ArticlePDF Available

Canadian Supportive Care Recommendations for the Management of Neutropenia in Patients with Cancer

Authors:

Abstract and Figures

Hematologic toxicities of cancer chemotherapy are common and often limit the ability to provide treatment in a timely and dose-intensive manner. These limitations may be of utmost importance in the adjuvant and curative intent settings. Hematologic toxicities may result in febrile neutropenia, infections, fatigue, and bleeding, all of which may lead to additional complications and prolonged hospitalization. The older cancer patient and patients with significant comorbidities may be at highest risk of neutropenic complications. Colony-stimulating factors (csfs) such as filgrastim and pegfilgrastim can effectively attenuate most of the neutropenic consequences of chemotherapy, improve the ability to continue chemotherapy on the planned schedule, and minimize the risk of febrile neutropenia and infectious morbidity and mortality. The present consensus statement reviews the use of csfs in the management of neutropenia in patients with cancer and sets out specific recommendations based on published international guidelines tailored to the specifics of the Canadian practice landscape. We review existing international guidelines, the indications for primary and secondary prophylaxis, the importance of maintaining dose intensity, and the use of csfs in leukemia, stem-cell transplantation, and radiotherapy. Specific disease-related recommendations are provided related to breast cancer, non-Hodgkin lymphoma, lung cancer, and gastrointestinal cancer. Finally, csf dosing and schedules, duration of therapy, and associated acute and potential chronic toxicities are examined.
Content may be subject to copyright.
KOUROUKIS et al.
9
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
Copyright © 2008 Multimed Inc.
ABSTRACT
Hematologic toxicities of cancer chemotherapy are
common and often limit the ability to provide treat-
ment in a timely and dose-intensive manner. These
limitations may be of utmost importance in the adju-
vant and curative intent settings. Hematologic toxici-
ties may result in febrile neutropenia, infections,
fatigue, and bleeding, all of which may lead to addi-
tional complications and prolonged hospitalization.
The older cancer patient and patients with significant
comorbidities may be at highest risk of neutropenic
complications. Colony-stimulating factors (
CSF
s) such
as filgrastim and pegfilgrastim can effectively attenu-
ate most of the neutropenic consequences of chemo-
therapy, improve the ability to continue chemotherapy
on the planned schedule, and minimize the risk of fe-
brile neutropenia and infectious morbidity and mor-
tality. The present consensus statement reviews the use
of
CSF
s in the management of neutropenia in patients
with cancer and sets out specific recommendations
based on published international guidelines tailored to
the specifics of the Canadian practice landscape. We
review existing international guidelines, the indications
for primary and secondary prophylaxis, the importance
of maintaining dose intensity, and the use of
CSF
s in
leukemia, stem-cell transplantation, and radiotherapy.
Specific disease-related recommendations are provided
related to breast cancer, non-Hodgkin lymphoma, lung
cancer, and gastrointestinal cancer. Finally,
CSF
dosing
and schedules, duration of therapy, and associated acute
and potential chronic toxicities are examined.
KEY WORDS
Canadian recommendations, neutropenia, febrile neu-
tropenia, supportive care, colony-stimulating factors,
chemotherapy-induced neutropenia, safety
1. INTRODUCTION
Advances in cancer treatment have led to the devel-
opment and administration of more-complex chemo-
therapy regimens in a wider spectrum of cancer pa-
tients, often resulting in increases in hematologic tox-
icities. Also, as the population ages and demographics
shift, greater numbers of older adults, some with sig-
nificant comorbidities, are being considered for che-
motherapy that may result in significant toxicity.
Agents to attenuate hematologic toxicities have
been in widespread use, particularly in primary and
secondary prevention of the neutropenia and febrile
neutropenia (FN) associated with chemotherapy and
stem-cell transplantation (SCT). In cancer patients, FN
has been shown to have a significant impact on pa-
tient outcomes and on the health care system 1. The
potential benefits of colony-stimulating factors (CSFs)
such as granulocyte colony–stimulating factor (G-CSF)
must be measured against their cost and potential
toxicities.
The consensus statement presented here was pre-
pared to summarize, from the Canadian perspective,
current guidelines on the use of
CSF
s, taking into ac-
count the available evidence and recently updated in-
ternational guidelines
2–4
. The present paper reviews
the updated guidelines, recommendations for primary
and secondary prophylaxis, and the use of growth fac-
tors in leukemia,
SCT
, and radiotherapy. In addition,
disease-specific recommendations are made for breast
cancer, lymphoma, lung cancer, and gastrointestinal
cancer. Finally, the latest safety information regard-
ing the use of growth factors is discussed.
2. SUMMARY OF EXISTING GUIDELINES
The American Society of Clinical Oncology (ASCO),
the European Organization for Research and Treat-
ment of Cancer (EORTC), and the National Compre-
hensive Cancer Network (NCCN) have all published
guidelines for the use of CSFs in patients with can-
cer 2–4 (Table I).
The 2005 ASCO guidelines were updated 2 from
the 2000 version; their evidence base included
MEDLINE and Cochrane Library searches up to and
including September 2005. The ASCO guidelines en-
dorse the importance of preventing FN as a clinical
Canadian supportive care
recommendations for the
management of neutropenia
in patients with cancer
C.T. Kouroukis
MD
MS
c,* S. Chia
MD
,
S. Verma
MD
,
D. Robson
MD
,
C. Desbiens
MD
,
§
C. Cripps
MD
,
||
and
J. Mikhael
MD
ME
d
#
PRACTICE GUIDELINE SERIES
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
10
outcome, regardless of other factors, particularly
when the FN rate associated with treatment is at least
20% and no other equally efficacious regimen that
would not require CSFs is available. Primary prophy-
laxis with CSFs is recommended for FN prevention in
patients at higher risk based on age, medical history,
disease characteristics, and myelotoxicity of the che-
motherapy regimen. Dose-dense chemotherapy re-
quiring CSFs is recommended only when clearly
supported by the available evidence or as part of a
clinical trial. Prophylactic CSF is suggested for older
patients (65 years of age or older) with aggressive-
histology lymphoma treated with curative intent. Sec-
ondary prophylaxis is recommended for patients who
experience a neutropenic complication associated
with an earlier chemotherapy cycle and in whom a
reduced dose in a subsequent cycle could compro-
mise disease-free survival (DFS), overall survival (OS),
or another treatment outcome.
In 2006, EORTC published its guidelines for the
use of CSFs in patients with cancer and chemotherapy-
induced neutropenia. Those guidelines were based
on literature published from 1996 through Septem-
ber 2005 3. In 2003, the EORTC Cancer in the Elderly
Task Force had published guidelines regarding the
use of CSFs in elderly patients with cancer 5. The EORTC
guidelines recommend prophylactic CSFs when the FN
rate of the proposed treatment is 20% or more. In the
case of regimens with FN rates of between 10% and
20%, the decision to use CSFs should be based on
patient-related risk factors, such as older age (over
65 years of age), advanced stage of disease, previ-
ous FN episodes, and lack of antibiotic prophylaxis.
As do the ASCO guidelines, the EORTC guidelines rec-
ommend CSFs when a reduction in chemotherapy is
associated with poorer prognosis and when dose-
dense regimens associated with a clinical and sur-
vival benefit are being used.
The updated NCCN guidelines for the management
of chemotherapy-induced neutropenia 4 are based on
a panel review of available evidence. The NCCN guide-
lines recommend routine CSF use to reduce the risk of
FN, the risk of hospitalization, and the use of intrave-
nous antibiotics in patients treated with a regimen
associated with a 20% risk of FN (category 1 evi-
dence). That recommendation encompasses patients
receiving curative or adjuvant treatment and treat-
ment to prolong survival or to improve quality of life.
For patients being treated with regimens associated
with a 10%–20% risk of FN, consideration should be
given to using a CSF in high-risk patients, but CSFs
should not be used in low-risk patients (those with a
less-than-10% risk of FN), unless a specific patient is
at significant risk of serious consequences of FN and
that patient is being treated with curative or adjuvant
intent.
Filgrastim (Neupogen: Amgen, Thousand Oaks,
CA, U.S.A.), pegfilgrastim (Neulasta: Amgen), and
ancestim (Stemgen: Amgen) are the only CSFs ap-
proved for use in Canada.
3. GUIDELINES FOR PROPHYLAXIS
3.1 Primary Prophylaxis
Updated international guidelines 2,3 have suggested
broadening the indication for CSF use for primary pro-
phylaxis in patients with solid tumours and hemato-
logic malignancies alike. The upfront use of G-CSFs is
suggested with the use of dose-dense chemotherapy
in some patients with breast cancer 6 and hematologic
malignancies 7. Figure 1 shows a combined EORTC and
ASCO algorithm (combined interpretation of the 2006
G-CSF guidelines of ASCO 2 and EORTC 3) for primary
G-CSF prophylaxis.
Based on the ASCO and EORTC updates, the thresh-
old to recommend primary prophylaxis with G-CSFs
was reduced from a 40% to a 20% risk of FN. The
initial estimate of 40% had been calculated from a
pharmacoeconomic study 8 based on the results of a
randomized study using G-CSF in patients with small-
cell lung cancer (SCLC) 9. The pharmacoeconomic
analysis indicated that prophylactic use of G-CSFs was
cost-effective when the FN rate was at least 40%. The
current lower value of 20% risk was arrived at using
clinical rather than economic evaluation. Addition-
ally, more recent studies have indicated that G-CSFs
can dramatically reduce the FN rate even in patients
with a baseline FN risk of about 20% 10,11.
The evaluation of neutropenia risk has been sum-
marized for several types of chemotherapy regimens
(see the EORTC guidelines’ Table 4 or the ASCO guide-
TABLE ICurrent guidelines for primary prophylaxis with granulocyte colony–stimulating factor (G-CSF) 2–4
Neutropenic event risk ASCO 2006 2 EORTC 2006 3 NCCN 2006 4
Moderate to high Use G-CSF (~20%) Use G-CSF (20%) Use G-CSF (>20%)
Intermediate Recommend G-CSF (<20%) Consider G-CSF (10%–20%) Consider G-CSF (10%–20%)
Low Not specified G-CSF not recommended (<10%) G-CSF not recommended
for most patients (<10%)
Risk factor assessment +++ +++ ++
ASCO = American Society of Clinical Oncology; EORTC = European Organization for Research and Treatment of Cancer; NCCN = National
Comprehensive Cancer Network.
KOUROUKIS et al.
11
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
lines’ Table 1 for a complete list). Although the rel-
evant data were taken from clinical trials, it is impor-
tant to realize that the risk of FN in practice could be
substantially higher, particularly if patients are older
or have comorbidities that may render them ineligible
for most clinical trials and increase their risk of com-
plications 1,5. The ASCO and EORTC guidelines have both
highlighted the higher risk of FN and infectious com-
plications in older cancer patients.
In a systematic review of randomized trials pub-
lished up to December 2006 that tested primary pro-
phylaxis with CSFs in patients with solid tumours or
lymphoma, significant improvements were noted in
infection-related mortality, early mortality, and FN 12.
Patients receiving CSFs experienced more bone pain
and a higher average relative dose intensity (DI). The
hospitalization rate and cancer-related survival data
were insufficient for a complete analysis.
In a 2004 Cochrane review that included studies
up to August 2003, use of CSFs as primary prophy-
laxis in patients with malignant lymphoma receiving
conventional chemotherapy was associated with a
reduction in the risk of severe neutropenia, FN, and
infection. No evidence of benefit was observed for a
reduction in the number of patients receiving intra-
venous antibiotics, a lower infection-related mortal-
ity, or any improvement in tumour response, freedom
from treatment failure, or OS 13.
No published Canadian economic models have
investigated the cost effectiveness of G-CSFs for pa-
tients with at least a 20% risk of FN, and economic
evaluations from other jurisdictions may not be ap-
plicable to Canadian practice 14. The ASCO recommen-
dations emphasize that the decision to use G-CSFs to
prevent FN should be based on clinical data rather
than on economics, looking at evidence of reduction
in infection-related endpoints.
A number of models that were developed to as-
sist in predicting neutropenic complications from
chemotherapy have previously been summa-
rized 15–17. In one example, targeted filgrastim therapy
based on the nadir absolute neutrophil count (ANC) in
the first cycle of adjuvant treatment for breast cancer
resulted in fewer hospitalizations, but no clinical out-
come advantages in survival or quality of life were
observed 18,19.
3.1.1 Summary of Guidelines for Primary Prophylaxis
The use of CSFs is recommended if the treatment
being contemplated is associated with a FN rate
of at least 20%, particularly in the curative or ad-
juvant setting.
The use of
CSF
s is recommended when risk factors
that may increase the toxicities of chemotherapy—
such as older age (65 years), comorbidities, and
previous neutropenic complications—are present.
When patients are being treated with regimens
associated with a 10%–20% risk of FN, clinical
judgment should be applied regarding the ben-
efits of CSFs, based on clinical, laboratory, patient
risk, and disease factors.
FIGURE 1 Combined European Organization for Research and Treatment of Cancer 3 and American Society of Clinical Oncology 2 algorithm
for primary prophylaxis with granulocyte colony–stimulating factor (G-CSF). FN = febrile neutropenia; NHL = non-Hodgkin lymphoma.
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
12
3.2 Secondary Prophylaxis
When maintaining chemotherapy
DI
is important,
G
-
CSF
s are recommended for patients who have al-
ready experienced a neutropenic complication (for
example,
FN
or neutropenia) resulting in a treatment
delay. Maintaining
DI
in such situations minimizes
treatment delay and infectious morbidity with the
intent of avoiding compromise to cancer-related sur-
vival. These criteria are expected to apply best to
patients receiving curative treatment who have al-
ready experienced a significant neutropenic event.
In palliative therapy, less myelotoxic regimens or
flexibility in the chemotherapy schedule to avoid sig-
nificant neutropenic events is preferable. Because
much of the recent evidence on the use of
G
-
CSF
s is
disease-specific, disease-specific situations [breast
cancer, gastrointestinal cancer, lung cancer, and non-
Hodgkin lymphoma (
NHL
)] are discussed later in this
paper.
As noted in the ASCO guidelines, no prospective
studies have specifically evaluated the efficacy of
secondary prophylaxis. Development of FN would be
considered a significant neutropenic event worthy of
future G-CSF prophylaxis.
3.2.1 G-CSF Use During FN
One randomized study 20 and two systematic re-
views 21,22 addressed the issue of CSF use during FN.
Although benefits were observed in terms of shorter
duration of neutropenia, shorter hospitalizations, and
perhaps less infectious burden, no differences in sur-
vival were seen. The authors felt that the use of G-CSFs
during FN would be reasonable if the patient was ex-
periencing a complicated FN episode—for example,
pneumonia, multi-organ dysfunction, or hypotension.
3.2.2 Summary of Guidelines for Secondary Prophylaxis
In patients with a previous neutropenic compli-
cation, CSFs should be used provided that the al-
ternative of dose reduction may impair tumour
response, survival, or treatment outcome.
Use of a CSF during FN should be reserved for
patients experiencing a complicated FN episode
(for example, pneumonia, multi-organ dysfunc-
tion, hypotension).
4. GUIDELINES FOR MAINTAINING DI
The clinical benefits of maintaining DI are perhaps
best demonstrated in adjuvant chemotherapy trials
in early-stage breast cancer. The concept of DI is de-
fined as the amount of drug delivered per unit time
(for example, milligrams per square meter delivered
per week or per cycle), and its impact on breast can-
cer outcomes has been the primary hypothesis in sev-
eral prospective randomized trials. The French
Adjuvant Study Group 05 trial compared high and
low (50%) DIs of epirubicin (E100 vs. E50 every
21 days) in a combination containing 5-fluorouracil
(5-FU), epirubicin, and cyclophosphamide (FEC) 23.
The higher DI arm yielded significant improvements
in DFS and OS. Similarly, the Cancer and Leukemia
Group B 8541 trial compared high, intermediate, and
low DIs of doxorubicin, in a combination of cyclo-
phosphamide, doxorubicin, and 5-FU 24. At high and
intermediate DIs, women experienced significantly
improved DFS and OS over those experienced by
women in the low DI group. Those two trials demon-
strated that, within the standard anthracycline dose
range, a threshold effect exists, meaning that adju-
vant chemotherapy delivered using a suboptimal DI
or lower cumulative anthracycline dose (or both) is
less efficacious. To maximally improve survival for
women with early-stage breast cancer, a critical
(“threshold”) DI or cumulative anthracycline dose (or
both) must be reached.
Other than 50% or lower, the exact threshold re-
duction in DI that adversely affects clinical outcomes
remains controversial. An analysis of the pivotal
Milan trial that used classical cyclophosphamide,
methotrexate, and 5-FU suggested that women who
received less than 85% of the scheduled dose had
worse clinical outcomes after 20 years of follow-
up 25,26. In addition, women who received less than
65% of the scheduled dose did no better than women
treated with surgery alone. Conversely, retrospective
data from larger cohorts of women treated with clas-
sical or intravenous cyclophosphamide, methotrex-
ate, and 5-FU have failed to demonstrate a statistically
significant correlation between chemotherapy DI and
clinical outcome 27,28.
Reduced DI of adjuvant chemotherapy because
of toxicity or poor treatment tolerance in primary
breast cancer is a common occurrence 29. In a study
of community practices across the United States in-
volving almost 20,000 women with early-stage breast
cancer treated with adjuvant chemotherapy, 55.5%
of patients received a DI below 85%. In a similar study
of approximately 4500 patients with aggressive NHL,
slightly more than half of all patients (53%) received
a relative DI below 85% 30.
4.1 Summary of Guidelines for CSFs in
Maintaining DI
The cumulative data suggest that reduced DI is a
common occurrence in the adjuvant systemic
therapy of early-stage breast cancer and in the
curative treatment of aggressive NHL.
Evidence suggests that a minimum DI is required
to maximize the benefit of chemotherapy; how-
ever, the exact threshold remains to be defined.
Therefore, when deciding to use a CSF, DI should
be considered, because CSF administration may
allow for a more optimal dose of chemotherapy
to be given.
KOUROUKIS et al.
13
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
5. GUIDELINES FOR SPECIFIC SETTINGS
5.1 Acute Leukemia
Colony-stimulating factors have been studied exten-
sively in acute myeloid (AML) and lymphoblastic (ALL)
leukemia, principally because the chemotherapeutic
regimens used are highly myelosuppressive, result-
ing in a high rate of morbidity and mortality attribut-
able to infection. Although the clinical trials differ,
various conclusions can be drawn from the existing
data:
In patients completing induction and consolida-
tion chemotherapy for
AML
,
CSF
s reduce the dura-
tion of neutropenia, but do not affect
treatment-related mortality or
OS 2
. The effect may
be more pronounced during consolidation therapy.
Long-term data on the use of CSFs in leukemia
demonstrate no adverse effect on disease status
or patient safety 31.
Cost analyses (in the United States) suggest that
the use of CSFs is cost effective in adult AML and
ALL 32.
When used as an adjunct in treatment of adult
ALL, CSFs reduce the incidence of severe infec-
tions 33,34.
Colony-stimulating factors may be beneficial
when used as “priming” therapy to enhance che-
motherapy in patients with AML 35.
5.1.1 Summary of Guidelines for CSFs in Acute
Leukemia
Colony-stimulating factors should be consid-
ered in patients with AML completing induction
or consolidation chemotherapy who experience
neutropenia.
Colony-stimulating factors should be considered
in patients undergoing chemotherapy for ALL who
experience neutropenia.
In patients with AML, CSFs as priming therapy con-
currently with chemotherapy may be useful, but
cannot be considered routine at the present time.
5.2 Stem-Cell Transplantation
Colony-stimulating factors—both G-CSF and granu-
locyte–macrophage CSF—are frequently used during
autologous and allogeneic hematopoietic SCT. Pre-
transplant, CSFs are used to assist in the mobilization
of stem cells from the marrow for peripheral collec-
tion. Post-transplant, they are used to reduce infec-
tion, shorten hospitalization, and possibly reduce
costs.
5.2.1 Mobilization
Growth factors are used in both autologous and allo-
geneic transplantation mobilization. Repeated stud-
ies have validated this collection approach and con-
firm its superiority over traditional bone-marrow har-
vest in yielding a better product that enhances
engraftment and reduces graft-versus-host disease
(GVHD) 36.
When used in combination with chemotherapy
or alone in high doses, CSFs promote enhanced mobi-
lization 37,38. Among the various regimens tested, the
one most commonly used is G-CSF 10 µg/kg daily for
7–10 days before apheresis, with or without chemo-
therapy (that is, high-dose cyclophosphamide).
Pegfilgrastim, although not yet approved for this in-
dication, has showed promise 39.
Another agent, ancestim (also known as “stem-
cell factor”), has been used to mobilize stem cells
and may even be more effective than G-CSF alone 40.
Ancestim is generally recommended only in patients
who do not successfully mobilize with a G-CSF–based
mobilization strategy 41.
5.2.2 Post SCT
Data from many randomized studies have showed
benefit with the use of CSFs in SCT, but the magnitude
of that benefit in yielding clinically important effects
has been questioned. A recent Canadian meta-analy-
sis 42 evaluated the use of CSFs post-transplant and
revealed that CSFs
reduce the risk of documented infection with a
risk ratio (RR) of 0.87 [95% confidence interval
(CI): 0.76 to 1.0; p = 0.05]. The absolute risk re-
duction was 8%, and the number needed to treat
to prevent 1 infection was 13. In allogeneic SCT,
the consequence may be reduced infection-related
mortality.
reduce the time to neutrophil recovery and to
platelet recovery to 50 × 109/L (p = 0.02), but
not to recovery to 20 × 109/L.
reduce hospitalization by 3 days (p < 0.00001).
reduce the duration of parenteral antibiotics (p =
0.02).
produce no differences in acute or chronic GVHD,
treatment-related mortality, or OS.
The heterogeneity of the available studies has left
the potential cost–benefit with the use of CSFs unclear.
However, to date, more studies than not have sug-
gested a positive benefit. Results from the recent
Canadian meta-analysis are consistent with other
published studies that have demonstrated a benefit
in infection reduction but not in OS 13,43,44.
The results from an analysis of a European data-
base raised concerns about the potential increase in
GVHD in patients receiving CSFs 45. However, a long-
term evaluation of data from the International Bone
Marrow Transplant Registry on the use of CSFs in more
than 500 patients treated with allogeneic SCT demon-
strated no long-term benefit or disadvantage with
regard to acute or chronic GVHD and OS 46.
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
14
5.2.3 Summary of Guidelines for G-CSF in SCT
For mobilization, 5–10 µg/kg daily can be used
for 7–10 days before apheresis, with or without
chemotherapy.
Post transplant, 5 µg/kg daily, starting on days 5–
7 can be used until the absolute neutrophil count
rises above 1.5 × 109/L.
5.3 Radiotherapy
The ASCO guidelines 2 indicate that CSFs should be
avoided in patients receiving chemotherapy and con-
comitant radiation, particularly radiation involving
the mediastinum. Therapeutic use of CSFs may be
considered in patients receiving radiotherapy alone
if prolonged delays secondary to neutropenia are ex-
pected. In practice, CSFs are not generally used in ra-
diotherapy because of the lack of evidence to suggest
an improvement in the rate of complication or sur-
vival. In Canada, CSFs are not approved for use with
radiotherapy.
6. DOSING AND FORMULATION OF CSFs
Currently two formulations of G-CSF are approved for
use in Canadian clinical practice. Filgrastim
(r-metHuG-CSF) stimulates the production of neutro-
phil precursors, enhances the function of mature neu-
trophils, and reduces the duration of neutropenia (and
thus its complications). Filgrastim is cleared by the
kidneys, and so its plasma half-life is 3–4 hours. Daily
administration of the drug is therefore required. With
the covalent binding of polyethylene glycol to the
N terminus of filgrastim (producing pegfilgrastim),
the plasma half-life of the drug is increased such that
pegfilgrastim levels as a function of the neutrophil
count become “self-regulating” 47. The net result is
that a single injection of pegfilgrastim is equivalent
to multiple daily injections of filgrastim.
Two large randomized controlled trials compared
single administration of pegfilgrastim with daily
filgrastim in patients receiving myelosuppressive che-
motherapy (an anthracycline–taxane regimen)
48,49
.
The larger of the two trials randomized 310 breast
cancer patients to either a single subcutaneous injec-
tion of pegfilgrastim 100 µg/kg on day 2 or to daily
subcutaneous injections of filgrastim at 5 µg/kg be-
ginning on day 2 and continuing until the
ANC
was
documented at 10 × 10
9
/L or higher after the expected
nadir or for up to 14 days, whichever occurred first
48
.
The second study randomized 157 patients in an iden-
tical design, except that a fixed dose of 6 mg of sub-
cutaneous pegfilgrastim was used
49
. The dose and
duration of the filgrastim in the standard arms was
identical across both studies. Both studies demon-
strated that pegfilgrastim was safe and well tolerated,
as filgrastim was. In regard to duration of severe neu-
tropenia and the depth of the
ANC
nadir, the effects of
pegfilgrastim were similar to those of filgrastim. How-
ever, in one study, the incidence of
FN
was signifi-
cantly lower in the pegfilgrastim arm
48
.
Pegfilgrastim and filgrastim both offer significant
and similar benefits following moderate-to-severe
myelosuppressive chemotherapy for the treatment of
cancer. The additional advantages of pegfilgrastim
include the single injection (convenience for patient
and health care provider) and also potentially a lower
rate of
FN
. Both formulations of
G
-
CSF
should be con-
sidered for patients with solid tumours or lymphomas
requiring a
CSF
for primary or secondary prophylaxis.
7. DURATION OF THERAPY WITH CSFs
As demonstrated in the studies mentioned in the pre-
vious subsection, and in the many studies contribut-
ing to a recent systematic review of primary
prophylaxis with G-CSF 12, the use of filgrastim should
be initiated soon after delivery of chemotherapy (most
studies started on day 2) and continued until a docu-
mented post-nadir ANC recovery to 1.5 × 109/L or
higher is reached. The key goal is to continue until
after the expected nadir. The exact ANC that it is clini-
cally important to achieve is debatable; 1.0–1.5 ×
109/L or higher is suggested. Unless daily blood
counts are being monitored, a conservative approach,
ensuring that the ANC rises well above the desired
level, is wise. Often, between the last filgrastim dose
and day 1 of the subsequent cycle of chemotherapy,
a significant gap occurs during which the ANC drops
to some degree. In the study that investigated sub-
cutaneous pegfilgrastim 49, the median time to ANC
recovery to 2.0 × 109/L or higher with anthracycline–
taxane chemotherapy was 9 days from the day of
chemotherapy.
The duration of filgrastim therapy will also de-
pend on the time to ANC nadir and the duration of
grade 4 neutropenia. Therefore, one additional ad-
vantage of pegfilgrastim is its “self-regulation” with
a single dose, obviating the need for blood count
monitoring and significantly reducing the risk of over-
shooting the target. Daily administration of filgrastim
is currently indicated, although other schedules have
been tested. Data from a nonrandomized observa-
tional study published by Papaldo et al. 50 showed
that a less frequent G-CSF dosing schedule was asso-
ciated with a benefit equivalent to that of daily ad-
ministration in women undergoing adjuvant
chemotherapy for breast cancer, although the rate of
FN in the control arm was only 7%.
8. DISEASE-SPECIFIC RECOMMENDATIONS
Given the prevalence and incidence of breast cancer,
lymphoma, and gastrointestinal and lung cancers, this
subsection presents a more focused analysis of the
available data on FN prevention and the use of CSFs in
those specific diseases.
KOUROUKIS et al.
15
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
8.1 Breast Cancer
Significant advances have been made in adjuvant
systemic therapy for early-stage breast cancer.
Those advances include the use of anthracyclines,
the advent and implementation of dose-dense che-
motherapy, and more recently, the addition of
taxanes. Although all of the foregoing therapeutic
approaches have resulted in improved patient out-
comes 51–56, it is important to recognize the related
toxicities and to ensure that appropriate support-
ive care measures are taken to mitigate the effects
of those toxicities.
Table
II
summarizes the adjuvant breast cancer
regimens commonly in use in Canada and the asso-
ciated rates of
FN
. These data indicate that most of
the adjuvant protocols have
FN
rates under 10%, but
that some protocols would have allowed for sec-
ondary
CSF
prophylaxis. Notably, however, clinical
trial populations tend to be healthier than the gen-
eral population with the same diagnosis, which
means that the rates of
FN
reported in clinical trials
may be lower than those seen in clinical practice.
The two adjuvant protocols in which primary pro-
phylaxis is definitely recommended are dose-dense
cyclophosphamide–doxorubicin (
AC
) followed by
paclitaxel, and docetaxel, doxorubicin, and cyclo-
phosphamide (
TAC
)
6,55
. Primary prophylaxis with
TAC
chemotherapy reduces the
FN
rate to 7.5% from
28.8%
56
.
Recent data have showed that
FEC
followed by
docetaxel is superior to
FEC
100 given for 6 cycles
and has a
FN
rate of 11.2%
54
. That finding has led to
wide adoption of the
FEC
protocol in Canada. Patients
who are being considered for
FEC
treatment followed
by docetaxel should be carefully assessed. Based on
current guidelines, primary prophylaxis should be
considered for patients with significant risk factors
for
FN
.
Breast cancer treatment regimens continue to
evolve, and new treatments are being developed.
Tools are also now available to determine which pa-
tients are likely to benefit from chemotherapy. In the
future, the best way to reduce chemotherapy-associ-
ated toxicities may be to sequester the patients who
will not benefit from chemotherapy.
8.2 Lymphoma
Aggressive-histology
NHL
, such as diffuse large
B cell, represent potentially curable neoplasms, even
in older adults. In a pivotal randomized trial,
CHOP
chemotherapy (cyclophosphamide–doxorubicin–
vincristine–prednisone) was shown to be as effec-
tive as, and less toxic than, more complex second-
and third-generation regimens
57
. Since then, the
addition of rituximab to chemotherapy in patients
with aggressive-histology CD20-positive lymphoma
has improved outcomes in both older
58
and younger
patients
59
. Additional studies have demonstrated the
potential benefits of dose-dense chemotherapy sup-
ported by primary prophylaxis in older adults
7
(that
is,
CHOP
given on a 14-day schedule as compared
with a 21-day schedule), but final publication of the
results of dose-dense chemotherapy with rituximab
(
CHOP
-
R
) are awaited
60–62
. In Canada,
CHOP
-
R
has
been the standard regimen for aggressive-histology
NHL
that expresses CD20. Administration of
CHOP
-
R
could be associated with a
FN
rate of 10% or less
59
or in the 10% to 20% range
58
, but the rate could be
much higher in elderly patients or in those with
comorbidities or poor performance status
63,64
.
Many NHL patients are older and therefore at
increased risk for chemotherapy-related toxici-
ties 63,64, particularly infectious and hematologic
toxicities. Several clinical trials have demonstrated
that a combination of CHOP-like chemotherapy with
rituximab or dose-dense CHOP can improve out-
comes for older adults with aggressive-histology
B cell NHL 58,65,66.
Providing chemotherapy on an accepted sched-
ule has become the standard of care for patients with
potential curable aggressive-histology NHL. Although
no prospective randomized studies have tested stan-
dard against less-than-standard DIs, results of pub-
lished studies have suggested that maintaining the DI
of chemotherapy in aggressive NHL is important 67–70.
Furthermore, regimens that were designed to be less
toxic than standard CHOP have produced inferior out-
comes in older adults with NHL 66,71,72.
Current international guidelines suggest primary
prophylaxis with G-CSF for all older patients (typi-
cally 65 years of age and older) with aggressive-his-
tology lymphoma who are receiving curative-style
(CHOP-R–like) chemotherapy 2,5. Given the importance
of maintaining DI, secondary prophylaxis is also valu-
able in patients of any age who are being treated for
NHL with curative intent.
TABLE II Common adjuvant breast cancer regimens and associated
rates of febrile neutropenia (FN)
Chemotherapy regimen FN incidence
(%)
Docetaxel, doxorubicin, cyclophosphamide 55 28.8
5-Fluorouracil, epirubicin, cyclophosphamide, docetaxel 54 11.2
Oral cyclophosphamide, epirubicin, 5-fluorouracil 51 9.0
5-Fluorouracil, epirubicin, cyclophosphamide 54 8.4
Docetaxel, doxorubicin, cyclophosphamide 56,a 7.5
Docetaxel, cyclophosphamide 53 5.0
5-Fluorouracil, doxorubicin, cyclophosphamide 55 4.4
Doxorubicin, cyclophosphamide, paclitaxel 6,52 3–6
Doxorubicin, cyclophosphamide 52 0–2.5
Dose-dense doxorubicin, cyclophosphamide, paclitaxel 56,a 2.0
Oral cyclophosphamide, methotrexate, 5-fluorouracil 51 1.0
aNecessitated primary prophylaxis with granulocyte colony–
stimulating factor.
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
16
8.3 Gastrointestinal Cancer
Colorectal cancer is the second leading cause of can-
cer death in Western countries
73
, and 50% of patients
who undergo surgery alone for cure ultimately relapse
and die of their disease
74
. In 2002, results of the
MOSAIC
trial were reported at the
ASCO
annual meeting. With
the use of
FOLFOX
(5-
FU
–leucovorin–oxaliplatin)
infusional therapy, 2% of patients relapsed or died as
compared with 26% in the 5-
FU
–leucovorin arms. This
improvement in survival was associated with a 41%
rate of grades 3 and 4 neutropenia in patients receiv-
ing oxaliplatin, but the neutropenia was complicated
by fever or infection in only 1.8% of patients. Adju-
vant therapy with 5-
FU
–leucovorin produced only a
4.7% rate of grades 3 and 4 neutropenia, and only a
0.2% rate of associated fever
75
. The recently revised
ASCO
guidelines
2
for the use of
CSF
s in patients with a
greater-than-20% risk of
FN
currently preclude the use
of those agents prophylactically.
8.4 Lung Cancer
The hematologic toxicities of the various chemo-
therapy regimens in patients with
SCLC
and non-small-
cell lung cancer (
NSCLC
) were included in the
EORTC 3
guideline summary tables (see that guideline’s
Table 4) and in the American Society of Hematology/
ASCO
guideline
2
(see that guideline’s Table 1). De-
pending on the characteristics of the lung cancer sub-
type and the regimen selected,
FN
rates in excess of
20% may be seen. For patients with
NSCLC
, few data
are available demonstrating any benefit in response
rate or survival from primary prophylaxis with a
G
-
CSF 76
. A meta-analysis of randomized trials evalu-
ated the role of
CSF
s in patients with
SCLC
both for
maintaining and for increasing
DI 22
. In the seven stud-
ies designed to maintain
DI
, the response rate was
higher in the groups that received
CSF
s (
RR
: 0.92; 95%
CI
: 0.87 to 0.97), but
OS
was not better [hazard ratio
(
HR
): 1.0; 95%
CI
: 0.94 to 1.13]. In five trials in which
CSF
s were used to increase
DI
, no detectable increase
was observed in either response rate (
RR
: 1.02; 95%
CI
:
0.94 to 1.09) or OS (HR: 0.82; 95% CI: 0.67 to 1.0).
In a more recently published randomized study 77 of
G-CSF prophylaxis in 175 patients with SCLC who were
treated with cyclophosphamide, doxorubicin, and
etoposide, and who were all given prophylactic anti-
biotics, G-CSF reduced the incidence of FN to 18% from
32% (RR: 0.57; 95% CI: 0.34 to 0.97). The difference
in the rate of FN in the first cycle was 24% as com-
pared with 10% (p = 0.01), indicating an early ben-
efit for treatment with G-CSF, despite the use of
prophylactic antibiotics.
9. SAFETY
Differences in the chemical structures of the CSFs have
produced various therapeutic agents. Filgrastim
(Neupogen) is identical to endogenous G-CSF except
that it has an added N-terminal methionine.
Pegfilgrastim (Neulasta) has a polyethylene glycol
molecule bound to the N-terminal methionine; this
structural difference imparts a different pharmacoki-
netic profile. Lenograstim (Granocyte: Chugai Phar-
maceutical, Bedminster, NJ, U.S.A.) is a glycosylated
product identical to the endogenous human mol-
ecule 78. Despite their chemical differences, all of
these molecules interact with the G-CSF receptor and
initiate downstream signalling through the JAKSTAT
(Janus kinase–signal transducers and activators of
transcription) intracellular pathway 79, thus enhanc-
ing the activity, production, and release of neutro-
phils into the peripheral blood.
With the expanded use of CSFs comes a growing
body of data and literature concerning safety and as-
sociated toxicities. The toxicities review that follows
focuses on post-chemotherapy toxicities. Data are
supplemented with the recorded toxicities for CSF use
in the treatment of myelodysplasia and the procure-
ment of stem cells in peripheral blood collection.
9.1 Acute Toxicity
The short-term side effects of CSFs are generally mild
and seldom require dose adjustments or drug cessa-
tion. Documented acute toxicities include bone pain
(25%–30%), headache (16%–55%), fatigue (6%–
33%), nausea (3%–18%), myalgia (5%–41%), insom-
nia (6%–30%), fever (2%–27%), and anorexia
(11%) 80. A multivariate analysis performed by
Murata et al. 81 on apheresis donors indicated that
G-CSF given at doses higher than 8 µg/kg daily was
associated with increased bone pain; headache was
more frequent in donors younger than 35 years of
age; and nausea or vomiting (or both) were more fre-
quent in female donors. Most acute toxicities of CSFs
can be controlled with conservative measures and
non-opioid or opioid analgesics. The administration
of dexamethasone did not seem to ameliorate G-CSF
related adverse events 82. Astemizole, an oral anti-
histamine, has been reported to reduce G-CSF–induced
bone pain unresponsive to acetaminophen 83.
Self-limiting laboratory abnormalities, including
elevated alkaline phosphatase, lactate dehydrogenase,
uric acid, alanine aminotransferase, and gamma-
glutamyl transpeptidase, and decreased potassium and
magnesium have also been reported. Although labo-
ratory coagulation abnormalities have been noted in
the literature 84, clinical thrombotic sequelae are rare
and do not suggest induction of a frank hypercoagu-
lable state.
The potential for CSFs to induce anemia has been
investigated. Papaldo et al. 85 evaluated an adjuvant
anthracycline regimen with or without G-CSF in early
breast cancer. In a recent exploratory hypothesis-gen-
erating analysis of that trial, the use of G-CSF was as-
sociated with a higher incidence of grade 2 anemia
KOUROUKIS et al.
17
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
(38.8% vs. 26.2%, p = 0.005) even though the che-
motherapy DI did not differ between the two study
arms 86. Nonetheless, no difference in clinical out-
comes (such as the need for red blood cell transfu-
sion) was detected between the study arms.
Three trials compared pegfilgrastim with fil-
grastim in the setting of prophylactic CSF support fol-
lowing chemotherapy 48,49,87 and showed similar acute
toxicity profiles.
9.2 Cutaneous Toxicity
Skin toxicities from CSFs can be categorized into three
patterns:
Injection site reactions are the most common, with
one case series reporting a 25% incidence of lo-
calized painful or pruritic wheals 88.
Generalized de novo skin toxicities are rare, but
reports of Sweet syndrome, bullous pyoderma
gangrenosum, leukocytoclastic vasculitis, and fol-
liculitis have all been published 89.
Isolated cases of CSFs exacerbating pre-existing
cutaneous inflammatory disorders such as vas-
culitis and psoriasis have also been documented.
9.3 Pulmonary Toxicity
Anecdotal accounts of CSF-induced pulmonary tox-
icity have been published, including cough, dyspnea,
pulmonary infiltrates 90, and acute respiratory distress
syndrome 91, which is thought to be mediated by neu-
trophil-induced alveolar capillary wall damage 92,
although such reports are exceedingly rare.
In 2001, a systematic review of all published cases
of
CSF
-related pulmonary toxicity uncovered 84 cases
93
.
These cases were further classified into three groups:
Pulmonary toxicity associated with CSF use alone
(group 1, n = 2)
Pulmonary toxicity with CSF used in combination
with other potentially pulmonotoxic agents
(group 2, n = 73)
Pulmonary toxicity during CSF-enhanced neutro-
penia recovery (group 3, n = 9)
The authors concluded that the evidence was in-
sufficient to categorically link the use of CSFs with
significant pulmonary toxicity, because only 2 of the
reported cases were directly linked to CSF use. How-
ever, they did argue that CSFs may interact with other
potentially pulmonotoxic drugs, especially in neu-
tropenic patients with pulmonary infiltrates, warrant-
ing close observation in that patient population.
9.4 Leukemogenicity
The leukemogenic potential of alkylating agents has
been well established in the cancer literature 94.
The National Surgical Adjuvant Breast and
Bowel Project (
NSABP
) experience of adjuvant, stan-
dard-dose,
AC
therapy in 4483 women with breast
cancer revealed an 8-year incidence of treatment-
induced leukemia of 0.27%
95
. Despite the rarity of
secondary hematologic malignancies, the newer regi-
mens enabled by
CSF
s may demonstrate an increase
in the risk of secondary myelodysplastic syndrome
(
MDS
) and acute myelogenous leukemia (
AML
) be-
cause patients receive higher doses of genotoxic
drugs. The better outcomes that result allow for
longer survival, during which secondary hematologic
malignancies may develop.
More recently, Patt et al.
96
evaluated 64,715 pa-
tients from the Surveillance Epidemiology and End
Results (
SEER
)–Medicare database and demonstrated
that the adjusted
HR
for developing
AML
was 1.53
(95%
CI
: 1.14 to 2.06) in patients who received adju-
vant chemotherapy as compared with those who did
not. The use of
G
-
CSF
s within the 1st year of breast
cancer diagnosis was not associated with an increased
risk for developing
AML
(
HR
: 1.14; 95%
CI
: 0.67 to
1.92).
Preclinical models have suggested a possible leu-
kemogenic effect of G-CSFs 97,98; however, to date,
clinical data have not confirmed it.
In the prospective randomized phase III adjuvant
breast trial by the Cancer and Leukemia Group B,
Citron et al. 6 compared a dose-dense regimen of che-
motherapy supported by G-CSF with the standard regi-
men. In an updated report after a median follow-up
of 69 months 99, the incidence of AML or MDS or a com-
bination was no higher in the dose-dense arms than
in the standard arms without routine CSF support
(0.70%). Other adjuvant breast cancer trials incor-
porating routine CSFs have also failed to reveal a sig-
nificantly increased risk of secondary leukemia 50,100.
Trials investigating CSF protocols for other disease
sites (SCLC, urothelial cancer, and sarcoma, for in-
stance) have not reported the incidence of AML or
MDS 101–103.
The magnitude of the additional risk of CSFs, if
present, to the incidence of treatment-related AML or
MDS may be outweighed by the benefit. Population-
based data and meta-analyses have attempted to elu-
cidate an answer to that question.
In a retrospective cohort study based on SEER
claims data, Hershman et al. 104 assessed women older
than 65 years of age who received adjuvant chemo-
therapy, with or without CSFs, for stages IIII breast
cancer between 1991 and 1999. Of the studied
women, 1.16% developed AML or MDS 18 months or
more after diagnosis. Of the 906 patients treated with
G-CSFs, 16 (1.77%) developed AML or MDS; of the 4604
patients not treated with G-CSF, 48 (1.04%) developed
AML or MDS. The risk of AML or MDS did not change
substantially when clinical, treatment, and demo-
graphic variables were accounted for (HR: 2.14; 95%
CI: 1.12 to 4.08).
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
18
This SEER database analysis is based on a large
numbers of patients, but as noted in an accompany-
ing editorial 105, the data are derived from non-vali-
dated health care claims that do not provide specific
data on cumulative dose or duration of either G-CSF
or chemotherapy. A study of this kind could equally
underestimate the incidence of treatment-related
malignancy in patients who died from breast cancer
in the first years of follow-up. The more dose-inten-
sive the adjuvant therapy regimen, the higher the risk
of secondary leukemia. Also, failure to recover mar-
row after exposure to chemotherapy is an indication
for G-CSF, but such failure may also be a marker of
marrow deficiency that may increase susceptibility
to malignant transformation.
Based on the analysis by Hershman et al., the use
of G-CSF was associated with a doubling of the risk
for subsequent AML or MDS in the studied population,
even though the absolute risk remained low. The au-
thors themselves suggested that even if the associa-
tion were to be confirmed, the benefits of G-CSF may
still outweigh its risks 104.
A similar analysis was performed on a population
of 182 French women who developed leukemia after
adjuvant chemotherapy for early breast cancer
106
.
Patients who received
G
-
CSF
(8.8% of the group) had
a significantly increased risk of
AML
or
MDS
(
RR
: 6.26;
95%
CI
: 1.89 to 20.7), although the reason for
G
-
CSF
treatment, the dose, and the duration were not sys-
tematically recorded in medical files. The authors also
noted that
G
-
CSF
was administered chiefly because of
poor hematologic tolerance to chemotherapy, which
could reflect chemotherapy drug accumulation as a
result of altered pharmacokinetics, metabolism, or
bone marrow sensitivity of the patients.
Smith et al. 95 retrospectively reviewed data from
six NSABP trials that were distinguished by differences
in cyclophosphamide intensity and dose, and by the
presence or absence of mandated prophylactic sup-
port with growth factors. As compared with patients
receiving standard chemotherapy, patients receiving
dose-intense chemotherapy with G-CSF support
showed cumulative incidences of MDS and AML of
1.01% (95% CI: 0.63% to 1.62%) and 0.21% (95%
CI: 0.11% to 0.41%) respectively at 5 years. Those
results should be interpreted with caution, because
increasing the chemotherapy dose is, in itself, a risk
factor for AML and MDS 107 and distinguishing the
leukemogenicity of intensified therapy from that of
G-CSF administration is often difficult.
In addition to the combined analysis, Smith
et al. 95 also attempted to analyze the final results of
NSABP B-25, a trial in which women were randomly
assigned to 4 cycles of AC chemotherapy with double
the cumulative dose of cyclophosphamide 108. Al-
though use of G-CSF was mandated for all patients,
total G-CSF dose varied considerably across patients.
Controlling for treatment arm, patient age, and sur-
gical procedure, the estimated risk for AML and MDS
in patients receiving more than the median dose of
G-CSF was 3.58 relative to patients receiving the me-
dian dose or less (p = 0.02). However, the authors
also noted that the result was not based on a random-
ized comparison and that the use of G-CSF was likely
correlated with other factors. Also, patients achiev-
ing an unusually high plasma level of doxorubicin or
cyclophosphamide, or both, are possibly at higher
risk simultaneously for AML or MDS and for FN and
severe infection. In that case, any suggested associa-
tion between the use of G-CSF and the subsequent in-
cidence of AML or MDS may have no causal basis.
Long-term data relating to CSF use in hematology
have revealed uncertain associations with secondary
AML and MDS. Data from the Severe Chronic Neutro-
penia International Registry revealed an association
between the use of CSFs and acquired cytogenetic
clonal abnormalities of the marrow. However, no
evidence definitively related the dose of G-CSF or the
duration of G-CSF therapy to clinical malignant trans-
formation 109. Thus far, registry studies have not iden-
tified an increased risk of malignancy among healthy
individuals who received G-CSF before harvesting of
stem cells from peripheral blood; however, the num-
bers are small, and more than 2000 donors would
have to be followed for 10 years to detect a rise by a
factor of 10 in the leukemia risk 110.
Finally, in a retrospective review of children un-
dergoing chemotherapy for ALL at a single institu-
tion, the cumulative incidence of therapy-associated
AML was significantly higher in the cohort who re-
ceived G-CSF in their treatment protocol than in the
cohort that did not 111.
The data from the dose-dense trials and hematol-
ogy reports are not conclusive. Mitigating factors such
as chemotherapy dose and inherited predispositions
to secondary cancers have not been fully explored.
The ambiguity demands further research with longer
follow-up. Clinical patterns of secondary leukemia
are emerging with corresponding molecular pro-
files 112, thus enabling more precise definition of iatro-
genic as compared with sporadic leukemia.
9.5 Safety Conclusions
A review of the reported toxicity data associated with
CSF
s reveals an acceptable pattern of short-term tox-
icities, manageable with conservative measures
alone. Further follow-up is necessary to elucidate
potential associations between
CSF
s and pulmonary
toxicities, and
CSF
s and secondary hematologic ma-
lignancies. Colony-stimulating factors should always
be used within the context of approved guidelines
and labelling.
10. SUMMARY
Neutropenia is a common complication of chemo-
therapy that can result in severe sequelae in cancer
KOUROUKIS et al.
19
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
patients. The management of neutropenia requires
a patient-specific approach, accounting for the ma-
lignancy, the chemotherapeutic regimen, and patient
risk factors such as age, comorbid illness, and past
history. The appropriate use of
CSF
s is critical to man-
aging these patients in situations of both primary
and secondary prophylaxis, especially in high-risk
situations in which chemotherapeutic regimens are
associated with a 20% or higher risk of
FN
. Benefits
include fewer infections, shorter hospitalizations,
and possibly lesser mortality. As in all aspects of
cancer care, the risks must be weighed against the
benefits, tailoring the treatment to each individual
patient.
11. ACKNOWLEDGMENTS
Amgen Canada provided an unrestricted educational
grant; Klick Communications provided editorial and
project management services.
12. REFERENCES
1. Kuderer NM, Dale DC, Crawford J, Cosler LE, Lyman GH.
Mortality, morbidity, and cost associated with febrile neutro-
penia in adult cancer patients. Cancer 2006;106:2258–66.
2. Smith TJ, Khatcheressian J, Lyman GH, et al. 2006 Update of
recommendations for the use of white blood cell growth fac-
tors: an evidence-based clinical practice guideline. J Clin Oncol
2006;24:3187–205.
3. Aapro MS, Cameron DA, Pettengell R, et al. EORTC guidelines
for the use of granulocyte-colony stimulating factor to reduce
the incidence of chemotherapy-induced febrile neutropenia in
adult patients with lymphomas and solid tumours. Eur J Can-
cer 2006;42:2433–53.
4. National Comprehensive Cancer Network (NCCN). NCCN Prac-
tice Guidelines in Oncology: Myeloid Growth Factors.
V.1.2007. Fort Washington, PA: NCCN; 2007.
5. Repetto L, Biganzoli L, Koehne CH, et al. EORTC Cancer in
the Elderly Task Force guidelines for the use of colony-stimu-
lating factors in elderly patients with cancer. Eur J Cancer
2003;39:2264–72.
6. Citron ML, Berry DA, Cirrincione C, et al. Randomized trial
of dose-dense versus conventionally scheduled and sequen-
tial versus concurrent combination chemotherapy as postop-
erative adjuvant treatment of node-positive primary breast
cancer: first report of Intergroup Trial C9741/Cancer and Leu-
kemia Group B Trial 9741. J Clin Oncol 2003;21:1431–9.
7. Pfreundschuh M, Trumper L, Kloess M, et al. Two-weekly or
3-weekly CHOP chemotherapy with or without etoposide for
the treatment of elderly patients with aggressive lymphomas:
results of the NHL-B2 trial of the DSHNHL. Blood 2004;104:
634–41.
8. Lyman GH, Lyman CG, Sanderson RA, Balducci L. Decision
analysis of hematopoietic growth factor use in patients receiv-
ing cancer chemotherapy. J Natl Cancer Inst 1993;85:488–93.
9. Crawford J, Ozer H, Stoller R, et al. Reduction by granulo-
cyte colony-stimulating factor of fever and neutropenia in-
duced by chemotherapy in patients with small-cell lung cancer.
N Engl J Med 1991;325:164–70.
10. Vogel CL, Wojtukiewicz MZ, Carroll RR, et al. First and sub-
sequent cycle use of pegfilgrastim prevents febrile neutrope-
nia in patients with breast cancer: a multicenter, double-blind,
placebo-controlled phase III study. J Clin Oncol 2005;23:
1178–84.
11. Timmer–Bonte JN, Adang EM, Smit HJ, et al. Cost-effective-
ness of adding granulocyte colony–stimulating factor to pri-
mary prophylaxis with antibiotics in small-cell lung cancer.
J Clin Oncol 2006;24:2991–7.
12. Kuderer NM, Dale DC, Crawford J, Lyman GH. Impact of
primary prophylaxis with granulocyte colony–stimulating fac-
tor on febrile neutropenia and mortality in adult cancer pa-
tients receiving chemotherapy: a systematic review. J Clin
Oncol 2007;25:3158–67.
13. Bohlius J, Reiser M, Schwarzer G, Engert A. Impact of granu-
locyte colony–stimulating factor (CSF) and granulocyte–mac-
rophage CSF in patients with malignant lymphoma: a systematic
review. Br J Haematol 2003;122:413–23.
14. Lyman GH, Kuderer NM. The economics of the colony-stimu-
lating factors in the prevention and treatment of febrile neu-
tropenia. Crit Rev Oncol Hematol 2004;50:129–46.
15. Scott S. Identification of cancer patients at high risk of febrile
neutropenia. Am J Health Syst Pharm 2002;59(suppl 4):
S16–19.
16. Lyman GH. Risk assessment in oncology clinical practice.
From risk factors to risk models. Oncology (Williston Park)
2003;17(suppl 11):8–13.
17. Lyman GH, Kuderer NM, Balducci L. Cost–benefit analysis
of granulocyte colony–stimulating factor in the management
of elderly cancer patients. Curr Opin Hematol 2002;9:207–14.
18. Rivera E, Erder MH, Moore TD, et al. Targeted filgrastim sup-
port in patients with early-stage breast carcinoma: toward the
implementation of a risk model. Cancer 2003;98:222–8.
19. Rivera E, Haim EM, Fridman M, Frye D, Hortobagyi GN.
First-cycle absolute neutrophil count can be used to improve
chemotherapy-dose delivery and reduce the risk of febrile neu-
tropenia in patients receiving adjuvant therapy: a validation
study. Breast Cancer Res 2003;5:R114–20.
20. Garcia–Carbonero R, Mayordomo JI, Tornamira MV, et al.
Granulocyte colony–stimulating factor in the treatment of high-
risk febrile neutropenia: a multicenter randomized trial. J Natl
Cancer Inst 2001;93:31–8.
21. Clark OA, Lyman GH, Castro AA, Clark LG, Djulbegovic B.
Colony-stimulating factors for chemotherapy-induced febrile
neutropenia: a meta-analysis of randomized controlled trials.
J Clin Oncol 2005;23:4198–214.
22. Berghmans T, Paesmans M, Lafitte JJ, Mascaux C, Meert AP,
Sculier JP. Role of granulocyte and granulocyte–macrophage
colony-stimulating factors in the treatment of small-cell lung
cancer: a systematic review of the literature with methodologi-
cal assessment and meta-analysis. Lung Cancer 2002;37:
115–23.
23. French Adjuvant Study Group. Benefit of a high-dose
epirubicin regimen in adjuvant chemotherapy for node-posi-
tive breast cancer patients with poor prognostic factors: 5-year
follow-up results of French Adjuvant Study Group 05 ran-
domized trial. J Clin Oncol 2001;19:602–11.
24. Budman DR, Berry DA, Cirrincione CT, et al. Dose and dose
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
20
intensity as determinants of outcome in the adjuvant treatment
of breast cancer. The Cancer and Leukemia Group B. J Natl
Cancer Inst 1998;90:1205–11.
25. Bonadonna G, Valagussa P. Dose-response effect of adjuvant
chemotherapy in breast cancer. N Engl J Med 1981;304:10–15.
26. Bonadonna G, Valagussa P, Moliterni A, Zambetti M,
Brambilla C. Adjuvant cyclophosphamide, methotrexate, and
fluorouracil in node-positive breast cancer: the results of 20
years of follow-up. N Engl J Med 1995;332:901–6.
27. Cameron DA, Massie C, Kerr G, Leonard RC. Moderate neu-
tropenia with adjuvant CMF confers improved survival in early
breast cancer. Br J Cancer 2003;89:1837–42.
28. Mayers C, Panzarella T, Tannock IF. Analysis of the prognos-
tic effects of inclusion in a clinical trial and of myelosuppres-
sion on survival after adjuvant chemotherapy for breast
carcinoma. Cancer 2001;91:2246–57.
29. Lyman GH, Dale DC, Crawford J. Incidence and predictors of
low dose-intensity in adjuvant breast cancer chemotherapy: a
nationwide study of community practices. J Clin Oncol 2003;
21:4524–31.
30. Lyman GH, Morrison VA, Dale DC, Crawford J, Delgado DJ,
Fridman M on behalf of the OPPS Working Group and ANC Study
Group. Risk of febrile neutropenia among patients with inter-
mediate-grade non-Hodgkin’s lymphoma receiving CHOP che-
motherapy. Leuk Lymphoma 2003;44:2069–76.
31. Heil G, Hoelzer D, Sanz MA, et al. Long-term survival data
from a phase 3 study of filgrastim as an adjunct to chemo-
therapy in adults with de novo acute myeloid leukemia.
Leukemia 2006;20:404–9.
32. Bennett CL, Stinson TJ, Laver JH, Bishop MR, Godwin JE,
Tallman MS. Cost analyses of adjunct colony stimulating fac-
tors for acute leukemia: can they improve clinical decision
making. Leuk Lymphoma 2000;37:65–70.
33. Ottmann OG, Hoelzer D, Gracien E, et al. Concomitant granu-
locyte colony-stimulating factor and induction chemoradio-
therapy in adult acute lymphoblastic leukemia: a randomized
phase III trial. Blood 1995;86:444–50.
34. Geissler K, Koller E, Hubmann E, et al. Granulocyte colony-
stimulating factor as an adjunct to induction chemotherapy
for adult acute lymphoblastic leukemia—a randomized
phase-III study. Blood 1997;90:590–6.
35. Lowenberg B, van Putten W, Theobald M, et al. Effect of prim-
ing with granulocyte colony–stimulating factor on the outcome
of chemotherapy for acute myeloid leukemia. N Engl J Med
2003;349:743–52.
36. Schmitz N, Linch DC, Dreger P, et al. Randomised trial of
filgrastim-mobilised peripheral blood progenitor cell transplan-
tation versus autologous bone-marrow transplantation in lym-
phoma patients. Lancet 1996;347:353–7.
37. Narayanasami U, Kanteti R, Morelli J, et al. Randomized trial
of filgrastim versus chemotherapy and filgrastim mobilization
of hematopoietic progenitor cells for rescue in autologous trans-
plantation. Blood 2001;98:2059–64.
38. Engelhardt M, Bertz H, Afting M, Waller CF, Finke J. High-
versus standard-dose filgrastim (rhG-CSF) for mobilization of
peripheral-blood progenitor cells from allogeneic donors and
CD34+ immunoselection. J Clin Oncol 1999;17:2160–72.
39. Johnston E, Crawford J, Blackwell S, et al. Randomized, dose-
escalation study of SD/01 compared with daily filgrastim in
patients receiving chemotherapy. J Clin Oncol 2000;18:
2522–8.
40. Hess DA, Levac KD, Karanu FN, et al. Functional analysis of
human hematopoietic repopulating cells mobilized with granu-
locyte colony–stimulating factor alone versus granulocyte
colony–stimulating factor in combination with stem cell fac-
tor. Blood 2002;100:869–78.
41. To LB, Bashford J, Durrant S, et al. Successful mobilization
of peripheral blood stem cells after addition of ancestim (stem
cell factor) in patients who had failed a prior mobilization with
filgrastim (granulocyte colony–stimulating factor) alone or
with chemotherapy plus filgrastim. Bone Marrow Transplant
2003;31:371–8.
42. Dekker A, Bulley S, Beyene J, Dupuis LL, Doyle JJ, Sung L.
Meta-analysis of randomized controlled trials of prophylactic
granulocyte colony–stimulating factor and granulocyte–
macrophage colony–stimulating factor after autologous and
allogeneic stem cell transplantation. J Clin Oncol 2006;24:
5207–15.
43. Lyman GH, Kuderer NM, Djulbegovic B. Prophylactic granu-
locyte colony–stimulating factor in patients receiving dose-
intensive cancer chemotherapy: a meta-analysis. Am J Med
2002;112:406–11.
44. Sung L, Nathan PC, Alibhai SM, Tomlinson GA, Beyene J.
Meta-analysis: effect of prophylactic hematopoietic colony-
stimulating factors on mortality and outcomes of infection.
Ann Intern Med 2007;147:400–11.
45. Ringden O, Labopin M, Gorin NC, et al. Treatment with granu-
locyte colony–stimulating factor after allogeneic bone mar-
row transplantation for acute leukemia increases the risk of
graft-versus-host disease and death: a study from the Acute
Leukemia Working Party of the European Group for Blood
and Marrow Transplantation. J Clin Oncol 2004;22:416–23.
46. Khoury HJ, Loberiza FR Jr, Ringden O, et al. Impact of
posttransplantation G-CSF on outcomes of allogeneic hemato-
poietic stem cell transplantation. Blood 2006;107:1712–16.
47. Layton JE, Hockman H, Sheridan WP, Morstyn G. Evidence
for a novel in vivo control mechanism of granulopoiesis: ma-
ture cell–related control of a regulatory growth factor. Blood
1989;74:1303–7.
48. Holmes FA, O’Shaughnessy JA, Vukelja S, et al. Blinded, ran-
domized, multicenter study to evaluate single administration
pegfilgrastim once per cycle versus daily filgrastim as an ad-
junct to chemotherapy in patients with high-risk stage II or
stage III/IV breast cancer. J Clin Oncol 2002;20:727–31.
49. Green MD, Koelbl H, Baselga J, et al. A randomized double-
blind multicenter phase III study of fixed-dose single-admin-
istration pegfilgrastim versus daily filgrastim in patients
receiving myelosuppressive chemotherapy. Ann Oncol 2003;
14:29–35.
50. Papaldo P, Lopez M, Marolla P, et al. Impact of five prophy-
lactic filgrastim schedules on hematologic toxicity in early
breast cancer patients treated with epirubicin and cyclophos-
phamide. J Clin Oncol 2005;23:6908–18.
51. Levine MN, Bramwell VH, Pritchard KI, et al. Randomized
trial of intensive cyclophosphamide, epirubicin, and fluorou-
racil chemotherapy compared with cyclophosphamide, meth-
otrexate, and fluorouracil in premenopausal women with
node-positive breast cancer. National Cancer Institute of
KOUROUKIS et al.
21
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
Canada Clinical Trials Group. J Clin Oncol 1998;16:2651–8.
52. Mamounas EP, Bryant J, Lembersky B, et al. Paclitaxel after
doxorubicin plus cyclophosphamide as adjuvant chemotherapy
for node-positive breast cancer: results from NSABP B-28. J Clin
Oncol 2005;23:3686–96.
53. Jones SE, Savin MA, Holmes FA, et al. Phase III trial compar-
ing doxorubicin plus cyclophosphamide with docetaxel plus
cyclophosphamide as adjuvant therapy for operable breast
cancer. J Clin Oncol 2006;24:5381–7.
54. Roche H, Fumoleau P, Spielmann M, et al. Sequential adju-
vant epirubicin-based and docetaxel chemotherapy for node-
positive breast cancer patients: the FNCLCC PACS 01 Trial. J Clin
Oncol 2006;24:5664–71.
55. Martin M, Pienkowski T, Mackey J, et al. Adjuvant docetaxel
for node-positive breast cancer. N Engl J Med 2005;352:
2302–13.
56. Martin M, Lluch A, Segui MA, et al. Toxicity and health-re-
lated quality of life in breast cancer patients receiving adju-
vant docetaxel, doxorubicin, cyclophosphamide (TAC) or
5-fluorouracil, doxorubicin and cyclophosphamide (FAC ): im-
pact of adding primary prophylactic granulocyte-colony stimu-
lating factor to the TAC regimen. Ann Oncol 2006;17:1205–12.
57. Fisher RI, Gaynor ER, Dahlberg S, et al. Comparison of a stan-
dard regimen (CHOP) with three intensive chemotherapy regi-
mens for advanced non-Hodgkin’s lymphoma. N Engl J Med
1993;328:1002–6.
58. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus
rituximab compared with CHOP alone in elderly patients with
diffuse large-B-cell lymphoma. N Engl J Med 2002;346:
235–42.
59. Pfreundschuh M, Trumper L, Osterborg A, et al. CHOP-like
chemotherapy plus rituximab versus CHOP-like chemotherapy
alone in young patients with good-prognosis diffuse large-
B-cell lymphoma: a randomised controlled trial by the
MabThera International Trial (MInT) Group. Lancet Oncol
2006;7:379–91.
60. Pfreundschuh M, Kloess M, Schmits R, et al. Six, not eight
cycles of bi-weekly CHOP with rituximab (R-CHOP-14) is the
preferred treatment for elderly patients with diffuse large B-cell
lymphoma (DLBCL): results of the RICOVER-60 trial of the Ger-
man High-Grade Non-Hodgkin Lymphoma Study Group
(DSHNHL) [abstract 13]. Blood 2005;106:9a.
61. Verdonck LF, van Imhoff GW, Raemakers JMM, et al. Six
courses of intensified CHOP plus G-CSF compared to eight courses
of standard CHOP in patients with intermediate-risk aggressive
non-Hodgkin lymphoma results of a prospective randomized
HOVON trial [abstract 14]. Blood 2005;106:9a.
62. Sonneveld P, van Putten W, Holte H, et al. Intensified CHOP
with rituximab for intermediate or high-risk non-Hodgkin’s
lymphoma: interim analysis of a randomized phase III trial in
elderly patients by the Dutch HOVON and Nordic Lymphoma
groups [abstract 16]. Blood 2005;106:10a.
63. Gómez H, Mas L, Casanova L, et al. Elderly patients with
aggressive non-Hodgkin’s lymphoma treated with CHOP che-
motherapy plus granulocyte–macrophage colony–stimulating
factor: identification of two age subgroups with differing he-
matologic toxicity. J Clin Oncol 1998;16:2352–8.
64. Gómez H, Hidalgo M, Casanova L, et al. Risk factors for treat-
ment-related death in elderly patients with aggressive non-
Hodgkin’s lymphoma: results of a multivariate analysis. J Clin
Oncol 1998;16:2065–9.
65. Kouroukis C, Browman GP, Esmail R, Meyer RM. A system-
atic review of chemotherapy for older patients with newly di-
agnosed, advanced stage, aggressive histology non-Hodgkin’s
lymphoma. Ann Intern Med 2002;136:144–52.
66. Sonneveld P, de Ridder M, Van Der Lelie H, et al. Compari-
son of doxorubicin and mitoxantrone in the treatment of eld-
erly patients with advanced diffuse non-Hodgkin’s lymphoma
using CHOP versus CNOP chemotherapy. J Clin Oncol 1995;13:
2530–9.
67. Epelbaum R, Haim N, Ben Shahar M, Ron Y, Cohen Y. Dose-
intensity analysis for CHOP chemotherapy in diffuse aggres-
sive large cell lymphoma. Isr J Med Sci 1988;24:533–8.
68. Epelbaum R, Faraggi D, Ben Arie Y, et al. Survival of diffuse
large cell lymphoma. A multivariate analysis including dose
intensity variables. Cancer 1990;66:1124–9.
69. Lee KW, Kim DY, Yun T, et al. Doxorubicin-based chemo-
therapy for diffuse large B-cell lymphoma in elderly patients:
comparison of treatment outcomes between young and eld-
erly patients and the significance of doxorubicin dosage.
Cancer 2003;98:2651–6.
70. Dixon D, Neilan B, Jones SE, et al. Effect of age on therapeu-
tic outcome in advanced diffuse histiocytic lymphoma: The
Southwest Oncology Group experience. J Clin Oncol 1986;
4:295–305.
71. Meyer RM, Browman GP, Samosh ML, et al. Randomized
phase II comparison of standard CHOP with weekly CHOP in eld-
erly patients with non-Hodgkin’s lymphoma. J Clin Oncol
1995;13:2386–93.
72. Bastion Y, Blay JY, Divine M, et al. Elderly patients with ag-
gressive non-Hodgkin’s lymphoma: disease presentation, re-
sponse to treatment, and survival—a Groupe d’Etude des
lymphomes de l’adulte study on 453 patients older than
69 years. J Clin Oncol 1997;15:2945–53.
73. International Agency for Research on Cancer (IARC). GLOBOCAN
2000: Cancer Incidence, Mortality, and Prevalence Worldwide
[CD ROM]. Lyon, France: IARC Press; 2001.
74. Obrand DI, Gordon PH. Incidence and patterns of recurrence
following curative resection for colorectal carcinoma. Dis
Colon Rectum 1997;40:15–24.
75. Andre T, Boni C, Mounedji–Boudiaf L, et al. Oxaliplatin, fluo-
rouracil, and leucovorin as adjuvant treatment for colon can-
cer. N Engl J Med 2004;350:2343–51.
76. Grossi F, Tiseo M. Granulocyte growth factors in the treat-
ment of non-small cell lung cancer (NSCLC). Crit Rev Oncol
Hematol 2006;58:221–30.
77. Timmer–Bonte JN, de Boo TM, Smit HJ, et al. Prevention of
chemotherapy-induced febrile neutropenia by prophylactic
antibiotics plus or minus granulocyte colony–stimulating fac-
tor in small-cell lung cancer: a Dutch randomized phase III
study. J Clin Oncol 2005;23:7974–84.
78. Martin–Christin F. Granulocyte colony stimulating factors:
how different are they? How to make a decision? Anticancer
Drugs 2001;12:185–91.
79. Kaushansky K. Lineage-specific hematopoietic growth fac-
tors. N Engl J Med 2006;354:2034–45.
80. Gutierrez–Delgado F, Bensinger W. Safety of granulocyte
colony-stimulating factor in normal donors. Curr Opin
PRACTICE GUIDELINE SERIES
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
22
Hematol 2001;8:155–60.
81. Murata M, Harada M, Kato S, et al. Peripheral blood stem cell
mobilization and apheresis: analysis of adverse events in 94
normal donors. Bone Marrow Transplant 1999;24:1065–71.
82. Liles W, Huang JE, Llewelyn C, Price TH, Dale DC. A com-
parative trial of granulocyte colony-stimulating factor (G-CSF)
and dexamethasone alone and in combination for the mobili-
zations of neutrophils in the peripheral blood of normal human
volunteers [abstract]. Blood 1995;86(suppl 1):609a.
83. Gudi R, Krishnamurthy M, Pachter BR. Astemizole in the treat-
ment of granulocyte colony–stimulating factor–induced bone
pain. Ann Intern Med 1995;123:236–7.
84. Falanga A, Marchetti M, Evangelista V, et al. Neutrophil acti-
vation and hemostatic changes in healthy donors receiving
granulocyte colony–stimulating factor. Blood 1999;93:
2506–14.
85. Papaldo P, Lopez M, Cortesi E, et al. Addition of either
lonidamine or granulocyte colony–stimulating factor does not
improve survival in early breast cancer patients treated with
high-dose epirubicin and cyclophosphamide. J Clin Oncol
2003;21:3462–8.
86. Papaldo P, Ferretti G, Di Cosimo S, et al. Does granulocyte
colony–stimulating factor worsen anemia in early breast can-
cer patients treated with epirubicin and cyclophosphamide?
J Clin Oncol 2006;24:3048–55.
87. Vose JM, Crump M, Lazarus H, et al. Randomized, multicenter,
open-label study of pegfilgrastim compared with daily
filgrastim after chemotherapy for lymphoma. J Clin Oncol
2003;21:514–19.
88. Mehregan DR, Fransway AF, Edmonson JH, Leiferman KM.
Cutaneous reactions to granulocyte–monocyte colony–stimu-
lating factor. Arch Dermatol 1992;128:1055–9.
89. Asnis LA, Gaspari AA. Cutaneous reactions to recombinant
cytokine therapy. J Am Acad Dermatol 1995;33:393–410.
90. Iki S, Yoshinaga K, Ohbayashi Y, Urabe A. Cytotoxic drug-
induced pneumonia and possible augmentation by G-CSF—clini-
cal attention. Ann Hematol 1993;66:217–18.
91. Couderc LJ, Stelianides S, Frachon I, et al. Pulmonary toxic-
ity of chemotherapy and G/GM-CSF: a report of five cases. Respir
Med 1999;93:65–8.
92. Tate RM, Repine JE. Neutrophils and the adult respiratory dis-
tress syndrome. Am Rev Respir Dis 1983;128:552–9.
93. Azoulay E, Attalah H, Harf A, Schlemmer B, Delclaux C.
Granulocyte colony–stimulating factor or neutrophil-induced
pulmonary toxicity: myth or reality? Systematic review of clini-
cal case reports and experimental data. Chest 2001;120:
1695–701.
94. Thirman MJ, Larson RA. Therapy-related myeloid leukemia.
Hematol Oncol Clin North Am 1996;10:293–320.
95. Smith RE, Bryant J, DeCillis A, Anderson S. Acute myeloid
leukemia and myelodysplastic syndrome after doxorubicin–
cyclophosphamide adjuvant therapy for operable breast can-
cer: the National Surgical Adjuvant Breast and Bowel Project
experience. J Clin Oncol 2003;21:1195–204.
96. Patt DA, Duan Z, Fang S, Hortobagyi GN, Giordano SH. Acute
myeloid leukemia after adjuvant breast cancer therapy in older
women: understanding risk. J Clin Oncol 2007;25:3871–6.
97. Brodsky RA, Bedi A, Jones RJ. Are growth factors leuke-
mogenic? Leukemia 1996;10:175–7.
98. Kaplinsky C, Trakhtenbrot L, Hardan I, et al. Tetraploid my-
eloid cells in donors of peripheral blood stem cells treated with
rhG-CSF. Bone Marrow Transplant 2003;32:31–4.
99. Hudis C, Citron M, Berry D, et al. Five year follow-up of
INT C9741: dose-dense (DD) chemotherapy (CRx) is safe and
effective [abstract 49]. Breast Cancer Res Treat 2005;94
(suppl 1):S20.
100. Crump M, Tu D, Shepherd L, Levine M, Bramwell V, Pritchard
K. Risk of acute leukemia following epirubicin-based adju-
vant chemotherapy: a report from the National Cancer Insti-
tute of Canada Clinical Trials Group. J Clin Oncol 2003;21:
3066–71.
101. Thatcher N, Girling DJ, Hopwood P, Sambrook RJ, Qian W,
Stephens RJ. Improving survival without reducing quality of
life in small-cell lung cancer patients by increasing the dose-
intensity of chemotherapy with granulocyte colony–stimulat-
ing factor support: results of a British Medical Research
Council Multicenter Randomized Trial. Medical Research
Council Lung Cancer Working Party. J Clin Oncol 2000;18:
395–404.
102. Le Cesne A, Judson I, Crowther D, et al. Randomized phase III
study comparing conventional-dose doxorubicin plus
ifosfamide versus high-dose doxorubicin plus ifosfamide plus
recombinant human granulocyte–macrophage colony–stimu-
lating factor in advanced soft tissue sarcomas: a trial of the
European Organization for Research and Treatment of Can-
cer/Soft Tissue and Bone Sarcoma Group. J Clin Oncol 2000;
18:2676–84.
103. Sternberg CN, de Mulder PH, Schornagel JH, et al. Random-
ized phase III trial of high-dose-intensity methotrexate, vin-
blastine, doxorubicin, and cisplatin (MVAC) chemotherapy and
recombinant human granulocyte colony-stimulating factor
versus classic MVAC in advanced urothelial tract tumors: Euro-
pean Organization for Research and Treatment of Cancer Pro-
tocol no. 30924. J Clin Oncol 2001;19:2638–46.
104. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid
leukemia or myelodysplastic syndrome following use of granu-
locyte colony-stimulating factors during breast cancer adju-
vant chemotherapy. J Natl Cancer Inst 2007;99:196–205.
105. Touw IP, Bontenbal M. Granulocyte colony-stimulating fac-
tor: key (f)actor or innocent bystander in the development of
secondary myeloid malignancy? J Natl Cancer Inst 2007;99:
183–6.
106. Le Deley MC, Suzan F, Cutuli B, et al. Anthracyclines,
mitoxantrone, radiotherapy, and granulocyte colony–stimulat-
ing factor: risk factors for leukemia and myelodysplastic syn-
drome after breast cancer. J Clin Oncol 2007;25:292–300.
107. Praga C, Bergh J, Bliss J, et al. Risk of acute myeloid leuke-
mia and myelodysplastic syndrome in trials of adjuvant
epirubicin for early breast cancer: correlation with doses of
epirubicin and cyclophosphamide. J Clin Oncol 2005;23:
4179–91.
108. Fisher B, Anderson S, DeCillis A, et al. Further evaluation of
intensified and increased total dose of cyclophosphamide for
the treatment of primary breast cancer: findings from National
Surgical Adjuvant Breast and Bowel Project B-25. J Clin Oncol
1999;17:3374–88.
109. Freedman MH, Bonilla MA, Fier C, et al. Myelodysplasia
syndrome and acute myeloid leukemia in patients with con-
KOUROUKIS et al.
23
CURRENT ONCOLOGYVOLUME 15, NUMBER 1
genital neutropenia receiving G-CSF therapy. Blood 2000;96:
429–36.
110. Tigue CC, McKoy JM, Evens AM, Trifilio SM, Tallman MS,
Bennett CL. Granulocyte-colony stimulating factor adminis-
tration to healthy individuals and persons with chronic neu-
tropenia or cancer: an overview of safety considerations from
the Research on Adverse Drug Events and Reports project.
Bone Marrow Transplant 2007;40:185–92.
111. Relling MV, Boyett JM, Blanco JG, et al. Granulocyte colony–
stimulating factor and the risk of secondary myeloid malig-
nancy after etoposide treatment. Blood 2003;101:3862–7.
112. Pedersen–Bjergaard J, Philip P. Two different classes of
therapy-related and de-novo acute myeloid leukemia? Cancer
Genet Cytogenet 1991;55:119–24.
Correspondence to: C. Tom Kouroukis, Juravinski
Cancer Centre, 699 Concession Street, Hamilton,
Ontario L8V 5C2.
E-mail: tom.kouroukis@hrcc.on.ca
* Juravinski Cancer Centre, Hamilton, ON.
BC Cancer Agency, Vancouver, BC.
Sunnybrook Health Sciences Centre–Odette Can-
cer Centre, Toronto, ON.
§Centre Hospitalier Affilie de Quebec–Hôpital du
St-Sacrement, Quebec City, QC.
|| Ottawa Hospital Regional Cancer Centre, Ottawa,
ON.
#Princess Margaret Hospital, Toronto, ON.
... It may lead to life-threatening infections requiring hospitalization, treatment with antibiotics, and reduction or delay of chemotherapy doses [1]. Risk factors for development of febrile neutropenia include intensive myelosuppressive chemotherapy regimens and patient clinical characteristics, such as advanced age, progressive stage of disease, prior episode of febrile neutropenia, and comorbidities [1,2]. According to evidence-based guidelines for the management of febrile neutropenia, granulocyte colony-stimulating factors (G-CSFs) are recommended for primary prophylaxis of febrile neutropenia in high-risk patients (defined as ≥ 20% risk of developing febrile neutropenia) [1][2][3][4][5][6][7]. ...
... Risk factors for development of febrile neutropenia include intensive myelosuppressive chemotherapy regimens and patient clinical characteristics, such as advanced age, progressive stage of disease, prior episode of febrile neutropenia, and comorbidities [1,2]. According to evidence-based guidelines for the management of febrile neutropenia, granulocyte colony-stimulating factors (G-CSFs) are recommended for primary prophylaxis of febrile neutropenia in high-risk patients (defined as ≥ 20% risk of developing febrile neutropenia) [1][2][3][4][5][6][7]. ...
Article
Full-text available
Purpose Lipegfilgrastim (Lonquex, Teva Pharma B.V.) is approved for reduction in neutropenia duration and febrile neutropenia incidence. In the framework of lipegfilgrastim regulatory approval in the EU, the Health Authorities requested a drug utilization study. This study was conducted to characterize prescribing patterns of lipegfilgrastim and quantify the extent of on- and off-label use of lipegfilgrastim in real-world setting in Europe. Methods Information on lipegfilgrastim use between January 2014 and March 2020 was abstracted from medical records in hospital and outpatient clinical settings. Indication for lipegfilgrastim was classified either as on-label or off-label use according to pre-determined criteria. The primary endpoint was the extent of lipegfilgrastim off-label use based on the most recent lipegfilgrastim cycle. Results Records of 481 patients were obtained from five European countries. Lipegfilgrastim was most commonly prescribed for prevention of neutropenia by oncologists and hematologists. Patients who were administered lipegfilgrastim were primarily ≥ 55 years old (65.1%) and female (65.7%). The most frequent underlying diagnosis was breast cancer (38.3%). For the most recent lipegfilgrastim cycle, on-label use was recorded in 452/459 patients with no missing data (98.5%), while off-label use was recorded in 7/459 patients (1.5%). The majority of off-label use was attributed to use with non-cytotoxic chemotherapy (57.1%). Off-label use of lipegfilgrastim across all treatment cycles with no missing data was 11/1547 cycles (0.7%). Conclusion Using real-world data, these findings confirm the low rate of lipegfilgrastim off-label use as reported in a preceding feasibility study, indicating very high adherence to the approved indication.
... Supportive Care Guidelines (Kouroukis et al., 2008) warrant the prophylactic use of G-CSF in patients when the risk of FN ≥ 20%, in patients having risk of FN < 20% and > 10% with other risk factors such as elderly age ≥ 65 years, previous history of FN and if chemotherapy dose reduction deemed pernicious to overall outcome. G-CSF dosage regimen has been documented with infrequent FN linked hospitalization, low length of hospital stay, decreased antibiotic use and mortality (Vanderpuye-Orgle et al., 2016). ...
... growth factor such as G-CSF. According to several guidelines [34][35][36][37], G-CSF may also be used as primary Fig. 3 Distribution of the absolute error for scenarios where the ANC was monitored until day 3, 4, 5, 6, 7, 10, 15 and 19. ANC ipred.,d,D is the individual predicted ANC at day D, given data available up to day d and ANC true,D is the true ANC at day D. Orange, blue and green boxes represent monitoring frequency every, every other and every third day. ...
Article
Full-text available
Purpose: To investigate whether a more frequent monitoring of the absolute neutrophil counts (ANC) during myelosuppressive chemotherapy, together with model-based predictions, can improve therapy management, compared to the limited clinical monitoring typically applied today. Methods: Daily ANC in chemotherapy-treated cancer patients were simulated from a previously published population model describing docetaxel-induced myelosuppression. The simulated values were used to generate predictions of the individual ANC time-courses, given the myelosuppression model. The accuracy of the predicted ANC was evaluated under a range of conditions with reduced amount of ANC measurements. Results: The predictions were most accurate when more data were available for generating the predictions and when making short forecasts. The inaccuracy of ANC predictions was highest around nadir, although a high sensitivity (≥90%) was demonstrated to forecast Grade 4 neutropenia before it occurred. The time for a patient to recover to baseline could be well forecasted 6 days (±1 day) before the typical value occurred on day 17. Conclusions: Daily monitoring of the ANC, together with model-based predictions, could improve anticancer drug treatment by identifying patients at risk for severe neutropenia and predicting when the next cycle could be initiated.
... In contrast, 34% of BRCA1 mutation carriers experienced febrile neutropenia, a significantly higher frequency than observed among BRCA2 mutation carriers (0%; p < 0.0001; Figure 1G). The BRCA1 and BRCA2 mutation carriers were relatively young patients (median age 41.5 and 46, respectively; Table 1), without co-morbidities and in whom febrile neutropenia would not be expected (Kouroukis et al., 2008). Consistent with the increased incidence of febrile neutropenia, we observed a trend toward an increased incidence of neutropenia among BRCA1 mutation carriers as compared to BRCA2 mutation carriers ( Figure 1I; p < 0.1). ...
Article
Full-text available
BRCA1 is a well-known DNA repair pathway component and a tissue-specific tumor suppressor. However, its role in hematopoiesis is uncertain. Here, we report that a cohort of patients heterozygous for BRCA1 mutations experienced more hematopoietic toxicity from chemotherapy than those with BRCA2 mutations. To test whether this reflects a requirement for BRCA1 in hematopoiesis, we generated mice with Brca1 mutations in hematopoietic cells. Mice homozygous for a null Brca1 mutation in the embryonic hematopoietic system (Vav1-iCre;Brca1F22–24/F22–24) developed hematopoietic defects in early adulthood that included reduced hematopoietic stem cells (HSCs). Although mice homozygous for a huBRCA1 knockin allele (Brca1BRCA1/BRCA1) were normal, mice with a mutant huBRCA1/5382insC allele and a null allele (Mx1-Cre;Brca1F22–24/5382insC) had severe hematopoietic defects marked by a complete loss of hematopoietic stem and progenitor cells. Our data show that Brca1 is necessary for HSC maintenance and normal hematopoiesis and that distinct mutations lead to different degrees of hematopoietic dysfunction.
... Neutropenia increases the risk of developing serious or life-threatening infections, and this risk predictably increases with increasing duration and severity of neutropenia [1,2]. Treatment with recombinant granulocyte colony-stimulating factors (G-CSFs) stimulates neutrophil production and reliably reduces the severity and duration of chemotherapy-induced neutropenia and febrile neutropenia [3][4][5][6][7][8][9][10]. Current treatment guidelines from the American Society of Clinical Oncology [11], the National Comprehensive Cancer Network [12], the European Organisation for Research and Treatment of Cancer [13], the European Society for Medical Oncology [14], and Canadian supportive care guidelines [15] recommend the prophylactic use of G-CSFs in patients receiving chemotherapy when the risk of febrile neutropenia is 20 % or higher. ...
Article
Full-text available
The recombinant human granulocyte colony-stimulating factor (G-CSF) known as filgrastim (Tevagrastim®, Ratiograstim®, Biograstim®) in Europe (approved in 2008) and tbo-filgrastim (Granix®) in the USA (approved in 2012; Teva Pharmaceutical Industries Ltd., Petach Tikva, Israel) is indicated to reduce the duration of severe neutropenia in patients with non-myeloid malignancies receiving myelosuppressive anti-cancer drugs associated with a clinically significant incidence of febrile neutropenia. This article presents pooled clinical data for tbo-filgrastim compared with Neupogen® (Amgen, Thousand Oaks, CA, USA) as well as tbo-filgrastim post-marketing safety data. The safety and efficacy of tbo-filgrastim were evaluated in three phase III studies in 677 patients receiving myelosuppressive chemotherapy and study drug (348 patients with breast cancer, 237 with lung cancer, 92 with non-Hodgkin lymphoma). In each study, the efficacy of tbo-filgrastim was similar to that of Neupogen. Overall, 633 (93.5 %) patients receiving the study drug experienced 6093 treatment-emergent adverse events (AEs), most of which were related to chemotherapy. Adverse events related to the study drug (tbo-filgrastim or Neupogen) were experienced by 185 (27.3 %) patients; 19 (2.8 %) had severe drug-related AEs, 5 (0.7 %) had drug-related serious AEs, and 6 (0.9 %) discontinued the study due to drug-related AEs. Overall, the most common drug-related AEs were bone pain (7.1 %), myalgia (4.0 %), and asthenia (4.4 %). The post-marketing safety profile of tbo-filgrastim was consistent with that observed during the clinical studies. The availability of tbo-filgrastim, a G-CSF with safety and efficacy comparable to those of Neupogen, provides physicians with an alternative treatment option for supportive care of patients with non-myeloid malignancies receiving myelosuppressive chemotherapy.
... Consequently, filgrastim or pegfilgrastim was used in 31.1% of the patients treated with bendamustine, similar to their rate of use (38%) in the study by Kahl et al. 24 . Cancer patients with severe myelosuppression during the first cycle of chemotherapy can be candidates for growth-factor support in subsequent cycles, when maintenance of dose intensity is important 36,40 . However, it should be noted that, in many provinces, granulocyte colony-stimulating factors cannot be accessed for noncurative therapies such as bendamustine, leaving dose reductions and delays as the only options for managing neutropenia in those provinces. ...
Article
Full-text available
Bendamustine is a bifunctional alkylating agent with unique properties that distinguish it from other agents in its class. Bendamustine is used as monotherapy or in combination with other agents to treat patients with non-Hodgkin lymphoma (nhl) and chronic lymphocytic leukemia (cll). The prospective interventional open-label bend-act trial evaluated bendamustine in patients with rituximab-refractory indolent nhl (inhl) and previously untreated cll. Study objectives were to assess the safety and tolerability of bendamustine monotherapy and to provide patients with access to bendamustine before Health Canada approval. The study aimed to enrol up to 100 patients. All patients with inhl received an intravenous dose of bendamustine 120 mg/m(2) over 60 minutes on days 1 and 2 for up to eight 21- or 28-day treatment cycles. All patients with cll received an intravenous dose of bendamustine 100 mg/m(2) over 30 minutes on days 1 and 2 for up to six 28-day treatment cycles. Of 90 patients treated on study (16 with cll and 74 with inhl), 35 completed the study (4 with cll and 31 with inhl). The most common treatment-emergent adverse events (teaes) were nausea (70%), fatigue (57%), vomiting (40%), and diarrhea (33%)-mostly grades 1 and 2. Ondansetron was the most common supportive medication used in the patients (63.5% of those with inhl and 68.8% of those with cll). Neutropenia (32%), anemia (23%), and thrombocytopenia (21%) were the most frequent hematologic teaes, with neutropenia being the most common grade 3 or 4 teae leading to dose modification. Dose delays occurred in 28 patients (31.3%) because of grade 3 or 4 teaes, with a higher incidence of dose delays being observed in inhl patients on the 21-day treatment cycle than in those on the 28-day treatment cycle (50.0% vs. 24.1%). During the study, 33 patients (36.7%) experienced at least 1 serious adverse event, and 4 deaths were reported (all in patients with inhl). The type and frequency of the teaes reported accorded with observations in earlier clinical trials and post-marketing experiences, thus confirming the acceptable and manageable safety profile of bendamustine.
Article
Background: Granulocyte colony-stimulating factors (G-CSFs) are widely used to prevent neutropenia in cancer patients undergoing myelosuppressive chemotherapy. Several biosimilar medicines of G-CSF are now available, with their development involving a step-wise series of comparisons to demonstrate similarity to reference biologics. Randomised clinical trials (RCTs) are considered confirmatory, and for G-CSF biosimilars, patients with breast cancer (BC) undergoing myelosuppressive chemotherapy are the most sensitive population in which to confirm similarity. This meta-analysis aimed to compare the clinical efficacy and safety of approved or proposed G-CSF biosimilars (filgrastim or pegfilgrastim) with reference G-CSF in patients with BC. Methods: A Medline literature search up to March 2017 identified RCTs comparing biosimilar G-CSF to reference in BC patients. The primary efficacy end-point was mean difference in duration of severe neutropenia (DSN). Secondary efficacy end-points were differences in depth of absolute neutrophil count (ANC) nadir, time to ANC recovery and incidence of febrile neutropenia. Safety analyses included calculation of risk ratios for bone pain events, myalgia events and serious adverse events. Random effect models were fitted to obtain the pooled estimates of the mean difference for continuous outcomes and the risk ratio for dichotomous outcomes and their corresponding 95% confidence intervals (CIs). Findings: Eight eligible RCTs were included in this meta-analysis. Overall difference in DSN between reference and biosimilar medicines was not statistically significant (0·06 d [95% CI -0·05, 0·17]). The analysis of secondary efficacy end-points showed no significant differences between reference biologics and biosimilar medicines, as well as the analysis of bone pain events, myalgia events and serious adverse events.
Article
Full-text available
A lo largo de las dos últimas décadas se han logrado notables avances en el tratamiento del paciente hematooncológico. Sin duda, uno de los más notables ha sido la reducción de la morbimortalidad por complicaciones infecciosas, así como la reducción del período de neutropenia, gracias al empleo de factores de crecimiento hematopoyéticos. A pesar de ello, la neutropenia febril (NF) es una consecuencia severa del uso de quimioterapia mielosupresora, que usualmente deriva en hospitalizaciones y en la necesidad de administrar antibióticos intravenosos, utilizando recursos importantes para el sistema de salud. La NF también está asociada a reducciones de dosis y retrasos o suspensión en la aplicación de quimioterapia, lo que puede afectar el resultado final del tratamiento. Entonces, es de suma importancia identificar a los pacientes que tienen riesgo elevado de desarrollar NF, para que puedan recibir una quimioterapia óptima y su riesgo de NF sea tratado de manera apropiada, así como que los pacientes que se presenten con NF reciban un diagnóstico y tratamiento oportunos.
Article
Full-text available
The objective of this study was to assess the impact of the primary prophylaxis of granulocyte colony-stimulating factor (G-CSF) in the management of childhood B-cell non-Hodgkin lymphoma (B-NHL). Patients with advanced-stage mature B-NHL were randomized to receive prophylactic G-CSF (G-CSF+) or not receive G-CSF (G-CSF-) based on protocols of the B-NHL03 study. The G-CSF group received 5 μg/kg/d Lenograstim from day 2 after each course of six chemotherapy courses. Fifty-eight patients were assessable, 29 G-CSF + and 29 G-CSF-. G-CSF + patients showed a positive impact on the meantime to neutrophil recovery and hospital stay. On the other hand, they had no impact in the incidences of febrile neutropenia, serious infections, stomatitis and total cost. Our study showed that administration of prophylactic G-CSF through all six chemotherapy courses for childhood B-NHL showed no clinical and economic benefits for the management of childhood B-NHL treatment.
Article
Background: Chemotherapy-induced severe neutropenia (SN) is the most common dose-limiting toxicity of chemotherapy (CT), its development can complicate the patients evolution. Additionally, SN forces us to reduce CT dosage or to delay its application. Pegfilgrastim (PgF) is a granulocyte colony-stimulating factor bound to a polyethylene glycol molecule. Objectives: To evaluate the efficacy and safety of PgF as primary prophylaxis of CT-induced SN. Patients and Methods: A retrospective study from October 2006 to June 2008. The Study included 28 patients > 18 years old with confirmed tumors (mean age 53.5 yrs, 22 of them women [76.8])), that were treated with CT and received 6 mg PgF fixed-dose, subcutaneously 24 hours after having finished each cycle. Complete blood count (CBC) performed on day one and nadir of each cycle. Age, gender, site of neoplasm, histology, stage, functional state (ECOG), number and types of risk factors (RF) for developing SN and FN, incidence of leucopenia and neutropenia G3-4, scheme and number of CT cycles, number of PgF applications, adverse events related to PgF, delays or dosage reductions were recorded. Results: Patients received 116 cycles and 116 PgF applications. AC and TAC were the most common treatment combinations. Main RF was feminine gender in 22 patients (78.6%). We recovered 95.7% (111/116) and 77.6% (90/116) basal CBCs and nadir, respectively. In basal counts, leucopenia or neutropenia G 3-4 were not detected, whereas at the nadir 3 cycles with leucopenia G3 and one G4 were observed; in addition, neutropenia G4 in four cycles (4.4%). There was a decrease of dosage in one patient (3,5%), delay in 9 cycles (8.1%), antibiotic use in 2 cases (7,1%), infection in one (3,5%), transfusions in three patients (10,7%) and symptoms related to PgF in one case (3,5%). Conclusions: Pegfilgrastim is effective as primary prophylaxis. The risk of chemotherapy induced severe neutropenia should be evaluated for all patients undergo this treatment modality.
Article
Full-text available
Granulocyte colony-stimulating factor (G-CSF) enhances neutrophil functions in vitro and in vivo. It is known that neutrophil-derived products can alter the hemostatic balance. To understand whether polymorphonuclear leukocyte (PMN) activation, measured as PMN degranulation and phenotypical change, may be associated to hemostatic alterations in vivo, we have studied the effect of recombinant human G-CSF (rHuG-CSF) administration on leukocyte parameters and hemostatic variables in healthy donors of hematopoietic progenitor cells (HPCs). Twenty-six consecutive healthy donors receiving 10 μg/kg/d rHuG-CSF subcutaneously for 5 to 7 days to mobilize HPCs for allogeneic transplants were included in the study. All of them responded to rHuG-CSF with a significant white blood cell count increase. Blood samples were drawn before therapy on days 2 and 5 and 1 week after stopping rHuG-CSF treatment. The following parameters were evaluated: (1) PMN activation parameters, ie, surface CD11b/CD18 antigen expression, plasma elastase antigen levels and cellular elastase activity; (2) plasma markers of endothelium activation, ie, thrombomodulin (TM) and von Willebrand factor (vWF) antigens; (3) plasma markers of blood coagulation activation, ie, F1+2, TAT complex, D-dimer; and (4) mononuclear cell (MNC) procoagulant activity (PCA) expression. The results show that, after starting rHuG-CSF, an in vivo PMN activation occurred, as demonstrated by the significant increment of surface CD11b/CD18 and plasma elastase antigen levels. Moreover, PMN cellular elastase activity, which was significantly increased at 1 day of treatment, returned to baseline at day 5 to 6, in correspondence with the elastase antigen peak in the circulation. This change was accompanied by a parallel significant increase in plasma levels of the two endothelial and the three coagulation markers. The PCA generated in vitro by unstimulated MNC isolated from rHuG-CSF–treated subjects was not different from that of control cells from untreated subjects. However, endotoxin-stimulated MNC isolated from on-treatment individuals produced significantly more PCA compared with both baseline and control samples. All of the parameters were decreased or normal 1 week after stopping treatment. These data show that rHuG-CSF induces PMN activation and transiently affects some hemostatic variables in healthy HPC donor subjects. The clinical significance of these findings remains to be established.
Article
Interval reduction from 3 (CHOP-21) to 2 weeks (CHOP-14; Pfreundschuh et al., Blood, 2004) and the addition of rituximab to CHOP-21 (R-CHOP-21; Coiffier et al., NEJM, 2002) improved outcome in elderly patients with DLBCL to a similar extent without increasing toxicity compared to CHOP-21. In the RICOVER-60 trial, elderly patients (61–80 years, stages I–IV) were randomized to receive 6 or 8 cycles of CHOP-14 with or without 8 applications of rituximab given on days 1, 15, 29, 43, 57, 71, 85, and 99. Radiotherapy was planned to sites of initial bulk and/or extranodal involvement. The primary endpoint was freedom from treatment failure (FFTF) with events defined as additional therapy, failure to achieve complete remission, progressive disease, relapse, or death. The trial was powered to show a 9% difference in FFTF rate after 3 years. Between 07/2000 and 06/2005, 1330 patients were recruited. A planned interim analysis was performed on 828 evaluable patients with CD20+ DLBCL (median age 68 years; IPI=1: 32%, IPI=2: 29%; IPI=3: 23%; IPI=4,5: 16%). As by intention to treat, there was no difference in FFTF between 6 (n=414) and 8 (n=413) cycles (p=0.23), but FFTF after R-CHOP-14 (n=414) was significantly better than after CHOP-14 (n=413) alone (p=0.000025). As the empirical p-value of the log rank test statistics for FFTF was considerably lower than the critical value for the interim analysis (pcrit=0.031), the formal criterion for stopping the trial according to the alpha spending function (O’ Brien and Fleming boundary) was met and the trial was stopped on June 17, 2005 with 50/1330 patients still under therapy. After a median observation time of 26 months, there was a trend for a better FFTF after 8 cycles of CHOP-14 (n=210) compared to 6 cycles (n=203; 58% vs. 53%; p=0.13), but this trend was neutralized after the addition of rituximab: 70% FFTF for both 6 (n=211) and 8 cycles R-CHOP-14 (n=203). The advantage of R-CHOP-14 over CHOP-14 with respect to overall survival after 26 months is not yet significant (74% vs. 78%; p=0.13). Excluding patients with stage I, the RICOVER-60 population is very similar to the one included in the GELA 98.5 trial; however, the projected 2.5-year survival rate for elderly stage II–IV patients after 6 x R-CHOP-14 (74%) compares favorably with 8 x R-CHOP-21 in the GELA trial (64%; Feugier et al. JCO 2005). The superiority of 6 x R-CHOP-14 over 8 x R-CHOP-21 is mostly due to the better 2.5-year survival of poor-prognosis patients (IPI=3,4,5: 64% in the RICOVER trial vs. 54% in the GELA trial). In conclusion, the results observed with 6 cycles of R-CHOP-14 in this largest randomized trial of DLBLC performed to date are the best ever reported for elderly patients with DLBCL. 6 x R-CHOP-14 should be considered as reference standard in future trials for elderly patients with DLBCL.
Article
High-risk Non-Hodgkin lymphoma (NHL) in elderly patients (> 60 yrs) is associated with a relatively low complete response (CR) rate and a poor overall survival (OS). Previous studies have shown that addition of Rituximab to standard CHOP chemotherapy may improve CR and/or OS. A similar improvement has been shown for intensification of CHOP from 21 day intervals to 14 day intervals. The Dutch HOVON group and the Nordic lymphoma group have performed a multi-center, randomized phase III trial to compare 8 cycles of intensified CHOP (CHOP14) with the same regimen plus 6 administrations of Rituximab in previously untreated elderly patients with intermediate or high-risk NHL. Inclusion criteria were diffuse large B-cell lymphoma, mantle cell lymphoma or follicular lymphoma grade III; intermediate or high-risk NHL according to age adjusted IPI score; CD20-positive NHL; age 65 yrs or higher. The target number was 400 patients to be accrued in 5 years based on an expected increase in event-free survival with hazard ratio HR=0.70. Aplanned interim analysis was performed after inclusion of 250 patients, restricted to 211 patients included from 1st September 2001 to 1st October 2004. Forty patients had to be excluded because of lack of treatment and evaluation data, leaving 171 patients for the analysis. The median time off protocol treatment was 119 days (range 4–468 days). The median age was 73 years (range 62–88 years). There was no difference between the two treatment arms regarding histology, age, WHO classification of NHL, age-adjusted IPI score, Ann Arbor stage, WHO performance status and serum LDH. There was no significant difference of toxicity or CR between the two treatment arms. However, a highly significant difference was observed for event-free survival and overall survival in favor of the treatment arm with CHOP plus Rituximab. Based on this interim analysis, the randomization was halted and patients in the control arm were further treated with CHOP plus Rituximab. Since the outcome of the interim analysis could be biased due to incomplete data, a final decision to stop the trial awaits a new analysis which is planned for 26th August 2005 after collection of missing or incomplete data. If the new analysis confirms the results of the interim analysis, i.e. that Rituximab significantly improves the outcome of treatment in elderly patients with intermediate or high-risk NHL, even when an intensified CHOP regimen is used, the study will be closed and outcomes will be presented.
Article
PURPOSE: This multicenter, randomized, double-blind, active-control study was designed to determine whether a single subcutaneous injection of pegfilgrastim (SD/01, sustained-duration filgrastim; 100 μg/kg) is as safe and effective as daily filgrastim (5 μg/kg/d) for reducing neutropenia in patients who received four cycles of myelosuppressive chemotherapy. PATIENTS AND METHODS: Sixty-two centers enrolled 310 patients who received chemotherapy with docetaxel 75 mg/m² and doxorubicin 60 mg/m² on day 1 of each cycle for a maximum of four cycles. Patients were randomized to receive on day 2 either a single subcutaneous injection of pegfilgrastim 100 μg/kg per chemotherapy cycle (154 patients) or daily subcutaneous injections of filgrastim 5 μg/kg/d (156 patients). Absolute neutrophil count (ANC), duration of grade 4 neutropenia, and safety parameters were monitored. RESULTS: One dose of pegfilgrastim per chemotherapy cycle was comparable to daily subcutaneous injections of filgrastim with regard to all efficacy end points, including the duration of severe neutropenia and the depth of ANC nadir in all cycles. Febrile neutropenia across all cycles occurred less often in patients who received pegfilgrastim. The difference in the mean duration of severe neutropenia between the pegfilgrastim and filgrastim treatment groups was less than 1 day. Pegfilgrastim was safe and well tolerated, and it was similar to filgrastim. Adverse event profiles in the pegfilgrastim and filgrastim groups were similar. CONCLUSION: A single injection of pegfilgrastim 100 μg/kg per cycle was as safe and effective as daily injections of filgrastim 5 μg/kg/d in reducing neutropenia and its complications in patients who received four cycles of doxorubicin 60 mg/m² and docetaxel 75 mg/m².
Article
Purpose: To determine the relative efficacy of an intensive cyclophosphamide, epirubicin, and fluorouracil (CEF) adjuvant chemotherapy regimen compared with cyclophosphamide, methotrexate, and fluorouracil (CMF) in node-positive breast cancer. Patients and methods: Premenopausal women with node-positive breast cancer were randomly allocated to receive either cyclophosphamide 100 mg/m2 orally days 1 through 14; methotrexate 40 mg/m2 intravenously (i.v.) days 1 and 8; and fluorouracil 600 mg/m2 i.v. days 1 and 8 or cyclophosphomide 75 mg/m2 orally days 1 through 14; epirubicin 60 mg/m2 i.v. days 1 and 8; and fluorouracil 500 mg/m2 i.v. days 1 and 8. Each cycle was administered monthly for 6 months. Patients administered CEF received antibiotic prophylaxis with cotrimoxazole two tablets twice a day for the duration of chemotherapy. Results: The median follow-up was 59 months. One hundred sixty-nine of the 359 CMF patients developed recurrence compared with 132 of the 351 CEF patients. The corresponding 5-year relapse-free survival rates were 53% and 63%, respectively (P = .009). One hundred seven CMF patients died compared with 85 CEF patients. The corresponding 5-year actuarial survival rates were 70% and 77%, respectively (P = .03). The rate of hospitalization for febrile neutropenia was 1.1% in the CMF group compared with 8.5% in the CEF group. There was one case of congestive heart failure in a patient who received CMF compared with none in the CEF group. Acute leukemia occurred in five patients in the CEF group. Conclusion: The results of this trial show the superiority of CEF over CMF in terms of both disease-free and overall survival in premenopausal women with axillary node-positive breast cancer.
Article
Neutrophil transfusion therapy has not been widely utilized clinically due to the low yield of functional neutrophils from healthy donors. In recent years, this obstacle to effective neutrophil transfusion therapy has been partially overcome by the administration of G-CSF to donors in an attempt to augment the yield of neutrophils obtained by leukapheresis. Optimal conditions for the mobilization of neutrophils in the peripheral blood of potential granulocyte donors was defined in our study, which compared the relative effects of G-CSF and dexamethasone alone and in combination for this purpose. Five subjects (age: 20-42 yrs.; wt: 60-78 kg) were enrolled. Each received the following single-dose regimens on 5 consecutive weeks: 1) G-CSF: 300 μg SC; 2) G-CSF: 600 pμg SC; 3) dexamethasone: 8 mg PO; 4) G-CSF: 300 MS SC + dexamethasone: 8 mg PO; and 5) G-CSF: 600 μg SC + dexamethasone: 6 mg PO. Venous blood was collected at 0, 6,12, and 24 hours following administration of drugs for determination of WBC, leukocyte differential, and absolute neutrophil count (ANC). Maximal ANC was achieved at 12 hours following each of the regimens studied except the regimen of dexamethasone alone (max: 24 hours). The addition of dexamethasone significantly increased the maximal ANC induced by either 300 μg or 600 μg of G-CSF alone (p<.05). The greatest mobilization of neutrophils occurred at 12 hours following the administration of G-CSF (600 μg) and dexamethasone (8 mg}, where the ANC increased 12-fold, from a mean baseline value of 3594/μL to 43, 017/μL However, the maximal ANC induced by the combination of 600 μg G-CSF and dexamethasone was not statistically different (within the limits of our study) from the combination of 300 μg G-CSF and dexamethasone (p>.05). There were no significant differences between the five treatment groups in regards to the severity and frequency of side effects (p<.05). Thus, dexamethasone significantly increases the level of neutrophilta induced in normal subjects by G-CSF. We conclude that the combination of dexamethasone and G-CSF (at the dosages used in this study) is a convenient and well-tolerated regimen that should be employed for optimal mobilization of neutrophils in the peripheral blood of granulocyte donors. Moreover, the optimal quantitative yield of neutrophils is likely to be achieved by leukapheresis 12 hours following drug administration.