You are on page 1of 9

Noncardiogenic Pulmonary Edema: An Unusual and

Serious Complication of Anticancer Therapy


EVANGELOS BRIASOULIS, NICHOLAS PAVLIDIS
Department of Medical Oncology, University of Ioannina, Ioannina, Greece
Key Words. Chemotherapy Interleukin-2 Pulmonary toxicity NCPE ARDS Noncardiogenic pulmonary edema Acute lung injury
Cytarabine Gemcitabine All-trans retinoic acid Arsenic trioxide (As203).

A BSTRACT
Noncardiogenic pulmonary edema (NCPE) is a rare
and less well-recognizable pulmonotoxic syndrome of
anticancer therapy than pneumonitis/fibrosis. NCPE is
a clinical syndrome characterized by simultaneous presence of severe hypoxemia, bilateral alveolar infiltrates
on chest radiograph, and no evidence of left atrial
hypertension/congestive heart failure. The diagnosis of
drug-related NCPE relies upon documented exclusion
of any infectious, metabolic, or cancer-related causes.
The time proximity to therapy with drugs that are
known to precipitate NCPE, any preceding episodes of
flu-like symptoms during previous chemotherapy
courses and possible response to corticosteroids may
further support such a diagnosis. Cancer therapeutic
agents clearly associated with NCPE are cytarabine,

gemcitabine, and interleukin-2, as well as all-trans


retinoic acid in acute promyelocytic leukemia patients,
while a few other compounds have rarely or occasionally
been implicated. The pathophysiology of lung injury in
drug-induced NCPE remains unclear. There are indications suggesting that both a direct cytotoxic insult to the
lung epithelial cells and induction of a cytokine-triggered inflammatory response may be involved in its
pathogenesis. By distinction to drug-induced pulmonary
pneumonitis that may lead to permanent pulmonary
fibrosis, NCPE if not fatal, can be reversed upon prompt
recognition, following immediate discontinuation of the
offensive drug and start of intensive supportive treatment and intravenous corticosteroids. The Oncologist
2001;6:153-161

INTRODUCTION
Several cancer therapeutic drugs are known to induce
pulmonary damage, which may result in a variety of clinicopathologic syndromes with minor to severe clinical consequences [1]. Clinical syndromes associated with drug-induced
pulmonary toxicity include pneumonitis/fibrosis, hypersensitivity lung disease, and noncardiogenic pulmonary edema
(NCPE)/acute respiratory distress syndrome (ARDS). These
syndromes share a similar symptomatology but differ in regard
to the time-relation to cancer treatment, the radiographic findings, the duration of pulmonary damage, and the long-term
outcome [2]. Non-specific symptomatology of cough, progressive dyspnea, and often a low-grade fever alert clinicians
confronted with the diagnostic challenge to recognize subclinical syndromes and differentiate fully developed drug-induced
pulmonary reactions from lung injuries caused by infectious,
cardiac, and neoplastic causes.

The most common chemotherapy-associated lung


injury is drug-induced pneumonitis. This type of pulmonary
toxicity has extensively been studied and attributed to a
majority of the pulmonotoxic antineoplastic agents, of
which bleomycin is the prototype and nitrosoureas rank
second [3-7]. Major concern with this pulmonary toxicity is
the potential to progress to irreversible chronic pulmonary
fibrosis.
Noncardiogenic pulmonary edema is a rarely cited and
less well-recognizable pulmonotoxic syndrome of anticancer therapy. Our recent experience with NCPE that
complicated gemcitabine plus docetaxel combination
chemotherapy [8] prompted us to investigate existing medical literature in regard to this unusual pulmonary toxicity
of anticancer therapy. To our knowledge, this is the first
review on anticancer-therapy-associated noncardiogenic
pulmonary edema.

Correspondence: Evangelos Briasoulis, M.D., Medical Oncology Department, Ioannina University, Ioannina, 45110, Greece.
Telephone and Fax: 30-651-99394; e-mail: ebriasou@otenet.gr. Received August 15, 2000; accepted for publication
November 30, 2000. AlphaMed Press 1083-7159/2001/$5.00/0

The Oncologist 2001;6:153-161 www.TheOncologist.com

Briasoulis, Pavlidis
METHODS
A computerized Internet PubMed search of English and
non-English language medical literature was conducted by
using Reference Manager Version 9.5 (ISI ResearchSoft;
PA; http://www.risinc.com/RShome.html) for the period
1965 to 2000, and manually for the period 1960 to 1965.
For our search, we used as all-field parameters the generic
names of pharmacologic anticancer agents, the words
chemotherapy, immunotherapy, and biotherapy, and the
key phrases anticancer treatment and drug-induced, combined with the words or key phrases noncardiogenic or primary pulmonary edema, NCPE, pulmonary toxicity,
pulmonary disease, respiratory distress syndrome, and
acute lung injury.
Reports of drug-induced pulmonary disease related to
anticancer treatment had to fit the most accepted definitions
for NCPE and ARDS in order to be considered for this
review [9-11]. NCPE, also termed as Acute Lung Injury
(ALI), is a clinical syndrome characterized by simultaneous
presence of severe hypoxemia, bilateral alveolar infiltrates
on chest radiograph, and no evidence of left atrial hypertension. The term ARDS is applied to the most severe cases
of acute respiratory failure. According to the criteria set by
the American-European Consensus Conference on ARDS,
a 200 mmHg value of quotient PaO2/FIO2 (arterial oxygen-tension/fractional inspired-oxygen) defines ARDS,
while 300 mmHg characterizes ALI.
ANTICANCER THERAPY-INDUCED NONCARDIOGENIC
PULMONARY EDEMA
Clinical PresentationDiagnosis
NCPE is a rare but major complication of anticancer
therapy. This typically presents as a sub-acute clinical syndrome characterized by severe dyspnea, cough, tachypnea,
fatigue, and a low-grade fever that occurs shortly after or
within a few days following the administration of the offensive pharmaceutical agent or agents [8, 12, 13]. Patients
with drug-induced NCPE are usually markedly hypoxemic,
with crackles on auscultation and chest radiography showing confluent alveolar consolidations almost identical to
those seen in congestive heart failure, and a normal-size
heart [14].
Diagnosing drug-induced NCPE is actually an exercise
of exclusion, as there is no diagnostic test available.
Diagnosis relies upon typical radiograph findings [15] and
the documented exclusion of heart dysfunction and any
infectious, metabolic, or cancer-related causes. The time
proximity to administration of drugs known to precipitate
NCPE, any preceding episodes of flu-like symptoms during
previous chemotherapy courses, and possible response to

154

corticosteroids may further support such a diagnosis. By distinction from drug-induced pulmonary pneumonitis that
potentially may evolve to pulmonary fibrosis, NCPE if not
fatal, can be fully reversed upon prompt recognition, following discontinuation of offensive drugs and immediate start of
intensive support treatment and intravenous corticosteroids.
Cancer therapeutic agents that clearly precipitate NCPE
are cytarabine, gemcitabine, and interleukin 2 (IL-2), and the
all-trans retinoic acid and arsenic trioxide (As2O3) in acute
promyelocytic leukemia patients, while a few more compounds have rarely or occasionally been implicated (Table 1).
Biotherapeutic Agents
Among biological agents, recombinant IL-2 (rIL-2) has
been clearly associated with the induction of NCPE toxicity,
while there is some weak evidence for a similar pulmonotoxic
potential of white blood cell growth-stimulating factors.
IL-2, a glycoprotein naturally produced by human T cells
during immune response, was introduced in cancer therapeutics 15 years ago as a novel approach to the treatment of solid
tumors [16, 17]. This biological response modifier is currently employed in the management of metastatic melanoma
and renal cell carcinoma, alone or in combination with
chemotherapy or biotherapy [18-21].
Unfortunately, the clinical development of rIL-2 has been
compromised by frequently associated toxicities and a narrow
therapeutic index [22-24]. Regarding pulmonary toxicity, rIL2 given intravenously is commonly complicated by NCPE.
This occurs in the context of a vascular leak syndrome, the
most frequent complication of IL-2, and is mediated via a
mechanism of increased microvascular permeability [25, 26].
It is now clear that rIL-2-induced pulmonary toxicity is a doserelated phenomenon that tends to resolve within a few days
upon treatment discontinuation [27].
Initial clinical testing identified malaise and weight gain
as the dose-limiting toxicities of systemic administration of
rIL-2 [28], whereas further clinical experience revealed a significant incidence of NCPE in patients treated with high-dose
intravenous rIL-2 [29]. The incidence rate as recorded in the
first large cohorts of treated patients ranged from 10%-20%,
with a considerable number of treated patients requiring intubation [27, 30, 31]. Interestingly, newest clinical data show a
decline in rIL-2-related pulmonary toxicity, obviously due to
improvement in key safety issues such as eligibility screening
and optimization of therapeutic conditions. Kammula et al.
reviewed safety data of high-dose bolus rIL-2 (720,000 IU/kg
every 8 h) administered in 1,241 cancer patients over a 12year period and found a clear improvement in rIL-2 safety
profile with a remarkable drop from 12% to 3% in the incidence of NCPE [32]. The feasibility and the safety of longterm administration of subcutaneous rIL-2 at conventional

Cancer-Therapy-Related Noncardiogenic Pulmonary Edema

155

Table 1. Cancer therapeutic agents associated with the development of noncardiogenic pulmonary edema
Therapy/agent

Reportedestimated
incidence

Cytarabine
(high dose)

high

Characteristics/comments

Ref.

Potentially fatal toxicity described in leukemia patients treated with moderate to high
[47-49, 104]
cytarabine doses (1-1.5 g/m2/continuous infusion, or >3 g/m2 as 2 h IV infusion per 12 h)
over 3 to 4 days. It develops 1-2 weeks post chemotherapy usually at initial course.
Pathophysiology: increased alveolar capillary permeability

Recombinant IL-2
3%-20%
(high dose intravenous)

Described in renal cancer and melanoma patients, usually in the context of generalized
vascular leak syndrome. Severe but reversible on discontinuation of IL-2. An incidence
decline is being recorded over the last decade reflecting an improvement in safety keys
as the screening of eligible patients and the optimization of therapeutic conditions.
Pathophysiology: a damaging effect on vascular endothelial cells by cytokines

[23, 27, 32]

Bone marrow
transplantation

moderate

Bone marrow transplant patients may develop NCPE in the context of systemic capillary [75]
leak syndrome 1-2 months following high-dose chemotherapy. A pivotal contribution by
circulating leukocytes is suggested.

All-trans-retinoic
acid (ATRA)

12%

It occurs in the context of ATRA syndrome that may develop in acute promyelocytic
leukemia patients undergoing remission induction treatment with ATRA and is
characterized by fever, respiratory distress, peripheral edema and pleural or
pericardial effusions. Treatment with corticosteroids effective if started early.

[82]

Arsenic trioxide
(As203)

15%

Same as with ATRA.

[83]

Gemcitabine

0.1%

It is usually building up over a number of courses, but it may also occur after a single
administration. Life-threatening but also reversible upon treatment discontinuation and
start of intensive supportive therapy and IV corticosteroids.

[54, 56-60]

Gemcitabine plus
docetaxel

rare

In patients with solid tumors. Similar clinical features and outcome as with single
gemcitabine.

[8, 66]

Vinblastine

anecdotal

Mitomycin plus
vinblastine

2%

It tends to occur shortly after chemotherapy administration.

[69, 71]

Intrathecal
methotrexate

occasional

Three cases have been reported of rapidly developing respiratory distress following
the administration of methotrexate into the cerebrospinal fluid.

[73, 74]

G-CSF

occasional

Dacarbazine plus
fotemustine

anecdotal

[68]

[38-40]
Reported at a sequential administration regimen.

[105]

Note: Characterizations high, moderate, rare, occasional, and anecdotal are given in cases of lack of clear incidence data and must be loosely interpreted.

doses of 4.5 million IU/day, three times weekly, has been


well established [33] while novel locoregional administration
strategies, such as the inhalation of nebulized rIL-2, are being
investigated with the aim of improving its therapeutic index
[34]. The more favorable toxicity profile of subcutaneous,
compared with intravenous, bolus administration of rIL-2, is
possibly attributed to a lower systemic absorption (30% of
the injected dose) and a better pharmacokinetic profile (sustained systemic exposure and lower peak levels) associated
with this route [22].
Hematopoietic growth factors G-CSF and GM-CSF
have both been associated as causative or contributing factors with the development of pulmonary toxicity. G-CSF
administered during bleomycin-containing chemotherapy

regimens may enhance subclinical bleomycin pulmonary


toxicity [35, 36], while a possible pulmonotoxic synergy
has been suggested for the combination of GM-CSF with
chemoradiotherapy in small cell lung cancer patients who
received this treatment [37]. Regarding pulmonary edema,
G-CSF and GM-CSF have occasionally been implicated
with the development of true NCPE, as a small number of
published case reports indicate, not all of them well documented [38-40]. It is most possible that the underlying
pathophysiological machinery in NCPE cases is the same as
with the capillary-leak syndrome that has more commonly
been associated with the clinical use of these hematopoietic
factors [41-43]. It should be recognized that a definite association of G-CSF and GM-CSF with pulmonary toxicity in

Briasoulis, Pavlidis
most of the clinical studies is rather poorly documented, but
the pulmonotoxic potential of G-CSF has been highlighted
in a single preclinical study. Hierholzer et al. have shown
that installing G-CSF into the lungs of rats by intratracheal
injection resulted in recruitment of neutrophils, lung injury,
and impaired pulmonary function [44].
Cytotoxic Drugs
Two cytotoxic antimetabolites, cytarabine, and gemcitabine, both analogues of the nucleoside cytidine, have clearly
been associated with the development of pulmonary toxicity of
the NCPE type. Cytarabine is used in the chemotherapy of
leukemia and lymphoma, and gemcitabine is used in the
chemotherapy of solid tumors. These agents compete with
deoxycytidine triphosphate for incorporation into the C sites of
the growing DNA strand and halt DNA synthesis in progress,
inducing cell apoptosis primarily at the S phase [45, 46].
Treatment of leukemia patients with high or moderate
doses of cytarabine has been associated with the development
of NCPE at a relatively high rate. Andersson et al. reported a
12.5% incidence of fatal pulmonary edema in 103 relapsed
leukemia patients at a median time of 16 days from the initiation of high-dose cytarabine therapy. Postmortem findings
in seven patients who died of ARDS showed a massive pulmonary edema with a highly proteinaceous intra-alveolar
infiltrate, and in one case diffuse alveolar damage. No inflammatory changes were detected [47]. Similar toxicity has also
been seen in pediatric patients. Shearer et al. described a fatal
pulmonary insufficiency in 5 of 22 pediatric patients treated
with high-dose cytarabine and G-CSF support for relapsed
acute myelogenous leukemia. Postmortem examination disclosed profound pulmonary edema [48]. In another study,
Haupt reviewed clinical and pathologic features of 181
patients with leukemia who had been autopsied at the Johns
Hopkins Hospital. They found a highly significant increase in
the frequency of pulmonary edema in 51 patients (it was massive in 24 patients) who were treated with cytarabine within
the last 30 days prior to their death as compared with noncytarabine-treated patients. These investigators failed to detect
any other cause possibly related to the observed pulmonary
edema in 67% of the cases [49]. An interesting syndrome characterized by pulmonary edema, fever, and diarrhea was
reported in 4 of 23 lymphoma patients who were treated with
high-dose cytarabine. High concentrations of tumor necrosis
factor-alpha and platelet activating factor activity were found
in the serum of two of those patients [50]. Finally, a review of
radiography findings in patients treated with cytarabine who
developed pulmonary complications demonstrated diffuse
consolidations of the alveolar pattern in both lungs that
regressed within 3 to 7 days in the majority of patients who
recovered [15].

156

Gemcitabine is currently employed in the treatment of


solid tumors, as a drug of a particularly modest toxicity profile with a low incidence of the side effects normally associated with cytotoxics [51]. In regard to pulmonary toxicity a
self-limiting dyspnea has been reported in 5%-8% of patients
treated with this agent [52] while true NCPE was recorded in
clinical trials at an incidence of about 0.1% [53]. Interestingly,
flu-like symptoms and fever have also been reported in 20%35%, and peripheral edema in up to 20% of the patients
treated with gemcitabine [51]. Gemcitabine-induced pulmonary toxicity usually upsurges after a number of repeated
drug administrations gradually building up, although occasionally it has also been reported to occur after a single-dose
administration [54-56]. A small number of NCPE cases associated with the gemcitabine administration have been reported
so far. In the first published report Pavlakis et al. presented
three cases of cancer patients who developed NCPE/ARDS
while being treated with gemcitabine. Two patients died, after
being repeatedly challenged with gemcitabine despite persistent episodes of dyspnea following first chemotherapy
courses. Corticosteroids provided maximum benefit in a third
patient in whom they were commenced early at diagnosis and
were coupled with discontinuation of gemcitabine [54].
Following this report, two fatal and three successful corticosteroid-managed cases were reported [57-60]. In most of
these cases a diffuse alveolar damage consistent with acute
respiratory distress syndrome was found and systemic
corticosteroids seemed to help if started early at diagnosis.
There are also a few other cytotoxic compounds that have
occasionally been associated with NCPE. Those are docetaxel, vinblastine and intrathecal methotrexate. Docetaxel, a
novel mitotic spindle poison that acts as a microtubule stabilizer, has only infrequently been associated with the development of pulmonary toxicity, that is usually of the type of
hypersensitivity pneumonitis not meeting the criteria for
NCPE [61, 62]. Notably, a fluid retention syndrome is known
to occur in as many as 20%-60% of nonpremedicated patients
treated with the agent, but this is typically peripheral, with
pleural effusions reported only occasionally [63-65].
Although no NCPE has been attributed to docetaxel as a
single agent to date, several cases have been reported with
its combination with gemcitabine and radiotherapy [8, 66,
67]. Vinblastine as a single agent has anecdotally been
associated with the development of NCPE [68], but this
toxicity seems to occur more often when combined with
mitomycin [69-71].
Although the most common pulmonary disorder connected with methotrexate is a gradually developing interstitial pneumonitis in rheumatoid arthritis patients [72], three
cases of NCPE have been reported that were associated with
intrathecal methotrexate. The clinical course, radiology

157

findings, and in one patient, the pathology report favor the


diagnosis of NCPE in these cases [73, 74].
A high incidence of NCPE has been reported to occur in
allogeneic and autologous bone marrow transplant patients in
a single study. In those patients, NCPE presented in the context of a capillary leak syndrome and was accompanied by
generalized organ dysfunction in many cases [75]. It is
thought that induction chemotherapy may also play some role
in the development of lung toxicity in these patients. Data
suggest that standard-dose chemotherapy produces asymptomatic pulmonary dysfunction and inflammation, which may
prime the lungs for further injury by high-dose chemotherapy.
In a prospective study, high-risk breast cancer patients developed a 12.6% decrease of diffusing capacity of the lungs for
carbon monoxide (DL(CO)) after three cycles of induction
chemotherapy with CAF (cyclophosphamide, doxorubicin,
5-fluorouracil) and prior to high-dose chemotherapy with
autologous bone marrow or peripheral blood progenitor cell
support [76].
ATRA Syndrome
The vitamin A derivative all-trans-retinoic acid (ATRA)
and As203 are two drugs with the potential to induce granulocytic differentiation and promote apoptosis of leukemic
promyelocytes [77-79]. Currently used in the treatment of
acute promyelocytic leukemia [80, 81], these agents have
both been associated with the development of NCPE/ARDS
in the context of ATRA syndrome. This is a life-threatening
complication that occurs in a quarter of patients with acute
promyelocytic leukemia who undergo remission induction
treatment with ATRA or As203, with or without chemotherapy [82, 83]. The ATRA syndrome characterizes a loosely
defined constellation of signs and symptoms that primarily
consist of fever and respiratory distress, but also of weight
gain, peripheral edema, pleural or pericardial effusions, and
hypotension [84]. The pathophysiology of the ATRA syndrome is uncertain, but in vitro data provide evidence that
the induction of inflammatory cytokines in acute promyelocytic leukemia cells may play an important role in its pathogenesis [77].
Pathophysiology
The exact mechanism of lung injury in the case of
NCPE related to cancer therapeutic agents varies with each
class of drugs involved, but overall, it remains poorly
defined. It must be emphasized that comorbidities existing
in cancer patients potentially complicate the attempt to
identify an unequivocal explanation for these clinical findings. Furthermore, in combination chemotherapies or highdose chemotherapies with G-CSF support, it is quite
difficult to mark out the major offensive compound,

Cancer-Therapy-Related Noncardiogenic Pulmonary Edema


although lung toxicity can obviously be amplified when
pulmonotoxic agents are used concurrently.
Regarding cytotoxic agents, a direct insult by the offensive agent to the alveolar type I pneumocytes or pulmonary
vascular endothelial cells and the triggering of a cytokinemediated inflammatory response appear to be the most frequently involved pathogenic mechanisms. Findings from
leukemia patients who died of ARDS following treatment
with high-dose cytarabine revealed massive edema and
partly diffuse alveolar damage without inflammatory reaction, probably indicating that a direct cytocidal result was a
most contributing pathogenetic mechanism in those cases
[47]. The hypothesis for the involvement of a cytokinemediated inflammatory response is suggested by the drugrelated fever that occurs in almost half of the patients treated
with cytarabine or gemcitabine [85] and the rapid response
to corticosteroids reported by several investigators [60, 86].
Additional data provide evidence indicating the involvement of several cytokines, such as tumor necrosis factoralpha and platelet activating factor in the development of
lung injury [50, 87, 88]. Regarding a possible implication of
the dosage in the development of NCPE, a dose/pulmonarytoxicity relationship has only been documented for cytarabine, while this does not seem to be the case with gemcitabine
[89, 90]. Whatever the exact mechanism of pathogenesis of
the lung injury in the case of chemotherapy-induced NCPE,
the end-result is an increase in vascular permeability
accompanied by extravasation of fluids and proteins into
the air space [9]. This has been clearly documented by
autopsy findings [47, 54]. Interestingly, two cancer therapeutic agents with pulmonotoxic potential are also known
to induce a capillary leak syndrome [51, 91].
The pathogenesis of the lung injury that is related to
intravenous administration of high doses of recombinant
IL-2 has been investigated extensively. Initial preclinical
studies suggested that microvasculature damage, causing a
generalized increase of vascular permeability to albumin,
was the basic mechanism of rIL-2-induced NCPE [25].
Subsequent studies went into more detail and associated the
systemic administration of rIL-2 with lesions of venous and
capillary endothelia, alveolar basement membrane, and
type I epithelial cells in animals, while leukocyte or platelet
activation, generation of free radicals, and activation of the
complement system have also been suggested to be
involved in its pathogenesis [92-97]. Despite a widening
evidence of the involvement of various cytokines released
by activated lymphoid cells in the pathogenesis of rIL-2induced NCPE, the exact mechanism of the damaging
events that drive this clinical syndrome remain unclear [23,
33]. Recent demonstration of functional IL-2 receptors on
type II pneumocytes indicates that these cells may also

Briasoulis, Pavlidis
become involved in the pathogenesis of rIL-2-induced
NCPE [98].
Regarding NCPE that presents in the context of the
ATRA syndrome (see above), activation of production of
inflammatory cytokines by acute promyelocytic leukemia
cells after exposure to ATRA or As203 seems to play an
important role in its pathogenesis. In vitro data suggest that
acute promyelocytic leukemia cells may produce an
increased expression of IL-1 beta, tumor necrosis factoralpha, IL-8, L-selectin, and intercellular adhesion molecule-1 after induction of differentiation with ATRA, an
effect considered related to the progress of the differentiation [77, 99, 100]. The similarity of the clinical phenomena
that characterize the ATRA syndrome with those observed
after increased production or administration of exogenous
cytokines, the overexpression of which has been shown in
vitro to be activated after exposure of acute promyelocytic
leukemia cells to ATRA, suggests a link between the
ATRA syndrome/NCPE and increased production of cytokines and surface integrins during the process of induction
of differentiation.
Finally, research on systemic capillary leak syndrome
(SCLS) that is a distinct clinical entity characterized by
attacks of a marked shift of plasma from the intravascular
to the extravascular space may offer an additional theoretical basis for the interpretational approach of the pathophysiology underlining chemotherapy-related NCPE. There is

158

evidence for a complement-mediated injury that leads to a


breakdown of the endothelial barrier and plasma leakage in
SCLS [101-103].
Treatment
Awareness of early recognition of evolving lung toxicity and withdrawal of the possibly offensive drug or drugs
are suggested as the most important steps toward successful
management of drug-induced NCPE. Intravenous corticosteroids, if started early in the course of drug-induced
NCPE, diuretic therapy, and respiratory support with or
without mechanical ventilation have reportedly salvaged a
number of patients from this pulmonary toxicity.
CONCLUSION
It is important for practicing oncologists to realize that a
number of cancer-therapeutic agents can produce, or interact
with other drugs to produce, noncardiogenic pulmonary
edema. Prompt evaluation of any new respiratory symptoms
that occur in patients who are treated with pharmaceutical
agents potentially inducing NCPE is important for early detection of this severe complication. Patients with pulmonary compromise should be hospitalized and can successfully be
managed with high-dose corticosteroids, diuretics, and oxygen
supplementation. Immediate discontinuation of chemotherapy
is recommended upon suspicion of lung toxicity, and in disputed cases, a lung biopsy should be taken into consideration.

R EFERENCES
1 Kreisman H, Wolkove N. Pulmonary toxicity of antineoplastic
therapy. Semin Oncol 1992;19:508-520.
2 Stover DE, Kaner RJ. Pulmonary toxicity. In: DeVita VT,
Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice
of Oncology, 5th ed. Philadelphia, PA: Lippincott-Raven,
1997:2729-2739.
3 Simpson AB, Paul J, Graham J et al. Fatal bleomycin pulmonary toxicity in the west of Scotland 1991-95: a review of
patients with germ cell tumours. Br J Cancer 1998;78:10611066.
4 ODriscoll BR, Hasleton PS, Taylor PM et al. Active lung
fibrosis up to 17 years after chemotherapy with carmustine
(BCNU) in childhood. N Engl J Med 1990;323:378-382.
5 Castro M, Veeder MH, Mailliard JA et al. A prospective study
of pulmonary function in patients receiving mitomycin. Chest
1996;109:939-944.
6 Gaetani P, Silvani V, Butti G et al. Nitrosourea derivativesinduced pulmonary toxicity in patients treated for malignant
brain tumors. Early subclinical detection and its prevention.
Eur J Cancer Clin Oncol 1987;23:267-271.
7 Jules-Elysee K, White DA. Bleomycin induced pulmonary
toxicity. Clin Chest Med 1990;11:1-20.

8 Briasoulis E, Froudarakis M, Milionis H et al. Chemotherapy


induced noncardiogenic pulmonary edema: case report
related to gemcitabine plus docetaxel combination with G-CSF
support. Respiration 2000;67:680-683.
9 Flick MR, Matthay MA. Pulmonary edema and acute lung
injury. In: Murray JF, Nadel JA, eds. Textbook of Respiratory
Medicine, 3rd ed. Philadelphia, PA: W.B. Saunders
Company, 2000:1575-1598.
10 Bernard GR, Artigas A, Brigham KL et al. The AmericanEuropean Consensus Conference on ARDS. Definitions,
mechanisms, relevant outcomes, and clinical trial coordination.
Am J Respir Crit Care Med 1994;149:818-824.
11 Rosenow EC III, Myers JL, Swensen SJ et al. Drug-induced
pulmonary disease. An update. Chest 1992;102:239-250.
12 Cooper JA Jr, White DA, Matthay RA. Drug-induced pulmonary disease. Part 1: Cytotoxic drugs. Am Rev Respir Dis
1986;133:321-340.
13 Andersson BS, Cogan BM, Keating MJ et al. Subacute pulmonary failure complicating therapy with high-dose Ara-C
in acute leukemia. Cancer 1985;56:2181-2184.
14 Reed CR, Glauser FL. Drug-induced noncardiogenic pulmonary
edema. Chest 1991;100:1120-1124.

159

Cancer-Therapy-Related Noncardiogenic Pulmonary Edema

15 Tham RT, Peters WG, de Bruine FT et al. Pulmonary complications of cytosine-arabinoside therapy: radiographic findings.
Am J Roentgenol 1987;149:23-27.

31 Mann H, Ward JH, Samlowski WE. Vascular leak syndrome


associated with interleukin-2: chest radiographic manifestations.
Radiology 1990;176:191-194.

16 Lotze MT, Robb RJ, Sharrow SO et al. Systemic administration of interleukin-2 in humans. J Biol Response Mod
1984;3:475-482.

32 Kammula US, White DE, Rosenberg SA. Trends in the safety


of high dose bolus interleukin-2 administration in patients
with metastatic cancer. Cancer 1998;83:797-805.

17 Rosenberg SA, Lotze MT, Mule JJ. NIH conference. New


approaches to the immunotherapy of cancer using interleukin2. Ann Intern Med 1988;108:853-864.

33 Guida M, Abbate I, Casamassima A et al. Long-term subcutaneous recombinant interleukin-2 as maintenance therapy:
biological effects and clinical implications. Cancer Biother
1995;10:195-203.

18 Keilholz U, Stoter G, Punt CJ et al. Recombinant interleukin2-based treatments for advanced melanoma: the experience of
the European Organization for Research and Treatment of
Cancer Melanoma Cooperative Group. Cancer J Sci Am
1997;3(suppl 1):S22-S28.
19 Dutcher JP, Atkins M, Fisher R et al. Interleukin-2-based therapy for metastatic renal cell cancer: the Cytokine Working
Group experience, 1989-1997. Cancer J Sci Am 1997;3(suppl
1):S73-S78.
20 Allen MJ, Vaughan M, Webb A et al. Protracted venous infusion 5-fluorouracil in combination with subcutaneous interleukin-2 and alpha-interferon in patients with metastatic renal
cell cancer: a phase II study. Br J Cancer 2000;83:980-985.
21 Legha SS. Durable complete responses in metastatic melanoma
treated with interleukin-2 in combination with interferon alpha
and chemotherapy. Semin Oncol 1997;24:S39-S43.
22 Anderson PM, Sorenson MA. Effects of route and formulation on clinical pharmacokinetics of interleukin-2. Clin
Pharmacokinet 1994;27:19-31.

34 Huland E, Heinzer H, Mir TS et al. Inhaled interleukin-2 therapy in pulmonary metastatic renal cell carcinoma: six years of
experience. Cancer J Sci Am 1997;3(suppl 1):S98-S105.
35 Lei KI, Leung WT, Johnson PJ. Serious pulmonary complications in patients receiving recombinant granulocyte colonystimulating factor during BACOP chemotherapy for aggressive
non-Hodgkins lymphoma. Br J Cancer 1994;70:1009-1013.
36 Matthews JH. Pulmonary toxicity of ABVD chemotherapy
and G-CSF in Hodgkins disease: possible synergy [Letter].
Lancet 1993;342:988.
37 Bunn PA Jr, Crowley J, Kelly K et al. Chemoradiotherapy with
or without granulocyte-macrophage colony-stimulating factor
in the treatment of limited-stage small-cell lung cancer: a
prospective phase III randomized study of the Southwest
Oncology Group. J Clin Oncol 1995;13:1632-1641.
38 Kitamura S, Kinouchi K, Fukumitsu K et al. A risk of pulmonary edema associated with G-CSF pretreatment. Masui
1997;46:946-950.

23 Vial T, Descotes J. Clinical toxicity of interleukin-2. Drug


Saf 1992;7:417-433.

39 Ruiz-Arguelles GJ, Arizpe-Bravo D, Sanchez-Sosa S et al.


Fatal G-CSF-induced pulmonary toxicity [Letter]. Am J
Hematol 1999;60:82-83.

24 Shulman KL, Stadler WM, Vogelzang NJ. High-dose continuous intravenous infusion of interleukin-2 therapy for
metastatic renal cell carcinoma: the University of Chicago
experience. Urology 1996;47:194-197.

40 Schilero GJ, Oropello J, Benjamin E. Impairment in gas


exchange after granulocyte colony stimulating factor (G-CSF)
in a patient with the adult respiratory distress syndrome. Chest
1995;107:276-278.

25 Fairman RP, Glauser FL, Merchant RE et al. Increase of rat pulmonary microvascular permeability to albumin by recombinant
interleukin-2. Cancer Res 1987;47:3528-3532.

41 Arning M, Kliche KO, Schneider W. GM-CSF therapy and


capillary-leak syndrome [Letter]. Ann Hematol 1991;62:83.

26 Saxon RR, Klein JS, Bar MH et al. Pathogenesis of pulmonary edema during interleukin-2 therapy: correlation of
chest radiographic and clinical findings in 54 patients. Am J
Roentgenol 1991;156:281-285.

42 Emminger W, Emminger-Schmidmeier W, Peters C et al.


Capillary leak syndrome during low dose granulocytemacrophage colony- stimulating factor (rh GM-CSF) treatment
of a patient in a continuous febrile state. Blut 1990;61:219-221.

27 Conant EF, Fox KR, Miller WT. Pulmonary edema as a


complication of interleukin-2 therapy. Am J Roentgenol
1989;152:749-752.

43 Heitger A, Maurer K, Neu N et al. Capillary leak syndrome


in a patient with septicemia and granulocyte-colony-stimulating factor (G-CSF)-induced accelerated granulopoiesis
[Letter]. Med Pediatr Oncol 1998;31:126-127.

28 Lotze MT, Matory YL, Rayner AA et al. Clinical effects and


toxicity of interleukin-2 in patients with cancer. Cancer
1986;58:2764-2772.

44 Hierholzer C, Kelly E, Lyons V et al. G-CSF instillation into


rat lungs mediates neutrophil recruitment, pulmonary
edema, and hypoxia. J Leukoc Biol 1998;63:169-174.

29 Rosenstein M, Ettinghausen SE, Rosenberg SA. Extravasation


of intravascular fluid mediated by the systemic administration
of recombinant interleukin 2. J Immunol 1986;137:1735-1742.

45 Gemcitabine. In: Dollery C, ed. Therapeutic Drugs, 2nd ed.


Edinburgh, UK: Churchill Livingstone, 1999:G25-G30.

30 Lee RE, Lotze MT, Skibber JM et al. Cardiorespiratory


effects of immunotherapy with interleukin-2. J Clin Oncol
1989;7:7-20.

46 Cytarabine. In: Dollery C, ed. Therapeutic Drugs, 2nd ed.


Edinburgh, UK: Churchill Livingstone, 1999:C373-C375.
47 Andersson BS, Luna MA, Yee C et al. Fatal pulmonary failure complicating high-dose cytosine arabinoside therapy in
acute leukemia. Cancer 1990;65:1079-1084.

Briasoulis, Pavlidis

160

48 Shearer P, Katz J, Bozeman P et al. Pulmonary insufficiency


complicating therapy with high dose cytosine arabinoside in
five pediatric patients with relapsed acute myelogenous
leukemia. Cancer 1994;74:1953-1958.

66 Dunsford ML, Mead GM, Bateman AC et al. Severe pulmonary toxicity in patients treated with a combination of
docetaxel and gemcitabine for metastatic transitional cell
carcinoma. Ann Oncol 1999;10:943-947.

49 Haupt HM, Hutchins GM, Moore GW. Ara-C lung: noncardiogenic pulmonary edema complicating cytosine arabinoside
therapy of leukemia. Am J Med 1981;70:256-261.

67 Hesse K, Heinrich B, Zimmermann F et al. Combined


radiochemotherapy with docetaxel in patients with unresectable locally advanced head and neck tumors. Strahlenther
Onkol 2000;176:67-72.

50 Chiche D, Pico JL, Bernaudin JF et al. Pulmonary edema and


shock after high-dose aracytine-C for lymphoma; possible role
of TNF-alpha and PAF. Eur Cytokine Netw 1993;4:147-151.
51 Aapro MS, Martin C, Hatty S. Gemcitabinea safety
review. Anticancer Drugs 1998;9:191-201.
52 Nelson R, Trassoff P. Dyspnea with gemcitabine is commonly
seen, often disease related, transient and rarely severe. Eur J
Cancer 1999;31(A):S197-S198.
53 Roychowdhury DF, Smith CA, Peterson P et al. A report on
serious pulmonary toxicity associated with gemcitabinebased therapy (36th Annual Meeting). Proc Am Soc Clin
Oncol 2000;19:196a.
54 Pavlakis N, Bell DR, Millward MJ et al. Fatal pulmonary
toxicity resulting from treatment with gemcitabine. Cancer
1997;80:286-291.
55 Linskens RK, Golding RP, van Groeningen CJ et al. Severe
acute lung injury induced by gemcitabine. Neth J Med
2000;56:232-235.
56 Ciotti R, Belotti G, Facchi E et al. Sudden cardio-pulmonary
toxicity following a single infusion of gemcitabine [Letter].
Ann Oncol 1999;10:997.
57 Tempero MA, Brand R. Fatal pulmonary toxicity resulting
from treatment with gemcitabine. Cancer 1998;82:1800-1801.
58 Sauer-Heilborn A, Kath R, Schneider CP et al. Severe nonhaematological toxicity after treatment with gemcitabine.
J Cancer Res Clin Oncol 1999;125:637-640.
59 Marruchella A, Fiorenzano G, Merizzi A et al. Diffuse alveolar damage in a patient treated with gemcitabine. Eur Respir J
1998;11:504-506.
60 Vander EN, Miller V. Successful treatment of gemcitabine
toxicity with a brief course of oral corticosteroid therapy.
Chest 1998;114:1779-1781.
61 Etienne B, Perol M, Nesme P et al. Acute diffuse interstitial
pneumopathy following docetaxel (Taxotere). Apropos of
2 cases. Rev Mal Respir 1998;15:199-203.
62 Merad M, Le CA, Baldeyrou P et al. Docetaxel and interstitial pulmonary injury. Ann Oncol 1997;8:191-194.
63 Schrijvers D, Wanders J, Dirix L et al. Coping with toxicities of docetaxel (Taxotere). Ann Oncol 1993;4:610-611.
64 Trudeau ME, Eisenhauer EA, Higgins BP et al. Docetaxel in
patients with metastatic breast cancer: a phase II study of the
National Cancer Institute of Canada-Clinical Trials Group.
J Clin Oncol 1996;14:422-428.
65 Semb KA, Aamdal S, Oian P. Capillary protein leak syndrome
appears to explain fluid retention in cancer patients who
receive docetaxel treatment. J Clin Oncol 1998;16:3426-3432.

68 Israel RH, Olson JP. Pulmonary edema associated with


intravenous vinblastine [Letter]. JAMA 1978;240:1585.
69 Hoelzer KL, Harrison BR, Luedke SW et al. Vinblastine-associated pulmonary toxicity in patients receiving combination
therapy with mitomycin and cisplatin. Drug Intell Clin Pharm
1986;20:287-289.
70 Konits PH, Aisner J, Sutherland JC et al. Possible pulmonary
toxicity secondary to vinblastine. Cancer 1982;50:2771-2774.
71 Rao SX, Ramaswamy G, Levin M et al. Fatal acute respiratory
failure after vinblastine-mitomycin therapy in lung carcinoma.
Arch Intern Med 1985;145:1905-1907.
72 Leduc D, De VP, Lheureux P et al. Pneumonitis complicating
low-dose methotrexate therapy for rheumatoid arthritis.
Discrepancies between lung biopsy and bronchoalveolar lavage
findings. Chest 1993;104:1620-1623.
73 Bernstein ML, Sobel DB, Wimmer RS. Noncardiogenic pulmonary edema following injection of methotrexate into the
cerebrospinal fluid. Cancer 1982;50:866-868.
74 Hamous JE, Guffy MM, Aschenbrener CA. Fatal acute respiratory failure following intrathecal methotrexate administration.
Cancer Treat Rep 1983;67:1025-1026.
75 Cahill RA, Spitzer TR, Mazumder A. Marrow engraftment
and clinical manifestations of capillary leak syndrome. Bone
Marrow Transplant 1996;18:177-184.
76 Bhalla KS, Wilczynski SW, Abushamaa AM et al. Pulmonary
toxicity of induction chemotherapy prior to standard or highdose chemotherapy with autologous hematopoietic support.
Am J Respir Crit Care Med 2000;161:17-25.
77 Chen ZX, Xue YQ, Zhang R et al. A clinical and experimental
study on all-trans retinoic acid-treated acute promyelocytic
leukemia patients. Blood 1991;78:1413-1419.
78 Puccetti E, Guller S, Orleth A et al. BCR-ABL mediates arsenic
trioxide-induced apoptosis independently of its aberrant kinase
activity. Cancer Res 2000;60:3409-3413.
79 Shao W, Fanelli M, Ferrara FF et al. Arsenic trioxide as an
inducer of apoptosis and loss of PML/RAR alpha protein in
acute promyelocytic leukemia cells. J Natl Cancer Inst
1998;90:124-133.
80 Tallman MS, Andersen JW, Schiffer CA et al. All-transretinoic acid in acute promyelocytic leukemia. N Engl J Med
1997;337:1021-1028.
81 Niu C, Yan H, Yu T et al. Studies on treatment of acute
promyelocytic leukemia with arsenic trioxide: remission
induction, follow-up, and molecular monitoring in 11 newly
diagnosed and 47 relapsed acute promyelocytic leukemia
patients. Blood 1999;94:3315-3324.

161

82 De Botton S, Dombret H, Sanz M et al. Incidence, clinical features, and outcome of all trans-retinoic acid syndrome in 413
cases of newly diagnosed acute promyelocytic leukemia. The
European APL Group. Blood 1998;92:2712-2718.
83 Camacho LH, Soignet SL, Chanel S et al. Leukocytosis and
the retinoic acid syndrome in patients with acute promyelocytic leukemia treated with arsenic trioxide. J Clin Oncol
2000;18:2620-2625.
84 Frankel SR, Eardley A, Lauwers G et al. The retinoic acid
syndrome in acute promyelocytic leukemia. Ann Intern Med
1992;117:292-296.
85 Pollera CF, Ceribelli A, Crecco M et al. Weekly gemcitabine in
advanced or metastatic solid tumors. A clinical phase I study.
Invest New Drugs 1994;12:111-119.
86 Barrios NJ, Tebbi CK, Freeman AI et al. Toxicity of high dose
Ara-C in children and adolescents. Cancer 1987;60:165-169.
87 Sleijfer S, Vujaskovic Z, Limburg PC et al. Induction of tumor
necrosis factor- as a cause of bleomycin-related toxicity.
Cancer 1998;82:970-974.
88 Powell BL, Zekan PJ, Muss HB et al. Ara-C syndrome during low-dose continuous infusion therapy. Med Pediatr
Oncol 1986;14:310-312.
89 Stentoft J. The toxicity of cytarabine. Drug Saf 1990;5:7-27.
90 Vermorken JB, Guastalla JP, Hatty SR et al. Phase I study of
gemcitabine using a once every 2 weeks schedule. Br J Cancer
1997;76:1489-1493.
91 Piccart MJ, Klijn J, Paridaens R et al. Corticosteroids significantly delay the onset of docetaxel-induced fluid retention: final
results of a randomized study of the European Organization for
Research and Treatment of Cancer Investigational Drug Branch
for Breast Cancer. J Clin Oncol 1997;15:3149-3155.
92 Dubinett SM, Huang M, Lichtenstein A et al. Tumor necrosis
factor-alpha plays a central role in IL-2-induced pulmonary
vascular leak and lymphocyte accumulation. Cell Immunol
1994;157:170-180.
93 Rabinovici R, Sofronski MD, Renz JF et al. Platelet activating factor mediates IL-2-induced lung injury in the rat. J Clin
Invest 1992;89:1669-1673.
94 Queluz TT, Brunda M, Vladutiu AO et al. Morphological
basis of pulmonary edema in mice with cytokine-induced
vascular leak syndrome. Exp Lung Res 1991;17:1095-1108.

Cancer-Therapy-Related Noncardiogenic Pulmonary Edema


95 Klausner JM, Paterson IS, Goldman G et al. IL-2-induced
lung injury is mediated by oxygen free radicals. Surgery
1991;109:169-175.
96 Lissoni P, Barni S, Cattaneo G et al. Activation of the complement system during immunotherapy of cancer with IL-2: a
possible explanation of the capillary leak syndrome. Int J Biol
Markers 1990;5:195-197.
97 Welbourn R, Goldman G, Kobzik L et al. Involvement of
thromboxane and neutrophils in multiple-system organ edema
with IL-2. Ann Surg 1990;212:728-733.
98 Lesur O, Arsalane K, Berard J et al. Functional IL-2 receptors are expressed by rat lung type II epithelial cells Am J
Physiol 1997;273:L495-L503.
99 Hsu HC, Tsai WH, Chen PG et al. In vitro effect of granulocyte-colony stimulating factor and all-trans retinoic acid on
the expression of inflammatory cytokines and adhesion molecules in acute promyelocytic leukemic cells. Eur J Haematol
1999;63:11-18.
100 Grande A, Manfredini R, Tagliafico E et al. All-transretinoic acid induces simultaneously granulocytic differentiation and expression of inflammatory cytokines in HL-60
cells. Exp Hematol 1995;23:117-125.
101 Atkinson JP, Waldmann TA, Stein SF et al. Systemic capillary leak syndrome and monoclonal IgG gammopathy; studies in a sixth patient and a review of the literature. Medicine
(Baltimore) 1977;56:225-239.
102 Lofdahl CG, Solvell L, Laurell AB et al. Systemic capillary
leak syndrome with monoclonal IgG and complement alterations. A case report on an episodic syndrome. Acta Med
Scand 1979;206:405-412.
103 Johansson BR, Lofdahl CG. Ultrastructure of the microvessels in skeletal muscle in a case of systemic capillary leak
syndrome. Acta Med Scand 1979;206:413-416.
104 Jehn U, Goldel N, Rienmuller R et al. Noncardiogenic pulmonary edema complicating intermediate and high-dose Ara
C treatment for relapsed acute leukemia. Med Oncol Tumor
Pharmacother 1988;5:41-47.
105 Gerard B, Aamdal S, Lee SM et al. Activity and unexpected
lung toxicity of the sequential administration of two alkylating agentsdacarbazine and fotemustinein patients with
melanoma. Eur J Cancer 1993;29A:711-719.

You might also like