You are on page 1of 11

Analytic Review

Journal of Intensive Care Medicine


1-11
Oncologic Emergencies—The Old, ª The Author(s) 2018
Article reuse guidelines:
sagepub.com/journals-permissions
the New, and the Deadly DOI: 10.1177/0885066618803863
journals.sagepub.com/home/jic

Krishna Thandra, MD1, Zuhair Salah, MD1, and Sanjay Chawla, MD1

Abstract
Cancer continues to be a leading cause of death despite a broader understanding of its biology and the development of novel
therapies. Nonetheless, with an increasing survival of this population, intensivists must be aware of the associated emergencies,
both old and new. Oncologic emergencies can be seen as an initial presentation of the disease or precipitated by its treatment. In
this review, we present key oncologic emergencies that may be encountered in daily practice, complications associated with
innovative therapies, and treatment-related adverse events.

Keywords
cancer, critical care, tumor lysis syndrome, oncologic emergency, cytokine release syndrome, CAR-T cell

Introduction ANC of <500/mL.5 Neutropenia can occur due to the adverse


effects of cytotoxic chemotherapy, tumor-infiltrating bone
Despite the development of numerous and innovative thera-
marrow, or direct interference with hematopoiesis as seen in
pies, cancer remains a leading cause of death in the United
some hematologic malignancies. 5 Febrile neutropenia is
States and the second leading cause of death globally. In
defined as a single oral temperature of 38.3 C (101 F) sus-
2015, cancer was responsible for nearly 8.8 million deaths
tained over a 1-hour period in a neutropenic patient.6 The major
worldwide as per the World Health Organization. In 2017,
risk factors for neutropenic complications include the rate of
approximately 1.6 million people were expected to be diag-
ANC decline, recent chemotherapy, current immunosuppres-
nosed with cancer in the United States alone.1 Nonetheless, the
sion, elevated liver enzymes prior to treatment, reduced glo-
evolution of cancer care over the past several decades has led to
merular filtration rate, and cardiovascular comorbidities.7
a decline in mortality rates by 1.5% per year, from 1991 to
Cytotoxic drugs that have the highest risk of inducing neutro-
2015, with a total decrease of 26%, resulting in about 2.4
million fewer deaths.2 penia include anthracyclines, taxanes, topoisomerase inhibi-
Patients with cancer account for 14% to 22% of all intensive tors, platinum, gemcitabine, vinorelbine, and certain
care unit (ICU) admissions.3,4 Therefore, intensivists must be alkylators such as cyclophosphamide and ifosfamide.7
knowledgeable of the unique emergencies associated with cancer The causative organism is identified only in a minority of
care. Commonly encountered cancer-related emergencies may patients, and in the rest, it is thought to be due to a variety of
arise as a result of the underlying cancer, which can be the initial organisms. The common causative organisms are listed in
presentation of the disease, occur during the disease course, or can Table 2. Patients with febrile neutropenia are at high risk for
be precipitated during treatment. Many of the most common rapid deterioration; thus, identifying the source of the infection
cancer-related emergencies are highlighted in Table 1. Due to the should be aggressively pursued.3 Common sites of infection in
scope of this review, however, we will focus on the etiology, decreasing order include the gastrointestinal tract, blood, skin,
pathophysiology, and management of selected key oncologic lung, and urinary tract.9 Empiric antibiotic therapy is the main-
emergencies. These include febrile neutropenia, hyperleukocyto- stay of management and is selected based on clinical status,
sis, tumor lysis syndrome (TLS), cardiac tamponade, and pulmon-
ary hemorrhage as well as unique complications associated with 1
Critical Care Medicine Service, Department of Anesthesiology & Critical
emerging cancer therapies such as chimeric antigen receptor-T Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
(CAR-T)-cell and immunotherapy-related adverse events.
Received June 25, 2018. Received revised September 05, 2018. Accepted
September 10, 2018.
White Blood Cells—Too Few or Too Many Corresponding Author:
Febrile Neutropenia Sanjay Chawla, Critical Care Medicine Service, Department of Anesthesiology
& Critical Care Medicine, Memorial Sloan Kettering Cancer Center, 1275 York
Neutropenia is defined as an absolute neutrophil count (ANC) Avenue, C-1179, New York, NY 10065, USA.
of less than 1500/mL, while severe neutropenia is defined as an Email: chawlas@mskcc.org
2 Journal of Intensive Care Medicine XX(X)

Table 1. Common Cancer-Related Emergencies.a on the isolated organism. Recommendations for infections due
to Staphylococcus aureus, Pseudomonas aeruginosa, fungi, or
Metabolic Hypocalcemia
Hypoglycemia
mycobacterial species include catheter removal and antibiotic
Tumor lysis syndrome treatment for at least 2 weeks.7 However, if coagulase-negative
Neurologic Spinal cord compression Staphylococcus is identified, the catheter can generally
Brain metastasis remain.7
Elevated ICP The role of granulocyte–colony stimulating factor (G-CSF)
Intracranial hemorrhage is limited once neutropenia occurs and has shown more benefit
Cardiovascular Cardiac tamponade if given immediately after receipt of chemotherapy. The Amer-
SVC syndrome
Hematologic Hyperviscosity syndrome
ican Society of Clinical Oncology (ASCO) recommends
Leukostasis against the routine use of a CSF unless there is a high risk for
DIC/TTP/AHUS infection-associated complications.10 A course of G-CSF is
Infectious Neutropenic sepsis/septic shock reasonable until the ANC rises above 1500/mL in high-risk
Sepsis patients who remain febrile after the initiation of antibiotics.
Typhlitis High-risk patients include those with expected prolonged
Pulmonary Malignant airway obstruction (>10 days) or severe neutropenia (<100/mL), age >65 years,
Hemoptysis
Pulmonary embolism/VTE
pneumonia, sepsis, invasive fungal infection, or hospitalization
Chemotherapy Extravasation at the time fever develops.11
Anaphylaxis
Surgery-related/requiring Bowel obstruction/perforation
surgical intervention Hemorrhage Leukostasis
Infection Leukostasis or symptomatic hyperleukocytosis is an extreme
Pathological fractures form of leukocytosis that is defined by a white blood cell
Tracheoesophageal fistula
HSCT related GVHD
(WBC) count >100 000/mL. Due to the high quantity, WBCs
Hepatic sinusoidal obstruction syndrome can clump together, inhibiting efficient microvascular flow
DAH leading to end-organ damage with secondary tissue hypoxia
and hemorrhage.12 Patients with hematologic malignancies,
Abbreviations: AHUS, atypical hemolytic uremic syndrome; DAH, diffuse
most commonly acute myeloid leukemia (AML) and acute
alveolar hemorrhage; DIC, disseminated intravascular coagulation; GVHD,
graft versus host disease; ICP, intracranial hemorrhage; SVC, superior vena lymphoid leukemia (ALL), are at highest risk. However, it
cava; TTP, thrombotic thrombocytopenic purpura; VTE, venous can also be seen with chronic myeloid leukemia during blast
thromboembolism.
a
crisis.12 The total WBC count is a prognostic factor with
Items that are in italics are the specific topics that are covered in the review.
commonly accepted cutoffs of 50 000/mL for AML and 400
000/mL for ALL, since blast cells are much larger than
lymphocytes.13
Table 2. Common Causative Organisms of Febrile Neutropenia.8
Two possible pathophysiologic mechanisms have been pro-
Gram-Negative Fungal posed. The first focuses on hyperviscosity that largely depends
Gram-Positive Organisms Organisms Organisms on cellular deformability and the volume of cell fraction in
the blood. Blast cells are less pliable than leukocytes, and thus,
Staphylococcus aureus Escherichia coli Candida species
Staphylococcus epidermidis Klebsiella pneumoniae
the increased cell fraction amplifies the overall viscosity of the
Streptococcus pneumoniae Pseudomonas aeruginosa blood leading to “sludging” and impaired flow in the micro-
Streptococcus pyogenes vasculature. 12 An alternative mechanism suggests that
Streptococci viridans endothelial cells are activated by blast cells which secrete
Enterococcus faecalis inflammatory cytokines leading to an adhesive interaction
Enterococcus faecium between them and thus precipitating leukostasis.12
Corynebacterium The diagnosis is made based on the cell counts and periph-
eral smear along with clinical signs and symptoms of end-organ
hypoxemia or hemorrhage. An X-ray or computed tomography
duration of fever, medical comorbidities, and other factors (CT) of chest usually shows bilateral interstitial or alveolar
(Figure 1). Patients with fever >48 hours should be hospitalized infiltrates. Computed tomography or magnetic resonance ima-
for intravenous (IV) antibiotics that should continue until the ging of the brain may reveal intracranial hemorrhage.14 Of
ANC is >500/mL or depending on the identified source of note, patients with leukostasis are at high risk for concomitant
infection.6 Addition of an antifungal agent should be consid- TLS, which is discussed in the next section.15
ered when fever persists despite 4 to 7 days of broad-spectrum Initial management for at-risk patients includes aggressive
antibiotics without an identified causative organism.6 When hydration with calcium- and potassium-free fluids with close
central line-associated bloodstream infection is the confirmed monitoring for the development of TLS. Definitive treatment
source of infection, the decision to remove the catheter depends focuses on chemotherapy to decrease tumor load.15 In cases of
Thandra et al 3

Fever ( 38.3oC/101oF) & neutropenia (ANC<500/µl)

Initiate infectious work-up


Clinically
unstable?

Yes No

Admit to ICU and start broad Inpatient vs outpatient


spectrum antibiotics : treatment depending on
piperacillin/tazobactam or clinical scenario
carbapenem (except ertapenem) or
ceftazidime or
cefepime
Plus
vancomycin or linezolid if at risk for MRSA
Consider PO vancomycin or IV metronidazole if
abdominal symptoms or suspected Clostridium
difficile infection
Fever persists for 4-7 days
despite antibiotics with no
identified causative agent?

Yes No

Consider addition of Continue antibiotics


antifungal agent: depending on site of
micafungin or infection or until
voriconazole or
posaconazole or ANC>500/µl
isavuconazonium

Figure 1. Initial management of febrile neutropenia.6

very high WBC counts (>200 000 to 300 000/mL) or in symp- Too Much Growth
tomatic patients, leukapheresis is the treatment of choice,
Tumor Lysis Syndrome
although there is little evidence on mortality reduction.16 Leu-
kapheresis is commonly performed through a central venous Tumor lysis syndrome is a constellation of metabolic abnorm-
catheter but can also be done through bilateral antecubital per- alities that occur secondary to the rapid lysis of tumor cells and
ipheral veins if adequate bore catheters can be placed. A single release of their intracellular contents. It is usually seen upon
leukapheresis session can deplete the blood of 20% to 50% of initiation of cytotoxic chemotherapy but can also occur spon-
its total WBCs; the remaining blood is then reinfused into the taneously in highly proliferative malignancies due to increased
patient.17 In asymptomatic patients with AML in whom induc- cell turnover. Tumor lysis syndrome is most commonly
tion chemotherapy cannot be started immediately, cytoreduc- reported in patients with high-grade non-Hodgkin lymphoma
tion with hydroxyurea is suggested.18 In patients where there is (NHL) and acute leukemias but can also occur during the treat-
severe anemia, and impending congestive cardiac failure, ment of large solid tumors (ie, hepatocellular carcinoma, meta-
exchange transfusion may be considered. Partial exchange static prostate cancer) that are highly chemosensitive.19
helps in correcting hyperleukocytosis and severe anemia with- The pathophysiology is mainly attributed to the release of
out increasing the viscosity and fluid overload, which are the intracellular components (potassium, phosphate, and uric acid)
main drawbacks of whole blood transfusion and hydration, into the bloodstream. The symptoms of TLS are nonspecific
respectively.15 Figure 2 depicts a flowchart detailing manage- and are largely attributed to the predominant metabolic
ment strategies in case of leukostasis. abnormalities and those related to acute kidney injury (AKI).20
4 Journal of Intensive Care Medicine XX(X)

guidelines is shown in Table 3.23 Management of TLS as out-


lined in Figure 3 starts with risk stratification and initiation of
interventions focused on preventing clinical TLS as it is asso-
ciated with morbidity and mortality. In patients who are at
high risk for TLS or with established TLS, aggressive hydra-
tion is provided along with rasburicase for the initial manage-
ment of hyperuricemia (Figure 3). 23 Rasburicase, a
recombinant form of urate oxidase metabolizes insoluble uric
acid to allantoin, may be considered if hyperuricemia devel-
ops despite allopurinol therapy or as an initial management in
pediatric patients. Rasburicase is known to be active ex vivo;
as such, blood samples should be sent on ice in order to avoid
a falsely normal level in the presence of true hyperuricemia.
To decrease the risk of calciphylaxis, it is recommended to
use aluminium hydroxide or sevelamer in place of calcium-
based phosphate binders.20

Too Much Fluid in the Wrong Places


Malignant Pericardial Effusion and Cardiac Tamponade
Figure 2. Management of leukostasis. The pericardial sac is easily distensible and can potentially
accommodate up to 2 L of fluid if the accumulation occurs
slowly.24 However, tamponade physiology may arise with as
Hyperkalemia occurs secondary to the release of intracellular
little as 100 mL of fluid if the accumulation is acute.25 As the
potassium during cell lysis.21 Uric acid is produced by the
intrapericardial pressure increases, it prevents the distension of
catabolism of purines by xanthine oxidase. Increased break-
cardiac chambers but to a higher degree on the right ventricle
down of nucleic acids produces large amounts of uric acid,
since it is a thin-walled structure. Venous return is thus ham-
which can crystallize in the renal tubules leading to AKI. pered resulting in compromised filling; diastolic pressures
This is further exacerbated by hyperphosphatemia, which begin to equalize with subsequent reduction in cardiac output
can deposit in the renal tubules as calcium phosphate crys- resulting in tamponade. The classic presentation of Beck’s
tals and thus lead to hypocalcemia. Independent of uric acid triad—hypotension, elevated jugular venous pressure (JVP),
precipitation, hyperuricemia by itself can lead to vasocon- and muffled heart sounds—may not be seen when accumula-
striction, decreased renal blood flow, and release of pro- tion is gradual, and patients may present with only chest dis-
inflammatory mediators due to vascular smooth muscle comfort or dyspnea.26 Pericardial effusion in patients with
irritation leading to AKI. cancer is due to metastasis, direct extension of a local tumor,
There are both clinical and laboratory criteria for the diag- or cancer therapy. Metastatic disease occurs most commonly
nosis of TLS according to the classification system of Cairo from noncontiguous breast or lung cancer and melanoma, while
and Bishop.22 Laboratory TLS is the presence of 2 or more primary neoplasms of the pericardium are extremely rare.27
metabolic abnormalities including hyperuricemia, hyperkale- Diagnosis of pericardial effusion occurs through clinical
mia, hyperphosphatemia, or hypocalcaemia (Figure 3) occur- examination plus either echocardiography or CT scan.27 Clin-
ring within 3 days prior to or 7 days after the initiation of ical presentation can be nonspecific and includes tachycardia,
treatment assuming that adequate hydration and a hypourice- hypotension, peripheral edema, pulsus paradoxus, elevated
mic agent are given.22 Clinical TLS is defined as the presence JVP, and muffled heart sounds. Chest X-ray (CXR) may show
of laboratory TLS along with AKI (increase in creatinine of a water bottle appearance of the cardiac silhouette. Electrocar-
0.3 mg/dL from baseline, or when no baseline is available, a diogram findings can include low voltage and electrical alter-
single value 1.5 times the upper limit of normal for the nans as the heart pendulates within the distended fluid-filled
patient’s age and sex), symptomatic hyperkalemia, or hypo- sac.27 Echocardiography confirms the diagnosis of pericardial
calcemia such as cardiac dysrhythmias, sudden death, tetany, effusion and, if present, tamponade physiology. Echocardio-
paraesthesia, or seizure.22 The American Society of Clinical graphic signs of tamponade physiology include early diastolic
Oncology stratifies patients into high, intermediate, and low right ventricular collapse, late diastolic atrial collapse with a
risk of developing TLS based on the type of cancer, leukocyte dilated IVC, and no respiratory variation.28
count, and rapidity of proliferation with the expected response The mainstay of emergent treatment is pericardiocentesis.
to chemotherapy.23 This may be performed under ultrasound guidance or via the
In each of the risk groups, the coexisting metabolic abnorm- blind subxiphoid approach, which is more prone to complica-
alities are treated accordingly, and a summary of the ASCO tions. A catheter can also be placed in the pericardial space if
Thandra et al 5

Figure 3. Management of tumor lysis syndrome.

Table 3. Management of Electrolyte Abnormalities in TLS.

Abnormality Severity Recommendations

Hyperphosphatemia Moderate 6.5mg/dL Administration of a phosphate binder


Symptomatic Initiation of renal replacement therapy (hemodialysis or CRRT)
Hypocalcaemia Asymptomatic No therapy
Symptomatic Calcium gluconate as 10% solution 1000 mg IV administered slowly with ECG
monitoring
Hyperkalemia Moderate and asymptomatic 6.0 mEq/L Potassium-binding resins along with cardiac monitoring
Severe (>7.0 mEq/L) and/or symptomatic Potassium-binding resins along with cardiac monitoring
Calcium gluconate 1000 mg by slow IV infusion
Short-acting human insulin 0.1 U/kg IV in dextrose-containing solution
Sodium bicarbonate 1 to 2 mEq/kg IV push to move potassium intracellularly
Renal replacement therapy
Abbreviations: CRRT, continuous renal replacement therapy; ECG, electrocardiogram; IV, intravenous; TLS, tumor lysis syndrome.

recurrence is anticipated. If the effusion is due to lung adeno- treatment strategies, prolonged catheter drainage has been
carcinoma, it is highly likely to recur and a pericardial window shown to have a clear advantage. A systematic review
or pericardiectomy should be considered.29 Of the various has shown that once a catheter is placed, the rates of
6 Journal of Intensive Care Medicine XX(X)

Table 4. Select Causes of Hemoptysis.

Pulmonary Vascular
Airway Diseases Pulmonary Parenchymal Diseases Diseases Disorders of Coagulation Miscellaneous

Bronchovascular fistula Infection: Pulmonary arteriovenous Anticoagulant and antiplatelet Drugs and toxins:
 Lung abscess malformation medications  Bevacizumab
 Fungal infections  Cocaine
Bronchial adenoma/  Necrotizing pneumonia Pulmonary embolism Disseminated intravascular Idiopathic
carcinoma  Tuberculosis or nontuberculous coagulation
Metastatic cancer to mycobacterial infection Pulmonary veno- Thrombocytopenia (including
bronchus or trachea  Viral occlusive disease ITP, TTP, HUS)
Abbreviations: HUS, hemolytic uremic syndrome; ITP, immune thrombocytopenic purpura; TTP, thrombotic thrombocytopenic purpura;

reaccumulation are similar with or without the use of sclerosing initial hemostasis is achieved. An initial CXR may help to
agents.30 After drainage, patients should be closely monitored localize the bleeding site; however, bronchoscopy will pro-
due to the risk of paradoxical hemodynamic instability (PHI).31 vide maximal information if any active bleeding site is
The exact physiology of PHI remains unclear but portends a detected.37 Computed tomography angiography may yield
grave prognosis with risk factors including tamponade, large additional information regarding the source of active bleed-
effusion, and evidence of malignant pericardial process.32 Pul- ing.38 Surgical exploration remains the treatment of choice
monary edema is another uncommon complication but can only in selected cases, such as pulmonary artery rupture and
present immediately after drainage of pericardial fluid. The risk other bleeding refractory to nonsurgical therapies.35 Endovas-
can be minimized by draining the fluid slowly in cases of large cular embolization has an 85% success in controlling hemop-
pericardial effusion. There are limited data supporting routine tysis and is considered the most effective and minimally
diuretics immediately after drainage if hemodynamically tol- invasive procedure for managing massive and recurrent
erated.33 However, diuresis should be employed in patients hemoptysis in almost all other cases.38 Paraplegia, vascular
who are at high risk for developing pulmonary edema, if hemo- perforation, and hemorrhage are some of the complications
dynamically feasible. associated with embolization. Early interventional pulmonary
involvement is strongly encouraged if expertise is readily
Pulmonary Hemorrhage available to potentially control bleeding from central or per-
ipheral lesions. This may be achieved with an endobronchial
Massive hemoptysis is the expectoration of a large amount of
blocker to temporarily occlude and tamponade the culprit
blood due to a rapid rate of bleeding, but the precise volumes
bronchus as a supportive measure or until therapeutic intent
remain controversial. Thresholds from 100 mL up to 1000 mL
is achieved. Additionally, central endoluminal lesions can be
over 24 hours have been proposed, but no single definition is
universally accepted.34 Hemoptysis has multiple causes which controlled with topical vasoconstriction, thermal, or direct
are categorized under parenchymal diseases, airway diseases, cautery.39 Independent predictors of mortality in patients with
vascular diseases, coagulation disorders, and miscellaneous massive hemoptysis include chronic alcoholism, neoplasm,
(Table 4). In patients with cancer, massive bleeding is gener- mechanical ventilation, multilobar infiltrates, and pulmonary
ally due to direct infiltration of the tumor cells into the main vascular involvement.40
blood vessels or due to massive tumor necrosis.35 Additionally, Conversely, diffuse alveolar hemorrhage (DAH) encom-
tumors in the lung have abundant and irregular vasculature. passes a variety of disorders in which significant bleeding
Asphyxia due to the flooding of the airways rather than exsan- occurs in the lungs, but not always with overt hemoptysis.
guination is usually the cause of death and is commonly accom- Causes of DAH include underlying autoimmune disorders,
panied by cardiovascular collapse.36 infection, toxins, drug reactions, coagulopathy, or malignancy.
Patients with hemoptysis should be managed based on the It is characterized by damage to the pulmonary microvascula-
rate and severity of bleeding (massive or nonmassive) and the ture. In patients with cancer, it is most commonly seen in
clinical condition.36 In addition to maintaining a patent airway hematologic malignancies or after hematopoietic stem cell
and ensuring adequate gas exchange, adequate resuscitation is transplantation (HSCT). Risk factors associated with DAH in
commonly necessary before any diagnostic investigation.35 HSCT patients include older age, myeloablative conditioning,
However, occasionally diagnostic bronchoscopy is required and severe acute GVHD.41 It typically presents within the first
in order to temporarily control bleeding, which may include month following HSCT; however, cases have been described
placement of a bronchial blocker, and allow for resuscitation. beyond that. The pathophysiology of DAH post HSCT is not
Ideally, once hemodynamic stability is achieved, patients clear but is thought to be related to extensive inflammation
should be placed in a lateral decubitus position with the bleed- secondary to uncontrolled cytokine release.
ing origin in the dependent location. Cough suppression or Diffuse alveolar hemorrhage typically presents as dyspnea,
paralysis can also be used to avoid recurrent bleeding once nonproductive cough, acute hypoxic respiratory failure with
Thandra et al 7

diffuse lung infiltrates, and drop in hemoglobin. The diagnosis axicabtagene ciloleucel complete Risk Evaluation and Mitiga-
is typically made through bronchoscopy demonstrating bloody tion Strategies programs. These programs aim to certify provi-
bronchoalveolar lavage that does not clear with repeated ders by requiring training and education regarding the
lavages or becomes progressively bloodier. Confirming the particular side effects and risks of these therapies.
diagnosis requires a biopsy where an accumulation of erythro- CAR-T-cell therapy is associated with a variety of specific
cytes, fibrin, or hemosiderin-laden macrophages can be seen in and unique toxicities; the 2 we will highlight are cytokine
the alveolar space.42 release syndrome (CRS) and neurotoxicity, either of which
Treatment is primarily supportive with correction of any may require ICU admission. Several cytokines including inter-
coagulopathies and mechanical ventilation while treating the feron g, tumor necrosis factor a, interleukin-6 (IL-6), IL-10,
underlying condition leading to DAH. In addition, since exten- granulocyte–macrophage CSF, soluble glycoprotein 130, solu-
sive inflammation is thought to be the underlying cause, corti- ble IL-6 receptor, and monocyte chemoattractant protein 1
costeroids are commonly utilized. Although dosing is not clear, have been implicated in causing toxicity with significant
pulse methylprednisolone (500-2000 mg/d) for the first 5 days increase in their respective levels.48 Patients with high pretreat-
followed by prednisone 1 mg/kg/d with gradual tapering has ment disease burden and those who received high doses of
been used. When DAH is suspected to be due to medications or CAR-T cells were at increased risk for more severe CRS.49
bleeding disorders, anticoagulant reversal or administration of
the appropriate blood products is indicated. There are no ade- Cytokine release syndrome. Patients with CRS initially develop
quate clinical trials showing efficacy and safety of recombinant constitutional symptoms, but this syndrome can progress rap-
factor VIIa use in pulmonary hemorrhage, but it has been used idly to multiple organ system dysfunction. It is difficult to
off-label in the pediatric population.43 Extracorporeal mem- differentiate from septic shock and macrophage activation syn-
brane oxygenation remains an option in refractory hypoxic drome/hemophagocytic lymphohistiocytosis, which can by
respiratory failure secondary to DAH but is challenging given itself be a complication of CAR-T-cell therapy. Concomitant
the need for anticoagulation in the context of active hemor- antimicrobials are generally employed while awaiting sepsis
rhage.44 There is no evidence available for use of antifibrino- workup results.
lytics in DAH currently, although such agents can be Procalcitonin is a biomarker that has gained popularity in
considered in extreme cases. Recipients of antifibrinolytics determining presence of bacterial sepsis, but its role in CRS has
have a higher risk of systemic thromboses if given IV. There not been established. Certain biomarkers including rapidly
are several case reports of inhaled delivery; however, dosing measurable C-reactive protein levels were thought to estimate
and delivery remain a challenge.45 the severity of the CRS toxicity as it correlated well with IL-6
levels.48 However, the utility of any specific biomarkers to
predict the severity of CRS remains unknown and results
Too Much Stress—All Revved Up should be used as an adjunct to management, which is primar-
ily based on clinical symptomatology.
Chimeric Antigen Receptor-T-Cell Complications
In addition to understanding the general management prin-
and Toxicity ciples of CRS, intensivists should also be familiar with the
Chimeric antigen receptor-T cells are a novel therapy that har- specific CAR-T-cell product being used since each varies in
nesses the immune system to target and kill cancer cells. Nor- symptoms and severity of CRS, timing of symptoms, and indi-
mally, when T cells encounter exogenous/foreign antigens, it vidual reaction to each product. Table 5 lists symptoms and
leads to T-cell proliferation and attack on the antigen-carrying signs in patients with CRS. Treatment of CRS is guided by its
cells to kill them. CAR-T-cell therapy utilizes a patient’s own T toxicity grading as detailed in Table 6.50 Cytokine release syn-
cells which are collected and genetically reprogrammed to tar- drome treatment primarily focuses on supportive ICU care and
get and kill cancer cells that express a selected antigen.46 Cur- pharmacologic treatments that may be targeted (tocilizumab)
rently, CAR-T-cell therapy remains in its early stages with or generalized (corticosteroids). It is not fully clear to what
multiple ongoing trials, and notable responses for complete extent immunosuppression interferes with expansion of CAR-
remission are reported to be 70% to 90% in short-term fol- T cells, which is necessary for effective tumor control. As such,
low-up.47 In 2017, the Food and Drug Administration (FDA) the goal of pharmacologic treatment is to control life-
approved the first CAR-T-cell therapy, tisagenlecleucel threatening symptoms.
(KYMRIAH), for relapsed/refractory B-cell ALL in children Tocilizumab is a monoclonal antibody which competitively
and young adults. Soon thereafter, the FDA also approved binds to the IL-6 receptor and thus can mitigate toxicity. Toci-
axicabtagene ciloleucel (YESCARTA) for relapsed/refractory lizumab administration in general is employed for grade 3 to
diffuse large B-cell lymphoma. There are several other ongoing grade 4 CRS but is variable depending on the specific CAR-T-
trials to evaluate CAR-T-cell efficacy in other malignancies cell product given and can be used from grade 1 to grade 4
including chronic lymphocytic leukemia, NHL, mesothelioma, CRS. A single dose of 8 mg/kg IV is given for patients >30 kg
ovarian cancer, breast cancer, neuroblastoma, and sarcoma. and can be repeated if there is no response within 6 hours.
The FDA has required that hospitals and health-care profes- Clinical symptoms and signs are used to initiate tocilizumab
sionals involved with patients who receive tisagenlecleucel or as cytokine levels are generally not available immediately.
8 Journal of Intensive Care Medicine XX(X)

Table 5. Signs and Symptoms Associated With CRS. Table 7. Sign, Symptoms, and Workup of Immune-Related Adverse
Events.
Organ System Signs and Symptoms
Organ System Signs and Symptoms Workup
Constitutional Fever, rigors, malaise, fatigue, anorexia, myalgias,
and arthralgias Skin Skin rash (can be >30% Complete skin examination
Skin Rash of BSA) Punch biopsy
Respiratory Tachypnea, hypoxia, pulmonary edema/vascular leak Skin sloughing
syndrome Erythema
Cardiovascular Tachycardia, hypotension, cardiac arrest, Purpura
cardiomyopathy Epidermal detachment
Gastrointestinal Nausea, vomiting, diarrhea, transaminitis, Endocrine Fatigue Visual fields
hyperbilirubinemia Headache TFTs
Hematologic/ Cytopenia, elevated D-dimer, hypofibrinogenemia, Visual disturbances ACTH level
coagulation bleeding Altered mental status FSH and LH levels
Renal Acute kidney injury, hypokalemia, hypophosphatemia Metabolic abnormalities Random cortisol levels
Tachycardia Serum chemistry
Abbreviation: CRS, cytokine release syndrome. Hypotension Brain MRI (pituitary protocol)
CNS Fluctuating muscle Full neurological
weakness assessment
Table 6. Grading of CRS. Motor and sensory Lumbar puncture
neuropathy Nerve conduction studies
Grade Toxicity Hyporeflexia EMG
Respiratory muscle Anti-acetylcholine receptor
Grade 1 Symptoms are not life-threatening and require symptomatic
weakness antibody
treatment only, eg, fever, nausea, fatigue, headache,
Ptosis
myalgias, malaise
Diplopia
Grade 2 Symptoms require and respond to moderate intervention
Dysphagia
Oxygen requirement <40% or
Cardiac Chest pain ECG
Hypotension responsive to fluids or low dose of one
Shortness of breath Echocardiogram
vasopressor or grade 2 organ toxicity
Presyncope Troponin
Grade 3 Symptoms require and respond to aggressive intervention
Syncope Infectious workup
Oxygen requirement 40% or
Arrhythmias Consider myocardial biopsy
Hypotension requiring high dose or multiple vasopressors or
Pulmonary Upper respiratory Infectious workup
Grade 3 organ toxicity or grade 4 transaminitis
infection Chest imaging (preferably
Grade 4 Life-threatening symptoms
Cough high-resolution CT in
Requirement for ventilator support or
Dyspnea severe cases)
Grade 4 organ toxicity (excluding transaminitis)
Chest pain Consider bronchoscopy þ/
Grade 5 Death
Hypoxia  BAL
Abbreviation: CRS, cytokine release syndrome. New findings on chest
imaging
Since tocilizumab is thought to be unlikely to affect T-cell Abbreviations: ACTH, adrenocorticotropic hormone; BAL, bronchoalveolar
expansion, it is usually used as first-line therapy. Steroids are lavage; BSA, body surface area; CNS, central nervous system; CT, computed
used as second-line therapy for patients who do not respond or tomography; ECG, electrocardiogram; EMG, electromyography; FSH, follicle-
stimulating hormone; LH, luteinizing hormone; MRI, magnetic resonance
have an inadequate response to tocilizumab as it could alter the imaging; TFT, thyroid function tests.
effectiveness of the CAR-T cells.51

Neurotoxicity. The complete pathophysiology of neurotoxicity corticosteroids, most commonly dexamethasone. There is a
remains unclear but is thought to be due to vascular instability suggested association of tocilizumab with worsening neuro-
with increased blood–brain barrier permeability leading to ele- toxicity when used for CRS; hence, there is no role for tocili-
vated concentrations of cytokines in the CSF.52 Some of the zumab in isolated neurotoxicity currently. The mainstay of
risk factors associated with neurotoxicity are higher CAR-T- treatment involves steroids, generally dexamethasone, with the
cell dose, severity of disease burden, and lymphodepletion dose depending on the CAR-T-cell construct and the severity of
prior to CAR-T-cell therapy.52 It is thought to be a separate symptoms. A grading severity scale is currently being devel-
phenomenon with variable and unusual presentations but is oped for neurotoxicity.
almost invariably associated with CRS.52
Presenting symptoms vary widely from a simple headache
to confusion, delirium, hallucinations, aphasia, blinking, star-
Immune Checkpoint Blockade–Related Adverse Events
ing, tremors or life-threatening seizures, and brain herniation. Immune checkpoint blockade is rapidly becoming a mainstay
Management of neurotoxicity includes close vigilance for of cancer treatment both as salvage therapy and as initial ther-
early development or progression and prompt treatment with apy, showing a remarkable benefit in the treatment of a wide
Thandra et al 9

Figure 4. Immune-related adverse events caused by immune checkpoint inhibitors.

range of cancer types.53 Immune checkpoint inhibitors (ICPis) the mucosal membranes is essential and a punch biopsy should
are monoclonal antibodies that liberate the host’s immune be performed. Some of the possible presenting signs and symp-
response in order to attack cancer cells. This is done by block- toms along with suggested diagnostic tests are listed in Table 7.
ing intrinsic downregulators of immunity such as cytotoxic T- No matter which organ system is involved, it is always impor-
lymphocyte antigen 4 (CTLA-4) and programmed cell death 1 tant to rule out and empirically treat for possible infections
(PD-1) or its ligand PD-L1.53 As such, these immunotherapies until the workup comes back negative.
expose patients to a wide array of new immune-related adverse As seen in Figure 4, irAEs can affect multiple organ sys-
effects (irAEs) that have the potential to affect any organ sys- tems. Pertinent critical care manifestations include Stevens-
tem and can lead to catastrophic events with prolonged mor- Johnson syndrome, toxic epidermal necrolysis, hypophysitis
bidity or in some cases death. (most commonly associated with anti-CTLA-4 therapy),54
The exact mechanism underlying irAEs is unknown, but it is multiple hormonal deficiencies related to the function of the
believed to be related to how these downregulators of immu- anterior pituitary gland, severe hypoadrenalism with severe
nity (CTLA-4, PD-1, PD-L1) maintain immunologic homeos- electrolyte abnormalities,55 myasthenia gravis, Guillain-Barré
tasis and how that is disrupted by the administration of ICPis.53 syndrome leading to respiratory failure,56 arrhythmias (includ-
Proposed mechanisms include increasing levels of inflamma- ing third-degree atrioventricular block), and cardiomyopathies
tory cytokines (similar to CRS), nonspecifically increasing T- which are associated with high morbidity and mortality if treat-
cell activity against a variety of antigens, increasing levels of ment is not initiated early.57 Immune-related pneumonitis has
preexisting autoantibodies, and enhanced complement- also been reported and can lead to hypoxic respiratory failure
mediated inflammation.53 and acute respiratory distress syndrome.55
Due to their far-reaching effects, it is difficult to devise a In addition to discontinuing the offending agent, the main-
protocol to detect all of the possible irAEs. Instead, the workup stay of treatment for all irAEs is high-dose steroids with con-
is dictated by the organ system involved, for example, if the sideration of additional immunosuppressive agents such as
skin is involved, thorough examination of the skin including infliximab, mycophenolate mofetil, cyclophosphamide, IV
10 Journal of Intensive Care Medicine XX(X)

immunoglobulin, or plasmapheresis in cases refractory to ster- 7. Lyman GH, Kuderer NM, Crawford J, et al. Predicting individual
oids.55 Figure 4 highlights some of the more catastrophic irAEs risk of neutropenic complications in patients receiving cancer
and their management. Recent evidence suggests that there is chemotherapy. Cancer. 2010;117(9):1917-1927.
no difference in clinical outcomes between patients who 8. Kanamaru A, Tatsumi Y. Microbiological data for patients with
require treatment for irAEs and those who do not.53 Addition- febrile neutropenia. Clin Infect Dis. 2004;39(suppl 1):S7-S10.
ally, retrospective data suggest that irAEs associated with one 9. Bow EJ. Management of the febrile neutropenic cancer patient:
class of inhibitor may not necessarily occur during subsequent lessons from 40 years of study. Clin Microbiol Infect. 2005;
treatment with another agent. 11(Suppl 5):24-29.
10. Smith TJ, Khatcheressian J, Lyman GH, et al. 2006 update of
recommendations for the use of white blood cell growth factors:
Conclusion an evidence-based clinical practice guideline. J Clin Oncol. 2006;
Despite overall improved cancer care, classic oncologic emer- 24(19):3187-3205.
gencies continue to occur and many require ICU management. 11. Smith TJ, Bohlke K, Lyman GH, et al. Recommendations for the
Cancer therapies have evolved with more focused tumor tar- use of WBC growth factors: American Society of Clinical Oncol-
geting; however, complications and side effects are not neces- ogy Clinical Practice Guideline update. J Clin Oncol. 2015;
sarily limited and unique emergencies have been identified. 33(28):3199-3212.
Although overall survival from cancer has improved, patients 12. Porcu P, Cripe LD, Ng EW, et al. Hyperleukocytic leukemias and
are commonly admitted to the ICU at some point during their leukostasis: a review of pathophysiology, clinical presentation
clinical course. Therefore, it is crucial for intensivists to have a and management. Leuk Lymphoma. 2000;39(1-2):1-18.
working knowledge of the common oncologic emergencies and 13. Inaba H, Fan Y, Pounds S, et al. Clinical and biologic features and
novel therapy–related complications as they are more likely to treatment outcome of children with newly diagnosed acute mye-
be seen in everyday critical care practice. loid leukemia and hyperleukocytosis. Cancer. 2008;113(3):
522-529.
Declaration of Conflicting Interests 14. Gong J, Wu B, Guo T, Zhou S, He B, Peng X. Hyperleukocytosis:
a report of five cases and review of the literature. Oncol Lett.
The author(s) declared no potential conflicts of interest with respect to
the research, authorship, and/or publication of this article. 2014;8(4):1825-1827. PMID: 25202418.
15. Jain R, Bansal D, Marwaha RK. Hyperleukocytosis: emergency
management. Indian J Pediatr. 2012;80(2):144-148.
Funding 16. Haase R, Merkel N, Diwan O, Elsner K, Kramm CM. Leukapher-
The author(s) disclosed receipt of the following financial support for esis and exchange transfusion in children with acute leukemia and
the research, authorship, and/or publication of this article: Department hyperleukocytosis. A single center experience. Klin Padiatr.
of Anesthesiology & Critical Care Medicine, MSK Cancer Center
2009;221(06):374-378.
Support Grant/Core Grant (P30 CA008748).
17. Chang MC, Chen TY, Tang JL, et al. Leukapheresis and cranial
irradiation in patients with hyperleukocytic acute myeloid leuke-
ORCID iD mia: no impact on early mortality and intracranial hemorrhage.
Sanjay Chawla, MD http://orcid.org/0000-0002-6840-7988 Am J Hematol. 2007;82(11):976-980.
18. Grund FM, Armitage JO, Burns C. Hydroxyurea in the prevention
References of the effects of leukostasis in acute leukemia. Arch Intern Med.
1. Halfdanarson TR, Hogan WJ, Madsen BE. Emergencies in hema- 1977;137(9):1246-1247.
tology and oncology. Mayo Clin Proc. 2017;92(4):609-641. 19. Howard SC, Jones DP, Pui C-H. The tumor lysis syndrome. N
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Can- Engl J Med. 2011;364(19):1844-1854.
cer J Clin. 2018;68(1):7-30. 20. Coiffier B, Riouffol C. Management of tumor lysis syndrome in
3. Taccone FS, Artigas AA, Sprung CL, Moreno R, Sakr Y, Vincent adults. Expert Rev Anticancer Ther. 2007;7(2):233-239.
JL. Characteristics and outcomes of cancer patients in European 21. Rampello E, Fricia T, Malaguarnera M. The management of
ICUs. Crit Care. 2009;13(1):R15. tumor lysis syndrome. Nat Clin Pract Oncol. 2006;3(8):438-447.
4. Soares M, Caruso P, Silva E, et al. Characteristics and outcomes 22. Cairo MS, Bishop M. Tumour lysis syndrome: new therapeutic
of patients with cancer requiring admission to intensive care units: strategies and classification. Br J Haematol. 2004;127(1):3-11.
a prospective multicenter study. Crit Care Med. 2010;38(1):9-15. 23. Coiffier B, Altman A, Pui C-H, Younes A, Cairo MS. Guidelines
5. Halfdanarson TR, Hogan WJ, Moynihan TJ. Oncologic emergen- for the management of pediatric and adult tumor lysis syndrome:
cies: diagnosis and treatment. Mayo Clin Proc. 2006;81(6): an evidence-based review. J Clin Oncol. 2008;26(16):2767-2778.
835-848. 24. Reddy PS, Curtiss EI, O’Toole JD, Shaver JA. Cardiac tampo-
6. Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical practice nade: hemodynamic observations in man. Circulation. 1978;
guideline for the use of antimicrobial agents in neutropenic 58(2):265-272.
patients with cancer: 2010 update by the Infectious Diseases Soci- 25. Spodick DH. Acute cardiac tamponade. N Engl J Med. 2003;
ety of America. Clin Infect Dis. 2011;52(4):e56-e93. 349(7):684-690.
Thandra et al 11

26. Sternbach G. Claude Beck: cardiac compression triads. J Emerg 43. Witmer CM, Huang YS, Lynch K, Raffini LJ, Shah SS. Off-label
Med. 1988;6(5):417-419. recombinant factor VIIa use and thrombosis in children: a multi-
27. Lewis MA, Hendrickson AW, Moynihan TJ. Oncologic emergen- center cohort study. J Pediatr. 2011;158(5):820.e1-825.e1.
cies: pathophysiology, presentation, diagnosis, and treatment. CA 44. Abrams D, Agerstrand CL, Biscotti M, Burkart KM, Bacchetta M,
Cancer J Clin. 2011;61(5):287-314. Brodie D. Extracorporeal membrane oxygenation in the manage-
28. Leimgruber PP, Klopfenstein HS, Wann LS, Brooks HL. The ment of diffuse alveolar hemorrhage. ASAIO J. 2015;61(2):
hemodynamic derangement associated with right ventricular dia- 216-218.
stolic collapse in cardiac tamponade: an experimental echocardio- 45. Solomonov A, Fruchter O, Zuckerman T, Brenner B, Yigla M.
graphic study. Circulation. 1983;68(3):612-620. Pulmonary hemorrhage: a novel mode of therapy. Respir Med.
29. Kim SH, Kwak MH, Park S, et al. Clinical characteristics of 2009;103(8):1196-1200.
malignant pericardial effusion associated with recurrence and 46. Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest.
survival. Cancer Res Treat. 2010;42(4):210. 2015;125(9):3392-3400.
30. Virk SA, Chandrakumar D, Villanueva C, Wolfenden H, Liou K, 47. Annesley CE, Summers C, Ceppi F, Gardner RA. The evolution
Cao C. Systematic review of percutaneous interventions for and future of CAR T cells for B-Cell acute lymphoblastic leuke-
malignant pericardial effusion. Heart. 2015;101(20):1619-1626. mia. Clin Pharmacol Ther. 2018;103(4):591-598.
31. Abdelsalam M, Moritz TA, Snyder JA, Cheriyath P, Spizzieri CL. 48. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predic-
Paradoxical hemodynamic instability complicating pericardial tive biomarkers for cytokine release syndrome after chimeric
window surgery for cardiac tamponade in a cancer patient. Tex antigen receptor T-cell therapy for acute lymphoblastic leukemia.
Heart Inst J. 2012;39(5):711-713. Cancer Discov. 2016;6(6):664-679.
32. Wagner PL, McAleer E, Stillwell E, et al. Pericardial effusions in 49. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells
the cancer population: prognostic factors after pericardial window
expressing CD19 chimeric antigen receptors for acute lympho-
and the impact of paradoxical hemodynamic instability. J Thorac
blastic leukaemia in children and young adults: a phase 1 dose-
Cardiovasc Surg. 2011;141(1):34-38.
escalation trial. Lancet. 2015;385(9967):517-528.
33. Philippakis G, Marinakis A, Manoloudakis N. Pericardial window
50. Lee DW, Gardner R, Porter DL, et al. Current concepts in the
procedures: implications on left ventricular function. Int J Surg
diagnosis and management of cytokine release syndrome. Blood.
Case Rep. 2013;4(4):403-405.
2014;124(2):188-195.
34. Ibrahim WH. Massive haemoptysis: the definition should be
51. Fitzgerald JC, Weiss SL, Maude SL, et al. Cytokine release
revised. Eur Respir J. 2008;32(4):1131-1132.
syndrome after chimeric antigen receptor T cell therapy for
35. Jean-Baptiste E. Clinical assessment and management of massive
acute lymphoblastic leukemia. Crit Care Med. 2017;45(2):
hemoptysis. Crit Care Med. 2000;28(5):1642-1647.
e124-e31.
36. Larici AR, Franchi P, Occhipinti M, et al. Diagnosis and manage-
52. Gust J, Hay KA, Hanafi L-A, et al. Endothelial activation and
ment of hemoptysis. Diagn Interv Radiol. 2014;20(4):299-309.
blood–brain barrier disruption in neurotoxicity after adoptive
37. Haponik EF, Chin R. Hemoptysis: clinicians’ perspectives. Chest.
1990;97(2):469-475. immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;
38. Yoon W, Kim JK, Kim YH, Chung TW, Kang HK. Bronchial and 7(12):1404-1419. PMID: 29025771.
nonbronchial systemic artery embolization for life-threatening 53. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse
hemoptysis: a comprehensive review. Radiographics. 2002; events associated with immune checkpoint blockade. N Engl J
22(6):1395-1409. Med. 2018;378(2):158-168.
39. Peralta AR, Chawla M, Lee RP. Novel bronchoscopic manage- 54. Michot JM, Bigenwald C, Champiat S, et al. Immune-related
ment of airway bleeding with absorbable Gelatin and Thrombin adverse events with immune checkpoint blockade: a comprehen-
Slurry. J Bronchology Interv Pulmonol. 2018;25(3):204-211. sive review. Eur J Cancer. 2016;54:139-148.
(PMID: 29351111). 55. Haanen JBAG, Carbonnel F, Robert C, et al. Management of
40. Fartoukh M, Khoshnood B, Parrot A, et al. Early prediction of in- toxicities from immunotherapy: ESMO Clinical Practice Guide-
hospital mortality of patients with hemoptysis: an approach to lines for diagnosis, treatment and follow-up. Ann Oncol. 2017;
defining severe hemoptysis. Respiration. 2012;83(2):106-114. 28(suppl_4):iv119-iv42.
41. Majhail NS, Parks K, Defor TE, Weisdorf DJ. Diffuse alveolar 56. Hasegawa Y, Kawai S, Ota T, Tsukuda H, Fukuoka M. Myasthe-
hemorrhage and infection-associated alveolar hemorrhage fol- nia gravis induced by nivolumab in patients with non-small-cell
lowing hematopoietic stem cell transplantation: related and lung cancer: a case report and literature review. Immunotherapy.
high-risk clinical syndromes. Biol Blood Marrow Transplant. 2017;9(9):701-707.
2006;12(10):1038-1046. 57. Jain V, Bahia J, Mohebtash M, Barac A. Cardiovascular compli-
42. Collard HR, Schwarz MI. Diffuse alveolar hemorrhage. Clin cations associated with novel cancer immunotherapies. Curr
Chest Med. 2004;25(3):583-92, vii. Treat Options Cardiovasc Med. 2017;19(5):36.

You might also like