Centers for Disease Control and Prevention. Immunization schedules.

http://www.cdc.gov/vaccines/schedules/hcp/imz/child-adolescent.html. Accessed June 10, 2015.

American Cancer Society. Diet and physical activity.

http://www.cancer.org/Cancer/CancerCauses/DietandPhysicalActivity/index. Accessed June 10, 2015.

Libutti SK et al. Cancer of the colon. In: DeVita VT, Jr et al, eds. DeVita, Hellman, and Rosenberg’s Cancer:

Principles & Practice of Oncology. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2011:768.

Scher HI et al. Cancer of the prostate. In: DeVita VT, Jr et al, eds. DeVita, Hellman, and Rosenberg’s Cancer:

Principles & Practice of Oncology. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2011:932.

Morrow M et al. Cancer of the breast: malignant tumors of the breast. In: DeVita VT, Jr et al, eds. DeVita,

Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 9th ed. Philadelphia, PA: Lippincott

Williams & Wilkins; 2011:1117.

Hamajima N et al. Alcohol, tobacco and breast cancer—collaborative reanalysis of individual data from 53

epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the disease. Br

J Cancer. 2002;87:1234.

Gilchrest BA et al. The pathogenesis of melanoma induced by ultraviolet radiation. N EnglJ Med. 1999;340:1341.

American Cancer Society. Skin cancer prevention and early detection.

http://www.cancer.org/acs/groups/cid/documents/webcontent/003184-pdf.pdf. Accessed June 10, 2015.

Christensen D. Data still cloudy on association between sunscreen use and melanoma risk. J Natl Cancer Inst.

2003;95:932.

NCCN Clinical Practice Guidelines In Oncology (NCCN Guidelines®)for Detection, Prevention, and Risk of

Cancer. http://www.nccn.org/professionals/physician’gls/fguidelines.asp. Accessed June 10, 2015.

American Cancer Society. Imaging (Radiology) Tests for Cancer.

http://www.cancer.org/treatment/understandingyourdiagnosis/examsandtestdescriptions/imagingradiologytests/iAccessed June 10, 2015.

Oken MM et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol.

1982;5:649.

Heron DE et al. Radiation medicine innovations for the new millennium. J Natl Med Assoc. 2003;95:55.

Durante M, Loeffler JS. Charged particles in radiation oncology. Nat Rev Clin Oncol. 2010;7:37.

National Cancer Institute. Dictionary of cancer terms. http://www.cancer.gov/dictionary/. Accessed June 10,

2015.

Facts & Comparisons eAnswers [online]. 2015. Alphen aan den Rijn, South Holland: Wolters Kluwer Health.

Accessed June 10, 2015.

Skipper HE et al. Experimental evaluation of potential anticancer agents: XIII. On the criteria and kinetics

associated with “curability” of experiment leukemia. Cancer Chemother Rep. 1964;35:1.

Skipper HE. Reasons for success and failure in treatment of murine leukemias with the drugs now employed in

treating human leukemias. In: Skipper HE, ed. Cancer Chemotherapy. Ann Arbor, MI: University Microfilms

International; 1978;1.

Sparreboom A, Baker SD. Cancer therapeutics: pharmacokinetics and pharmacodynamics of anticancer drugs.

In: DeVita VT, Jr et al, eds. DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 9th

ed. Philadelphia, PA: Lippincott Williams & Wilkins. 2011:174.

Wood WC et al. Dose and dose intensity of adjuvant chemotherapy for stage II, node-positive breast carcinoma

[published correction appears in N EnglJ Med. 1994;331:139]. N EnglJ Med. 1994;330:1253.

Lorigan P et al. Randomized phase III trial of dose-dense chemotherapy supported by whole-blood hematopoietic

progenitors in better-prognosis small-cell lung cancer [published correction appears in J Natl Cancer Inst.

2005;97: 941]. J Natl Cancer Inst. 2005;97:666.

Balducci L et al. Management of adverse effects of treatment: neutropenia and thrombocytopenia. In: DeVita

VT, Jr et al, eds. DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 9th ed.

Philadelphia, PA: Lippincott Williams & Wilkins; 2011:1960.

Lazarus HM et al. Stem cell transplantation: autologous stem cell transplantation. In: DeVita VT, Jr et al, eds.

DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 9th ed. Philadelphia, PA:

Lippincott Williams & Wilkins; 2011:1907.

Mormont MC, Levi F. Cancer chronotherapy: principles, applications, and perspectives. Cancer. 2003;97:155.

Goldie JH, Coldman AJ. A mathematic model for relating the drug sensitivity of tumors to the spontaneous

mutation rate. Cancer Treat Rep. 1979;63:1727.

Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med. 2002;53:615.

Leonard GD et al. The role of ABC transporters in clinical practice. Oncologist. 2003;8:411.

Pessina A et al. Altered DNA-cleavage activity of topoisomerase from WEHI-3B leukemia cells with specific

resistance to ciprofloxacin. Anticancer Drugs. 2001;12:441.

DeVita VT, Jr, Chu E. Principles of medical oncology: basic principles. In: DeVita VT, Jr et al, eds. DeVita,

Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 8th ed. Philadelphia, PA: Lippincott

Williams & Wilkins; 2008:338.

Ali S, Coombes RC. Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev

Cancer. 2002;2:101.

Higano CS et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active

cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670.

Kirkwood J. Cancer immunotherapy: the interferon-alpha experience. Semin Oncol. 2002;29(3, Suppl 7):18.

Atkins MB. Interleukin-2: clinical applications. Semin Oncol. 2002;29(3, Suppl 7):12.

Lotze MT et al. Lysis of fresh and cultured autologous tumor by lymphocytes cultured in T-cell growth factor.

Cancer Res. 1981;41:4420.

Rayner AA et al. Lymphokine-activated killer (LAK) cells: analysis of factors relevant to the immunotherapy of

human cancer. Cancer. 1985;55:1327.

Rosenberg SA et al. Aprogress report on the treatment of 157 patients with advanced cancer using lymphokineactivated killer cells and interleukin-2 or high-dose interleukin-2 alone. N EnglJ Med. 1987;316:889.

Rosenberg SA et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using

high-dose bolus interleukin 2. JAMA. 1994;271:907.

Eisenhauer EA et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).

Eur J Cancer. 2009;45:228.

National Cancer Institute. Common Terminology Criteria for Adverse Events Version 4.0.

http://evs.nci.nih.gov/ftp1/CTCAE. Accessed June 10, 2015.

Cella D et al. Advances in quality of life measurements in oncology patients. Semin Oncol. 2002;29(3, Suppl

8):60.

Fojo AT, Bates SE. Cancer therapeutics: assessment of clinical response. In: DeVita VT, Jr et al, eds. DeVita,

Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 9th ed. Philadelphia, PA: Lippincott

Williams & Wilkins; 2011:311.

American Society of Health-System Pharmacists. ASHP Guidelines on Preventing Medication Errors With

Antineoplastic Agents. http://www.ashp.org/DocLibrary/BestPractices/MedMisGdlAntineo.aspx.

Accessed June 10, 2015.

Neuss MN et al. 2013 Updated American Society of Clinical Oncology/Oncology Nursing Society Chemotherapy

Administration Safety Standards Including Standards for the Safe Administration and Management of Oral

Chemotherapy. J Oncol Practice. 2013;9(2s):5–13.

Cohen MR et al. Preventing medication errors in cancer chemotherapy. Am J Health Syst Pharm. 1996;53:737.

Attilio RM. Caring enough to understand: the road to oncology medication error prevention. Hosp Pharm.

1996;31:17.

Goldspiel BR et al. Preventing chemotherapy errors: updating guidelines to meet new challenges. Am J Health

Syst Pharm. 2015;72:668–669.

American Society of Health-System Pharmacists. ASHP guidelines on handling hazardous drugs. Am J Health

Syst Pharm. 2006;63:1172.

Selevan SG et al. A study of occupational exposure to antineoplastic drugs and fetal loss in nurses. N EnglJ Med.

1985;313:1173.

National Institute for Occupational Safety and Health. NIOSH alert: preventing occupational exposures to

antineoplastic and other hazardous drugs in health care settings. http://www.cdc.gov/niosh/docs/2004-

165/pdfs/2004-165.pdf. Accessed June 10, 2015.

Hemminki K et al. Spontaneous abortions and malformations in the offspring of nurses exposed to anaesthetic

gases, cytostatic drugs, and other potential hazards in hospitals, based on registered information of outcome. J

Epidemiol Community Health. 1985;39:141.

Goodin S et al. Safe handling of oral chemotherapeutic agents in clinical practice: recommendations from an

international pharmacy panel. J Oncol Pract. 2011;7:7.

American Society of Health-Systems Pharmacists. The ASHP Discussion Guide on USP Chapter <797> for

Compounding Sterile Preparations. Summary of Revisions to USP Chapter <797>.

http://www.ashp.org/s_ashp/docs/files/discguide797-2008.pdf. Accessed June 10, 2015.

p. 1967

Myelosuppression is one of the most common toxicities of cytotoxic

anticancer therapy. Cytotoxic therapy may affect any one or all of the

bone marrow cell lines including erythrocytes, neutrophils, and platelets.

Complications from anemia, neutropenia, and thrombocytopenia can

cause significant morbidity and mortality, including complications from

bleeding and infections. Prophylactic administration of growth factors

may protect against the myelosuppressive effects of chemotherapy.

Case 94-1 (Questions 1–4)

The upper and lower gastrointestinal (GI) tract is highly susceptible to

cytotoxic chemotherapy, and toxicities can include nausea and vomiting,

mucositis, xerostomia, constipation, and diarrhea. Efficacy of preventive

strategies is limited; thus, supportive treatment is a foundation for

managing adverse effects in patients.

Case 94-2 (Questions 1–3),

Case 94-3 (Question 1)

Dermatologic toxicities from anticancer therapies include alopecia, nail

changes, hyperpigmentation, radiation sensitivities, hand-foot syndrome,

dry skin, and papular–pustular (acneiform) rash. Most of the toxicities

are cosmetic and resolve upon discontinuation of the agent. Onset and

duration of dermatologic toxicities depend on the causative agent.

Treatment is supportive.

Case 94-4 (Questions 1–4),

Tables 94-1 and 94-2

Extravasation is the unintended infiltration of an agent into the tissue

area surrounding the vein during an infusion. Several anticancer agents

demonstrate vesicant properties that can cause tissue necrosis and

permanent damage to the extravasated area. Cases of extravasation

require emergency treatment that is agent-specific, including elevation

of the extremity, extraction of the agent, hot or cold treatment, and

potential antidotes.

Case 94-4 (Questions 5–7),

Tables 94-3–94-5

Many anticancer therapies are associated with immunoglobulin-Emediated hypersensitivity reactions. Monoclonal antibodies including

rituximab, trastuzumab, cetuximab, and ofatumumab are among some of

the agents most commonly associated with reactions. Reactions

commonly occur with the first dose, and they may be minimized with

premedications such as acetaminophen, diphenhydramine, and

corticosteroids.

Case 94-5 (Question 1),

Tables 94-6 and 94-7

Multiple types of central nervous system (CNS) toxicity, including

encephalopathy, cerebellar toxicity, and peripheral neuropathy, are

Case 94-6 (Questions 1–4),

Table 94-8

associated with anticancer agents and differ in presentation between

various agents. Most of the symptoms of neurotoxicity are reversible

over time, but modifications to regimens including discontinuation of

agents or reduction of doses may be necessary.

Cardiomyopathy, arrhythmias, and hypertension are common types of

cardiotoxicity observed in patients receiving infusion and oral anticancer

therapies. Anthracycline-induced cardiomyopathy is closely linked to a

patient’s cumulative dose and may be treated with current heart failure

medications.

Case 94-7 (Questions 1–4)

p. 1968

p. 1969

Several anticancer agents carry severe risks of nephrotoxicity and

bladder toxicity for which preventive measures are necessary. Cisplatin

is one of the most nephrotoxic agents. Preventive strategies for cisplatin

nephrotoxicity include normalsaline hydration, mannitol, and amifostine,

and methotrexate-induced nephrotoxicity may be prevented with

alkalinization of the urine and leucovorin rescue. Ifosfamide-induced

hemorrhagic cystitis may be prevented with concomitant use of mesna.

Case 94-8 (Questions 1, 2),

Case 94-9 (Questions 1–3),

Table 94-9

Many anticancer medications demonstrate organ-specific toxicities, such

as pulmonary fibrosis caused by bleomycin and transaminitis caused by

cytarabine. Specific treatments for resolution of these toxicities do not

exist; rather, supportive care for symptoms is necessary. If adverse

effects do not resolve, discontinuation of therapy or dose modifications

may be warranted.

Case 94-10 (Questions 1–4),

Case 94-11 (Question 1),

Tables 94-10–94-13

Many anticancer agents cause long-term complications after therapy,

including treatment-related acute myeloid leukemia, lymphomas, bladder

cancer, and bone sarcoma. The risks of secondary malignancies should

be considered in assessing the adverse effect profile within the risks and

benefits of specific types of therapy.

Case 94-12 (Questions 1, 2)

Cytotoxic chemotherapy is potentially gonadotoxic with use of specific

agents. Sex, age, agent, and cumulative dose are factors in determining

the risk of infertility. Methods of preserving fertility should be discussed

with patients before initiating therapy.

Case 94-13 (Question 1),

Case 94-14 (Questions 1–3)

Cytotoxic, targeted anticancer and immunotherapy agents are toxic to cancer cells and

also to various host tissues and organs. The adverse effects of anticancer therapies

are considered for both infusional and oral therapies and can be classified as

common and acute toxicities, specific organ toxicities, and long-term complications.

Common and acute toxicities generally occur as a result of inhibition of host-cell

division. Host tissues most susceptible to cytotoxic agents include tissues with

renewal cell populations, such as lymphoid tissues, bone marrow, and epithelium of

the GI tract and skin. Some other common and acute toxicities (e.g., nausea and

vomiting, hypersensitivity reactions) frequently occur in patients shortly after

therapy. Specific organ toxicities often are attributed to a unique uptake or a selective

toxicity of the anticancer agent to the organ. Long-term complications are toxicities

that occur months to years after anticancer therapy. These long-term toxicities occur

secondary to continued immunodeficiencies or from permanent damage to the organ

cells from the specific therapy. Regardless of the type of toxicities observed, most

are classified for severity by the National Cancer Institute (NCI) Common

Terminology Criteria for Adverse Events. This classification creates a common

method for classification of events in clinical trials and for management of toxicities

that occur for patients receiving standard of care regimens.

1 These criteria may be

accessed at the NCI website

(http://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm).

The toxicities associated with anticancer therapies are the most important factors

limiting the use of potentially curative doses. Therefore, all discussions regarding the

benefits of anticancer agents must include a discussion of toxicities associated with

their use. Concerns regarding the toxicities of therapy include the incidence,

predictability, severity, and reversibility of the adverse effects. In addition, the

specific agent, dose intensity, and treatment duration can influence the incidence of

several adverse effects. Although the incidence and predictability may be well

defined in specific patient populations, the incidence often varies depending on

individual susceptibility. The specific adverse effects that an individual patient will

experience may be difficult to predict. Because several toxicities have well-defined

characteristics, clinicians should be aware of the most common adverse effects.

Clinicians should also be aware of patient-specific factors, such as the stage of

disease, concomitant illnesses, and concurrent medications, which could cause signs

or symptoms that mimic the adverse effects associated with anticancer therapy. Many

patients have disease involvement which may impair organ function. In addition,

most patients with cancer receive many other medications, including antibiotics and

analgesics, which may cause additional adverse effects or interact with anticancer

agents. When a patient reports a new symptom, it may be difficult to determine

whether it is secondary to anticancer therapy, concurrent medications, or disease

progression.

COMMON AND ACUTE TOXICITIES

Hematologic Toxicities

The bone marrow contains a population of pluripotent stem cells capable of selfrenewal and differentiation into any mature blood cell. Their progeny commits to

either the myeloid or the lymphoid cell line. The myeloid stem cell further commits to

developing into an erythrocyte (red blood cell (RBC)), megakaryocyte (platelet), or

granulocyte (white blood cell (WBC)). Multiple types of granulocytes exist including

neutrophils, basophils, and eosinophils, although neutrophils are the most common

type.

After committing to a particular cell line, bone marrow precursor cells undergo a

series of divisions (mitosis) to increase the number of cells. The cells then undergo

several developmental stages to mature and differentiate into their final forms

(postmitotic) and leave the bone marrow. The total time required for a cell to pass

through the mitotic and postmitotic pool under normal resting conditions is

approximately 10 to 14 days. This process is regulated by several cytokines; although

many cytokines have been identified, only a few growth factors are now produced

through recombinant DNA technology. These growth factors can expand the mitotic

pool and accelerate maturation and differentiation. Ultimately, these growth factors

decrease the total time spent in these stages to approximately 5 to 7 days.

The development and circulating life span of hematopoietic cell lines determine

the severity of the depression of that cell line (nadir, lowest point) and the time

course of peripheral cytopenias. Because RBCs survive approximately 120 days in

the peripheral blood, clinically significant anemia is unlikely if production is

p. 1969

p. 1970

impaired for a short period of time. Instead, anemia usually develops slowly after

several courses of cytotoxic therapy. In contrast, platelets survive approximately 10

days, and granulocytes survive only 6 to 8 hours. Hence, neutropenia generally

occurs before thrombocytopenia, but both may be observed after the first or

subsequent courses of cytotoxic chemotherapy. The clinician may have to adjust the

subsequent chemotherapy dosage based on nadir depth and slow recovery. Lifethreatening neutropenia or thrombocytopenia often necessitates some action to

minimize the risk of adverse effects with additional courses of cytotoxic

chemotherapy. To diminish these effects, one can reduce the dose, delay therapy until

recovery, or administer colony-stimulating factors (CSFs). The availability of CSFs

provides an alternative approach to preventing severe neutropenia.

MYELOSUPPRESSION

CASE 94-1

QUESTION 1: J.T., a 68-year-old, 59-kg man with no significant past medical history, presents to the

university hospital with complaints of cough and shortness of breath (SOB). Chest radiograph reveals a lesion in

the right upper lobe; surgical resection and cytologic examination are positive for non-small-cell lung cancer

(NSCLC). A workup for metastases is negative. J.T. is diagnosed with early-stage (stage II) NSCLC. His

physicians plan to initiate adjuvant chemotherapy of carboplatin targeted to an area under the concentration–

time curve (AUC) of 6 mg/mL × minute and paclitaxel 135 mg/m

2 on day 1. Discuss the toxicities that might be

expected to occur with this regimen. What effects on the bone marrow can be anticipated and how might they

clinically appear in J.T.? What factors can influence the incidence and severity of these adverse effects? When

can J.T. expect these effects to occur?

1.

2.

3.

4.

Although several toxicities are commonly associated with carboplatin and

paclitaxel, the most predictable and severe toxicity associated with this regimen is

myelosuppression. This chemotherapy regimen can significantly affect any cell line,

including RBCs, neutrophils, and platelets, and the cytopenias can cause significant

morbidity or mortality. Decreased RBCs can cause anemia, and patients usually

present with fatigue and decreased exercise tolerance. Having low neutrophil counts

significantly increases a patient’s risk for bacterial infections. Moreover, reduced

platelets can cause thrombocytopenia, which can cause bleeding from the GI and

genitourinary tracts.

Both patient-related and agent-related factors can significantly influence the degree

of cytopenia a patient faces after cytotoxic therapy. Agent-related factors include the

specific agent, dose intensity, and dose density. Because most anticancer treatments

are not given as a single agent, the effects of concurrent cytotoxic therapies may

intensify the myelosuppressive effect of an individual agent. Host factors that

specifically may affect the cellularity of the bone marrow compartment also influence

the degree of cytopenia. They include the following:

Patient age. Younger patients are generally better able to tolerate cytotoxic

chemotherapy than elderly patients because they have a more cellular marrow with

a decreased percentage of marrow fat.

Bone marrow reserve. Certain diseases might present with tumor cells in the bone

marrow, such as leukemias and some lymphomas, in which case the bone marrow

does not have a healthy reserve of normal hematopoietic cells to help in the

recovery process.

The degree of myelosuppression from previous cytotoxic chemotherapy, radiation

therapy, or both. Prior cytotoxic chemotherapy and radiation therapy to fields

involving marrow-producing bone (pelvic bone and sternum) reduce bone marrow

reserves.

The ability of the liver or kidney to metabolize and excrete the compounds

administered. If agents are administered to patients with specific organ

insufficiencies (i.e., renal or hepatic), slower clearance, resulting in increased

systemic exposure, can occur. This can cause greater toxicities, including longer

cytopenias.

These factors, along with the kinetics of the stem cells, can help clinicians predict

the severity and duration of cytopenia observed after therapy.

With most myelosuppressive agents, the patient’s WBC and platelet counts begin

to fall within 5 to 7 days of cytotoxic therapy administration, reach a nadir within 7

to 10 days, and recover within 14 to 26 days. Phase-specific cytotoxic chemotherapy

agents, such as the vinca alkaloids and antimetabolites, cause a fairly rapid onset of

cytopenia that recovers faster than those occurring after treatment with phasenonspecific agents, such as alkylating agents and anthracyclines. For poorly

understood reasons, nitrosoureas typically produce severe, delayed neutropenia and

thrombocytopenia 4 to 6 weeks after therapy. Other agents that exhibit this pattern

include mitomycin and mechlorethamine. All of these anticancer agents exert their

cytotoxic effects during the resting phase of the cell cycle. The nitrosoureas, as well

as mitomycin and mechlorethamine, can cause two neutropenic nadirs; the first nadir

occurs at the conventional time expected for phase-nonspecific agents and the second

nadir occurs approximately 4 to 6 weeks after therapy. Many combination regimens

with these agents are therefore given for 6-week cycles to avoid treatment before the

second nadir. However, most other myelosuppressive regimens can be safely given

every 3 to 4 weeks. The majority of the targeted therapies do not suppress bone

marrow production, because they are designed to inhibit a specific molecular

pathway rather than proliferating cells in general. Because of their minimal

myelosuppressive effects, they may be desirable agents to add to regimens that are

known to cause cytopenias.

Each of the agents included in J.T.’s regimen has marked myelosuppressive

activity. His elderly age may place him at higher risk for exhibiting

myelosuppression. J.T. should be carefully counseled to contact his physician or

report to the emergency department if he experiences signs or symptoms of an

infection (including fever) or bleeding. Typically, these symptoms occur 10 to 14

days after the first day of chemotherapy.

Prevention of Neutropenia

CASE 94-1, QUESTION 2: About 9 days after the first course of cytotoxic chemotherapy, J.T. experienced

a severe sore throat and fever. He was admitted to the hospital and treated with intravenous (IV) antibiotics. At

the time, his WBC count was 300 cells/μL; absolute neutrophil count (ANC), 50 cells/μL; platelets, 102,000

cells/μL; and hemoglobin (Hgb), 11 g/dL. His fever resolved after 3 days, and all cultures were negative for

bacterial growth. It is now 3 weeks after chemotherapy, and he is scheduled to receive a second course. Should

he receive the same doses he was given initially?

One option would be to reduce (usually by 25%) the dose of each agent for all

subsequent cycles. Although a dose reduction can clearly cause less neutropenia, it

can also compromise the response and survival of patients with chemotherapysensitive tumors. Because J.T.’s cancer (i.e., early-stage NSCLC) is both

chemosensitive and potentially curable, a dosage reduction is undesirable. To

minimize the risk of neutropenia with future therapy, CSFs can be administered to

J.T. to prevent potential complications associated with neutropenia.

p. 1970

p. 1971

Prophylactic administration of CSFs can be used to reduce the myelosuppressive

effects of cytotoxic chemotherapy. Several CSFs—granulocyte colony-stimulating

factors (G-CSFs [filgrastim, tbo-filgrastim, and filgrastim-sndz]), granulocytemacrophage colony-stimulating factor (GM-CSF [sargramostim]), and a pegylated

long-acting form of filgrastim, pegfilgrastim—are available in the United States.

Pegfilgrastim was developed with the aim of providing the same pharmacologic

benefit as filgrastim while offering the advantage and convenience of fewer

injections. The FDA approved filgrastim-sndz as the first biosimilar in the United

States in March 2015. Although tbo-filgrastim was FDA-approved in 2012, it is not

considered a biosimilar because it was filed as a Biologics License Application,

prior to the establishment of the FDA biologics approval pathway. Clinical safety,

particularly related to immunogenicity, is a concern with new approval of

biosimilars, and assessment of tolerability and safety should occur in clinical

practice.

2 The evidence-based clinical practice guidelines for the use of CSFs have

been developed by the American Society of Clinical Oncology (ASCO).

3 These

guidelines recommend primary prophylaxis for all patients receiving chemotherapy

regimens that have been previously reported to cause a febrile neutropenia incidence

of approximately 20%. A CSF used in these patients can reduce both the incidence of

febrile neutropenia and need for hospitalizations and broad-spectrum antibiotics.

However, CSF usage has not been shown to lead to better tumor response or higher

overall survival. Two randomized Phase III clinical trials have shown that the risk of

neutropenic fever is reduced when primary prophylaxis is used in regimens with a

known neutropenia incidence of approximately 20%. In one trial, 928 patients with

breast cancer receiving docetaxel 100 mg/m2 every 21 days were randomly assigned

to receive placebo or pegfilgrastim 6 mg subcutaneously (SC) 24 hours after

chemotherapy. Patients who received pegfilgrastim had a lower incidence of febrile

neutropenia (1% vs. 17%, respectively) and hospitalizations (1% vs. 14%,

respectively).

4 A trial in patients (n = 171) with small-cell lung cancer (SCLC)

receiving a dose-intense regimen containing cyclophosphamide 1,000 mg/m2 on day

1, doxorubicin 45 mg/m2 on day 1, and etoposide 100 mg/m2 on days 1 to 3 every 21

days was conducted. Patients were randomly assigned to receive prophylactic

antibiotics with or without filgrastim. The rate of febrile neutropenia over all five

cycles was 32% with prophylactic antibiotics without filgrastim versus 18% with

antibiotics and filgrastim.

5 A meta-analysis of 17 randomized trials including 3,493

adult patients with solid tumors and lymphomas showed that the use of filgrastim as

primary prophylaxis reduced the risk of febrile neutropenia and improved the rate of

full-dose cytotoxic chemotherapy given on schedule. Additionally, the investigators

in the meta-analysis observed a significant reduction in the risk of infection-related

mortality.

6 The use of CSFs for secondary prophylaxis is recommended by ASCO for

those patients who experience a neutropenic complication from the prior cycle,

where primary CSF was not used, when a reduced dose or treatment delay may

negatively impact the survival/treatment outcome.

3 Because the regimen J.T. received

does not typically produce a 20% incidence of febrile neutropenia, a CSF was not

recommended for him after his first course of cytotoxic chemotherapy. Now that J.T.

has experienced febrile neutropenia and he has a potentially curable malignancy, a

CSF is indicated with subsequent courses of chemotherapy to prevent additional

febrile episodes.

Dosing of Colony-Stimulating Factors

CASE 94-1, QUESTION 3: How should a CSF be dosed in J.T. to reduce the severity of chemotherapyinduced neutropenia?

The recommended initial dose of filgrastim, tbo-filgrastim or filgrastim-sndz, is 5

mcg/kg/day as a single daily subcutaneous (SC) injection, and of sargramostim, 250

mcg/m2

/day SC beginning 24 to 72 hours following the administration of myelotoxic

chemotherapy. The ASCO guidelines state that rounding the dose of either weightbased filgrastim or sargramostim to the nearest vial size may enhance patient

convenience and reduce cost without clinical detriment. Because commercially

available vials or syringes contain either 300 or 480 mcg of filgrastim, adult patients

weighing less than 75 kg should receive 300 mcg daily and adult patients weighing

more than 75 kg should receive 480 mcg daily.

3 Because of differences in

commercially available vial sizes, the weight break point for sargramostim is slightly

different; patients who weigh more than 60 kg should receive 500 mcg daily and

patients who weigh less than 60 kg should receive 250 mcg daily. Pegfilgrastim is

given once per cycle, 24 to 72 hours following administration of myelotoxic

chemotherapy and not less than 14 days prior to the next treatment, as 6 mg SC in

adult patients regardless of patient weight. A new formulation of pegfilgrastim was

recently approved that is a timed automated-inject device that delivers the

medication 27 hours after it is activated.

7

The ASCO guidelines also recommend a shorter duration of treatment than the

manufacturers. The manufacturers recommend that therapy with filgrastim or

sargramostim continues until the patient’s neutrophil count is greater than 10,000

cells/μL after the expected chemotherapy nadir. This is based on the observation that

the neutrophil count falls roughly 50% after discontinuing a CSF. The risk of

bacterial infection is highest, however, in patients with neutrophil counts of less than

500 to 1,000 cells/μL; patients with neutrophil counts greater than that are not thought

to be at high risk for experiencing bacterial infections. Thus, many clinicians elect to

discontinue the CSF when the neutrophil count reaches 2,000 to 4,000 cells/μL after

the chemotherapy nadir. This reduces the number of treatment days and the cost

associated with therapy while concurrently reducing the excessive risk for bacterial

infections. The ASCO guidelines support this recommendation to discontinue CSF

earlier.

In summary, J.T. should be given either filgrastim, tbo-filgrastim or filgrastim-sndz

300 mcg/day, or sargramostim 250 mcg/day SC beginning the day after his last dose

of chemotherapy. Treatment should continue until the ANC is greater than 2,000 to

4,000 cells/μL. Filgrastim is used much more widely than sargramostim.

Alternatively, J.T. could receive a single 6-mg injection of pegfilgrastim the day after

chemotherapy administration. This more convenient administration is due to

pegfilgrastim’s favorable pharmacokinetics. Self-regulation of pegfilgrastim serum

levels, related to its pegylation, is almost entirely dependent on neutrophil receptor-

mediated clearance. Serum levels of pegfilgrastim will remain elevated during

neutropenia induced by chemotherapy, and decline on recovery of neutrophil counts.

8

Aside from high cost and inconvenience, the only negative effect of filgrastim or

sargramostim therapy is mild transient bone pain. Bone pain is most commonly

experienced when patients begin to recover peripheral blood cells after their nadir.

The proposed mechanism suggests the stimulatory effect of CSF on granulopoiesis

causes the pain. Most patients commonly report pain in bone marrow-rich areas, such

as the sternum and pelvic regions. They should be advised that the bone pain

experienced during marrow recovery is normal and usually is relieved with

analgesic agents.

Treatment of Fever and Neutropenia with a Colony-Stimulating Factor

CASE 94-1, QUESTION 4: If J.T. was not given filgrastim and presented with febrile neutropenia, would

CSF therapy be helpful?

p. 1971

p. 1972

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more