets >75 × 109/L), patients were treated with daily SC injections of Chiron rIL-2 (Proleukin) (3 to 6 × 106 international units (IU)/m2/d), combined with IFN-α (Roferon A; Hoffman La Roche, Switzerland) 3 × 106 U/d, for 5 consecutive days each week for 4 weeks. The median time from ABSCT until the onset of immunotherapy was 4 (2.5 to 10.5) months. Patients received two identical cycles of 4 weeks of immunotherapy consisting of a combination of rIL-2 and IFN-α followed by a 4-week rest period (a total of 40 injections given over a 3-month period). Toxicities were evaluated according to the World Health Organization (WHO) toxicity grading scale.

Supportive care.Patients received paracetamol (500 mg/6 hours) as prophylaxis and therapy for controlling fever, myalgia, and muscle stiffness. Promethazine (25 mg/8 hours) was prescribed to control chills and allergic reaction and ondansetron hydrochloric dihydrate (Zofran) (8 mg × 3 days) or Navoben (5 mg × 1 day) to control nausea and vomiting. For other adverse events, treatment was given according to clinical symptoms.

Table 3.

Effect of CMI on Actuarial Survival and DFS in ML Patients who Received Immunotherapy Versus Historical Controls

CMISurvival: Follow-Up Range (mo)No. of PatientsPDFS: No. of PatientsP
HD (8-78) 24/24 .02 21/24 .042 
 − (8-84) 17/25  17/25  
NHL (7-78) 29/32 .01 24/32 .01 
 − (5-84) 18/36  15/36  
ML (7-78) 53/56 .01 45/56 .01 
 − (5/84) 35/61  32/61 
CMISurvival: Follow-Up Range (mo)No. of PatientsPDFS: No. of PatientsP
HD (8-78) 24/24 .02 21/24 .042 
 − (8-84) 17/25  17/25  
NHL (7-78) 29/32 .01 24/32 .01 
 − (5-84) 18/36  15/36  
ML (7-78) 53/56 .01 45/56 .01 
 − (5/84) 35/61  32/61 

rIL-2 (Proleukin).Human rIL2 was kindly provided by Chiron BV, Amsterdam, The Netherlands. Specific activity was approximately 1.5 × 107 U/mg protein. αIL-2 was supplied as a lyophilized powder in 1-mg vials (18 × 106 IU) and reconstituted in sterile water. IU are used throughout the report.

IFN-α (Roferon-A).Recombinant human IFN-α (Roferon A) was provided by Hoffmann La Roche. Specific activity was 2 × 108 U/mg protein. It was supplied as a lyophilized powder and reconstituted in sterile water.

Statistical evaluation.The clinical and demographic characteristics of ML, HD, and NHL patient groups who received immunotherapy and patients belonging to the historical control group who did not receive immunotherapy were analyzed with “Fisher-Irwin exact test.”

A multivarianate Cox proportional hazards analysis model was used to analyze improved DFS in patients who received or did not receive immunotherapy and other risk factors.50 

The Kaplan-Meier (KM) method was used to calculate the probability of survival and DFS as a function of time in the ML, HD, and NHL groups of patients with and without immunotherapy.51 

The “Log Rank test” was used to compare between pairs of KM curves.52 

We compared the clinical outcome of patients receiving cytokine-mediated immunotherapy (CMI) post-ABSCT with that of patients who did not receive CMI post-ABSCT for the same malignancies in our institution. In particular, there were no significant differences among the groups with respect to patient sex, diagnosis, and stage of disease at BMT. There was significant difference among the groups in age, the NHL historical controls were significantly younger (P < .02) (Table 1). There was no difference in ablative regimen between the groups. In addition, risk factors associated with improved DFS post-ABSCT in patients who received immunotherapy, were analyzed with the multivariant Cox proportional hazards analysis model. The independent variables that were included in the analysis were: age, sex, systemic symptoms, and stage of disease; disease status at BMT showed that of all the independent variables analyzed above, only age was found to be significant in the development of relapse, with a relative risk of 1.14 (P < .02) (ie, older patients tend to relapse more than younger patients) (Table 2). Other than this, we found no candidate variables among patient characteristics or clinical disease factors that differed between the study group and the control group.

Survival.The overall survival of ML patients who received immunotherapy was significantly higher than that of ML patients not receiving immunotherapy. The survival at 48 months was 90% (95% confidence interval 70% to 97%) for the immunotherapy patients, and 46% (95% confidence interval 30% to 60%) for the historical controls (P < .01) (Fig 1A) (Table 3).

Fig. 1.

The effect of IL2/IFN-α immunotherapy on actuarial survival (A), DFS (B), and relapse (C) in ML patients who received immunotherapy (n = 56) versus historical controls (n = 61, TRT + R) after autologous bone marrow transplantation (P < .01, P < .01, and P < .01, respectively).

Fig. 1.

The effect of IL2/IFN-α immunotherapy on actuarial survival (A), DFS (B), and relapse (C) in ML patients who received immunotherapy (n = 56) versus historical controls (n = 61, TRT + R) after autologous bone marrow transplantation (P < .01, P < .01, and P < .01, respectively).

Close modal

Similarly, the overall survival was significantly higher for the HD and NHL patients who received immunotherapy as compared with the historical controls. The survival rates at 48 months were 100% and 80% (95% confidence interval 43% to 95%) versus 57% (95% confidence interval 31% to 76%) and 42% (95% confidence interval 24% to 58%), respectively (P < .02) (Fig 2A, P < .01, and Fig 3A, P < .02) (Table 3).

Fig. 2.

The effect of rIL-2/IFN-α immunotherapy on actuarial survival (A), DFS (B), and relapse (C) in NHL patients who received immunotherapy (n = 32) versus controls (n = 36, TRT + R) after autologous bone marrow transplantation (P < .01, P < .01, and P < .01, respectively).

Fig. 2.

The effect of rIL-2/IFN-α immunotherapy on actuarial survival (A), DFS (B), and relapse (C) in NHL patients who received immunotherapy (n = 32) versus controls (n = 36, TRT + R) after autologous bone marrow transplantation (P < .01, P < .01, and P < .01, respectively).

Close modal
Fig. 3.

The effect of rIL-2/IFN-α immunotherapy on acturial survival (A), DFS (B), and relapse (C) in Hodgkin's patients who received immunotherapy (n = 24) versus historical controls (n = 25, TRT + R) after autologous bone marrow transplantation (P < .02, P < .042, and NS, respectively).

Fig. 3.

The effect of rIL-2/IFN-α immunotherapy on acturial survival (A), DFS (B), and relapse (C) in Hodgkin's patients who received immunotherapy (n = 24) versus historical controls (n = 25, TRT + R) after autologous bone marrow transplantation (P < .02, P < .042, and NS, respectively).

Close modal
Table 4.

Adverse Events in 56 Lymphoma Patients Treated With CMI With Roferon-A and rlL-2 Following ABMT

EventToxicity (WHO Grading)Total No. and (%) of Patients
Grade IGrade IIGrade IIIGrade IV
Nausea/vomiting 14 30   —  43 (77) 
Diarrhea 23  —  28 (50) 
Anorexia  —  19 26 (46) 
Weight loss  —  10  —   —  10 (18) 
Fever 37  —  48 (86) 
Chills  23  —  32 (57) 
Fatigue  —  25 20 46 (82) 
Loss of taste  —   —  6 (11) 
Stomatitis  —  14  —  17 (30) 
Dyspnea (bronchitis)  —   —  4 (7) 
Dry cough  —  18  —  19 (34) 
Neuromood 11  —  19 (34) 
Dry skin  —   —  8 (14) 
Contact dermatitis in the injection site 16  —  19 (34) 
Cardiotoxicity  —   —   —  1 (∼2) 
Myalgia  —  13  —  15 (26) 
Alopecia  —   —  3 (∼6) 
Thrombocytopenia  —  10 25 10 45 (80) 
Anemia  —  10 18  —  28 (50) 
Elevation of liver enzymes (ALKP, GGTP)  —  10 18  —  28 (50) 
EventToxicity (WHO Grading)Total No. and (%) of Patients
Grade IGrade IIGrade IIIGrade IV
Nausea/vomiting 14 30   —  43 (77) 
Diarrhea 23  —  28 (50) 
Anorexia  —  19 26 (46) 
Weight loss  —  10  —   —  10 (18) 
Fever 37  —  48 (86) 
Chills  23  —  32 (57) 
Fatigue  —  25 20 46 (82) 
Loss of taste  —   —  6 (11) 
Stomatitis  —  14  —  17 (30) 
Dyspnea (bronchitis)  —   —  4 (7) 
Dry cough  —  18  —  19 (34) 
Neuromood 11  —  19 (34) 
Dry skin  —   —  8 (14) 
Contact dermatitis in the injection site 16  —  19 (34) 
Cardiotoxicity  —   —   —  1 (∼2) 
Myalgia  —  13  —  15 (26) 
Alopecia  —   —  3 (∼6) 
Thrombocytopenia  —  10 25 10 45 (80) 
Anemia  —  10 18  —  28 (50) 
Elevation of liver enzymes (ALKP, GGTP)  —  10 18  —  28 (50) 

DFS.The overall DFS of ML patients who received immunotherapy was significantly higher than that of comparable ML patients in the historical control group who did not receive immunotherapy. The actuarial DFS at 48 months was 70% (95% confidence interval 50% to 84%) and 48% (95% confidence interval 32% to 61%), respectively (P < .01) (Fig 1B) (Table 3).

Similarly, the actuarial DFS was significantly higher for the NHL and HD patients after immunotherapy as compared with the historical controls. The actuarial DFS with NHL receiving immunotherapy at 48 months was 64% (95% confidence interval 36% to 80%) and 41% (95% confidence interval 25% to 48%) for patients who did not receive immunotherapy (P < .01) (Fig 2B). The actuarial DFS of patients with HD receiving immunotherapy at 48 months was 88% (95% confidence interval 50% to 96%) and 60% (95% confidence interval 34% to 78%) for patients who did not receive immunotherapy (P < .042) (Fig 3B) (Table 3).

The relapse rate was significantly lower for ML patients who received immunotherapy in comparison to a similar cohort of patients belonging to the historical controls. Of the 56 patients who received immunotherapy, 11 (20%) relapsed (8 NHL and 3 HD patients), while of the 61 patients who did not receive immunotherapy 29 (46%) relapsed (21 NHL and 8 HD patients) (P < .01) (Fig 1C, P < .01, Fig 2C, P < .01, Fig 3C, not significant [NS] ).

Toxicity.All patients were evaluated for treatment-related adverse events. Most of the toxic effects improved gradually with therapy and were manageable with standard antipyretic and analgesic agents. Most of the patients continued their regular activities while receiving treatment. WHO grade II-III fever, chills, and fatigue were very common and occurred in 86% of the patients. The intensity of fever, chills, and fatigue tended to decrease throughout the treatment course (fewer patients experienced these side effects on the second cycle of therapy than on the first). Anorexia (46.5%), nausea with or without vomiting (77%), and diarrhea were common adverse events. A mild erythematous, pruritic maculopapular rash was observed in 34% of the patients. Three patients had grade II-III hair loss. Mild neurotoxicity, consisting of depression, insomnia, and nervousness was observed in 34% of the patients. Mild to moderate anemia (8 to 11 g%) was observed in 50% of the patients, while mild to severe thrombocytopenia without bleeding tendency occurred in 80% of the patients. Only nine patients required red blood cell or platelet transfusions. Grade II-III liver enzyme elevation was observed in half of the patients. The SC administration of IFN-α and rIL-2 resulted in transient inflammation and local induration of the injection sites, which persisted for up to 2 weeks after treatment. Allergic manifestations, including contact dermatitis53 and bronchial asthma (one patient), were observed. One patient developed severe cardiotoxicity with cardiogenic shock responding to intravenous fluid and dopamine support.54,55 All side effects improved gradually and resolved after termination of the treatment. In only two cases did treatment have to be discontinued or interrupted because of toxicity (Table 4).

This report describes results of a nonrandomized out-patient treatment program based on administration of IFN-α and rIL-2 for the treatment of MRD in patients with lymphoma post-ABSCT. Combined IFN-α and rIL-2 immunotherapy resulted in a significant enhancement of survival and DFS as a result of reduced relapse rates in both NHL and HD patients post-ABSCT, in comparison to historical controls. Of 56 patients with HD and NHL treated with CMI, 45 patients are in CR (80.4%), after a follow-up of 7 to 78 months (median, 34 months). In comparison, of the 61 historical control patients, 32 are in CR (52.5%), after a follow-up of 4 to 84 (median, 23) months.

Prevention of relapse postautologous transplant may be achieved either by intensifying pretransplant chemoradiotherapy which, however, has been shown to augment organ toxicity and predisposition to early transplant-related death, or by intensifying the patient's immunocompetent effector cells against residual tumor cells by cytokine administration.36 There is evidence that tumor cell lines that have become resistant to chemotherapeutic agents are still susceptible to lysis by rIL-2 activated killer cells.56 Moreover, the synergism of IFN-α and rIL-2 shown to be effective in our murine model of lymphoid leukemia/lymphoma (unpublished data), as well as in other tumors in experimental animals and man,43,46 may increase the antitumor effect by various mechanisms.

While IFN-α has a direct antiproliferative cytoreductive effect, it may also help amplify the antitumor effects of the host's immune cells by augmentation of the expression of class I and class II cell-surface molecules.57 rIL-2 manifests its antitumor effects through stimulation of T-cell–dependent and mostly NK cell-dependent immune reactivity, as has been documented in vitro in man and in vivo in murine leukemia.27,28,39-42,47,48 Both IFN-α and rIL-2 have been previously administered to patients with malignant lymphoma with encouraging results.23-26,31-35 rIL-2 was previously shown by us to be effective in the setting of MRD30 particularly following syngeneic BMT,23 in analogy with ABSCT in man.

Abnormal T-cell parameters have been observed to linger following autologous transplant, which may account for the immunodeficiency observed up to a year or more posttransplant. Quantifiably, abnormal T-cell parameters following ABMT include a depressed absolute number of CD4+ T cells, a decreased response to mitogens, antigens, or allogeneic cells in lymphoproliferative assays, decrease in vitro T-cell colony formation, and a profound impairment of IL-2 production.58-62 Peripheral blood NK activity, although rapidly restored to pretransplant values, remains lower than normal control values.62 

Because endogenous IL-2 production is decreased for about 1 year following ABSCT, administration of exogenous rIL-2 to these patients might be expected to enhance immune reconstitution and reduce the risk of relapse. Accordingly, rIL-2 has been administered recently postautologous transplant to leukemic patients in an attempt to provide an antileukemic effect, demonstrating some response in acute myeloid leukemia (AML) patients.63 Interestingly, no such positive data have been reported for patients with acute lymphoblastic leukemia (ALL).64 Fefer et al44,45 administered rIL-2 + lymphokine activated killer (LAK) cell therapy soon after ABMT to 16 high-risk ML patients (NHL, 12; HD, 4). Of these patients, five have relapsed, while 11 remain in complete remission 6 to 21 (median, 10) months post-ABMT.44,45 Others have demonstrated similar results with continuous high-dose rIL-2 administrated to ML patients resulting in in vitro reconstitution of NK and T-cell functions.65-67 

LAK precursor activity has been found to reconstitute rapidly after ABSCT in lymphoma patients. This may provide the biological rationale for rIL-2 consolidative immunotherapy post-ABSCT for ML patients.68 Our immunological evaluation of patients with hematologic malignancies treated with IFN-α + rIL-2 SC following ABSCT, demonstrated that cytokine therapy could stimulate the host immune system by amplifying cytotoxic activity of K562 and Daudi cell lines, as well as T-cell–dependent mitogenic response.36 Similarly, Higuchi et al69 demonstrated that in lymphoma patients who received high-dose rIL-2 post-ABMT, a higher percentage of the NK cell markers CD16 and CD56 was found in blood cells. Augmented lysis of K562 and Daudi cell lines was also found, which may reflect the induction of NK and circulating LAK effector activities.

Bosly et al70 demonstrated an increase of CD8 T cells and CD56+ NK cells and the correction of functional T-cell defects in lymphoma patients who received rIL-2 post-ABMT. IFN-α has also been shown to intensify NK cell-mediated cytotoxicity27,35 and to have a therapeutic effect in lymphoma patients, either alone or in combination with chemotherapy.23-26,71 Because rIL-2 and IFN-α have demonstrated their antitumor effect when administered alone,25,26,31-35 we took advantage of their synergistic effect to use them in combination. The rationale behind this combination was that IFN-α, in addition to providing a direct antiproliferative effect, may also be able to enhance the expression of specific antigens on target cells. These could then be more efficiently recognized and lysed by T cells activated and expanded by rIL-2. Vivancos et al72 have also recently administered IFN-α and rIL-2 in combination at relatively high doses to 16 patients (mostly leukemic) post-ABMT with tolerable toxicity.

Combination cytokine therapy with IFN-α and rIL-2 has been explored in solid tumor malignancies.73-77 The rationale for combination therapy with IFN-α and IL-2 derived from both in vitro and in vivo observation.78 Out-patient combination cytokine therapy has been successfully used in patients with solid tumor malignancy. Based on this clinical data, we administered IFN-α and rIL-2 combined, SC, on an out-patient basis to 56 ML patients 2.5 to 10 (median, 4) months post-ABSCT with very encouraging results. Combined ambulatory CMI with IFN-α and r-IL2 was found to be feasible and safe, with tolerable side effects.

Side effects were those expected from IFN-α and rIL-2 when given as single agents, or recently in combination for patients with other malignancies.73-77 They included fever, chills, fatigue, flu-like symptoms, anorexia, nausea, vomiting, and diarrhea and were transient and reversible. Most of the patients were able to continue their regular activities with the help of symptomatic treatment. The overall side effects of the combined treatment modality were less toxic than those published on intravenous administration of high-dose rIL-2 only.65 

In summary, our study on lymphoma patients demonstrated that combined IFN-α and rIL-2 immunotherapy at the stage of MRD post-ABSCT intensifies remission and reduces relapse rates in comparison to historical controls. Cytokine-mediated immunotherapy activates the cytologic activity of the host immune system, thus mediating possible antitumor response, which may result in increased survival and DFS in comparison to historical controls. Prospective randomized studies are being planned to confirm our encouraging results.

The authors thank Prof A. Polliack, Prof E. Rachmilewitz, and all of the physicians for referring patients who were included in this study.

Address reprint requests to A. Nagler, MD, MSc, Bone Marrow Transplantation Department, Hadassah University Hospital, POB 12000, Jerusalem 91120, Israel.

1
McMillan
AK
Goldstone
AH
Autologous bone marrow transplantation non-Hodgkin's lymphoma.
Eur J Haematol
46
1991
129
2
Gale
RP
Armitage
JO
Dicke
KA
Autotransplants: Now and in the future.
Bone Marrow Transplant
7
1991
153
3
Freedman
AS
Takvorian
T
Neuberg
D
Mauch
P
Rabinowe
SN
Anderson
KC
Soiffer
RJ
Spector
N
Grossbard
M
Robertson
MJ
Blake
K
Coral
F
Canellos
GP
Ritz
J
Nadler
LM
Autologous bone marrow transplantation in poor-prognosis intermediate-grade and high-grade B-cell non-Hodgkin's lymphoma in first remission: A pilot study.
J Clin Oncol
11
1993
931
4
Phillips
GL
Fay
JW
Herzig
RH
Lazarus
HM
Wolff
SN
Lin
HS
Shina
DC
Glasgow
GP
Griffith
RC
Lamb
CW
Herzig
GP
The treatment of progressive non-Hodgkin's lymphoma with intensive chemoradiotherapy and autologous marrow transplantation.
Blood
75
1990
831
5
Loiseau
HA
Hartmann
O
Valteau
D
McDowell
H
Brugieres
L
Vassal
G
Kalifa
C
Patte
C
Lemerle
J
High-dose chemotherapy containing busulfan followed by bone marrow transplantation in 24 children with refractory or relapsed non-Hodgkin's lymphoma.
Bone Marrow Transplant
8
1991
465
6
Chopra
R
Goldstone
AH
Pearce
R
Philip
T
Petersen
F
Appelbaum
F
De Vol
E
Ernst
P
Autologous versus allogeneic bone marrow transplantation for non-Hodgkin's lymphoma: A case-controlled analysis of the European Bone Marrow Transplant Group Registry Data.
J Clin Oncol
10
1992
1690
7
Shipp
MA
Prognostic factors in aggressive non-Hodgkin's lymphoma: Who has “high-risk” disease?
Blood
83
1994
1165
8
Freedman
A
Nadler
LM
Bone marrow transplantation in low-grade non-Hodgkin's lymphoma.
Marrow Transplant Rev
2
1992
39
9
Yahalom
J
Gulati
S
Shank
B
Clarkson
B
Fuks
Z
Total lymphoid irradiation, high-dose chemotherapy and autologous bone marrow transplantation for chemotherapy-resistant Hodgkin's disease.
Int J Radiat Oncol Biol Phys
17
1989
915
10
Reece
DE
Connors
JM
Spinelli
JJ
Barnett
MJ
Fairey
RN
Klingemann
HG
Nantel
SH
O'Reilly
S
Shepherd
JD
Sutherland
HJ
Voss
N
Chan
KW
Phillips
GL
Intensive therapy with cyclophosphamide, carmustine, etoposide + cisplatin, and autologous bone marrow transplantation for Hodgkin's disease in first relapse after combination chemotherapy.
Blood
83
1994
1193
11
Chopra
R
McMillan
AK
Linch
DC
Yuklea
S
Taghipour
G
Pearce
R
Patterson
KG
Goldstone
AH
The place of high-dose BEAM therapy and autologous bone marrow transplantation in poor-risk Hodgkin's disease. A single-center eight-year study of 155 patients.
Blood
81
1993
1137
12
Bierman
PJ
Vose
JM
Armitage
JO
Autologous transplantation for Hodgkin's disease: Coming of age?
Blood
83
1994
1161
13
Slavin
S
Nagler
A
New developments in bone marrow transplantation.
Curr Opin Oncol
3
1991
254
14
Slavin
S
Ackerstein
A
Vourka-Karussis
U
Nagler
A
Or
R
Naparstek
E
Weiss
L
Control of relapse due to minimal residual disease (MRD) by cell-mediated cytokine-activated immunotherapy in conjunction with bone marrow transplantation.
Bailliere's Clin Haematol
4
1991
715
15
Mehta
J
Graft-versus-leukemia reactions in clinical bone marrow transplantation.
Leuk Lymphoma
10
1993
427
16
Slavin
S
Ackerstein
A
Naparstek
E
Or
R
Weiss
L
The graft-versus-leukemia (GVL) phenomenon: Is GVL separable from GVHD?
Bone Marrow Transplant
6
1990
155
17
Moscovitch
M
Slavin
S
Anti tumor effects of allogeneic bone marrow transplantation in (NZB × NZW) F1 hybrids with spontaneous lymphosarcoma.
J Immunol
132
1984
997
18
Phillips
GL
Herzig
RH
Lazarus
HM
Fay
JW
Griffith
R
Herzig
GP
High dose chemotherapy, fractionated total body irradiation and allogeneic marrow transplantation for malignant lymphoma.
J Clin Oncol
4
1986
480
19
Ernst P, Devol E: Allogeneic bone marrow transplantation in malignant lymphoma. The European Bone Marrow Transplant Group experience. Proceedings of the 4th International Conference Lymphoma, Lugano, Switzerland, June 9-12, 1990 (abstr P35)
20
Jones
RJ
Ambinder
RF
Piantadosi
S
Santos
GW
Evidence of a graft-versus-lymphoma effect associated with allogeneic bone marrow transplantation.
Blood
77
1991
649
21
Fujimiya
Y
Bakke
A
Chang
WC
Linker-Israeli
M
Udis
B
Horwitz
D
Pattengale
PK
Natural killer-cell immunodeficiency in patients with chronic myelogenous leukemia: I. Analysis of the defect using the monoclonal antibodies HNK-I (LEU-7) and B73.1.
Int J Cancer
37
1986
639
22
Dutcher
JP
Creekmore
S
Weiss
GR
Margolin
K
Markowitz
AB
Roper
M
Parkinson
D
Ciobanu
N
Fisher
RI
Boldt
DDH
Doroshow
JH
Rayner
AA
Hawkins
M
Atkins
M
A phase II study of interleukin-2 and lymphokine-activated killer cells in patients with metastatic malignant melanoma.
J Clin Oncol
7
1989
477
23
Gilewski
TA
Richards
JM
Biologic response modifiers in non-Hodgkin's lymphomas.
Semin Oncol
17
1990
74
24
Quesada
JR
Talpaz
M
Rios
A Kurzrock R
Gutterman
JU
Clinical toxicity of interferons in cancer patients: A review.
J Clin Oncol
4
1986
234
25
McLaughlin P: The role of interferon in the therapy of low grade lymphoma. Leuk Lymphoma 10:17, 1993 (suppl)
26
Smalley
RV
Andersen
JW
Hawkins
MJ
Bhide
V
O'Connell
MJ
Oken
MM
Borden
EC
Interferon alfa combined with cytotoxic chemotherapy for patients with non-Hodgkin's lymphoma.
N Engl J Med
327
1992
1336
27
Nagler
A
Lanier
LL
Cwirla
S
Philips
JH
Comparative studies of human FcRIII-positive and negative NK cells.
J Immunol
143
1989
3183
28
Nagler
A
Lanier
L
Phillips
JH
Constitutive expression of high affinity interleukin-2 receptors on human CD16-natural killer cells in vivo.
J Exp Med
171
1990
1527
29
Ackerstein
A
Kedar
E
Slavin
S
Use of recombinant interleukin-2 in conjunction with syngeneic bone marrow transplantation in mice as a model for control of minimal residual disease in malignant hematological disorders.
Blood
78
1991
1212
30
Slavin
S
Ackerstein
A
Weiss
L
Adoptive immunotherapy in conjunction with bone marrow transplantation amplification of natural host defence mechanisms against cancer by recombinant IL2.
Natl Immun Cell Growth Regul
7
1988
180
31
Dutcher
JP
Wiernik
PH
The role of recombinant interleukin-2 therapy for hematologic malignancies.
Semin Oncol
20
1993
33
32
Duggan
DB
Santarell
MT
Zamkoff
K
Lichtman
S
Ellerton
J
Cooper
R
Poiesz
B
Anderson
JR
Bloomfield
CD
Peterson
BA
A phase II study of recombinant interleukin-2 with or without recombinant interferon-β in non-Hodgkin's lymphoma. A study of cancer and leukemia group B.
Br J Immunother
12
1992
115
33
Bernstein
ZP
Vaickus
L
Friedman
N
Goldrosen
MH
Watanabe
H
Rahmam
R
Arbuck
SG
Sweeney
J
Vesper
D
Henderson
ED
Interleukin-2 lymphokine-activated killer cell therapy of non-Hodgkin's lymphoma and Hodgkin's disease.
J Immunother
10
1991
141
34
Lim
SH
Worman
CP
Callaghan
T
Jewell
A
Smith
MP
Goldstone
AH
Continuous intravenous infusion of high dose recombinant interleukin-2 for advanced lymphomas: A phase II study.
Leuk Res
15
1991
435
35
Weber
JS
Yang
JC
Topalian
SL
Schwartzentruben
DJ
White
DE
Rosenberg
SA
The use of Interleukin-2 and lymphokine-activated killer cells for the treatment of patients with non-Hodgkin's lymphoma.
J Clin Oncol
10
1992
33
36
Gisselbrecht
C
Maraninchi
D
Pico
JL
Milpied
N
Coiffier
B
Divine
M
Tiberghien
P
Bosly
A
Tilly
H
Boulat
O
Brandely
M
Interleukin-2 treatment in lymphoma: A phase II multicenter study.
Blood
83
1994
2081
37
Morecki
S
Nagler
A
Puyesky
Y
Nabet
C
Condiotti
R
Pick
M
Gan
S
Slavin
S
Effect of various cytokine combinations on induction of non-MHC-restricyted cytotoxicity.
Lymphokine Cytokine Res
12
1993
159
38
Morecki
S
Revel-Vilk
S
Nabet
C
Pick
M
Ackerstein
A
Nagler
A
Naparstek
E
Ben
Shahar M
Slavin
S
Immunological evaluation of patients with hematological malignancies receiving ambulatory cytokine-mediated immunotherapy with recombinant human interferon-α2a and interleukin-2.
Cancer Immunol Immunother
35
1992
401
39
Brunda
MJ
Tarnowski
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
Sulich
V
In vivo antitumor 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
Iigio
M
Sakurai
M
Tamura
T
Saijo
N
In vivo antitumor 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 reactivity.
J Immunol
126
1981
2321
43
Nagler
A
Ackerstein
A
Barak
V
Slavin
S
Treatment of chronic myelogenous leukemia with recombinant human interleukin-2 and interferon-α2a.
J Hematother
3
1994
75
44
Fefer A, Benyunes M, Higuchi C, York A, Massumoto C, Lindgren C, Buckner CD, Thompson JA: Interleukin-2 +/- lymphocytes as consolidative immunotherapy after autologous bone marrow transplantation for hematologic malignancies. Acta Hematol 89:2, 1993 (suppl 1)
45
Fefer
A
Benyunes
M
Massumoto
C
Higuchi
C
York
A
Buckner
CD
Thompson
JA
Interleukin-2 therapy after autologous bone marrow transplantation for hematologic malignancies.
Semin Oncol
20
1993
41
46
Slavin
S
Ackerstein
A
Weiss
L
Nagler
A
Or
R
Naparstek
E
Immunotherapy of minimal residual disease by immunocompetent lymphocytes and their activation by cytokines.
Cancer Invest
10
1992
221
47
Weiss
L
Reich
S
Slavin
S
Use of recombinant human interleukin-2 in conjunction with bone marrow transplantation as a model for control of minimal residual disease in malignant hematological disorders: I. Treatment of murine leukemia in conjunction with allogeneic bone marrow transplantation and IL-2-activated cell-mediated immunotherapy.
Cancer Invest
10
1992
19
48
Cohen
P
Vourka-Karussis
U
Weiss
L
Slavin
S
Spontaneous and IL-2-induced anti-leukemic and anti-host effects against tumor- and host-specific alloantigens.
J Immunol
151
1993
4803
49
Vourka-Karussis
U
Karussis
D
Ackerstein
A
Slavin
S
Enhancement of GVL effect with rhIL-2 following BMT in a murine model for acute myeloid leukemia in SJL/J mice.
Exp Hematol
23
1995
196
50
Cox
DR
Regression models and life tables.
JR Stat Soc B
34
1972
187
51
Kadar
E
Meier
P
Non-parametric estimation from incomplete observations.
J Am Stat Assoc
53
1958
457
52
Mantel
N
Evaluation of survival data and two new rank order statistics arising in its consideration.
Cancer Chemother Rep
50
1966
163
53
Klapholz
L
Ackerstein
A
Goldenhersh
MA
Vardy
D
Nagler
A
Local cutaneous reaction induced by subcutaneous interleukin-2 and interferon alpha-2a immunotherapy following ABMT.
Bone Marrow Transplant
11
1993
443
54
Schechter
D
Nagler
A
Ackerstein
A
Nassar
H
Admon
D
Naparstek
E
Rein
AJ
Recombinant interleukin-2 and interferon alpha immunotherapy following autologous bone marrow transplantation. A case report.
Cardiology
80
1992
168
55
Schechter D, Nagler A: Recombinant interleukin-2 and recombinant alpha immunotherapy cardiovascular toxicity. Am Heart J 123:1736, 1992 (editorial)
56
Slavin
S
Naparstek
E
Nagler
A
Ackerstein
A
Kapelushnik
J
Or
R
Allogeneic cell therapy for relapsed leukemia after bone marrow transplantation with donor peripheral blood lymphocytes.
Exp Hematol
23
1995
1553
57
Basham
TY
Merigan
TC
Recombinant interferon-gamma increases HLA-DR synthesis and expression.
J Immunol
130
1983
1492
58
Lum
LG
The kinetics of immune reconstitution after human marrow transplantation.
Blood
69
1987
369
59
Azogui
O
Gluckman
E
Fradelizi
D
Inhibition of IL-2 production after human allogeneic bone marrow transplantation.
J Immunol
131
1983
1205
60
Welte
K
Ciobanu
N
Moore
MA
Gulati
S
O'Reilly
RJ
Mertelsmann
R
Defective interleukin-2 production in patients after bone marrow transplantation and in vitro restoration of defective T lymphocyte proliferation by highly purified interleukin-2.
Blood
64
1984
380
61
Cayeux
S
Meuer
S
Pezzutto
A
Korbling
M
Haas
R
Schulz
R
Dorken
B
T-cell ontogeny after autologous bone marrow transplantation: Failure to synthesize interleukin-2 (IL-2) and lack of CD2- and CD3-mediated proliferation by both CD4- and CD8+ cells even in the presence of exogeneous IL-2.
Blood
74
1989
2270
62
Bosly
A
Brice
P
Humblet
Y
Doyen
C
Faille
A
Chatelain
B
Franks
C
Gisselbrecht
C
Symann
M
Interleukin-2 after autologous bone marrow transplantation as consolidative immunotherapy against minimal residual disease.
Nouv Rev Fr Hematol
32
1990
13
63
Klingemann
H-G
Phillips
GL
Is there a place for immunotherapy with interleukin-2 to prevent relapse after autologous stem cell transplantation for acute leukemia?
Leuk Lymphoma
16
1995
397
64
Weisdorf
DJ
Anderson
PM
Blazar
BR
Uckun
FM
Kersey
JH
Ramsay
NK
Interleukin-2 immediately after autologous bone marrow transplantation for acute lymphoblastic leukemia. A phase I study.
Transplantation
55
1993
61
65
Negrier
S
Ranchere
JY
Philip
I
Merrouche
Y
Biron
P
Blaise
D
Attal
M
Rebattu
P
Clavel
M
Pourreau
C
Palmer
P
Favrot
M
Jasmin
C
Maraninchi
D
Philip
T
Intravenous interleukin-2 just after high dose BCNU and autologous bone marrow transplantation. Report of a multicentric French pilot study.
Bone Marrow Transplant
8
1991
259
66
Blaise
D
Olive
D
Stoppa
AM
Viens
P
Pourreau
C
Lopez
M
Attal
M
Jasmin
D
Mognes
G
Mawas
C
Mannoni
P
Palmer
P
Franks
C
Philip
T
Maraninch
D
Hematologic and immunologic effects of the systemic administration of recombinant interleukin-2 after autologous bone marrow transplantation.
Blood
76
1990
1092
67
Blaise
D
Viens
P
Olive
D
Stoppa
AM
Gabert
J
Pourreau
CN
Attal
M
Gaspard
MH
Mannoni
P
Jasmin
C
Palmer
P
Franks
C
Michel
G
Mawas
C
Baume
D
Philip
T
Maraninchi
D
Recombinant interleukin-2 (rIL-2) after autologous bone marrow transplantation (BMT): A pilot study in 19 patients.
Eur Cytokine Netw
2
1991
121
68
Higuchi
CM
Thompson
JA
Cox
T
Lindgren
CG
Buckner
D
Fefer
A
Lymphokine-activated killer function following autologous bone marrow transplantation for refractory hematological malignancies.
Cancer Res
59
1989
5509
69
Higuchi
CM
Thompson
JA
Petersen
FB
Buckner
CD
Fefer
A
Toxicity and immunomodulatory effects of interleukin-2 after autologous bone marrow transplantation for hematologic malignancies.
Blood
77
1991
2561
70
Bosly
A
Guillaume
T
Brice
P
Humblet
Y
Staquet
P
Doyen
C
Chatelain
B
Franks
CR
Gisselbrecht
C
Symann
M
Effects of escalating doses of recombinant human interleukin-2 in correcting functional T-cell defects following autologous bone marrow transplantation for lymphomas and solid tumors.
Exp Hematol
20
1992
962
71
Klingemann
HG
Grigg
AP
Wilkie-Boyd
K
Barnett
MJ
Eaves
AC
Reece
DE
Shepherd
JD
Phillips
GL
Treatment with recombinant interferon (α-2b) early after bone marrow transplantation in patients at high risk for relapse.
Blood
78
1991
3306
72
Vivancos P, Granena A, Garcia J, Abad B, Valls A: Post-ABMT treatment with high doses of IL2 and IFN. Results of a phase II study. Proceedings of the 19th Annual Meeting of the EBMT and 9th Meeting of the Nurses Group. Garmisch-Partenkirchen, Germany, January 17, 1993, p 123 (abstr)
73
Atzpodien
J
Korfer
A
Franks
CR
Poliwoda
H
Kirchrer
H
Home therapy with recombinant interleukin-2 and interferon-α2b in advanced human malignancies.
Lancet
335
1990
1509
74
Hirsh
M
Lipton
A
Harvey
H
Givant
E
Hopper
K
Jones
G
Zeffren
J
Levitt
D
Phase I study of interleukin-2 and interferon alfa-29 as outpatient therapy for patients with advanced malignancy.
J Clin Oncol
8
1990
1657
75
Lee
KH
Talpaz
M
Rotherberg
JM
Murray
JL Papadopoulos N
Plager
C
Benjamin
R
Levitt
D
Gutterman
J
Concomitant administration of recombinant human interleukin-2 and recombinant interferon α-2A in cancer patients: A phase I study.
J Clin Oncol
17
1989
1726
76
Radhakrishna
Pillai M
Balaram
P
Padmanabhan
TK
Abraham
T
Krishnan
Nair M
Interleukin 2 and alpha interferon induced in vitro modulation of spontaneous cell mediated cytotoxicity in patients with cancer of the uterine cervix undergoing radiotherapy.
Acta Oncol
28
1989
39
77
Rosenberg
SA
Lotze
MT
Yang
JC
Linehan
WM
Seipp
C
Calabro
S
Karp
SE
Sherry
RM
Steinberg
S
White
DE
Combination therapy with interleukin-2 and alpha-interferon for the treatment of patients with advanced cancer.
J Clin Oncol
7
1989
1863
78
Rabinowitz
J
Petros
W
Stuart
A
Peters
W
Characterization of endogenous cytokine concentration after high dose chemotherapy with autologous bone marrow support.
Blood
81
1993
2452
Sign in via your Institution