ArticlePDF Available

Results of a multicenter randomized study to evaluate the safety and efficacy of combined immunotherapy with interleukin-2, interferon- 2b and histamine dihydrochloride versus dacarbazine in patients with stage IV melanoma

Authors:

Abstract and Figures

The safety and efficacy of immunotherapy with histamine dihydrochloride (HDC), interleukin-2 (IL-2) and interferon-alpha2b (IFN) compared with dacarbazine (DTIC) in adult patients with stage IV melanoma was evaluated. Two hundred and forty-one patients were randomized to either receive repeated 4-week cycles of IFN [3 MIU, s.c., once daily for 7 days], IL-2 (2.4 MIU/m(2), s.c., twice a day for 5 days) and HDC (1 mg, s.c., twice a day for 5 days) or DTIC 850 mg/m(2) i.v. every 3 weeks. The primary endpoint was overall survival. Median survival was longer for patients receiving HDC/IL-2/IFN (271 days) than for patients receiving DTIC (231 days), but this did not achieve statistical significance. Four patients receiving HDC/IL-2/IFN and nine receiving DTIC experienced at least one grade 4 adverse event. Striking differences in overall survival were observed between countries participating in the study. Treatment with HDC/IL-2/IFN was safely administered on an outpatient basis, but this immunotherapeutic regimen did not improve upon the response rate and overall survival seen with DTIC.
Content may be subject to copyright.
Annals of Oncology
doi:10.1093/annonc/mdm331
original article
Results of a multicenter randomized study to evaluate
the safety and efficacy of combined immunotherapy
with interleukin-2, interferon-a2b and histamine
dihydrochloride versus dacarbazine in patients
with stage IV melanoma
M. Middleton
1
, A. Hauschild
2
, D. Thomson
3
, R. Anderson
4
, S. Burdette-Radoux
5à
, K. Gehlsen
,
K. Hellstrand
7
& P. Naredi
8
*
1
Department of Medical Oncology, Christie Hospital, Manchester, UK;
2
Department of Dermatology, University of Kiel, Kiel, Germany;
3
Department of Medical Oncology,
Princess Alexandra Hospital, Woolloongabba, Queensland, Australia;
4
Departments of Oncology, Umea
˚University Hospital, Umea
˚, Sweden;
5
Department of Oncology,
McGill University, Montreal, Canada;
6
EpiCept Corporation, San Diego, CA, USA;
7
Dept of Virology, University of Go
¨teborg, Go
¨teborg;
8
Departments of Surgery, Umea
˚
University Hospital, Umea
˚, Sweden
Received 23 February 2007; revised 14 May 2007; accepted 20 May 2007
Background: The safety and efficacy of immunotherapy with histamine dihydrochloride (HDC), interleukin-2 (IL-2)
and interferon-a2b (IFN) compared with dacarbazine (DTIC) in adult patients with stage IV melanoma was evaluated.
Patients and methods: Two hundred and forty-one patients were randomized to either receive repeated 4-week
cycles of IFN [3 MIU, s.c., once daily for 7 days], IL-2 (2.4 MIU/m
2
, s.c., twice a day for 5 days) and HDC (1 mg, s.c.,
twice a day for 5 days) or DTIC 850 mg/m
2
i.v. every 3 weeks. The primary endpoint was overall survival.
Results: Median survival was longer for patients receiving HDC/IL-2/IFN (271 days) than for patients receiving DTIC
(231 days), but this did not achieve statistical significance. Four patients receiving HDC/IL-2/IFN and nine receiving
DTIC experienced at least one grade 4 adverse event. Striking differences in overall survival were observed between
countries participating in the study.
Conclusion: Treatment with HDC/IL-2/IFN was safely administered on an outpatient basis, but this
immunotherapeutic regimen did not improve upon the response rate and overall survival seen with DTIC.
Key words: clinical trials, dacarbazine, histamine dihydrochloride, immunotherapy, interferon alpha, interleukin-2,
melanoma, phase III
introduction
Stage IV melanoma is associated with a 5–11 month median
survival duration and, despite response rates ranging from
10–50%, available treatments have failed to improve upon this
and are associated with significant toxicity. The lack of
efficacious treatments for advanced melanoma requires that
new approaches be tested.
Currently, there are two agents approved for use in
stage IV melanoma patients: dacarbazine (DTIC) in Europe
and the USA and high-dose intravenous interleukin-2 (IL-2)
in the USA. DTIC provides a 10–17% tumor response rate,
but these responses are generally of short duration, and
there is no evidence suggesting that DTIC has a significant
impact on overall survival in this patient population
[1–3].
The impact of high-dose IL-2 on the overall survival of
patients with stage IV melanoma is not known. Pooled data
from eight studies report a median survival of 11.4 months.
IL-2 administered at high doses is associated with severe
systemic side-effects [4]. Single-agent interferon-alpha (IFNa)
therapy, as well as single-agent IL-2 treatment, yields
response rates of approximately 15% in metastatic melanoma
with infrequent long-term responders and occasional long-term
survivors [5]. The combination of IFNaand IL-2 has been
reported to increase response rates [6] but this has not been
confirmed in a randomized phase III trial [7].
Immunosuppression within and around tumors has been
implicated as a major factor in preventing better clinical
outcomes using cytokines such as IL-2 and IFNa[8, 9]. One
original
article
*Correspondence to: Peter Naredi, Department of Surgery, Umea
˚University Hospital,
S-90185 Umea
˚, Sweden. Tel: +46 90 7851153; Fax: +46 90 7851156;
E-mail: peter.naredi@surgery.umu.se
Now at Cancer Research UK Medical Oncology Unit, Churchill Hospital, Oxford, UK
à
Now at Vermont Cancer Center, University of Vermont, Burlington, VT, USA
§
Now at Research Corporation Technologies, Tucson, AZ, USA
ª2007 European Society for Medical Oncology
Annals of Oncology Advance Access published August 20, 2007
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
mechanism by which tumors may suppress T-cell and natural
killer (NK)-cell function is through the recruitment of
monocyte/macrophages (MO) to the tumor site. These cells
produce reactive oxygen species (ROS), which can cause
down-regulation of the CD3ftransduction antigen in NK and
T cells along with significant NK-cell and T-cell apoptosis.
Histamines inhibit ROS production in MO, and thereby
protect NK-cells and T-cells from functional inhibition and
apoptosis. The use of histamine dihydrochloride (HDC) as
a counter-suppressive agent in combination with IL-2 and/or
IFNahas been under investigation in several phase 2 and
3 clinical trials [10–16].
The basis for this trial was in vitro and in vivo studies
suggesting that the combination of HDC and IL-2 yields
a more efficient lymphocyte-mediated destruction of several
human malignant target cells [17–21] than either compound
used as a single agent; these in vitro findings are supported by
clinical studies in melanoma demonstrating that cytotoxic
lymphocytes are more efficiently activated by systemic
treatment with HDC/IL-2 in vivo than by monotherapy with
IL-2 [22]. A recent randomized trial in acute myeloid leukemia
(AML) demonstrated a statistically significant improvement
in long-term leukemia-free survival for patients treated
post-consolidation with HDC and IL-2 compared with the
no-treatment control patients [16].
Clinical experience from a randomized phase 3 study in
stage IV melanoma patients reported that the combination of
histamine plus IL-2 improved the overall survival of stage IV
melanoma patients with liver metastases versus IL-2 alone
(9.4 vs. 5.1 months) but there was no survival benefit for the
overall population [12]. A second confirmatory phase 3 study
failed to show any survival benefit for patients with liver
metastases (P. Naredi). In a phase II study in stage IV
melanoma patients combining HDC/IL-2/IFN, the median
survival exceeded 11 months and complete responses were seen,
including in patients with liver metastases [14]. The present
study was designed to test this outpatient combination
immune-based treatment regimen containing low-dose IL-2,
low-dose IFNaand histamine compared with DTIC alone in
stage IV melanoma patients.
patients and methods
patients and eligibility
Inclusion criteria were: age >18 years; histologically proven American
Joint Committee on Cancer (AJCC) stage IV melanoma; life expectancy
of 3 months; clinically adequate bone marrow, kidney, cardiac and liver
function (hemoglobin >10.0 g/dl, white blood cell count (WBC) >2.5 ·10
9
cells/L, absolute neutrophil count (ANC) >1.5 ·10
9
cells/L, platelet count
>100 ·10
9
/L, partial thromboplastin time (PTT or activated partial
thromboplastin time in Australia) within normal limits, creatinine
clearance >60 ml/min, serum bilirubin <1.5 times the upper limit of normal
(ULN), alanine aminotransferase (ALT) or aspartate aminotransferase
(AST) <3 times the ULN, fasting serum glucose <160 mg/dl; Karnofsky
status score of 70). All patients signed an informed consent.
Patients were excluded if any of the following conditions applied: prior
treatment with DTIC or IL-2 (except IL-2 used as an adjuvant >1 year
before enrollment in this study); abnormal cardiac function assessed by
resting ECG; any other concurrent systemic anti-neoplastic therapy;
ongoing active other malignancy (except in situ carcinoma of the cervix,
localized squamous or basal cell skin carcinomas); metastatic central
nervous system (CNS) malignancy at randomization (patients with stable,
treated brain metastases were eligible); documented history of asthma
treated within the past 5 years; medical, sociological or psychological
impediment to compliance with protocol; pregnancy or breast-feeding;
positive serum HIV test or history of autoimmune disease; active peptic
ulcer disease or a history of prior bleeding ulcer disease.
study design
Patients were stratified on the basis of liver metastases at baseline
(present or absent) and then randomized to either HDC/IL-2/IFN or to
DTIC. Site-specific randomization codes were produced electronically for
each stratified group, and site personnel called a central randomization
desk. Randomization was conducted in consecutive sequence within each
center. This study was not blinded because of the physiologic effects of
HDC (such as flushing). The study was approved by the Institutional Ethics
Review Boards.
Histamine dihydrochloride 1 mg (Ceplene, Maxim Pharmaceuticals,
San Diego, CA, USA) was administered twice daily on days 1–5 of each
week of the 4-week treatment cycle. HDC was administered by slow
subcutaneous injection over a period of not less than 10 min and not
more than 30 min. IL-2 (Proleukin, Chiron Corporation, Emeryville, CA,
USA) was administered subcutaneously at a dosage of 2.4 MIU/m
2
twice
daily for days 1–5 of weeks 1 and 2 of each treatment cycle. IFN
(interferon-a2b, Intron-A, Schering Plough, Kenilworth, NJ, USA) was
administered subcutaneously at a daily dose of 3 MIU for days 1–7 of
each week of the treatment cycle. Before the first cycle a priming dose of
IFN for 5–7 days was given. Intravenous DTIC 850 mg/m
2
was delivered
on day 1 of each 3-week cycle. DTIC was administered according to
standard practice in each institution and was obtained from commercial
stock.
The duration of treatment for patients receiving HDC/IL-2/IFN was
expected to be 12 months, except in the event of progressive disease or
adverse effect (AE) resulting in study discontinuation. Duration of
treatment for patients who received DTIC and achieved a response was
left to the discretion of the investigator.
study endpoints
The intent-to-treat (ITT) population comprised all randomized patients
and the intent-to-treat liver metastases (ITT-LM) subgroup was defined
as all randomized patients who had liver metastases at baseline. The
primary efficacy endpoint was overall survival in the ITT population
calculated as the number of days from randomization to death. Those
patients without a documented date of death at the time of data cut-off
were censored at the last date on which the patient was known to be alive.
Secondary efficacy endpoints were overall survival in the ITT-LM
population, progression-free survival, objective response rate and duration
of response. Progression-free survival was calculated as the number of
days from randomization to the first documentation of progressive disease
(PD) or death. Those patients without documented PD or death at the
time of data cut-off were censored at the date of the last assessment of
disease response. Objective tumor response rate was calculated as the
number of patients with a confirmed complete response (CR) or partial
response (PR) divided by the number of patients randomized. Assessment
for a response occurred every 12 weeks after randomization with
confirmation of the response 4 weeks later. The duration of the objective
response was calculated as the number of days from the first documentation
of CR or PR until the first documentation of PD or death. Only patients
with a confirmed CR or PR were included in the analysis.
statistics
The primary analysis compared the overall survival in the two treatment
groups using the stratified log-rank test, stratified by the presence or
original article Annals of Oncology
2|Middleton et al.
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
absence of liver metastases at baseline. The null hypothesis was that
distribution of overall survival be the same in the two treatment groups. All
Pvalues reported are two-sided. Kaplan-Meier curves were used to
summarize the survival distributions in the treatment groups, and the
median of the survival distributions with 95% confidence interval (CI) was
tabulated by treatment group.
A sample size of 96 patients in each group was required to provide 80%
power to detect a 50% increase in median survival time (7.0–10.5 months)
between the two treatment groups, with a type I error of 0.05. To allow
for an expected drop-out rate of 20%, enrollment of 240 patients
(120 per group) was planned to maintain the desired power.
Kaplan-Meier estimates of the survival rate in each treatment group
were calculated at 6, 12, 18, 24, 30 and 36 months, along with 95% CI.
The treatment groups were compared at each time point by using an
approximate ztest. The secondary endpoints of progression-free survival
and duration of objective response were analyzed by using the same
methods as for the primary analysis of overall survival. For duration of
response, only those patients with an objective tumor response
(confirmed CR or PR) were included in the analysis. The investigator’s
assessment of response was used to determine each patient’s best overall
response. The Fisher exact test was used to compare the treatment groups
for objective tumor response rates and for comparison of prognostic
factors between countries.
safety analysis
All patients who received at least 1 dose of study drug were included in
the safety analyses. Endpoints for assessment of safety included monitoring
the occurrence, frequency and severity of AE [according to Cancer and
Leukemia Group B (CALGB) expanded common toxicity criteria] and
their causal relationship to study drugs. Laboratory values, vital signs,
results of physical examinations and ECG, dose adjustments and treatment
withdrawals for safety reasons were included in the assessment of safety.
results
patient characteristics
A total of 241 patients were randomized from February 1998
to October 2000 (119 to HDC/IL-2/IFN and 122 to DTIC) at
43 centers in Australia (66 patients), Canada (23 patients),
Germany (52 patients), Israel (4 patients), Sweden (45 patients)
and the United Kingdom (51 patients). Follow-up continued
until June 2002, after which the trial database was locked.
The two treatment groups were similar with regard to sex,
race, age, WHO performance status, lactate dehydrogenase
(LDH) and location of disease. There were no significant
differences in key prognostic factors or demographic
characteristics between the two study groups (Table 1).
Patient participation by treatment cycle was defined as
receipt of at least one dose in a given cycle. Participation by
cycle was similar between the two treatment groups after the
first evaluation at week 12. However, there was a higher
drop-out rate in the HDC/IL-2/IFN group after the first cycle
(29% vs. 12% for DTIC). Five patients in the HDC/IL-2/IFN
and one patient in the DTIC arm completed 12 months of
treatment and three patients (all in the HDC/IL-2/IFN arm)
went on with extended treatment for a further 6 months.
efficacy
The primary measure of efficacy was the duration of survival
in the ITT population.
Table 1. Demographic summary: intent-to-treat (ITT) population
Characteristic ITT population
DTIC HDC/IL-2/IFN
N=122 N=119
Age (years)
Median 53 52
Range 23–74 20–73
Sex [n(%)]
Male 77 (63) 74 (62)
Female 45 (37) 45 (38)
WHO PS [n(%)]
0 75 (62) 71 (60)
1 46 (38) 48 (40)
2 1 (1) 0 (0)
Disease sites [n(%)]
a
Skin 31 (25) 29 (24)
Lymph nodes 55 (45) 61 (51)
Bone 17 (14) 9 (8)
Lung 68 (56) 61 (51)
Liver 59 (48) 60 (50)
Other 36 (30) 47 (40)
Number of organ sites
1 38 (31) 32 (27)
2 44 (36) 39 (33)
>2 40 (33) 47 (40)
Adjuvant therapy [n(%)] 6 (5) 5 (4)
LDH group [n(%)]
<ULN 54 (44) 58 (49)
ULN 50 (41) 47 (40)
Missing 18 (15) 14 (12)
DTIC, dacarbazine; HDC, histamine dihydrochloride; IL-2, interleukin-2;
IFN, interferon a2b; ITT, intent-to-treat population; N, total number of
patients in the specified population; n, number of patients with the
specified characteristic in that population; ULN, upper limit of normal;
WHO PS, WHO performance status.
a
Patients may be counted in multiple categories.
Figure 1. Kaplan–Meier survival distribution curves for the
intention-to-treat population (n=241) demonstrated no significant
difference in survival between patients treated with histamine
dihydrochloride/IL-2/interferon a2b (HDC/IL-2/IFN, red) or
dacarbazine (DTIC, blue) (P=0.94).
Annals of Oncology original article
doi:10.1093/annonc/mdm331 |3
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
Although median survival was longer in the group receiving
HDC/IL-2/IFN [271 (216–330, 95% CI) days versus 231 (157–
291) days for the DTIC group], no statistically significant
differences were seen in the Kaplan-Meier estimates for the ITT
population (P=0.94, stratified log-rank test, Fig. 1).
Analysis comparing the Kaplan-Meier estimate of the
percentage of ITT patients alive in the two treatment groups
revealed a statistically significant higher survival rate at 6
months after randomization (P=0.028) in the group receiving
HDC/IL-2/IFN. However, this trend was not retained for later
time periods.
The ITT-LM population was prospectively identified as
a subset of the overall study population that would be
considered in the primary analysis of the study. Although
median survival was longer in the group receiving HDC/IL-2/
IFN [211 (175–272) days versus 154 (114–256) days for the
DTIC group], no statistically significant differences were seen
(P=0.70, Fig. 2).
The results of the multivariate regression analyses for the ITT
and ITT-LM populations are presented in Table 2. In the ITT
population, baseline performance status and serum LDH were
statistically significant predictors of survival. In the ITT-LM
population, only serum LDH was a statistically significant
predictor of survival. In neither the ITT nor ITT-LM
populations did the treatment effect vary according to LDH
level (LDH >ULN P=0.43, LDH £ULN P=0.47 for ITT;
LDH >ULN P=0.55 and for LDH £ULN P=0.90 for
ITT-LM).
Of the 241 patients in the ITT population, 235 (98%) had
either died or experienced disease progression at the time of
data cut-off. The difference between the Kaplan-Meier
estimates of duration of progression-free survival for the two
treatment groups showed a trend but was not statistically
significant; 92 (88–95) days versus 83 (76–87) days for HDC/
IL-2/IFN and DTIC, respectively, P=0.18).
In the ITT population, no statistically significant differences
were seen between the two treatment groups in best tumor
response (Table 3). Fifteen (13%) of the patients receiving
HDC/IL-2/IFN and 17 (15%) of the patients receiving DTIC
achieved a CR or PR. The duration of objective response in
the ITT population was longer for HDC/IL-2/IFN at 310
(178–NA) days than for DTIC [219 (171–290), P=0.44].
Striking differences in median survival were seen among
the participating countries (P=0.003, log-rank test, Table 4,
Fig. 3). Germany (median 482 days, P=0.001), Canada
(336 days, P=0.045) and Australia (230 days, P=0.045) all had
longer median survival than the United Kingdom (178 days).
In Canada, Sweden and the United Kingdom, the median
duration of survival was higher for the group receiving
HDC/IL-2/IFN; however, in Germany and Australia, the
median duration of survival was higher for the group
receiving DTIC, with large variances in the medians between
countries. Despite uniform entry criteria patients in different
countries were entered at different stages of their disease
Figure 2. Kaplan–Meier survival distribution curves for the intention-to-
treat population with liver metastases demonstrated no significant
difference in survival between patients treated with histamine
dihydrochloride/IL-2/interferon a2b (HDC/IL-2/IFN, red, n=60) or
dacarbazine (DTIC, blue, n=59) (P=0.70).
Table 2. Duration of survival: multivariate Cox proportional hazards model: intent-to-treat population
Covariate ITT ITT-LM
Hazard ratio 95% CI Pvalue
a
Hazard ratio 95% CI Pvalue
a
Treatment (HDC/IL-2/IFN versus DTIC) 0.95 0.71–1.28 0.7424 0.87 0.57–1.33 0.5213
Number of disease sites
2 versus 1 0.95 0.63–1.43 0.8001 0.97 0.50–1.90 0.9346
3 versus 1 1.58 0.90–2.79 0.1110 1.52 0.63–3.64 0.3481
Baseline WHO PS (0 vs. 1) 0.58 0.42–0.79 0.0005* 0.66 0.43–1.01 0.0576
Sex (male vs. female) 1.18 0.86–1.61 0.3003 1.38 0.90–2.12 0.1426
Serum LDH (ULN vs. <ULN) 2.53 1.82–3.52 <0.0001* 2.15 1.37–3.36 0.0008*
Site of metastatic disease
Lymph metastases (yes vs. no) 0.82 0.56–1.18 0.2808 1.04 0.59–1.82 0.9058
Lung metastases (yes vs. no) 1.04 0.71–1.51 0.8552 1.09 0.63–1.90 0.7603
Liver metastases (yes vs. no) 1.05 0.73–1.50 0.8049
Bone metastases (yes vs. no) 1.04 0.64–1.70 0.8648 1.01 0.50–2.01 0.9867
CI, confidence interval; DTIC, dacarbazine; HDC, histamine dihydrochloride; IL-2, interleukin-2; IFN, interferon a2b; ITT, intent-to-treat population; ITT-
LM, intent-to-treat liver metastases population; ULN, upper limit of normal; WHO PS, World Health Organization performance status.
a
From Wald chi-square test. Differences that reached statistical significance are marked with an asterisk(*).
original article Annals of Oncology
4|Middleton et al.
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
(Table 4). Australia and Germany had significantly fewer
patients with elevated LDH than Sweden and Canada. Germany
also had significantly fewer patients with stage M1c than
Sweden and the United Kingdom. Performance status also
varied with Australia and Germany, despite the lowest
frequency of patients with elevated LDH and/or stage M1c,
having the highest frequency of patients considered to have
performance status 1.
In contrast to other phase 3 trials in melanoma we
permitted the entry of patients with ocular melanoma. In this
group of 37 patients median survival was longer for those
receiving HDC/IL-2/IFN [325 (174–611) days versus 220
(118–283) days for the DTIC group, P=0.14].
safety
The percentage of patients experiencing any AE was similar
between the two treatment groups (99% for the HDC/IL-2/IFN
group and 97% for the DTIC group), as was the percentage
of patients who died on study or within 28 days of the last
dose of study drug (10% for the HDC/IL-2/IFN group and 10%
for the DTIC group, all patients died from melanoma). Most
AE experienced by patients in both groups were mild or
moderate in severity. The percentage of patients who
discontinued because of an AE was also similar between the
two groups (23% for the HDC/IL-2/IFN group and 20% for
the DTIC group). Fatigue, lethargy, nausea and vomiting
were the most common AEs in the HDC/IL-2/IFN arm while
abdominal pain and hepatobiliary abnormalities were the
most common in the DTIC population (Table 5).
discussion
Stage IV melanoma patients have a poor prognosis and DTIC
remains the standard treatment despite no demonstrable
overall survival advantage. In this randomized study comparing
DTIC to an outpatient immunotherapy regimen, we did not
show statistically significant differences in terms of overall
survival, tumor response or duration of response. The duration
of response was 90 days longer in the combination group than
in the DTIC group, albeit this difference did not attain
statistical significance. The results for other secondary
endpoints were similar between groups.
The similarity in outcomes between the two treatment arms
may be explained by their being equally ineffective against
metastatic melanoma. However, a number of factors need to be
considered before coming to this conclusion. The survival
difference that this trial was powered to detect, at 50%, is very
large. A more modest advantage in favor of immunotherapy
may have been missed. At the time that this trial was conceived
Table 3. Tumor response: intent-to-treat population
Best tumor response DTIC,
N=122
HDC/IL-2/IFN,
N=119
n(%) n(%)
Complete remission 3 (3) 3 (3)
Partial remission 14 (12) 12 (10)
Stable disease 34 (29) 34 (29)
Progressive disease 65 (56) 69 (58)
DTIC, dacarbazine; HDC, histamine dihydrochloride; IL-2, interleukin-2;
IFN, interferon a2b; ITT, intent-to-treat population; N, number of patients
in specified treatment group; n, number of patients with specified response.
Table 4. Duration of survival, by country analyses: intent-to-treat population
Country
a
Elevated LDH (%) Stage M1c (%) WHO PS 1 (%) DTIC HDC/IL-2/IFN
NKaplan-Meier
estimate of
median duration
in days
b
(95% CI)
NKaplan-Meier
estimate of
median duration
in days
b
(95% CI)
Australia 38
c
70 55
h,i,j
32 239 (139–354) 34 216 (175–300)
Canada 52 74 26 11 318 (112–468) 12 353 (310–423)
Germany 25
d,e
65
f,g
40 28 540 (256–674) 24 459 (275–600)
Sweden 61 84 32 23 188 (129–343) 22 276 (167–611)
United Kingdom 44 82 30 26 125 (94–214) 25 208 (145–359)
CI, confidence interval; DTIC, dacarbazine; HDC, histamine dihydrochloride; IL-2, interleukin-2; IFN, interferon a2b; N, number of patients enrolled in
specified country; LDH, lactate dehydrogenase; ULN, upper limit of normal; Stage M1c, visceral metastases other than lung and/or LDH >ULN; WHO PS,
WHO performance status.
a
Israel is not included in these analyses because only four patients were enrolled at sites in that country.
b
Number of days from date of randomization to date of death (or to last date known to be alive).
c
P=0.012 vs. Sweden.
d
P=0.028 vs. Canada.
e
P=0.0004 vs. Sweden.
f
P=0.032 vs. Sweden.
g
P=0.048 vs. the United Kingdom.
h
P=0.015 vs. Canada.
i
P=0.016 vs. Sweden.
j
P=0.0071 vs. the United Kingdom.
Annals of Oncology original article
doi:10.1093/annonc/mdm331 |5
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
the sample size was on a par with many melanoma phase
3 trials. The drop-out rate by the end of cycle 1 matched the
20% we anticipated in the trial design. However, the lower
percentage of patients in the immunotherapy arm able to go
beyond the first cycle of treatment may also have contributed to
an underestimate of its potential. In contrast, the treatment
duration of the similar HDC/IL-2/IFN regimen used in the
earlier single site phase II study was a median 5 months [14].
This was probably a reflection of the relative complexity of the
regimen, and of physicians’ unfamiliarity with it and the
attendant side-effects.
The dose of IL-2 administered in this study, 144 MIU/m
2
during a 12-week period, is lower than in the two randomized
studies where the combination of HDC and IL-2 showed
a survival benefit over IL-2 alone. In the previously mentioned
study in melanoma [12] 224 MIU/m
2
of IL-2 was given
during a 12-week period. In a phase 2 study in renal cell
carcinoma a flat dose of 540 MIU IL-2 was administered
during a 10-week period [15].
The large intercountry differences in median survival
observed for this relatively small cohort of 241 patients
complicates interpretation of the trial. It is clear that, despite
uniform entry criteria, patients in different countries were
entered at different stages of their disease, as shown by the
variations in proportions of patients with elevated LDH and/or
M1c disease between countries. This may reflect differing care
pathways, local predispositions towards particular types of
therapy or competing studies available only in some countries.
Whatever the reason, it highlights an important issue in the
design of clinical trials in melanoma. The need for studies with
several hundred patients means that international
collaborations will be the norm for future studies, and careful
attention will be required to ensure that baseline assumptions
with respect to outcome apply across trials. For example, we
assumed a 7-month median survival in our control arm, but
in Germany DTIC-treated patient survival, at 17.7 months, was
more than double that estimate.
This randomized, phase 3 study of combination
immunotherapy with HDC/IL-2/IFN for AJCC stage IV
malignant melanoma demonstrated similar efficacy with
regards to overall survival, tumor response rates, duration of
response and progression-free survival compared to DTIC.
The lack of statistical difference between the treatment
groups in overall survival may be explained by an
insufficient biological activity of the combination treatment
or, alternatively, by the study being insufficiently powered,
intercountry differences with respect to survival, the lower
percentage of patients in the HDC/IL-2/IFN group who
completed more than one cycle of therapy and by the
relatively low dosage of IL-2. Risk as measured by percentage
of patients who died, had a severe AE or discontinued
because of an AE was similar between the two groups.
Immunotherapy regimens may yet provide treatment
alternatives for patients with stage IV melanoma, but this
immunotherapeutic regimen did not improve upon the
response rate and overall survival seen with DTIC.
Figure 3. Kaplan–Meier survival distribution curves for patients among
participating countries demonstrated a significant difference in survival
between countries (P=0.003, log-rank test): Australia (blue, n=66),
Sweden (green, n=45), United Kingdom (UK, red, n=51), Germany
(black, n=52) and Canada (pink, n=23). Israel (n=4) excluded.
Table 5. Incidence of most common and most severe adverse events:
safety population
Toxicity [n(%)] DTIC, N=116 HDC/ IL-2/IFN, N=114
Grade 3 Grade 4 Grade 3 Grade 4
Patients with
at least one AE
42(36) 9(8) 68(60) 4(4)
Hematological
Anemia 1 (1) 1 (1) 2 (2) 1 (1)
Granulocytopenia 2 (2)
Neutropenia 2 (2) 1 (1)
Thrombocytopenia 1 (1) 1 (1)
Cardiac 0 0 0 0
Gastrointestinal 17 (15) 1 (1) 26 (23)
Abdominal pain 10 (9) 12 (10)
Bleeding 3 (3) 1 (1)
Nausea 5 (4) 10 (9)
Vomiting 2 (2) 12 (10)
General
Fatigue 1 (1) 16 (14)
Lethargy 11 (10)
Multiorgan failure 3 (3)
Hepatobiliary 7 (6) 3 (3) 2 (2)
Infections 1 (1) 1 (1) 4 (4)
Anorexia/cachexia 1 (1) 1 (1) 2 (2)
Musculoskeletal pain 7 (6) 7 (6)
Headache 1 (1) 7 (6)
Depression 2 (2)
Brain edema 1 (1)
Renal 1 (1) 1 (1)
Respiratory 5 (4) 3 (3)
Skin 3 (3) 3 (3)
Flushing 4 (4)
DTIC, dacarbazine; HDC, histamine dihydrochloride; IL-2, interleukin-2;
IFN, interferon a2b; N, total number of patients in specified treatment
group; AE, adverse event; n, number of patients with specified severe
adverse event.
original article Annals of Oncology
6|Middleton et al.
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
acknowledgements
We thank the other investigators participating in this study:
M. Millward, P. Hersey, I. Davis, G. Toner, M. Byrne, I. Olver,
J. Trotter, R. Kefford, D. Kotesak, D. Wyld, J. Mattsson,
U. Stierner, C. Ingvar, A. Ha
˚kansson, J. Hansson, C. Lindholm,
M. Rande
´n, JH. Svensson, G. Wagenius, M. Breivald,
T. Hamblin, P. Lorigan, C. Price, C. Garbe, EB Bro
¨cker,
H. Na
¨her, R. Kaufmann, D. Schadendorf, C. Peschel,
P. von den Driesch, W. Sterry, W. Tilgen, K. Belanger,
Y. Drolet, F. Patenaude, R. Rajan, M. Smylie, T. Peretz,
R. Weitzen and R. Isacson. We thank the people at Maxim
Pharmaceuticals, Covance, CroMedica Global Inc. and
Synteract who managed this clinical trial. Acknowledgment
of research support: EpiCept Corporation (former Maxim
Pharmaceuticals), San Diego, CA, USA.
Conflict of interest statement. Dr A. Hauschild has served as
consultant to Maxim Pharmaceuticals and Schering Plough
(less than $10,000) and research funding from Schering
Plough (less than $100,000). No stock ownership and others.
Dr S. Burdette-Radoux had research funding from Maxim
Pharmaceuticals (less than $100,000). Dr K. Gehlsen was
Clinical Senior Officer of Maxim Pharmaceuticals at time of
the study. Dr K. Hellstrand served as consultant to Maxim
Pharmaceuticals (less than $100,000) and had research
funding from Maxim Pharmaceuticals (more than $100,000).
Dr P. Naredi served as consultant to Maxim Pharmaceuticals
(less than $100,000) and had research funding from Maxim
Pharmaceuticals (less than $100,000). Dr M. Middleton served
as consultant to Maxim Pharmaceuticals (less than $10,000).
Dr D. Thomson, none declared; Dr R. Andersson, none
declared.
references
1. Eggermont AM, Kirkwood JM. Re-evaluating the role of dacarbazine in
metastatic melanoma: what have we learned in 30 years? Eur J Cancer 2004;
40: 1825–1836.
2. Middleton MR, Grob JJ, Aaronson N et al. Randomized phase III study of
temozolomide versus dacarbazine in the treatment of patients with advanced
metastatic malignant melanoma. J Clin Oncol 2000; 18: 158–166.
3. Avril MF, Aamdal S, Grob JJ et al. Fotemustine compared with dacarbazine in
patients with disseminated malignant melanoma: a phase III study. J Clin Oncol
2004; 22: 1118–1125.
4. Atkins MB, Lotze MT, Dutcher JP et al. High-dose recombinant interleukin 2
therapy for patients with metastatic melanoma: analysis of 270 patients treated
between 1985 and 1993. J Clin Oncol 1999; 17: 2105–2116.
5. Kirkwood JM. Studies of interferons in the therapy of melanoma. Semin Oncol
1991; 18 (Suppl 7): 83–90.
6. Kruit WH, Punt CJ, Goey SH et al. Dose efficacy study of two schedules of high-
dose bolus administration of interleukin 2 and interferon-alpha in metastatic
melanoma. Br J Cancer 1996; 74: 951–955.
7. Sparano JA, Fisher RI, Sunderland M et al. Randomized phase III trial of
treatment with high-dose interleukin-2 either alone or in combination with
interferon- alfa-2a in patients with advanced melanoma. J Clin Oncol 1993; 11:
1969–1977.
8. Kono K, Salazar-Onfray F, Petersson M et al. Hydrogen peroxide secreted by
tumor-derived macrophages down-modulates signal-transducing zeta molecules
and inhibits tumour-specific T cell-and natural killer cell-mediated cytotoxicity.
Eur J Immunol 1996; 26: 1308–1313.
9. Jorkov AS, Donskov F, Steiniche T et al. Immune response in blood and
tumour tissue in patients with metastatic malignant melanoma treated with
IL-2, IFN alpha and histamine dihydrochloride. Anticancer Res 2003; 23:
537–542.
10. Hellstrand K, Naredi P, Lindner P et al. Histamine in immunotherapy of advanced
melanoma: a pilot study. Cancer Immunol Immunother 1994; 39: 416–419.
11. Brune M, Hellstrand K. Remission maintenance therapy with histamine and
interleukin-2 in acute myelogenous leukaemia. Br J Haematol 1996; 92:
620–626.
12. Agarwala SS, Glaspy J, O’Day SJ et al. Results from a randomized phase III study
comparing combined treatment with histamine dihydrochloride plus interleukin-2
versus interleukin-2 alone in patients with metastatic melanoma. J Clin Oncol
2002; 20: 125–133.
13. Donskov F, von der Maase H, Henriksson R et al. Outpatient treatment with
subcutaneous histamine dihydrochloride in combination with interleukin-2 and
interferon-alpha in patients with metastatic renal cell carcinoma: results of an
open single-armed multicentre phase II study. Ann Oncol 2002; 13: 441–449.
14. Lindner P, Rizell M, Mattsson J et al. Combined treatment with histamine
dihydrochloride, interleukin-2 and interferon-alpha in patients with metastatic
melanoma. Anticancer Res 2004; 24: 1837–1842.
15. Donskov F, Middleton M, Fode K et al. Two randomised phase II trials of
subcutaneous interleukin-2 and histamine dihydrochloride in patients with
metastatic renal cell carcinoma. Br J Cancer 2005; 93: 757–762.
16. Brune M, Castaigne S, Catalano J et al. Improved leukemia-free survival after
postconsolidation immunotherapy with histamine dihydrochloride and interleukin-
2 in acute myeloid leukemia: results of a randomized phase 3 trial. Blood 2006;
108: 88–96.
17. Hellstrand K, Kylefjord H, Asea A et al. Regulation of the natural killer cell
response to interferon-alpha by biogenic amines. J Interferon Res 1992; 12:
199–206.
18. Brune M, Hansson M, Mellqvist UH et al. NK cell-mediated killing of AML blasts:
role of histamine, monocytes and reactive oxygen metabolites. Eur J Haematol
1996; 57: 312–319.
19. Hansson M, Hermodsson S, Brune M et al. Histamine protects T cells and natural
killer cells against oxidative stress. J Interferon Cytokine Res 1999; 19:
1135–1344.
20. Mellqvist UH, Hansson M, Brune M et al. Natural killer cell dysfunction and
apoptosis induced by chronic myelogenous leukemia cells: role of reactive
oxygen species and regulation by histamine. Blood 2000; 96: 1961–1968.
21. Romero AI, Thoren FB, Brune M et al. NKp46 and NKG2D receptor expression in
NK cells with CD56dim and CD56bright phenotype: regulation by histamine and
reactive oxygen species. Br J Haematol 2006; 132: 91–98.
22. Asemissen AM, Scheibenbogen C, Letsch A et al. Addition of histamine to
interleukin 2 treatment augments type 1 T-cell responses in patients with
melanoma in vivo: immunologic results from a randomized clinical trial of
interleukin 2 with or without histamine (MP 104). Clin Cancer Res 2005; 11:
290–297.
Annals of Oncology original article
doi:10.1093/annonc/mdm331 |7
by guest on June 4, 2013http://annonc.oxfordjournals.org/Downloaded from
... Among the recently used monoclonal antibody preparations, ecromeximab and ipilimumab proved to be safe in combination with the HDI regimen, whereas no significant improvement in OS was observed [176][177][178]. The addition of anticancer cytokine preparations, e.g., IL-2, granulocyte-monocyte colony stimulating factor (GM-CSF) and endostatin, failed to induce the expected amplification of the immune response and increased the incidence of adverse reactions in metastatic melanoma patients [174,179,180]. Some putative therapeutic regimens composed of traditional chemotherapeutic drugs were found ineffective in clinical trials, e.g., the IFN-α2b + dacarbazine + cisplatin + fotemustine regimen and the IFN-α2b + dacarbazine + cisplatin + carmustine + tamoxifen regimen [181,182]. ...
Article
Full-text available
Interferon-α2b (IFN-α2b) is a highly active cytokine that belongs to the interferon-α (IFN-α) family. IFN-α2b has beneficial antiviral, antitumour, antiparasitic and immunomodulatory activities. Direct and indirect antiproliferative effects of IFN-α2b have been found to occur via multiple pathways, mainly the JAK-STAT pathway, in certain cancers. This article reviews mechanistic studies and clinical trials on IFN-α2b. Potential regulators of the function of IFN-α2b were also reviewed, which could be utilized to relieve the poor response to IFN-α2b. IFN-α2b can function not only by enhancing the systematic immune response but also by directly killing tumour cells. Different parts of JAK-STAT pathway activated by IFN-α2b, such as interferon alpha and beta receptors (IFNARs), Janus kinases (JAKs) and IFN‐stimulated gene factor 3 (ISGF3), might serve as potential target for enhancing the pharmacological action of IFN-α2b. Despite some issues that remain to be solved, based on current evidence, IFN-α2b can inhibit disease progression and improve the survival of patients with certain types of malignant tumours. More efforts should be made to address potential adverse effects and complications.
... The dramatic efficacy of ICIs was first demonstrated in metastatic melanoma in 2011 with the FDA approval of ipilimumab, an anti-CTLA4 monoclonal antibody (7). A 1 year overall survival of 25-35% with previous standard of care chemotherapy (8), increased to 73% with use of combination ICI therapies nivolumab (anti-PD1) and ipilimumab (anti-CTLA4) (9). ICIs provide a robust long term benefit with a 52% 5 year overall survival in patients with advanced melanoma after combination ipilimumab and nivolumab treatment (9). ...
Article
Full-text available
Immune checkpoint inhibitors have transformed the landscape of oncological therapy, but at the price of a new array of immune related adverse events. Among these is β-cell failure, leading to checkpoint inhibitor-related autoimmune diabetes (CIADM) which entails substantial long-term morbidity. As our understanding of this novel disease grows, parallels and differences between CIADM and classic type 1 diabetes (T1D) may provide insights into the development of diabetes and identify novel potential therapeutic strategies. In this review, we outline the knowledge across the disciplines of endocrinology, oncology and immunology regarding the pathogenesis of CIADM and identify possible management strategies.
... Another study performed with 241 patients shows that the treatment with HDC/IL-2/IFNα was safely administered on an outpatient basis (3 MIU IFNα s.c., once daily for 7 days, 2.4 MIU/m 2 IL-2 s.c., twice a day for 5 days, and 1 mg HDC s.c., twice a day for 5 days or dacarbazine 850 mg/m i.v. every 3 weeks), but this immunotherapeutic regimen did not improve the response rate or OS compared with dacarbazine.266 A significant increase in the production of IFNγ-producing T lymphocytes was observed in patients with melanoma and liver metastases treated with HDC/IL-2 (HDC 1 mg s.c. ...
Article
Full-text available
Cancer is the second leading cause of death globally and its incidence and mortality are rapidly increasing worldwide. The dynamic interaction of immune cells and tumor cells determines the clinical outcome of cancer. Immunotherapy comes to the forefront of cancer treatments, resulting in impressive and durable responses but only in a fraction of patients. Thus, understanding the characteristics and profiles of immune cells in the tumor microenvironment (TME) is a necessary step to move forward in the design of new immunomodulatory strategies that can boost the immune system to fight cancer. Histamine produces a complex and fine-tuned regulation of the phenotype and functions of the different immune cells, participating in multiple regulatory responses of the innate and adaptive immunity. Considering the important actions of histamine-producing immune cells in the TME, in this review we first address the most important immunomodulatory roles of histamine and histamine receptors in the context of cancer development and progression. In addition, this review highlights the current progress and foundational developments in the field of cancer immunotherapy in combination with histamine and pharmacological compounds targeting histamine receptors.
... BCDT, carmustine, cisplatin, dacarbazine, tamoxifen (Dartmouth regimen); DTIC, dacarbazine; IFN, interferon; IL2, interleukin-2. Summarised from:(12)(13)(14)(15)(16)(17)(18)(19)(20)(21)(22)(23)(24)(25)(26)(27)(28)(29)(30) and(31)(32)(33)(34)(35)(36)(37)(38)(39)(40). ...
Article
Full-text available
Metastatic melanoma is a fatal malignancy with a high mortality and morbidity. Since the early 1970s, available medical therapies were limited in improving survival. Surgery represented the best chance for a cure. However, surgery could only be offered to selected patients. The current landscape of treatment has radically evolved since the introduction of targeted and immunotherapies including BRAF and MEK inhibitors, and checkpoint blockers, like PD-1 and CTLA-4 antibodies. These new therapies have seen survival rates matching, and in some cases surpassing, that of surgery. Anti-PD1 and CTLA-4 combination treatments are associated with severe side effects and BRAF and MEK inhibitor combinations may trigger initial tumour responses but prolonged use have resulted in the development of resistant tumour clones and disease relapse. This review examines the role of pulmonary metastasectomy for lung metastasis from malignant melanoma in the current landscape of effective targeted therapy and immunotherapy.
... Prior to this dacarbazine was regarded as standard chemotherapy. Overall response rates were only 10-17% and for patients with metastatic melanoma one-year overall survival was in the order of 25%-35% [45]. ...
Article
Introduction: Melanoma in-transit metastases (ITMs) occur between the primary tumor site and the regional node field. Most patients with ITMs have a poor prognosis. The treatment of ITMs can be simple if surgical excision is possible, but challenging when ITMs are advanced, multiple or recurrent and impacting quality of life. Areas covered: The management of ITM is still evolving. Patients who are disease-free after surgical excision of ITMs are today candidates for adjuvant systemic treatment to reduce recurrence risk. Some who have multiple or advanced ITMs may achieve long-term survival or disease control with systemic therapies alone. Those who do not remain candidates for the numerous locoregional therapies used to treat ITMs, including electrocautery, intralesional injection, topical therapy, electrochemotherapy, isolated limb infusion, isolated limb perfusion, radiation therapy and, rarely, amputation. Relevant publications identified in Pubmed and MEDLINE databases are reviewed. Expert opinion: Patients with multiple ITMs can benefit from use of systemic therapies as primary treatment, in the adjuvant setting and in neoadjuvant trials. Multiple alternative treatment modalities exist for patients unsuitable for systemic therapies or in whom systemic therapies have failed. Trials assessing combined systemic and locoregional therapies for ITMs are in progress.
Article
Marching into the second decade after the approval of ipilimumab, it is clear that immune checkpoint inhibitors (ICIs) have dramatically improved the prognosis of melanoma. Although the current edge is already high, with a 4-year OS% of 77.9% for adjuvant nivolumab and a 6.5-year OS% of 49% for nivolumab/ipilimumab combination in the metastatic setting, a high proportion of patients with advanced melanoma have no benefit from immunotherapy, or experience an early disease relapse/progression in the first few months of treatment, surviving much less. Reasonably, the primary and acquired resistance to ICIs has entered into the focus of clinical research with positive (e.g., nivolumab and relatlimab combination) and negative feedbacks (e.g., nivolumab with pegylated-IL2, pembrolizumab with T-VEC, nivolumab with epacadostat, and combinatorial triplets of BRAF/MEK inhibitors with immunotherapy). Many intrinsic (intracellular or intra-tumoral) but also extrinsic (systematic) events are considered to be involved in the development of this resistance to ICIs: i) melanoma cell immunogenicity (e.g., tumor mutational burden, antigen-processing machinery and immunogenic cell death, neoantigen affinity and heterogeneity, genomic instability, melanoma dedifferentiation and phenotypic plasticity), ii) immune cell trafficking, T-cell priming, and cell death evasion, iii) melanoma neovascularization, cellular TME components(e.g., Tregs, CAFs) and extracellular matrix modulation, iv) metabolic antagonism in the TME(highly glycolytic status, upregulated CD39/CD73/adenosine pathway, iDO-dependent tryptophan catabolism), v) T-cell exhaustion and negative immune checkpoints, and vi) gut microbiota. In the present overview, we discuss how these parameters compromise the efficacy of ICIs, with an emphasis on the lessons learned by the latest melanoma studies; and in parallel, we describe the main ongoing approaches to overcome the resistance to immunotherapy. Summarizing this information will improve the understanding of how these complicated dynamics contribute to immune escape and will help to develop more effective strategies on how anti-tumor immunity can surpass existing barriers of ICI-refractory melanoma.
Article
Full-text available
Forty-three patients with metastatic melanoma were treated with a 5 day (18 patients) and a 3 day (25 patients) schedule of high-dose IL-2 11.7 MIU m2 and IFN-alpha 3 MIU m2 i.v. by bolus administration every 8 h, repeated every 21 days for a total of three courses. The 5 day schedule resulted in a high response rate of 41% (CI 18-67%), but was accompanied by severe cardiotoxicity (41%) and central nervous system toxicity (28%). The 3 day schedule was associated with manageable toxicity, but yielded a moderate response rate of 20% (CI 7-43%).
Article
Natural killer (NK) cells are deficient in patients with chronic myelogenous leukemia (CML), but the mechanisms responsible for the dysfunction are not completely understood. This study reports that CML cells effectively inhibit the baseline and interleukin-2 (IL-2)-induced NK cell cytotoxicity against a CML cell-derived line (K562). A sizable fraction of NK cells subsequently acquired features characteristic of programmed cell death/apoptosis. The CML cell-mediated inhibition of NK cells required triggering of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-mediated formation of reactive oxygen species (ROS) and was prevented by catalase, a scavenger of ROS, and by histamine, acting via H2-receptor–mediated inhibition of ROS production in CML cells. In contrast, nonmalignant neutrophilic granulocytes inhibited NK cells via ROS production without the requirement of exogenous NADPH oxidase-triggering stimuli. We propose that paracrine production of ROS may contribute to the dysfunction of NK cells in CML and that histamine may serve as an autocrine inhibitor of ROS formation in leukemic granulocytes.
Article
Peripheral blasts recovered from patients with acute myelogenous leukaemia (AML) were efficiently lysed by interleukin-2 (IL-2)-activated heterologous natural killer (NK) cells in vitro. The IL-2-induced killing of AML blasts was inhibited by monocytes, recovered from peripheral blood by centrifugal elutriation. Histamine, of concentrations within the micromolar range, abrogated the monocyte-induced inhibition of NK-cells; thereby, histamine and IL-2 synergistically induced NK-cell-mediated killing of AML blasts. The effect of histamine was apparently mediated by H2-type histamine receptors (H2R), since the H2R antagonist ranitidine completely blocked the response. Based on these in vitro findings, seven patients with AML in first (n=2), second (n=3) or third (n=2) complete remission (CR) were given home therapy with interleukin-2 (IL-2; 0.9×106 IU×2 s.c.) and histamine (0.4–0.7 mg×2 s.c.) in cycles of 21 d, separated by 6-week intervals. The patients also received treatment with low-dose cytarabine and thioguanine between cycles of histamine/IL-2. Toxicity was moderate and included local reactions to IL-2 at the site of injection and short-lasting flush, hypotension, and headache to histamine. The addition of histamine to treatment with IL-2 significantly enhanced the accumulation of CD25+ T cells in peripheral blood as compared to treatment with IL-2 alone (P<0.003). Five patients remain in complete remission at 9, 18, 21, 24 and 26 months; the two patients in CR3 relapsed after 8 and 33 months, respectively. In the five patients with earlier relapse, the duration of remission after treatment with histamine/IL-2 has in each case exceeded that of previous remissions. We conclude that (i) histamine and IL-2 synergize to kill human AML blasts in vitro, and (ii) histamine/IL-2 is a safe and feasible approach to immunotherapy of AML which merits further investigation.
Article
Monocytes, recovered from human peripheral blood by counter-current centrifugal elutriation (CCE), suppressed baseline natural killer (NK) cell cytotoxicity (NKCC) and rendered NK cells resistant to activation of cytotoxicity by human recombinant interferon-alpha (IFN-alpha) by a cell contact-dependent mechanism. Monocyte-induced suppression of resting and IFN-activated NK cells was abrogated by the biogenic amines histamine [via H2-type receptors (H2R)] and serotonin [via 5-HT1A-type receptors (5-HT1AR)]. Our data are suggestive of a monocyte/NK cell interaction that is subject to regulation by biogenic amines.
Article
Melanoma does not respond meaningfully to systemic chemotherapy. No significant improvement in overall survival has been observed with any therapy in the setting of advanced disease, nor in the adjuvant setting. The most active single drugs achieve 14% to 22% response rates in larger phase II series, and drug combinations have not in general improved true response rates by 15% or more. Experience with recombinant interferon alpha-2 (rIFN alpha 2) administered by a variety of schedules and routes has demonstrated tumor response rates of 12% to 22% in advanced melanoma. Administered combined with chemotherapy, rIFN alpha 2 has improved response rates in some, but not all trials to date. On the basis of increased responsiveness noted to be inversely related to tumor size, IFN alpha has been explored over the past 7 years as an adjuvant to definitive surgery for melanoma. The North Central Cancer Treatment Group (NCCTG) and Eastern Cooperative Oncology Group (ECOG) have recently completed trials of 3 and 12 months' duration of maximal dosages of rIFN alpha 2 in subjects with high-risk resected deep primary or lymph node metastatic melanoma. These randomized controlled studies completed accrual in 1990. The ECOG trial EST 1684 has tested the effect of rIFN alpha 2b, given intravenously daily for 5 days per week for 4 weeks at 20 x 10(6) IU/m2/d, then subcutaneously three times a week at 10 x 10(6) IU/m2/d for 11 months. A series of three analyses is planned for the ECOG trial, and the first of these interim analyses was reported to the ECOG in June, 1990, revealing an encouraging divergence of survival curves that does not achieve statistical significance at this early time. A randomized controlled study of 3 months' duration of rIFN alpha 2a at 12 x 10(6) IU/m2/d intramuscularly three times a week performed by the NCCTG is also in too early a stage to allow definitive conclusions yet. The toxicity of IFN alpha administered at maximum tolerated doses has been significant in terms of constitutional symptoms and organ dysfunction. There have been two instances of fatal toxicity observed in the ECOG study (286 subjects). Consideration of lower, more tolerable dosage regimens has been derived from the desire to evaluate longer periods of IFN alpha 2 therapy, and long-term maintenance treatment with IFN alpha 2 in adjuvant melanoma therapy. In addition, immunologic studies suggest that dosages of 3 x 10(6) IU/m2/d may be more immunologically active in vivo.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Sixteen patients with advanced metastatic malignant melanoma were treated with a high-dose infusion of interleukin-2 (IL-2; 18 x 10(6) IU/m-2 day-1) together with daily subcutaneous (s.c.) injections of interferon alpha (IFN alpha; 3 x 10(6) U/m-2 day-1) in 5-day cycles. Nine of these patients were given histamine (1 mg s.c.) twice daily during treatment with IL-2 and IFN alpha. In the seven patients who did not receive histamine, one partial response (that is a reduction of more than 50% in the total tumour burden) was observed in a patient with skin and lymph node melanoma. In the eight histamine-treated patients evaluable for response, four partial responses were observed. Two other patients showed regression at one site of metastasis but tumours remained unchanged at other sites. Two histamine-treated patients showed complete resolution of extensive liver metastasis. Sites of response in histamine-treated patients also included the subcutis, lymph nodes, skeleton, spleen and muscle. Lung melanoma did not respond to histamine/IL-2/IFN alpha. Three patients with lung tumours responded with significant (more than 50%) reduction of the volume of soft-tissue tumours, suggesting that the response to histamine may be organotropic. Survival was significantly prolonged in patients receiving histamine. Our data suggest that treatment with histamine may improve the antitumour efficacy of immunotherapy in metastatic melanoma.
Article
To compare the response rate, survival, and toxicity of treatment with high-dose intravenous (IV) bolus interleukin-2 (IL-2) plus interferon alfa-2a (IFN-alpha) with high-dose IL-2 alone in patients with advanced melanoma in a randomized phase III trial design. Eighty-five patients with advanced melanoma were randomly assigned to receive IL-2 6 X 10(6) U/m2 per dose every 8 hours as tolerated for a maximum of 14 doses on days 1 through 5 and 15 through 19, or IL-2 4.5 X 10(6) U/m2 per dose, plus IFN-alpha 3 X 10(6) U/m2 using an identical schedule. A planned interim analysis was performed after 85 patients were entered, which forms the basis for this report. Partial response (PR) occurred in two of 44 patients (5%; 95% confidence interval, 1% to 15%) receiving IL-2 alone, compared with four of 41 patients (10%; 95% confidence interval, 3% to 23%) receiving IL-2/IFN-alpha (P = .30). There were no complete responses (CRs). The median duration of response was 11.5 months (range, 2.0 to 15.7+). There was no significant difference in the median survival duration for patients receiving IL-2 alone (10.2 months) compared with patients receiving IL-2/IFN-alpha (9.7 months). The median and mean number of doses of IL-2 were equivalent in both groups, as was toxicity. There were three treatment-related deaths, two in the IL-2-alone arm and one in the IL-2/IFN-alpha arm. The trial was terminated after the first interim analysis based on predefined early-stopping rules, which included termination if the response rate in the IL-2/IFN-alpha arm was less than 25%. Using the preparation, dose, and schedule of IL-2 in our trial, IFN-alpha failed to enhance significantly the response rate to high-dose IL-2 in the treatment of patients with advanced melanoma.
Article
Although alterations in CD3-associated signal-transducing molecules in tumor-infiltrating T cells of patients with advanced cancer have been previously described, the mechanism behind these changes is not known. We demonstrate that macrophages isolated from metastatic lymph nodes of patients with malignant melanoma down-regulate levels of CD3 zeta in autologous peripheral blood T cells. Lipopolysaccharide (LPS)- or phorbol 12-myristate 13-acetate (PMA)-stimulated monocytes derived from peripheral blood of healthy donors also induced decreased expression of CD3 and CD16-associated zeta chains similar to that observed in T cells and natural killer (NK) cells of patients with advanced cancer. Co-culture with activated monocytes impaired Ca2+ mobilization in peripheral blood derived-T cells when stimulated with monoclonal antibodies to CD3 and also strongly inhibited melanoma-specific cytotoxic T lymphocyte (CTL) activity and NK activity. The presence of catalase, a scavenger of H2O2, during co-culture almost totally abrogated the inhibitory effect of activated monocytes on melanoma-specific CTL lines and on NK cells. Pre-treatment of CTL or NK cells with nontoxic concentrations (1 x 10(-5) M) of H2O2 also severely reduced their cytotoxic activity which could be prevented by catalase. The decrease in CD3 zeta and in CD16 zeta expression, induced by macrophages isolated from metastatic lymph nodes or by LPS-stimulated monocytes, was also prevented by catalase when maintained throughout the co-culture period. The possibility that monocyte/macrophage-derived reactive oxygen metabolites contribute directly to alterations in signal transducing molecules of T cells and NK cells and to the mechanism of immunosuppression in individuals with cancer should be considered.