Your activity: 2 p.v.

Cytarabine (conventional): Drug information

Cytarabine (conventional): Drug information
(For additional information see "Cytarabine (conventional): Patient drug information" and see "Cytarabine (conventional): Pediatric drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
ALERT: US Boxed Warning
Experienced physician:

Only health care providers experienced in cancer chemotherapy should use cytarabine.

Drug toxicities:

For induction therapy, patients should be treated in a facility with laboratory and supportive resources sufficient to monitor drug tolerance and protect and maintain a patient compromised by drug toxicity. The main toxic effect of cytarabine is bone marrow suppression with leukopenia, thrombocytopenia, and anemia. Less serious toxicity includes nausea, vomiting, diarrhea and abdominal pain, oral ulceration, and hepatic dysfunction.

The health care provider must judge possible benefit to the patient against known toxic effects of this drug in considering the advisability of therapy with cytarabine. Before making this judgment or beginning treatment, the health care provider should be familiar with the following text.

Brand Names: Canada
  • PMS-Cytarabine
Pharmacologic Category
  • Antineoplastic Agent, Antimetabolite;
  • Antineoplastic Agent, Antimetabolite (Pyrimidine Analog)
Dosing: Adult

Note: Antiemetics may be recommended to prevent nausea and vomiting; IV doses >1,000 mg/m2 are associated with a moderate emetic potential (ASCO [Hesketh 2020]; MASCC/ESMO [Roila 2016]). Consider hydration and antihyperuricemic therapy to prevent tumor lysis syndrome.

Acute lymphoblastic leukemia

Acute lymphoblastic leukemia (off-label dosing):

Induction regimen, relapsed or refractory: IV: 3,000 mg/m2 over 3 hours daily for 5 days (in combination with idarubicin [day 3]) (Weiss 2002).

Dose-intensive regimen: IV: 3,000 mg/m2 over 2 hours every 12 hours days 2 and 3 (4 doses/cycle) of even numbered cycles (in combination with methotrexate; alternates with Hyper-CVAD) (Kantarjian 2000).

CALGB 8811 regimen (Larson 1995):

Early-intensification phase: SUBQ: 75 mg/m2/dose days 1 to 4 and 8 to 11 (4-week cycle; repeat once).

Late-intensification phase: SUBQ: 75 mg/m2/dose days 29 to 32 and 36 to 39.

Linker protocol: Adults <50 years of age: IV: 300 mg/m2/day days 1, 4, 8, and 11 of even numbered consolidation cycles (in combination with teniposide) (Linker 1991).

CALGB 10403 regimen (Stock 2019; as part of multi-agent, multicourse chemotherapy; refer to protocol for further details):

Adults <40 years of age:

Remission consolidation phase (course 2): IV, SUBQ: 75 mg/m2 on days 1 to 4, 8 to 11, 29 to 32, and 36 to 39.

Delayed intensification phase (course 4): IV, SUBQ: 75 mg/m2 on days 29 to 32 and 36 to 39.

Acute myeloid leukemia remission induction

Acute myeloid leukemia remission induction: Standard-dose (manufacturer's labeling; in combination with other chemotherapy agents): IV: 100 mg/m2/day continuous infusion for 7 days or 200 mg/m2/day continuous infusion (as 100 mg/m2 over 12 hours every 12 hours) for 7 days.

7 + 3 regimens (a second induction course may be administered if needed; refer to specific references): IV: 100 mg/m2/day continuous infusion for 7 days (in combination with daunorubicin or idarubicin or mitoxantrone) (Arlin 1990; Dillman 1991; Fernandez 2009; Vogler 1992; Wiernik 1992) or (Adults <60 years) 200 mg/m2/day continuous infusion for 7 days (in combination with daunorubicin) (Dillman 1991).

Low-intensity therapy (off-label dosing):

Adults ≥65 years of age: SUBQ: 20 mg/m2/day for 14 days out of every 28-day cycle for at least 4 cycles (Fenaux 2010) or 10 mg/m2 every 12 hours for 21 days; if complete response not achieved, may repeat a second course after 15 days (Tilly 1990).

Adults ≥60 years of age (and ineligible for intensive chemotherapy): SUBQ: 20 mg/m2 once daily on days 1 to 10 every 28 days (in combination with venetoclax) until disease progression or unacceptable toxicity (Wei 2020).

Adults ≥55 years of age (and unsuitable for intensive therapy): SUBQ: 20 mg (flat dose) twice daily on days 1 to 10 every 28 days (in combination with glasdegib) until disease progression or unacceptable toxicity (Cortes 2019).

Acute myeloid leukemia consolidation

Acute myeloid leukemia consolidation (off-label use):

5 + 2 regimens: IV: 100 mg/m2/day continuous infusion for 5 days (in combination with daunorubicin or idarubicin or mitoxantrone) (Arlin 1990; Wiernik 1992).

5 + 2 + 5 regimen: IV: 100 mg/m2/day continuous infusion for 5 days (in combination with daunorubicin and etoposide) (Bishop 1996).

Single-agent: Adults ≤60 years of age: IV: 3,000 mg/m2 over 3 hours every 12 hours on days 1, 3, and 5 (total of 6 doses); repeat every 28 to 35 days for 4 courses (Mayer 1994).

Acute myeloid leukemia salvage treatment

Acute myeloid leukemia salvage treatment (off-label use):

CLAG regimen: IV: 2,000 mg/m2/day over 4 hours for 5 days (in combination with cladribine and G-CSF); may repeat once if needed (Wrzesień-Kuś 2003).

CLAG-M regimen: IV: 2,000 mg/m2/day over 4 hours for 5 days (in combination with cladribine, G-CSF, and mitoxantrone); may repeat once if needed (Wierzbowska 2008).

FLAG regimen: IV: 2,000 mg/m2/day over 4 hours for 5 days (in combination with fludarabine and G-CSF); may repeat once if needed (Montillo 1998).

GCLAC regimen: Adults 18 to 70 years (Becker 2011):

Induction: IV: 2,000 mg/m2 over 2 hours once daily for 5 days (in combination with clofarabine and filgrastim; administer 4 hours after initiation of clofarabine); may repeat induction once if needed.

Consolidation: IV: 1,000 mg/m2 over 2 hours once daily for 5 days (in combination with clofarabine and filgrastim; administer 4 hours after initiation of clofarabine) for 1 or 2 cycles.

HiDAC (high-dose cytarabine) ± an anthracycline: IV: 3,000 mg/m2 over 1 hour every 12 hours for 6 days (total of 12 doses) (Herzig 1985).

MEC regimen: IV: 1,000 mg/m2/day over 6 hours for 6 days (in combination with mitoxantrone and etoposide) (Amadori 1991) or

Adults <60 years of age: IV: 500 mg/m2/day continuous infusion days 1, 2, and 3 and days 8, 9, and 10 (in combination with mitoxantrone and etoposide); may administer a second course if needed (Archimbaud 1991; Archimbaud 1995).

Acute promyelocytic leukemia induction

Acute promyelocytic leukemia induction (off-label dosing): IV: 200 mg/m2/day continuous infusion for 7 days beginning on day 3 of treatment (in combination with tretinoin and daunorubicin) (Ades 2006; Ades 2008; Powell 2010).

Acute promyelocytic leukemia consolidation

Acute promyelocytic leukemia consolidation (off-label use):

In combination with idarubicin and tretinoin: High-risk patients (WBC ≥10,000/mm3) (Sanz 2010): Adults ≤60 years of age:

First consolidation course: IV: 1,000 mg/m2/day for 4 days.

Third consolidation course: IV: 150 mg/m2 every 8 hours for 4 days.

In combination with idarubicin, tretinoin, and thioguanine: High-risk patients (WBC >10,000/mm3) (Lo Coco 2010): Adults ≤61 years of age:

First consolidation course: IV: 1,000 mg/m2/day for 4 days.

Third consolidation course: IV: 150 mg/m2 every 8 hours for 5 days.

In combination with daunorubicin (Ades 2006; Ades 2008):

First consolidation course: IV: 200 mg/m2/day for 7 days.

Second consolidation course:

Age ≤60 years and low risk (WBC <10,000/mm3): IV: 1,000 mg/m2 every 12 hours for 4 days (8 doses).

Age <50 years and high risk (WBC ≥10,000/mm3): IV: 2,000 mg/m2 every 12 hours for 5 days (10 doses).

Age 50 to 60 years and high risk (WBC ≥10,000/mm3): IV: 1,500 mg/m2 every 12 hours for 5 days (10 doses) (Ades 2008).

Age >60 years and high risk (WBC ≥10,000/mm3): IV: 1,000 mg/m2 every 12 hours for 4 days (8 doses).

Chronic lymphocytic leukemia

Chronic lymphocytic leukemia (off-label use): OFAR regimen: IV: 1,000 mg/m2/dose over 2 hours days 2 and 3 every 4 weeks for up to 6 cycles (in combination with oxaliplatin, fludarabine, and rituximab) (Tsimberidou 2008).

Hodgkin lymphoma, relapsed or refractory

Hodgkin lymphoma, relapsed or refractory (off-label use):

DHAP regimen: IV: 2,000 mg/m2 over 3 hours every 12 hours day 2 (total of 2 doses/cycle) for 2 cycles (in combination with dexamethasone and cisplatin) (Josting 2002).

ESHAP regimen: IV: 2,000 mg/m2 day 5 (in combination with etoposide, methylprednisolone, and cisplatin) every 3 to 4 weeks for 3 or 6 cycles (Aparicio 1999).

Mini-BEAM regimen: IV: 100 mg/m2 every 12 hours days 2 to 5 (total of 8 doses) every 4 to 6 weeks (in combination with carmustine, etoposide, and melphalan) (Colwill 1995; Martin 2001).

BEAM regimen (transplant preparative regimen): IV: 200 mg/m2 twice daily for 4 days beginning 5 days prior to transplant (in combination with carmustine, etoposide, and melphalan) (Chopra 1993).

Non-Hodgkin lymphomas

Non-Hodgkin lymphomas (off-label use):

BEAM regimen (transplant-preparative regimen): IV: 200 mg/m2 twice daily for 3 days beginning 4 days prior to transplant (in combination with carmustine, etoposide, and melphalan) (Linch 2010) or 100 mg/m2 over 1 hour every 12 hours for 4 days beginning 5 days prior to transplant (in combination with carmustine, etoposide, and melphalan) (van Imhoff 2005).

Burkitt lymphoma:

CALGB 9251 regimen: Cycles 2, 4, and 6: IV: 150 mg/m2/day continuous infusion days 4 and 5 (Lee 2001; Rizzieri 2004).

CODOX-M/IVAC regimen:

Adults ≤60 years of age: Cycles 2 and 4 (IVAC): IV: 2,000 mg/m2 every 12 hours days 1 and 2 (total of 4 doses/cycle) (IVAC is combination with ifosfamide, mesna, and etoposide; IVAC alternates with CODOX-M) (Magrath 1996).

Adults ≤65 years of age: Cycles 2 and 4 (IVAC): IV: 2,000 mg/m2 over 3 hours every 12 hours days 1 and 2 (total of 4 doses/cycle) (IVAC is combination with ifosfamide, mesna, and etoposide; IVAC alternates with CODOX-M) (Mead 2008).

Adults >65 years of age: Cycles 2 and 4 (IVAC): IV: 1,000 mg/m2 over 3 hours every 12 hours days 1 and 2 (total of 4 doses/cycle) (IVAC is combination with ifosfamide, mesna, and etoposide; IVAC alternates with CODOX-M) (Mead 2008).

Hyper-CVAD alternating with high-dose methotrexate/cytarabine regimen:

Adults <60 years of age: Cycles 2, 4, 6, and 8: IV: 3,000 mg/m2 every 12 hours days 2 and 3 (total of 4 doses/cycle) of a 21-day cycle (in combination with methotrexate and leucovorin), alternating with Hyper-CVAD administered on odd-numbered cycles (cyclophosphamide, vincristine, doxorubicin, and dexamethasone) plus rituximab (in cycles 1 to 4) and CNS prophylaxis (Thomas 2006).

Adults ≥60 years of age: Cycles 2, 4, 6, and 8: IV: 1,000 mg/m2 every 12 hours days 2 and 3 (total of 4 doses/cycle) of a 21-day cycle (in combination with methotrexate and leucovorin) alternating with Hyper-CVAD administered on odd-numbered cycles (cyclophosphamide, vincristine, doxorubicin, and dexamethasone) with rituximab (in cycles 1 to 4) and CNS prophylaxis (Thomas 2006).

Mantle cell lymphoma:

R-DHAP regimen: Adults ≤65 years of age: IV: 2,000 mg/m2 every 12 hours on day 2 (total of 2 doses/cycle) every 3 weeks (in combination with rituximab plus dexamethasone and cisplatin) for 4 cycles (Le Gouill 2017) or 2,000 mg/m2 every 12 hours on day 2 (total of 2 doses/cycle; in combination with rituximab plus dexamethasone and cisplatin) alternating with R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) for 6 cycles (3 cycles each of R-CHOP and R-DHAP) (Hermine 2016).

RBAC regimen: IV: 500 to 800 mg/m2 over 2 hours (starting 2 hours after bendamustine) on days 2 through 4 every 28 days for up to 6 cycles (in combination with rituximab and bendamustine) (Visco 2013; Visco 2017).

Nordic regimen:

Adults ≤60 years of age: IV: 3,000 mg/m2 over 3 hours every 12 hours for a total of 4 doses (in combination with rituximab) for 2 cycles alternating with Maxi-CHOP (dose-intensified CHOP) for 3 cycles (total of 5 cycles) (Geisler 2008).

Adults >60 years of age: IV: 2,000 mg/m2 over 3 hours every 12 hours for a total of 4 doses (in combination with rituximab) for 2 cycles alternating with Maxi-CHOP for 3 cycles (total of 5 cycles) (Geisler 2008).

Hyper-CVAD alternating with high-dose methotrexate/cytarabine regimen:

Adults ≤60 years of age: Cycles 2, 4, 6, and 8: IV: 3,000 mg/m2 every 12 hours days 3 and 4 (total of 4 doses/cycle) of a 21-day cycle (in combination with methotrexate, leucovorin, and rituximab), alternating with Hyper-CVAD administered on odd-numbered cycles (cyclophosphamide, vincristine, doxorubicin, dexamethasone, and rituximab) (Romaguera 2005).

Adults >60 years of age: Cycles 2, 4, 6, and 8: IV: 1,000 mg/m2 every 12 hours days 3 and 4 (total of 4 doses/cycle) of a 21-day cycle (in combination with methotrexate, leucovorin, and rituximab) alternating with Hyper-CVAD administered on odd-numbered cycles (cyclophosphamide, vincristine, doxorubicin, dexamethasone, and rituximab) (Romaguera 2005).

Relapsed or refractory non-Hodgkin lymphomas:

DHAP regimen:

Adults ≤70 years of age: IV: 2,000 mg/m2 over 3 hours every 12 hours day 2 (total of 2 doses/cycle) every 3 to 4 weeks for 6 to 10 cycles (in combination with dexamethasone and cisplatin) (Velasquez 1988).

Adults >70 years of age: IV: 1,000 mg/m2 over 3 hours every 12 hours day 2 (total of 2 doses/cycle) every 3 to 4 weeks for 6 to 10 cycles (in combination with dexamethasone and cisplatin) (Velasquez 1988).

ESHAP regimen: IV: 2,000 mg/m2 over 2 hours day 5 every 3 to 4 weeks for 6 to 8 cycles (in combination with etoposide, methylprednisolone, and cisplatin) (Velasquez 1994).

Primary CNS lymphoma

Primary CNS lymphoma (off-label use): IV: 2,000 mg/m2 over 1 hour every 12 hours days 2 and 3 (total of 4 doses) every 3 weeks (in combination with methotrexate and followed by whole brain irradiation) for a total of 4 courses (Ferreri 2009) or 3,000 mg/m2 (maximum dose of 6,000 mg) over 3 hours on days 1 and 2 every 4 weeks for 2 cycles (administer cytarabine after 5 to 7 cycles of the induction R-MPV regimen [rituximab, methotrexate, leucovorin, vincristine, and procarbazine] and whole brain radiation therapy) (Shah 2007) or 2,000 mg/m2 over 2 hours every 12 hours days 1 to 4 (total of 8 doses) as consolidation therapy (in combination with etoposide); cytarabine/etoposide is administered following remission induction with methotrexate, leucovorin, temozolomide, and rituximab (Rubenstein 2013).

Meningeal leukemia

Meningeal leukemia: Intrathecal therapy: Note: Optimal intrathecal chemotherapy dosing should be based on age rather than on body surface area (BSA); CSF volume correlates with age and not to BSA (Bleyer 1983; Kerr 2001). Dosing provided in the manufacturer's labeling is BSA-based.

Off-label uses or doses for intrathecal therapy:

CNS prophylaxis (ALL): Intrathecal: 100 mg weekly for 8 doses, then every 2 weeks for 8 doses, then monthly for 6 doses (high-risk patients) or 100 mg on day 7 or 8 with each chemotherapy cycle for 4 doses (low risk patients) or 16 doses (high-risk patients) (Cortes 1995) or 70 mg on day 1 of remission induction cycle 1 (adults <40 years of age) (Stock 2019).

or as part of intrathecal triple therapy (TIT): Intrathecal: 40 mg days 0 and 14 during induction, days 1, 4, 8, and 11 during CNS therapy phase, every 18 weeks during intensification and maintenance phases (Storring 2009).

CNS prophylaxis (APL, as part of TIT): Intrathecal: 50 mg per dose; administer 1 dose prior to consolidation and 2 doses during each of 2 consolidation phases (total of 5 doses) (Ades 2006; Ades 2008).

CNS prophylaxis (Burkitt lymphoma; component of CODOX-M/IVAC regimen): Intrathecal: 70 mg on days 1 and 3 of cycles 1 and 3 (CODOX-M cycle) (Magrath 1996; Mead 2008).

CNS prophylaxis (Burkitt lymphoma; component of Hyper-CVAD alternating with cytarabine/methotrexate regimen): Intrathecal: 100 mg on day 7 of each 21-day treatment cycle (Thomas 2006).

CNS leukemia treatment (ALL, as part of TIT): Intrathecal: 40 mg twice weekly until CSF cleared (Storring 2009).

CNS lymphoma treatment: Intrathecal: 50 mg twice a week for 4 weeks, then weekly for 4 to 8 weeks, then every other week for 4 weeks, then every 4 weeks for 4 doses (Glantz 1999).

CNS treatment (Burkitt lymphoma; component of CODOX-M/IVAC regimen): Intrathecal: 70 mg on days 1, 3, and 5 of cycles 1 and 3 (CODOX-M cycle) and 70 mg on days 7 and 9 of cycles 2 and 4 (IVAC cycle) (Magrath 1996; Mead 2008).

Leptomeningeal metastases treatment: Intrathecal: 25 to 100 mg twice weekly for 4 weeks, then once weekly for 4 weeks, then a maintenance regimen of once a month (Chamberlain 2010) or 40 to 60 mg per dose (DeAngelis 2005).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

Altered kidney function may increase the incidence of CNS toxicity with high-dose cytarabine treatment; use with caution and consider reduced doses. There are no dosage adjustments provided in the manufacturer's labeling; however, the following adjustments have been recommended:

Aronoff 2007 (cytarabine 100 to 200 mg/m2): No adjustment necessary.

Kintzel 1995 (high-dose cytarabine 1 to 3 g/m2):

CrCl 46 to 60 mL/minute: Administer 60% of dose.

CrCl 31 to 45 mL/minute: Administer 50% of dose.

CrCl <30 mL/minute: Consider use of alternative drug.

Krens 2019 (cytarabine doses ≥1 g/m2):

GFR ≥60 mL/minute/1.73 m2: No dosage adjustment necessary.

GFR 31 to 59 mL/minute/1.73 m2: Administer 50% of original dose.

GFR ≤30 mL/minute/1.73 m2: Use is not recommended.

Smith 1997 (high-dose cytarabine; ≥2 g/m2/dose):

Serum creatinine 1.5 to 1.9 mg/dL or increase (from baseline) of 0.5 to 1.2 mg/dL: Reduce dose to 1 g/m2/dose.

Serum creatinine ≥2 mg/dL or increase (from baseline) of >1.2 mg/dL: Reduce dose to 0.1 g/m2/day as a continuous infusion.

Romaguera 2005; Thomas 2006 (cytarabine 3 g/m2/dose [studied in Burkitt or mantle cell lymphoma]):

Serum creatinine ≥1.5 mg/dL: Reduce dose to 1 g/m2/dose.

Hemodialysis: In 4-hour dialysis sessions (with high flow polysulfone membrane) 6 hours after cytarabine 1 g/m2 over 2 hours, 63% of the metabolite ARA-U was extracted from plasma (based on a single adult case report) (Radeski 2011).

Dosing: Hepatic Impairment: Adult

Dose may need to be adjusted in patients with liver failure since cytarabine is partially detoxified in the liver. Hepatic dysfunction may increase the incidence of CNS toxicity with high-dose cytarabine treatment; use with caution. There are no dosage adjustments provided in the manufacturer's labeling; however, the following adjustments have been recommended:

Floyd 2006: Transaminases (any elevation): Administer 50% of dose; may increase subsequent doses in the absence of toxicities.

Koren 1992 (dose level not specified): Bilirubin >2 mg/dL: Administer 50% of dose; may increase subsequent doses in the absence of toxicities.

Dosing: Pediatric

(For additional information see "Cytarabine (conventional): Pediatric drug information")

Note: Dosing and frequency may vary by protocol and/or treatment phase; refer to specific protocol. In pediatric patients, IV dosing may be based on either BSA (mg/m2) or weight (mg/kg); use extra precaution to verify dosing parameters during calculations. Intrathecal dosing is based on the age of the patient. Antiemetics may be recommended to prevent nausea and vomiting. In pediatrics, a 75 mg/m2 IV dose is associated with a moderate emetic potential and IV doses ≥3,000 mg/m2 are associated with a high emetic potential (POGO [Paw Cho Sing 2019]).

Acute lymphocytic leukemia, B-cell

Acute lymphocytic leukemia, B-cell (B-ALL):

High risk B-ALL: Limited data available, multiple regimens reported:

Vrooman 2018 (DFCI 05-001): Intensified consolidation in very high-risk patients:

Consolidation IB cycle: Children and Adolescents: IV: 75 mg/m2 once daily on days 2 through 5 and days 9 through 12 in combination with cyclophosphamide, 6-mercaptopurine, and intrathecal methotrexate (Vrooman 2018).

Consolidation IC cycle: Children and Adolescents: IV: 2,000 mg/m2 every 12 hours for 4 doses beginning on day 1; administered in combination with etoposide, dexamethasone, and asparaginase (Place 2015; Vrooman 2018).

Larsen 2016: Consolidation and delayed intensification I and II: Children and Adolescents: IV, SubQ: 75 mg/m2 over 1 to 30 minutes or subcutaneous once daily for 4 days every 7 days for 2 courses; specific days depends on protocol phase.

Steinherz 1993: NYII Protocol: Consolidation: Children and Adolescents: IV: 3,000 mg/m2 over 3 hours for 4 doses on Days 28, 29, 35, and 36; experts at some centers have suggested fewer doses (eg, 2 doses).

Isolated CNS; relapse: Limited data available; multiple regimens reported: Infants, Children, and Adolescents: IV: 300 mg/m2/dose; frequency dependent upon protocol.

Ribeiro 1995, Rivera 1996: Children and Adolescents ≤14 years: Induction: Administer on Days 8, 22, 36; for Maintenance administer every 5 weeks for 52 weeks in combination with teniposide.

Winick 1993: Continuation therapy: Infants, Children, and Adolescents: Administer on day 1 and 4 of week 7, 14, 21, 28, 35, 42, 49, and 56 in conjunction with teniposide.

Acute myeloid leukemia

Acute myeloid leukemia (AML):

Remission induction:

ADE regimen: Limited data available (Gamis 2014): Note: Some aspects of protocol dosing based on previous reports (Cooper 2012; Gibson 2011; Guest 2014; Woods 1990).

Induction 1 ADE (10 + 3 + 5) and Induction 2 ADE (8 + 3 + 5):

Infants and Children with BSA <0.6 m2: IV: 3.3 mg/kg over 1 to 30 minutes every 12 hours for 10 days (induction 1) or 8 days (induction 2) in combination with daunorubicin and etoposide.

Children and Adolescents with BSA ≥0.6 m2: IV: 100 mg/m2 over 1 to 30 minutes every 12 hours for 10 days (induction 1) or 8 days (induction 2) in combination with daunorubicin and etoposide.

7 + 3 regimen: Limited data available (Woods 1990):

Infants and Children <3 years: IV: 3.3 mg/kg/day continuous infusion for 7 days; minimum of 2 courses (in combination with daunorubicin).

Children ≥3 years and Adolescents: IV: 100 mg/m2/day continuous infusion for 7 days; minimum of 2 courses (in combination with daunorubicin).

Manufacturer's labeling: Manufacturer's labeling may not be reflective of current practice. Infants, Children, and Adolescents: IV: Standard dose: 100 mg/m2/day continuous infusion for 7 days or 200 mg/m2/day continuous infusion (as 100 mg/m2/dose over 12 hours every 12 hours) for 7 days.

Intensification: Limited data available (Gamis 2014): Note: Some aspects of protocol dosing based on previous reports (Cooper 2012; Gibson 2011; Guest 2014; Woods 1990).

Intensification 1: AE therapy.

Infants and Children with BSA <0.6 m2: IV: 33 mg/kg over 1 hour every 12 hours for 5 days (in combination with etoposide).

Children and Adolescents with BSA ≥0.6 m2: IV: 1,000 mg/m2 over 1 hour every 12 hours for 5 days (in combination with etoposide).

Intensification 2 (for patients not undergoing stem cell transplant): MA therapy:

Infants and Children with BSA <0.6 m2: IV: 33 mg/kg over 1 hour every 12 hours for 4 days (in combination with mitoxantrone).

Children and Adolescents with BSA ≥0.6 m2: IV: 1,000 mg/m2 over 1 hour every 12 hours for 4 days (in combination with mitoxantrone).

Intensification 3 (for patients not undergoing stem cell transplant):

Infants and Children with BSA <0.6 m2: IV: 99 mg/kg every 12 hours on days 1, 2, 8 and 9 in combination with E. coli L-asparaginase; Note: Some centers have substituted Erwinia asparaginase due to E. coli L-Asparaginase no longer being available.

Children and Adolescents with BSA ≥0.6 m2: IV: 3,000 mg/m2 every 12 hours on Days 1, 2, 8, and 9 in combination with E. coli L-asparaginase; Note: Some centers have substituted Erwinia asparaginase due to E. coli L-Asparaginase no longer being available.

Consolidation or refractory disease: Limited data available: Capizzi II regimen (high-dose cytarabine [fixed]): Children ≥6 years and Adolescents: IV: 3,000 mg/m2 over 3 hours every 12 hours for 4 doses, followed by E. coli L-Asparaginase (some centers have substituted Erwinia asparaginase due to E. coli L-Asparaginase no longer being available) 6 hours after the last dose of cytarabine (Capizzi 1984).

Salvage treatment for refractory/recurrent disease: Limited data available:

Clofarabine/cytarabine regimen: Children and Adolescents: IV: 1,000 mg/m2 over 2 hours once daily for 5 days (in combination with clofarabine; administer cytarabine 4 hours after initiation of clofarabine) for up to 2 induction cycles (Cooper 2014).

FLAG regimen: Children ≥11 years and Adolescents: IV: 2,000 mg/m2 over 4 hours once daily for 5 days (in combination with fludarabine and G-CSF); may repeat once if needed (Montillo 1998).

MEC regimen:

Children ≥5 years and Adolescents: IV: 1,000 mg/m2 over 6 hours for 6 days (in combination with etoposide and mitoxantrone) (Amadori 1991).

Adolescents ≥15 years: IV: 500 mg/m2/day continuous infusion days 1, 2, and 3 and days 8, 9, and 10 (in combination with mitoxantrone and etoposide); may administer a second course if needed (Archimbaud 1991; Archimbaud 1995).

Non-Hodgkin lymphomas

Non-Hodgkin lymphomas: Limited data available:

Intermediate-risk B-cell NHL (Group B): Consolidation: R-CYM 1 and 2 regimen (Goldman 2013; Patte 2007): Children and Adolescents: IV: 100 mg/m2/day as a continuous infusion over 24 hours on Days 2 through 6 (5 days; total dose: 500 mg/m2 as a 120-hour infusion) (in combination with rituximab and methotrexate).

High-risk B-cell NHL (Group C): Consolidation: R-CYVE 1 and 2 regimen (Cairo 2007; Patte 2001): Infants ≥6 months, Children, and Adolescents: IV: 50 mg/m2 infused over 12 hours (2000 to 0800) for 5 doses (Days 1 to 5); follow each 12-hour infusion dose with 3,000 mg/m2 infused over 3 hours which starts at the end of the 12-hour infusion (0800 - 1100) x 5 doses (days 2 to 6) in combination with etoposide.

B-lineage lymphoblastic lymphoma (B-LLy) (Termuhlen 2013):

Consolidation phase: Children and Adolescents: IV, SubQ: 75 mg/m2 once daily for 4 days every 7 days for 4 courses (Days 0 to 3, 7 to 10, 14 to 17, 21 to 24).

Delayed intensification: Children and Adolescents: IV, SubQ: 75 mg/m2 once daily for 4 days every 7 days for 2 courses (Days 35 to 38 and 42 to 45).

Meningeal leukemia

Meningeal leukemia: Infants, Children, and Adolescents: Note: Optimal intrathecal chemotherapy dosing should be based on age (see following information) rather than on body surface area (BSA); CSF volume correlates with age and not to BSA (Bleyer 1983; Kerr 2001).

Age-based intrathecal fixed dosing: Intrathecal:

CNS prophylaxis (AML) (Woods 1990):

<1 year: 20 mg per dose.

1 to <2 years: 30 mg per dose.

2 to <3 years: 50 mg per dose.

≥3 years: 70 mg per dose.

CNS prophylaxis (ALL): Dosing regimens variable, age-specific regimens reported from literature:

Gaynon 1993: Administer on day 0 of induction therapy: Children and Adolescents:

<2 years: 30 mg per dose.

2 to <3 years: 50 mg per dose.

≥3 years: 70 mg per dose.

Pieters 2007: Administer on day 15 of induction therapy, days 1 and 15 of reinduction phase; and day 1 of cycle 2 of maintenance 1A phase: Infants and young Children (≤12 months at enrollment):

<1 year: 15 mg per dose.

≥1 year: 20 mg per dose.

Lin 2008: Administer as part of triple intrathecal therapy (TIT) on days 1 and 15 of induction therapy; days 1, 15, 50, and 64 (standard risk patients) or days 1, 15, 29, and 43 (high-risk patients) during consolidation therapy; day 1 of reinduction therapy, and during maintenance therapy (very high-risk patients receive on days 1, 22, 45, and 59 of induction, days 8, 22, 36, and 50 of consolidation therapy, days 8 and 38 of reinduction therapy, and during maintenance): Infants, Children, and Adolescents:

<1 year: 18 mg per dose.

1 to <2 years: 24 mg per dose.

2 to <3 years: 30 mg per dose.

≥3 years: 36 mg per dose.

Treatment, CNS leukemia (ALL), Very high-risk: Administer as part of triple intrathecal therapy (TIT) weekly until CSF remission, then every 4 weeks throughout continuation treatment (Lin 2008): Infants, Children, and Adolescents:

<1 year: 18 mg per dose.

1 to <2 years: 24 mg per dose.

2 to <3 years: 30 mg per dose.

≥3 years: 36 mg per dose.

BSA-directed intrathecal dosing: Manufacturer's labeling (based on BSA): Manufacturer's labeling may not reflect current practice: Intrathecal: 30 mg/m2 every 4 days; range: 5 to 75 mg/m2 once daily for 4 days or once every 4 days until CNS findings normalize, followed by one additional treatment.

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Pediatric

IV: There are no dosage adjustments provided in the manufacturer's labeling; however, the following guidelines have been used by some clinicians:

Infants, Children, and Adolescents:

Standard dosing range (100 to 200 mg/m2): No adjustment necessary (Aronoff 2007)

High-dose cytarabine (1,000 to 3,000 mg/m2): There are no pediatric-specific recommendations; refer to specific protocols if available. Based on experience in adult patients, dosing adjustment suggested.

Dosing: Hepatic Impairment: Pediatric

IV: Dose may need to be adjusted in patients with liver failure since cytarabine is partially detoxified in the liver. There are no dosage adjustments provided in the manufacturer's labeling (refer to protocols); however, the following guidelines have been used by some clinicians: All patients:

Floyd 2006: Transaminases (any elevation): Administer 50% of dose; may increase subsequent doses in the absence of toxicity, based primarily on experience in adult patients.

Koren 1992 (dose not specified): Bilirubin >2 mg/dL: Administer 50% of dose; may increase subsequent doses in the absence of toxicity, based on experience in adult patients.

Dosing: Older Adult

Refer to adult dosing.

Dosing: Obesity: Adult

American Society of Clinical Oncology guidelines for appropriate systemic therapy dosing in adults with cancer with a BMI ≥30 kg/m2: Utilize patient's actual body weight (full weight) for calculation of BSA- or weight-based dosing; manage regimen-related toxicities in the same manner as for patients with a BMI <30 kg/m2; if a dose reduction is utilized due to toxicity, may consider resumption of full weight-based dosing (or previously tolerated dose level) with subsequent cycles only if dose escalations are allowed in the prescribing information, if contributing underlying factors (eg, hepatic or kidney impairment) are sufficiently resolved, AND if performance status has markedly improved or is considered adequate (ASCO [Griggs 2021]).

American Society for Blood and Marrow Transplantation (ASBMT) practice guideline committee position statement on chemotherapy dosing in obesity: Utilize actual body weight (full weight) for calculation of body surface area in cytarabine dosing for hematopoietic stem cell transplant conditioning regimens (ASBMT [Bubalo 2014]).

Dosing: Adjustment for Toxicity: Adult

Cytarabine/ARA-c syndrome (fever, myalgia, bone pain, occasional chest pain, maculopapular rash, conjunctivitis, and malaise and generally occurs 6 to 12 hours following cytarabine administration): May be managed with corticosteroids.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Generic: 20 mg/mL (25 mL)

Solution, Injection [preservative free]:

Generic: 20 mg/mL (5 mL, 50 mL); 100 mg/mL (20 mL)

Generic Equivalent Available: US

Yes

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Generic: 20 mg/mL (5 mL, 25 mL); 100 mg/mL (1 mL, 10 mL, 20 mL)

Administration: Adult

IV: Infuse standard dose therapy for AML (100 to 200 mg/m2/day) as a continuous infusion. Infuse high-dose therapy (off-label) over 1 to 3 hours (usually). Other rates have been used, refer to specific reference.

Antiemetics may be recommended to prevent nausea and vomiting; in adults, IV doses >1,000 mg/m2 are associated with a moderate emetic potential (ASCO [Hesketh 2020]; MASCC/ESMO [Roila 2016]).

Intrathecal: Intrathecal doses should be administered as soon as possible after preparation.

May also be administered SubQ.

Administration: Pediatric

Parenteral: Note: Antiemetics may be recommended to prevent nausea and vomiting. In pediatrics, a 75 mg/m2 IV dose is associated with a moderate emetic potential and IV doses ≥3,000 mg/m2 are associated with a high emetic potential (POGO [Paw Cho Sing 2019]).

IV infusion: May infuse as a continuous infusion over several hours (eg, 12 to 24 hours has been reported in pediatric trials) (Cairo 2007; Patte 2001). The manufacturer suggests continuous infusion of standard dose: 100 to 200 mg/m2/day. High-dose cytarabine has been infused over 1 to 3 hours according to protocols (refer to protocol) (Capizzi 1984).

IV Push: Administration over 15 to 30 minutes; has been reported in pediatric trials

SubQ: May administer at concentration not to exceed 100 mg/mL (Allwood 2003); rotate injection sites; some centers have used thigh, abdomen, and flank regions

Intrathecal administration: Antiemetic therapy should be administered prior to intrathecal doses of cytarabine. Administer as soon as possible after preparation.

Hazardous Drugs Handling Considerations

Hazardous agent (NIOSH 2016 [group 1]).

Use appropriate precautions for receiving, handling, storage, preparation, dispensing, transporting, administration, and disposal. Follow NIOSH and USP 800 recommendations and institution-specific policies/procedures for appropriate containment strategy (NIOSH 2016; USP-NF 2020).

Use: Labeled Indications

Acute lymphoblastic leukemia: Treatment of acute lymphoblastic leukemia.

Acute myeloid leukemia: Remission induction (in combination with other chemotherapy medications) in acute myeloid leukemia in adult and pediatric patients.

Chronic myeloid leukemia: Treatment of chronic myeloid leukemia in blast phase.

Meningeal leukemia: Prophylaxis and treatment of meningeal leukemia.

Use: Off-Label: Adult

Acute myeloid leukemia (consolidation); Acute myeloid leukemia (salvage); Acute promyelocytic leukemia (consolidation); Acute promyelocytic leukemia (induction); Chronic lymphocytic leukemia (refractory); Hodgkin lymphoma (relapsed or refractory); Non-Hodgkin lymphoma; Primary central nervous system lymphoma

Medication Safety Issues
Sound-alike/look-alike issues:

Cytarabine may be confused with cladribine, clofarabine, Cytosar, Cytoxan, daunorubicin and cytarabine (liposomal), vidarabine, vinorelbine

Cytarabine (conventional) may be confused with cytarabine liposomal

Cytarabine (conventional) (100 mg/5 mL; Hospira manufacturer) may be confused with vinorelbine (50 mg/5 mL; Hospira manufacturer) due to similar packaging; potential for inadvertent intrathecal administration of vinorelbine may occur (ISMP [Smetzer 2015]).

Cytosar-U may be confused with cytarabine, Cytovene, Cytoxan, Neosar

High alert medication:

This medication is in a class the Institute for Safe Medication Practices (ISMP) includes among its list of drugs classes which have a heightened risk of causing significant patient harm when used in error.

Administration issues:

Intrathecal medication safety: The American Society of Clinical Oncology (ASCO)/Oncology Nursing Society (ONS) chemotherapy administration safety standards (ASCO/ONS [Neuss 2016]) recommend the following safety measures for intrathecal chemotherapy:

• Intrathecal medication should not be prepared during the preparation of any other agents.

• After preparation, store in an isolated location or container clearly marked with a label identifying as "intrathecal" use only.

• Delivery to the patient should only be with other medications also intended for administration into the central nervous system.

• Administer immediately after a time-out/double-check procedure.

Adverse Reactions

The following adverse drug reactions are derived from product labeling unless otherwise specified. Reported adverse reactions are for IV, SUBQ, or intrathecal administration of cytarabine with concomitant chemotherapeutic agents.

Frequency not defined:

Cardiovascular: Chest pain, thrombophlebitis

Dermatologic: Alopecia, dermal ulcer, ephelis, pruritus, urticaria

Gastrointestinal: Abdominal pain, anorexia, diarrhea, esophageal ulcer, esophagitis, intestinal necrosis, nausea, oral inflammation, oral mucosa ulcer, proctocolitis, rectal ulcer, sore throat, vomiting

Genitourinary: Urinary retention

Hematologic & oncologic: Anemia, bone marrow depression, hemorrhage, leukopenia, megaloblastic anemia, reticulocytopenia, thrombocytopenia

Hepatic: Hepatic insufficiency, jaundice

Hypersensitivity: Allergic angioedema

Infection: Sepsis

Local: Cellulitis at injection site, inflammation at injection site, pain at injection site

Nervous system: Dizziness, headache, leukoencephalopathy (necrotizing), neuritis, paralysis, paraplegia

Ophthalmic: Blindness

Renal: Renal insufficiency

Respiratory: Dyspnea, pneumonia

Postmarketing (includes adverse reactions with intermediate or high dose cytarabine):

Cardiovascular: Acute myocarditis (Pallazola 2018), pericarditis (Woods 1999)

Dermatologic: Acute generalized exanthematous pustulosis (Chiu 2002), maculopapular rash (Sahu 2019), skin rash (Forghieri 2007), skin toxicity (including ear redness [red ear syndrome] and palmar-plantar erythrodysesthesia) (Sahu 2019)

Endocrine & metabolic: Hyperuricemia (Rudnick 1979)

Gastrointestinal: Increased serum amylase (Yang 2020), increased serum lipase (Yang 2020), pancreatitis (including acute pancreatitis) (Yang 2020)

Hepatic: Increased liver enzymes (Forghieri 2007)

Hypersensitivity: Anaphylaxis (Rassiga 1980)

Nervous system: Aseptic meningitis (Beaumont 2020), cerebellar syndrome (including ataxia, confusion, drowsiness, dysarthria, dysmetria, encephalitis, impaired consciousness, nystagmus disorder) (Lenfant 2021; Sainz de la Maza Cantero 2016), malaise (Sahu 2019), neurotoxicity (including peripheral neuropathy) (Malhotra 2004), reversible posterior leukoencephalopathy syndrome (Saito 2007)

Neuromuscular & skeletal: Myalgia (Sahu 2019), ostealgia (Sahu 2019), rhabdomyolysis (Truica 2002)

Ophthalmic: Conjunctivitis (Rudnick 1979), corneal toxicity (epithelial keratopathy) (Özcan 2021), keratoconjunctivitis (Cho 2018), uveitis (including anterior uveitis) (Cho 2018)

Respiratory: Pulmonary toxicity (including bronchiolitis obliterans organizing pneumonia, hypoxemia, pulmonary edema, pulmonary infiltrates) (Forghieri 2007)

Miscellaneous: Fever (Rudnick 1979)

Contraindications

Hypersensitivity to cytarabine or any component of the formulation

Warnings/Precautions

Concerns related to adverse effects:

• Bone marrow suppression: Myelosuppression (leukopenia, thrombocytopenia, and anemia) is the major toxicity of cytarabine. Use with caution in patients with prior drug-induced bone marrow suppression. Infection or neutropenic fever due to neutropenia or bleeding due to thrombocytopenia may occur.

• Cytarabine syndrome: Cytarabine (ARA-C) syndrome is characterized by fever, myalgia, bone pain, chest pain (occasionally), maculopapular rash, conjunctivitis, and malaise; generally occurs 6 to 12 hours following administration.

• GI toxicities: Toxicities (less serious) include nausea, vomiting, diarrhea, abdominal pain, oral ulcerations, and hepatic dysfunction.

• Hypersensitivity: Anaphylaxis resulting in acute cardiopulmonary arrest has been reported (rare).

• Pancreatitis: There have been reports of acute pancreatitis in patients receiving continuous infusion cytarabine and in patients receiving cytarabine who were previously treated with L-asparaginase.

• Sudden respiratory distress syndrome: Sudden respiratory distress, rapidly progressing to pulmonary edema and cardiomegaly, has been reported with high-dose cytarabine. May present as severe dyspnea with a rapid onset and refractory hypoxia with diffuse pulmonary infiltrates, leading to respiratory failure; may be fatal (Morgan 2011).

• Tumor lysis syndrome: Tumor lysis syndrome and subsequent hyperuricemia may occur.

Concurrent drug therapy issues:

• Drug-drug interactions: There have been case reports of fatal cardiomyopathy when high-dose cytarabine was used in combination with cyclophosphamide as a preparation regimen for transplantation.

Special populations:

• Pediatric: Delayed progressive ascending paralysis has been reported in two children who received combination chemotherapy with IV and intrathecal cytarabine at conventional doses for the treatment of acute myeloid leukemia (was fatal in one patient).

Dosage form specific issues:

• Benzyl alcohol: Some products may contain benzyl alcohol; do not use products containing benzyl alcohol or products reconstituted with bacteriostatic diluent intrathecally or for high-dose cytarabine regimens. Benzyl alcohol is associated with gasping syndrome in premature infants.

Other warnings/precautions:

• Experienced providers: Administer under the supervision of experienced cancer chemotherapy providers. Due to the potential toxicities, induction treatment with cytarabine should be in a facility with sufficient laboratory and supportive resources.

• High-dose treatment: High-dose regimens have been associated with GI, CNS, pulmonary, ocular (reversible corneal toxicity and hemorrhagic conjunctivitis; prophylaxis with ophthalmic corticosteroids is recommended) toxicities, and cardiomyopathy. Neurotoxicity associated with high-dose treatment may present as acute cerebellar toxicity (with or without cerebral impairment), personality changes, or may be severe with seizure and/or coma; may be delayed, occurring up to 3 to 8 days after treatment has begun; possibly irreversible. Risk factors for neurotoxicity include cumulative cytarabine dose, prior CNS disease and renal impairment (incidence may be up to 55% in patients with renal impairment); high-dose therapy (>18 g/m2 per cycle) and age >50 years also increase the risk for cerebellar toxicity (Herzig 1987).

• Intrathecal safety: When used for intrathecal administration, should not be prepared during the preparation of any other agents. After preparation, store intrathecal medications in an isolated location or container clearly marked with a label identifying as "intrathecal" use only. Delivery of intrathecal medications to the patient should only be with other medications also intended for administration into the CNS and administer immediately after a time-out/double-check procedure (ASCO/ONS [Neuss 2016]).

Metabolism/Transport Effects

None known.

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

5-Aminosalicylic Acid Derivatives: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Abrocitinib: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk X: Avoid combination

Baricitinib: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Baricitinib. Risk X: Avoid combination

BCG (Intravesical): Myelosuppressive Agents may diminish the therapeutic effect of BCG (Intravesical). Risk X: Avoid combination

BCG Products: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of BCG Products. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of BCG Products. Risk X: Avoid combination

Brincidofovir: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Brincidofovir. Risk C: Monitor therapy

Cedazuridine: May increase the serum concentration of Cytidine Deaminase Substrates. Risk X: Avoid combination

Chloramphenicol (Ophthalmic): May enhance the adverse/toxic effect of Myelosuppressive Agents. Risk C: Monitor therapy

Cladribine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk X: Avoid combination

Cladribine: Agents that Undergo Intracellular Phosphorylation may diminish the therapeutic effect of Cladribine. Risk X: Avoid combination

Cladribine: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Cladribine. Risk X: Avoid combination

CloZAPine: Myelosuppressive Agents may enhance the adverse/toxic effect of CloZAPine. Specifically, the risk for neutropenia may be increased. Risk C: Monitor therapy

Coccidioides immitis Skin Test: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the diagnostic effect of Coccidioides immitis Skin Test. Management: Consider discontinuing cytotoxic chemotherapy several weeks prior to coccidioides immitis skin antigen testing to increase the likelihood of accurate diagnostic results. Risk D: Consider therapy modification

COVID-19 Vaccine (Adenovirus Vector): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of COVID-19 Vaccine (Adenovirus Vector). Management: Administer a 2nd dose using an mRNA COVID-19 vaccine (at least 4 weeks after the primary vaccine dose) and a bivalent booster dose (at least 2 months after the additional mRNA dose or any other boosters). Risk D: Consider therapy modification

COVID-19 Vaccine (Inactivated Virus): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of COVID-19 Vaccine (Inactivated Virus). Risk C: Monitor therapy

COVID-19 Vaccine (mRNA): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of COVID-19 Vaccine (mRNA). Management: Give a 3-dose primary series for all patients aged 6 months and older taking immunosuppressive medications or therapies. Booster doses are recommended for certain age groups. See CDC guidance for details. Risk D: Consider therapy modification

COVID-19 Vaccine (Subunit): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of COVID-19 Vaccine (Subunit). Risk C: Monitor therapy

COVID-19 Vaccine (Virus-like Particles): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of COVID-19 Vaccine (Virus-like Particles). Risk C: Monitor therapy

Deferiprone: Myelosuppressive Agents may enhance the neutropenic effect of Deferiprone. Management: Avoid the concomitant use of deferiprone and myelosuppressive agents whenever possible. If this combination cannot be avoided, monitor the absolute neutrophil count more closely. Risk D: Consider therapy modification

Dengue Tetravalent Vaccine (Live): Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Dengue Tetravalent Vaccine (Live). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Dengue Tetravalent Vaccine (Live). Risk X: Avoid combination

Denosumab: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Management: Consider the risk of serious infections versus the potential benefits of coadministration of denosumab and cytotoxic chemotherapy. If combined, monitor patients for signs/symptoms of serious infections. Risk D: Consider therapy modification

Deucravacitinib: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk X: Avoid combination

Dipyrone: May enhance the adverse/toxic effect of Myelosuppressive Agents. Specifically, the risk for agranulocytosis and pancytopenia may be increased Risk X: Avoid combination

Fexinidazole: Myelosuppressive Agents may enhance the myelosuppressive effect of Fexinidazole. Risk X: Avoid combination

Filgotinib: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk X: Avoid combination

Flucytosine: Cytarabine (Conventional) may diminish the therapeutic effect of Flucytosine. Risk C: Monitor therapy

Inebilizumab: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Inebilizumab. Risk C: Monitor therapy

Influenza Virus Vaccines: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Influenza Virus Vaccines. Management: Administer influenza vaccines at least 2 weeks prior to initiating chemotherapy if possible. If vaccination occurs less than 2 weeks prior to or during chemotherapy, revaccinate at least 3 months after therapy discontinued if immune competence restored. Risk D: Consider therapy modification

Leflunomide: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Leflunomide. Management: Increase the frequency of chronic monitoring of platelet, white blood cell count, and hemoglobin or hematocrit to monthly, instead of every 6 to 8 weeks, if leflunomide is coadministered with immunosuppressive agents, such as cytotoxic chemotherapy. Risk D: Consider therapy modification

Lenograstim: Antineoplastic Agents may diminish the therapeutic effect of Lenograstim. Management: Avoid the use of lenograstim 24 hours before until 24 hours after the completion of myelosuppressive cytotoxic chemotherapy. Risk D: Consider therapy modification

Lipegfilgrastim: Antineoplastic Agents may diminish the therapeutic effect of Lipegfilgrastim. Management: Avoid concomitant use of lipegfilgrastim and myelosuppressive cytotoxic chemotherapy. Lipegfilgrastim should be administered at least 24 hours after the completion of myelosuppressive cytotoxic chemotherapy. Risk D: Consider therapy modification

Natalizumab: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Natalizumab. Risk X: Avoid combination

Ocrelizumab: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Ocrelizumab. Risk C: Monitor therapy

Ofatumumab: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Ofatumumab. Risk C: Monitor therapy

Olaparib: Myelosuppressive Agents may enhance the myelosuppressive effect of Olaparib. Risk C: Monitor therapy

Palifermin: May enhance the adverse/toxic effect of Antineoplastic Agents. Specifically, the duration and severity of oral mucositis may be increased. Management: Do not administer palifermin within 24 hours before, during infusion of, or within 24 hours after administration of myelotoxic chemotherapy. Risk D: Consider therapy modification

Pidotimod: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Pidotimod. Risk C: Monitor therapy

Pimecrolimus: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk X: Avoid combination

Pneumococcal Vaccines: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Pneumococcal Vaccines. Risk C: Monitor therapy

Poliovirus Vaccine (Live/Trivalent/Oral): Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Poliovirus Vaccine (Live/Trivalent/Oral). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Poliovirus Vaccine (Live/Trivalent/Oral). Risk X: Avoid combination

Polymethylmethacrylate: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the potential for allergic or hypersensitivity reactions to Polymethylmethacrylate. Management: Use caution when considering use of bovine collagen-containing implants such as the polymethylmethacrylate-based Bellafill brand implant in patients who are receiving immunosuppressants. Consider use of additional skin tests prior to administration. Risk D: Consider therapy modification

Promazine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Rabies Vaccine: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Rabies Vaccine. Management: Complete rabies vaccination at least 2 weeks before initiation of immunosuppressant therapy if possible. If combined, check for rabies antibody titers, and if vaccination is for post exposure prophylaxis, administer a 5th dose of the vaccine. Risk D: Consider therapy modification

Ropeginterferon Alfa-2b: Myelosuppressive Agents may enhance the myelosuppressive effect of Ropeginterferon Alfa-2b. Management: Avoid coadministration of ropeginterferon alfa-2b and other myelosuppressive agents. If this combination cannot be avoided, monitor patients for excessive myelosuppressive effects. Risk D: Consider therapy modification

Rubella- or Varicella-Containing Live Vaccines: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Rubella- or Varicella-Containing Live Vaccines. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Rubella- or Varicella-Containing Live Vaccines. Risk X: Avoid combination

Ruxolitinib (Topical): Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Ruxolitinib (Topical). Risk X: Avoid combination

Sipuleucel-T: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Sipuleucel-T. Management: Consider reducing the dose or discontinuing the use of immunosuppressants, such as cytotoxic chemotherapy, prior to initiating sipuleucel-T therapy. Risk D: Consider therapy modification

Sphingosine 1-Phosphate (S1P) Receptor Modulator: May enhance the immunosuppressive effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk C: Monitor therapy

Tacrolimus (Topical): Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Tacrolimus (Topical). Risk X: Avoid combination

Talimogene Laherparepvec: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Talimogene Laherparepvec. Specifically, the risk of infection from the live, attenuated herpes simplex virus contained in talimogene laherparepvec may be increased. Risk X: Avoid combination

Tertomotide: Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Tertomotide. Risk X: Avoid combination

Tofacitinib: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Tofacitinib. Risk X: Avoid combination

Typhoid Vaccine: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Typhoid Vaccine. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Typhoid Vaccine. Risk X: Avoid combination

Upadacitinib: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the immunosuppressive effect of Upadacitinib. Risk X: Avoid combination

Vaccines (Inactivated/Non-Replicating): Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Vaccines (Inactivated/Non-Replicating). Management: Give inactivated vaccines at least 2 weeks prior to initiation of chemotherapy when possible. Patients vaccinated less than 14 days before initiating or during chemotherapy should be revaccinated at least 3 months after therapy is complete. Risk D: Consider therapy modification

Vaccines (Live): Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Vaccines (Live). Specifically, the risk of vaccine-associated infection may be increased. Vaccines (Live) may diminish the therapeutic effect of Immunosuppressants (Cytotoxic Chemotherapy). Risk X: Avoid combination

Yellow Fever Vaccine: Immunosuppressants (Cytotoxic Chemotherapy) may enhance the adverse/toxic effect of Yellow Fever Vaccine. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Cytotoxic Chemotherapy) may diminish the therapeutic effect of Yellow Fever Vaccine. Risk X: Avoid combination

Reproductive Considerations

Patients who could become pregnant should avoid becoming pregnant during treatment and be advised of the potential risks if exposure during pregnancy would occur.

Pregnancy Considerations

Based on the mechanism of action and findings from animal reproduction studies, fetal harm may occur if cytarabine is administered during pregnancy. Limb and ear defects have been noted in case reports of cytarabine exposure during the first trimester of pregnancy. The following have also been noted in the neonate: Pancytopenia, WBC depression, electrolyte abnormalities, prematurity, low birth weight, decreased hematocrit or platelets. Risk to the fetus is decreased if treatment can be avoided during the first trimester.

Breastfeeding Considerations

It is not known if cytarabine is present in breast milk. Due to the potential for serious adverse reactions in the breastfed infant, the decision to discontinue cytarabine or to discontinue breastfeeding should take into account the importance of treatment to the mother.

Monitoring Parameters

LFTs (periodic), CBC with differential and platelet count frequently; once blasts are no longer apparent in the peripheral blood, bone marrow should be monitored frequently, serum creatinine and BUN (periodic), serum uric acid. Monitor for signs/symptoms of bleeding, infection, neutropenic fever, and/or tumor lysis syndrome.

The American Society of Clinical Oncology hepatitis B virus (HBV) screening and management provisional clinical opinion (ASCO [Hwang 2020]) recommends HBV screening with hepatitis B surface antigen, hepatitis B core antibody, total Ig or IgG, and antibody to hepatitis B surface antigen prior to beginning (or at the beginning of) systemic anticancer therapy; do not delay treatment for screening/results. Detection of chronic or past HBV infection requires a risk assessment to determine antiviral prophylaxis requirements, monitoring, and follow-up.

Mechanism of Action

Cytarabine inhibits DNA synthesis. Cytarabine gains entry into cells by a carrier process, and then must be converted to its active compound, aracytidine triphosphate. Cytarabine is a pyrimidine analog and is incorporated into DNA; however, the primary action is inhibition of DNA polymerase resulting in decreased DNA synthesis and repair. The degree of cytotoxicity correlates linearly with incorporation into DNA; therefore, incorporation into the DNA is responsible for drug activity and toxicity. Cytarabine is specific for the S phase of the cell cycle (blocks progression from the G1 to the S phase).

Pharmacokinetics

Absorption: Not effective when administered orally; less than 20% absorbed orally.

Distribution: Vd: 3 ± 11.9 L/kg; total body water; widely and rapidly since it enters the cells readily; crosses blood-brain barrier with CSF levels of 40% to 50% of plasma level.

Bioavailability: SubQ: 89% to 100% (Leveque 2014).

Protein binding: 13%.

Metabolism: Primarily hepatic; metabolized by deoxycytidine kinase and other nucleotide kinases to aracytidine triphosphate (active); about 86% to 96% of dose is metabolized to inactive uracil arabinoside (ARA-U); intrathecal administration results in little conversion to ARA-U due to the low levels of deaminase in the cerebral spinal fluid.

Half-life elimination: IV: Initial: 7 to 20 minutes; Terminal: 1 to 3 hours; Intrathecal: 2 to 6 hours.

Time to peak, plasma: SubQ: 20 to 60 minutes.

Excretion: Urine (~80%; 90% as metabolite ARA-U) within 24 hours.

Pricing: US

Solution (Cytarabine (PF) Injection)

20 mg/mL (per mL): $1.52

100 mg/mL (per mL): $1.10 - $1.25

Solution (Cytarabine Injection)

20 mg/mL (per mL): $0.75

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Brand Names: International
  • Alexan (AE, AT, BG, CH, CL, CZ, DK, EC, EG, GB, HK, HN, HR, HU, ID, IE, IL, IT, JO, LB, MT, PL, PT, RU, SA, SE, SI, SK, TH, TR, UA, VN, ZA);
  • ARA-Cell (IL);
  • Arabine (DK, FI, FR);
  • Aracytin (CO, GR, IT, UY);
  • Aracytine (FR);
  • Cancyt (PH, ZW);
  • Citarabina (ES);
  • Cyclocide N (JP);
  • Cytabin (BD);
  • Cytarabin (DE, NO);
  • Cytarabine Injection (GB, NZ);
  • Cytarabinum-Delta West (LU);
  • Cytarine (EG, IN, SG, TH, TW, VN, ZW);
  • Cytonal (RU, TR);
  • Cytosa U (KR);
  • Cytosar (AE, AT, BE, BG, BH, CH, CN, CY, CZ, EE, FI, GB, GH, HK, HN, HR, HU, IL, IQ, IR, JO, KE, KW, LB, LU, LV, LY, NL, OM, PE, PK, PT, QA, RO, SA, SY, TH, TW, TZ, UA, UG, VN, YE, ZM);
  • Cytosar-U (ID, KR, MY, VE);
  • Cytox (PH);
  • Cytrostar (LK);
  • Darbin (BR, PY);
  • Laracit (CO, CR, DO, GT, HN, MX, PA, SV);
  • Medsara (MX);
  • Mokcell (CR, DO, GT, HN, PA, PE, SV);
  • Mox (DO);
  • Oncotar (LK);
  • Saidewei (CN);
  • Tabine (EG, PH);
  • Tarabin (BD)


For country code abbreviations (show table)
  1. <800> Hazardous Drugs–Handling in Healthcare Settings. United States Pharmacopeia and National Formulary (USP 43-NF 38). Rockville, MD: United States Pharmacopeia Convention; 2020:74-92.
  2. Adès L, Chevret S, Raffous, et al. Is Cytarabine Useful in the Treatment of Acute Promyelocytic Leukemia? Results of a Randomized Trial From the European Acute Promyelocytic Leukemia Group. J Clin Oncol. 2006;24(36):5703-5710. [PubMed 17116939]
  3. Adès L, Sanz MA, Chevret S, et al. Treatment of Newly Diagnosed Acute Promyelocytic Leukemia (APL): A Comparison of French-Belgian-Swiss and PETHEMA Results. Blood. 2008;111(3):1078-1084. [PubMed 17975017]
  4. Ahlfors CE. Benzyl alcohol, kernicterus, and unbound bilirubin. J Pediatr. 2001;139(2):317-319. [PubMed 11487763]
  5. Allwood M, Stanley A. The Cytotoxics Handbook. 4th edition. Abingdon: Radcliffe Medical Press; 2003.
  6. Amadori S, Arcese W, Isacchi G, et al. Mitoxantrone, etoposide, and intermediate-dose cytarabine: an effective and tolerable regimen for the treatment of refractory acute myeloid leukemia. J Clin Oncol. 1991;9(7):1210-1214. [PubMed 2045861]
  7. American Academy of Pediatrics Committee on Drugs. "Inactive" ingredients in pharmaceutical products: update (subject review). Pediatrics. 1997;99(2):268-278. [PubMed 9024461]
  8. Aparicio J, Segura A, Garcerá S, et al. ESHAP is an Active Regimen for Relapsing Hodgkin's Disease. Ann Oncol. 1999;10(5):593-595. [PubMed 10416011]
  9. Archimbaud E, Leblond V, Michallet M, et al. Intensive Sequential Chemotherapy With Mitoxantrone and Continuous Infusion Etoposide and Cytarabine for Previously Treated Acute Myelogenous Leukemia. Blood. 1991;77(9):1894-1900. [PubMed 2018832]
  10. Archimbaud E, Thomas X, Leblond V, et al. Timed Sequential Chemotherapy for Previously Treated Patients With Acute Myeloid Leukemia: Long-Term Follow-Up of the Etoposide, Mitoxantrone, and Cytarabine-86 Trial. J Clin Oncol. 1995;13(1):11-18. [PubMed 7799010]
  11. Arlin Z, Case DC Jr, Moore J, et al. Randomized Multicenter Trial of Cytosine Arabinoside With Mitoxantrone or Daunorubicin in Previously Untreated Adult Patients With Acute Nonlymphocytic Leukemia (ANLL). Lederle Cooperative Group. Leukemia. 1990;4(3):177-183. [PubMed 2179638]
  12. Aronoff GR, Bennett WM, Berns JS, et al. Drug Prescribing in Renal Failure: Dosing Guidelines for Adults and Children. 5th ed. American College of Physicians; 2007:98, 170.
  13. Basch E, Prestrud AA, Hesketh PJ, et al. Antiemetics: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2011;29(31):4189-4198. [PubMed 21947834]
  14. Beaumont AL, Suner L. Drug-induced aseptic meningitis in a Ph+ ALL patient with meningeal involvement. Blood. 2020;136(4):520. doi:10.1182/blood.2019004195 [PubMed 32702128]
  15. Becker PS, Kantarjian HM, Appelbaum FR, et al. Clofarabine with high dose cytarabine and granulocyte colony-stimulating factor (G-CSF) priming for relapsed and refractory acute myeloid leukaemia. Br J Haematol. 2011;155(2):182-189. doi:10.1111/j.1365-2141.2011.08831.x [PubMed 21848522]
  16. Bishop JF, Matthews JP, Young GA, et al. A Randomized Study of High-Dose Cytarabine in Induction in Acute Myeloid Leukemia. Blood. 1996;87(5):1710-1717. [PubMed 8634416]
  17. Bleyer WA, Coccia PF, Sather HN, et al. Reduction in Central Nervous System Leukemia With a Pharmacokinetically Derived Intrathecal Methotrexate Dosage Regimen. J Clin Oncol. 1983;1(5):317-325. [PubMed 6366138]
  18. Bowman WP, Shuster JJ, Cook B, et al. Improved survival for children with B-cell acute lymphoblastic leukemia and stage IV small noncleaved-cell lymphoma: a pediatric oncology group study. J Clin Oncol. 1996; 14(4):1252-1261. [PubMed 8648381]
  19. Bubalo J, Carpenter PA, Majhail N, et al. Conditioning chemotherapy dose adjustment in obese patients: a review and position statement by the American Society for Blood and Marrow Transplantation practice guideline committee. Biol Blood Marrow Transplant. 2014;20(5):600-616. doi:10.1016/j.bbmt.2014.01.019 [PubMed 24462742]
  20. Burnett AK, Russell NH, Hills RK, et al. Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial. J Clin Oncol. 2013;31(27):3360-3368. doi:10.1200/JCO.2012.47.4874 [PubMed 23940227]
  21. Cairo MS, Gerrard M, Sposto R, et al. Results of a randomized international study of high-risk central nervous system B non-Hodgkin lymphoma and B acute lymphoblastic leukemia in children and adolescents. Blood. 2007;109(7):2736-2743. [PubMed 17138821]
  22. Capizzi RL, Poole M, Cooper MR, et al. Treatment of poor risk acute leukemia with sequential high-dose ARA-C and asparaginase. Blood. 1984;63(3):694-700. [PubMed 6696996]
  23. Capizzi RL, White JC, Powell BL, et al. Effect of Dose on the Pharmacokinetic and Pharmacodynamic Effects of Cytarabine. Semin Hematol. 1991;28(3)(suppl 4):54-69. [PubMed 1780754]
  24. Cassileth PA, Harrington DP, Appelbaum FR, et al. Chemotherapy Compared With Autologous or Allogeneic Bone Marrow Transplantation in the Management of Acute Myeloid Leukemia in First Remission. N Engl J Med. 1998;339(23):1649-1656. [PubMed 9834301]
  25. Centers for Disease Control (CDC). Neonatal deaths associated with use of benzyl alcohol - United States. MMWR Morb Mortal Wkly Rep. 1982;31(22):290-291. http://www.cdc.gov/mmwr/preview/mmwrhtml/00001109.htm [PubMed 6810084]
  26. Chamberlain MC. Leptomeningeal metastasis. Curr Opin Oncol. 2010;22(6):627-635. [PubMed 20689429]
  27. Cheung YW, Vishnuvajjala BR, Flora KP. Stability of cytarabine, methotrexate sodium, and hydrocortisone sodium succinate admixtures. Am J Hosp Pharm. 1984;41(9):1802-1806. [PubMed 6496516]
  28. Chiu A, Kohler S, McGuire J, et al. Cytarabine-induced acute generalized exanthematous pustulosis. J Am Acad Dermatol. 2002;47(4):633-635. [PubMed 12271319]
  29. Cho AR, Yoon YH, Kim JG, Kim YJ, Lee JY. Uveoretinal adverse effects presented during systemic anticancer chemotherapy: a 10-year single center experience. J Korean Med Sci. 2018;33(7):e55. doi:10.3346/jkms.2018.33.e55 [PubMed 29359539]
  30. Chopra R, McMillan AK, Linch DC, et al .The Place of High-Dose BEAM Therapy and Autologous Bone Marrow Transplantation in Poor-Risk Hodgkin's Disease. A Single-Center Eight-Year Study of 155 Patients. Blood. 1993;81(5):1137-1145. [PubMed 8443375]
  31. Colwill R, Crump M, Couture F, et al. Mini-BEAM as Salvage Therapy for Relapsed or Refractory Hodgkin's Disease Before Intensive Therapy and Autologous Bone Marrow Transplantation. J Clin Oncol. 1995;13(2):396-402. [PubMed 7844600]
  32. Cooper TM, Alonzo TA, Gerbing RB, et al. AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia. Cancer. 2014;120(16):2482-2489. doi:10.1002/cncr.28674 [PubMed 24771494]
  33. Cooper TM, Franklin J, Gerbing RB, et al. AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination with chemotherapy for newly diagnosed childhood acute myeloid leukemia: a report from the Children's Oncology Group. Cancer. 2012;118(3):761-769. [PubMed 21766293]
  34. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33(2):379-389. doi:10.1038/s41375-018-0312-9 [PubMed 30555165]
  35. Cortes J, O’Brien SM, Pierce S, et al. The Value of High-Dose Systemic Chemotherapy and Intrathecal Therapy for Central Nervous System Prophylaxis in Different Risk Groups of Adult Acute Lymphoblastic Leukemia. Blood. 1995;86(6):2091-2097. [PubMed 7662956]
  36. Cytarabine [prescribing information]. Lake Forest, IL: Hospira; June 2018.
  37. Cytarabine bulk package [prescribing information]. Lake Forest, IL: Hospira; December 2017.
  38. DeAngelis LM, Boutros D. Leptomeningeal Metastasis. Cancer Invest. 2005;23(2):145-154. [PubMed 15813508]
  39. de Lemos ML, Monfared S, Denyssevych T, et al. Evaluation of Osmolality and pH of Various Concentrations of Methotrexate, Cytarabine, and Thiotepa Prepared in Normal Saline, Sterile Water for Injection, and Lactated Ringers’s Solution for Intrathecal Administration. J Oncol Pharm Pract. 2009;15(1):45-52. [PubMed 18772215]
  40. Dillman RO, Davis RB, Green MR, et al. A Comparative Study of Two Different Doses of Cytarabine for Acute Myeloid Leukemia: A Phase III Trial of Cancer and Leukemia Group B. Blood. 1991;78(10):2520-2526. [PubMed 1824249]
  41. Döhner H, Estey EH, Amadori S, et al. Diagnosis and Management of Acute Myeloid Leukemia in Adults: Recommendations From an International Expert Panel, on Behalf of the European LeukemiaNet. Blood. 2010;115(3):453-474. [PubMed 19880497]
  42. Dupuis LL, Boodhan S, Sung L, et al; Pediatric Oncology Group of Ontario. Guideline for the classification of the acute emetogenic potential of antineoplastic medication in pediatric cancer patients. Pediatr Blood Cancer. 2011;57(2):191-198. [PubMed 21465637]
  43. Dupuis LL, Boodhan S, Holdsworth M, et al. Guideline for the prevention of acute nausea and vomiting due to antineoplastic medication in pediatric cancer patients. Pediatr Blood Cancer. 2013;60(7):1073-1082. [PubMed 23512831]
  44. Fenaux P, Mufti GJ, Hellström-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28(4):562-569. doi:10.1200/JCO.2009.23.8329 [PubMed 20026804]
  45. Fernandez HF, Sun Z, Yao X, et al. Anthracycline Dose Intensification in Acute Myeloid Leukemia. N Engl J Med. 2009;361(13):1249-1259. [PubMed 19776406]
  46. Ferreri AJ, Reni M, Foppoli M, et al. High-dose cytarabine plus high-dose methotrexate versus high-dose methotrexate alone in patients with primary CNS lymphoma: a randomised phase 2 trial. Lancet. 2009;374(9700):1512-1520. [PubMed 19767089]
  47. Floyd J, Mirza I, Sachs B, et al. Hepatotoxicity of Chemotherapy. Semin Oncol. 2006;33(1):50-67. [PubMed 16473644]
  48. Floyd JD, Nguyen DT, Lobins RL, et al. Cardiotoxicity of Cancer Therapy. J Clin Oncol, 2005;23(30):7685-7696. [PubMed 16234530]
  49. Forghieri F, Luppi M, Morselli M, Potenza L. Cytarabine-related lung infiltrates on high resolution computerized tomography: a possible complication with benign outcome in leukemic patients. Haematologica. 2007;92(9):e85-e90. doi:10.3324/haematol.11697 [PubMed 17768138]
  50. Gamis AS, Alonzo TA, Meshinchi S, et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol. 2014;32(27):3021-3032. [PubMed 25092781]
  51. Gardin C, Turlure P, Fagot T, et al. Postremission Treatment of Elderly Patients With Acute Myeloid Leukemia in First Complete Remission After Intensive Induction Chemotherapy: Results of the Multicenter Randomized Acute Leukemia French Association (ALFA) 9803 Trial. Blood. 2007;109(12):5129-5135. [PubMed 17341661]
  52. Gaynon PS, Steinherz PG, Bleyer WA, et al. Improved Therapy for Children With Acute Lymphoblastic Leukemia and Unfavorable Presenting Features: A Follow-Up Report of the Childrens Cancer Group Study CCG-106. J Clin Oncol. 1993;11(11):2234-2242. [PubMed 8229139]
  53. Geisler CH, Kolstad A, Laurell A, et al; Nordic Lymphoma Group. Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo-purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group. Blood. 2008;112(7):2687-2693. doi:10.1182/blood-2008-03-147025 [PubMed 18625886]
  54. Gibson BE, Webb DK, Howman AJ, et al. Results of a randomized trial in children with acute myeloid leukaemia: medical research council AML12 trial. Br J Haematol. 2011;155(3):366-376. [PubMed 21902686]
  55. Glantz MJ, LaFollette S, Jaeckle KA, et al. Randomized Trial of a Slow-Release Versus a Standard Formulation of Cytarabine for the Intrathecal Treatment of Lymphomatous Meningitis. J Clin Oncol. 1999;17(10):3110-3116. [PubMed 10506606]
  56. Goldman S, Smith L, Anderson JR, et al. Rituximab and FAB/LMB 96 chemotherapy in children with Stage III/IV B-cell non-Hodgkin lymphoma: a Children's Oncology Group report. Leukemia. 2013;27(5):1174-1177. [PubMed 22940833]
  57. Griggs JJ, Bohlke K, Balaban EP, et al. Appropriate systemic therapy dosing for obese adult patients with cancer: ASCO guideline update. J Clin Oncol. 2021;39(18):2037-2048. doi:10.1200/JCO.21.00471 [PubMed 33939491]
  58. Guest EM, Aplenc R, Sung L, et al. Gemtuzumab ozogamicin (GO) in infants with acute myeloid leukemia (AML) - combined results from the Children's Oncology Group (COG) trials. AAML03P1 and AAML0531. Blood. 2014;124:377.
  59. Guilhot F, Chastang C, Michallet M, et al. Interferon Alfa-2b Combined With Cytarabine Versus Interferon Alone in Chronic Myelogenous Leukemia. French Chronic Myeloid Leukemia Study Group. N Engl J Med. 1997;337(4):223-229. [PubMed 9227927]
  60. Hermine O, Hoster E, Walewski J, et al; European Mantle Cell Lymphoma Network. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet. 2016;388(10044):565-575. doi:10.1016/S0140-6736(16)00739-X [PubMed 27313086]
  61. Herzig RH, Hines JD, Herzig GP, et al. Cerebellar Toxicity With High-Dose Cytosine Arabinoside. J Clin Oncol. 1987;5(6):927-932. [PubMed 3585447]
  62. Herzig RH, Lazarus HM, Wolff SN, et al. High-Dose Cytosine Arabinoside Therapy With and Without Anthracycline Antibiotics for Remission Reinduction of Acute Nonlymphoblastic Leukemia. J Clin Oncol. 1985;3(7):992-997. [PubMed 3894588]
  63. Hesketh PJ, Kris MG, Basch E, et al. Antiemetics: ASCO guideline update. J Clin Oncol. 2020;38(24):2782-2797. doi:10.1200/JCO.20.01296 [PubMed 32658626]
  64. Hiddemann W. Cytosine Arabinoside in the Treatment of Acute Myeloid Leukemia: The Role and Place of High-Dose Regimens. Ann Hematol. 1991;62(4):119-128. [PubMed 2031974]
  65. Hwang JP, Feld JJ, Hammond SP, et al. Hepatitis B virus screening and management for patients with cancer prior to therapy: ASCO provisional clinical opinion update. J Clin Oncol. 2020;38(31):3698-3715. doi:10.1200/JCO.20.01757 [PubMed 32716741]
  66. Josting A, Rudolph C, Reiser M, et al. Time-Intensified Dexamethasone/Cisplatin/Cytarabine: An Effective Salvage Therapy With Low Toxicity in Patients With Relapsed and Refractory Hodgkin's Disease. Ann Oncol. 2002;13(10):1628-1635. [PubMed 12377653]
  67. Kantarjian HM, O'Brien S, Smith TL, et al. Results of Treatment With Hyper-CVAD, A Dose-Intensive Regimen, in Adult Acute Lymphocytic Leukemia. J Clin Oncol. 2000;18(3):547-561. [PubMed 10653870]
  68. Kerr JZ, Berg S, Blaney SM. Intrathecal Chemotherapy. Crit Rev Oncol Hematol. 2001;37(3):227-236. [PubMed 11248578]
  69. Kintzel PE, Dorr RT. Anticancer Drug Renal Toxicity and Elimination: Dosing Guidelines for Altered Renal Function. Cancer Treat Rev. 1995;21(1):33-64. [PubMed 7859226]
  70. Koren G, Beatty K, Seto A, et al. The Effects of Impaired Liver Function on the Elimination of Antineoplastic Agents. Ann Pharmacother. 1992;26(3):363-371. [PubMed 1554959]
  71. Krens SD, Lassche G, Jansman FGA, et al. Dose recommendations for anticancer drugs in patients with renal or hepatic impairment. Lancet Oncol. 2019;20(4):e200-e207. doi:10.1016/S1470-2045(19)30145-7 [PubMed 30942181]
  72. Land VJ, Shuster JJ, Crist WM, et al. Comparison of Two Schedules of Intermediate-Dose Methotrexate and Cytarabine Consolidation Therapy for Childhood B-Precursor Cell Acute Lymphoblastic Leukemia: A Pediatric Oncology Group Study. J Clin Oncol. 1994;12(9):1939-1945. [PubMed 8083715]
  73. Larsen EC, Devidas M, Chen s, et al. Dexamethasone and high-dose methotrexate improve outcome for children and young adults with high-risk B-acute lymphoblastic leukemia: a report from Children's Oncology Group Study AALL0232. J Clin Oncol. 2016;34(20):2380-2388. [PubMed 27114587]
  74. Larson RA, Dodge RK, Burns CP, et al. A Five-Drug Remission Induction Regimen With Intensive Consolidation for Adults With Acute Lymphoblastic Leukemia: Cancer and Leukemia Group B Study 8811. Blood. 1995;85(8):2025-2037. [PubMed 7718875]
  75. Le Gouill S, Thieblemont C, Oberic L, et al; LYSA Group. rituximab after autologous stem-cell transplantation in mantle-cell lymphoma. N Engl J Med. 2017;377(13):1250-1260. doi:10.1056/NEJMoa1701769 [PubMed 28953447]
  76. Lee EJ, Petroni GR, Schiffer CA, et al. Brief-Duration High-Intensity Chemotherapy for Patients With Small Noncleaved-Cell Lymphoma or FAB L3 Acute Lymphocytic Leukemia: Results of Cancer and Leukemia Group B Study 9251. J Clin Oncol. 2001;19(20):4014-4022. [PubMed 11600602]
  77. Lenfant C, Greiner N, Duprez T. Cytarabine-induced encephalitis. J Belg Soc Radiol. 2021;105(1):46. doi:10.5334/jbsr.2492 [PubMed 34611580]
  78. Leveque D. Subcutaneous administration of anticancer agents. Anticancer Res. 2014;34(4):1579-1586. [PubMed 24692685]
  79. Lin WY, Liu HC, Yeh TC, et al. Triple Intrathecal Therapy Without Cranial Irradiation for Central Nervous System Preventive Therapy in Childhood Acute Lymphoblastic Leukemia. Pediatr Blood Cancer. 2008;50(3):523-527. [PubMed 17455314]
  80. Linch DC, Yung L, Smith P, et al. Final Analysis of the UKLG LY02 Trial Comparing 6-8 Cycles of CHOP With 3 Cycles of CHOP Followed by a BEAM Autograft in Patients <65 Years With Poor Prognosis Histologically Aggressive NHL. Br J Haematol. 2010;149(2):237-243. [PubMed 20201949]
  81. Linker CA, Levitt LJ, O'Donnell M, et al. Treatment of Adult Acute Lymphoblastic Leukemia With Intensive Cyclical Chemotherapy: A Follow-up Report. Blood. 1991;78(11):2814-2822. [PubMed 1835410]
  82. Lo Coco F, Avvisati G, Vignetti M, et al. Front-Line Treatment of Acute Promyelocytic Leukemia With AIDA Induction Followed by Risk-Adapted Consolidation for Adults Younger Than 61 Years: Results of the AIDA-2000 Trial of the GIMEMA Group. Blood. 2010;116(17):3171-3179. [PubMed 20644121]
  83. Löwenberg B, Pabst T, Vellenga E, et al. Cytarabine Dose for Acute Myeloid Leukemia. N Engl J Med. 2011;364(11):1027-1036. [PubMed 21410371]
  84. Magrath I, Adde M, Shad A, et al. Adults and children with small non-cleaved-cell lymphoma have a similar excellent outcome when treated with the same chemotherapy regimen. J Clin Oncol. 1996;14(3):925-934. doi:10.1200/JCO.1996.14.3.925 [PubMed 8622041]
  85. Malhotra P, Mahi S, Lal V, Kumari S, Jain S, Varma S. Cytarabine-induced neurotoxicity responding to methyl prednisolone. Am J Hematol. 2004;77(4):416. doi:10.1002/ajh.20171 [PubMed 15551283]
  86. Martín A, Fernández-Jiménez MC, Caballero MD, et al. Long-Term Follow-Up in Patients Treated With Mini-BEAM as Salvage Therapy for Relapsed or Refractory Hodgkin's Disease. Br J Haematol. 2001;113(1):161-171. [PubMed 11328296]
  87. Matloub Y, Lindemulder S, Gaynon PS, et al. Intrathecal Triple Therapy Decreases Central Nervous System Relapse but Fails to Improve Event-Free Survival When Compared With Intrathecal Methotrexate: Results of the Children's Cancer Group (CCG) 1952 Study for Standard-Risk Acute Lymphoblastic Leukemia, Reported by the Children's Oncology Group. Blood. 2006;108(4):1165-1173. [PubMed 16609069]
  88. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive Postremission Chemotherapy in Adults With Acute Myeloid Leukemia, Cancer and Leukemia Group B. N Engl J Med. 1994;331(14):896-903. [PubMed 8078551]
  89. Mead GM, Barrans SL, Qian W, et al; UK National Cancer Research Institute Lymphoma Clinical Studies Group; Australasian Leukaemia and Lymphoma Group. A prospective clinicopathologic study of dose-modified CODOX-M/IVAC in patients with sporadic Burkitt lymphoma defined using cytogenetic and immunophenotypic criteria (MRC/NCRI LY10 trial). Blood. 2008;112(6):2248-2260. doi:10.1182/blood-2008-03-145128 [PubMed 18612102]
  90. Milligan DW, Wheatley K, Littlewood T, et al. Fludarabine and Cytosine are Less Effective than Standard ADE Chemotherapy in High-Risk Acute Myeloid Leukemia, and Addition of G-CSF and ATRA are not Beneficial: Results of the MRC AML-HR Randomized Trial. Blood. 2006;107(12):4614-4622. [PubMed 16484584]
  91. Montillo M, Mirto S, Petti MC, et al. Fludarabine, Cytarabine, and G-CSF (FLAG) for the Treatment of Poor Risk Acute Myeloid Leukemia. Am J Hematol. 1998;58(2):105-109. [PubMed 9625576]
  92. Morgan C, Tillett T, Braybrooke J, et al. Management of Uncommon Chemotherapy-Induced Emergencies. Lancet Oncol. 2011;12(8):806-814. [PubMed 21276754]
  93. Neuss MN, Gilmore TR, Belderson KM, et al. 2016 updated American Society of Clinical Oncology/Oncology Nursing Society chemotherapy administration safety standards, including standards for pediatric oncology. J Oncol Pract. 2016;12(12):1262-1271. doi:10.1200/JOP.2016.017905 [PubMed 27868581]
  94. Özcan G, Uçakhan ÖÖ. Cytarabine-induced corneal toxicity: clinical features and relief of symptoms with loteprednol etabonate 0.5% in two patients. Turk J Ophthalmol. 2021;51(2):114-117. doi:10.4274/tjo.galenos.2020.99248 [PubMed 33951900]
  95. Pallazola VA, Murray JC, Al Harthy M, Zimmerman SL, Webster J, Gondek LP. Anthracycline-induced acute myocarditis and ventricular fibrillation arrest. Am J Hematol. 2018;93(3):469-470. doi:10.1002/ajh.24989 [PubMed 29194712]
  96. Patte C, Auperin A, Gerrard M, et al. Results of the randomized international FAB/LMB96 trial for intermediate risk B-cell non-Hodgkin lymphoma in children and adolescents: it is possible to reduce treatment for the early responding patients. Blood. 2007;109(7):2773-2780. [PubMed 17132719]
  97. Patte C, Auperin A, Michon J, et al. The Société Française d'Oncologie Pédiatrique LMB89 protocol: highly effective multiagent chemotherapy tailored to the tumor burden and initial response in 561 unselected children with B-cell lymphomas and L3 leukemia. Blood. 2001;97(11):3370-3379. [PubMed 11369626]
  98. Paw Cho Sing E, Robinson PD, Flank J, et al. Classification of the acute emetogenicity of chemotherapy in pediatric patients: a clinical practice guideline. Pediatr Blood Cancer. 2019;66(5):e27646. doi:10.1002/pbc.27646 [PubMed 30729654]
  99. Pease CL, Horton TM, McClain KL, et al. Aseptic Meningitis in a Child After Systemic Treatment With High Dose Cytarabine. Pediatr Infect Dis J. 2001;20(1):87-89. [PubMed 11176579]
  100. Pegram AA, Kennedy LD. Prevention and Treatment of Veno-Occlusive Disease. Ann Pharmacother. 2001;35(7-8):935-942. [PubMed 11485147]
  101. Pieters R, Schrappe M, De Lorenzo P, et al. A Treatment Protocol for Infants Younger Than 1 Year With Acute Lymphoblastic Leukaemia (Interfant-99): An Observational Study and a Multicentre Randomised Trial. Lancet. 2007;370(9583):240-250. [PubMed 17658395]
  102. Place AE, Stevenson KE, Vrooman LM, et al. Intravenous pegylated asparaginase versus intramuscular native Escherichia coli L-asparaginase in newly diagnosed childhood acute lymphoblastic leukaemia (DFCI 05-001): a randomised, open-label phase 3 trial. Lancet Oncol. 2015;16(16):1677-1690. [PubMed 26549586]
  103. Powell BL, Moser B, Stock W, et al. Arsenic Trioxide Improves Event-Free and Over-All Survival for Adults With Acute Promyelocytic Leukemia: North American Leukemia Intergroup Study C9710. Blood. 2010;116(19):3751-3757. [PubMed 20705755]
  104. Radeski D, Cull GM, Cain M, Hackett LP, Ilett KF. Effective clearance of Ara-U the major metabolite of cytosine arabinoside (Ara-C) by hemodialysis in a patient with lymphoma and end-stage renal failure. Cancer Chemother Pharmacol. 2011;67(4):765-768. doi:10.1007/s00280-010-1373-2 [PubMed 20532508]
  105. Rassiga AL, Schwartz HJ, Forman WB, Crum ED. Cytarabine-induced anaphylaxis. Demonstration of antibody and successful desensitization. Arch Intern Med. 1980;140(3):425-426. doi:10.1001/archinte.140.3.425 [PubMed 6928748]
  106. Ribeiro RC, Rivera GK, Hudson M, et al. An intensive re-treatment protocol for children with an isolated CNS relapse of acute lymphoblastic leukemia. J Clin Oncol. 1995;13(2):333-338. doi:10.1200/JCO.1995.13.2.333 [PubMed 7844594]
  107. Rivera GK, Hudson MM, Liu Q, et al. Effectiveness of intensified rotational combination chemotherapy for late hematologic relapse of childhood acute lymphoblastic leukemia. Blood. 1996;88(3):831-837. [PubMed 8704238]
  108. Rizzieri DA, Johnson JL, Niedzwiecki D, et al. Intensive Chemotherapy With and Without Cranial Radiation for Burkitt Leukemia and Lymphoma: Final Results of Cancer and Leukemia Group B Study 9251. Cancer. 2004;100(7):1438-1448. [PubMed 15042678]
  109. Roila F, Herrstedt J, Aapro M, et al; ESMO/MASCC Guidelines Working Group. Guideline update for MASCC and ESMO in the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting: results of the Perugia consensus conference. Ann Oncol. 2010;21(suppl 5):v232-v243. [PubMed 20555089]
  110. Roila F, Molassiotis A, Herrstedt J, et al; participants of the MASCC/ESMO Consensus Conference Copenhagen 2015. 2016 MASCC and ESMO guideline update for the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting and of nausea and vomiting in advanced cancer patients. Ann Oncol. 2016;27(suppl 5):v119-v133. [PubMed 27664248]
  111. Romaguera JE, Fayad L, Rodriguez MA, et al. High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine. J Clin Oncol. 2005;23(28):7013-7023. doi:10.1200/JCO.2005.01.1825 [PubMed 16145068]
  112. Rubenstein JL, Hsi ED, Johnson JL, et al. Intensive chemotherapy and immunotherapy in patients with newly diagnosed primary CNS lymphoma: CALGB 50202 (Alliance 50202). J Clin Oncol. 2013;31(25):3061-3068. doi:10.1200/JCO.2012.46.9957 [PubMed 23569323]
  113. Rudnick SA, Cadman EC, Capizzi RL, Skeel RT, Bertino JR, McIntosh S. High dose cytosine arabinoside (HDARAC) in refractory acute leukemia. Cancer. 1979;44(4):1189-1193. doi:10.1002/1097-0142(197910)44:4<1189::aid-cncr2820440404>3.0.co;2-o [PubMed 498009]
  114. Sahu KK, Yanamandra U, Malhotra P. Ara-C related red ear syndrome. Ear Nose Throat J. 2019;98(3):169-170. doi:10.1177/0145561319831270 [PubMed 30836786]
  115. Sainz de la Maza Cantero S, Jiménez Martín A, de Felipe Mimbrera A, Corral Corral Í. Cerebellar toxicity due to cytarabine: a series of 4 cases. Toxicidad cerebelosa por citarabina: serie de 4 casos. Neurologia. 2016;31(7):491-492. doi:10.1016/j.nrl.2014.12.017 [PubMed 25728956]
  116. Saito B, Nakamaki T, Nakashima H, et al. Reversible posterior leukoencephalopathy syndrome after repeat intermediate-dose cytarabine chemotherapy in a patient with acute myeloid leukemia. Am J Hematol. 2007;82(4):304-306. doi:10.1002/ajh.20772 [PubMed 16947320]
  117. Sanz MA, Grimwade D, Tallman MS, et al. Management of Acute Promyelocytic Leukemia: Recommendations From an Expert Panel on Behalf of the European LeukemiaNet. Blood. 2009;113(9):1875-1891. [PubMed 18812465]
  118. Sanz MA, Montesinos P, Rayón C, et al. Risk-Adapted Treatment of Acute Promyelocytic Leukemia Based on all-trans Retinoic Acid and Anthracycline With Addition of Cytarabine in Consolidation Therapy for High-Risk Patients: Further Improvements in Treatment Outcome. Blood. 2010;115(25):5137-5146. [PubMed 20393132]
  119. Shah GD, Yahalom J, Correa DD, et al. Combined immunochemotherapy with reduced whole-brain radiotherapy for newly diagnosed primary CNS lymphoma. J Clin Oncol. 2007;25(30):4730-4735. doi:10.1200/JCO.2007.12.5062 [PubMed 17947720]
  120. Smith G, Damon LE, Rugo HS, et al. High-Dose Cytarabine Dose Modification Reduces the Incidence of Neurotoxicity in Patients With Renal Insufficiency. J Clin Oncol. 1997;15(2):833-839. [PubMed 9053511]
  121. Stasi R, Venditti A, Del Poeta G, et al. High-Dose Chemotherapy in Adult Acute Myeloid Leukemia: Rationale and Results. Leuk Res. 1996;20(7):535-549. [PubMed 8795687]
  122. Steinherz PG, Redner A, Steinherz L, Meyers P, Tan C, Heller G. Development of a new intensive therapy for acute lymphoblastic leukemia in children at increased risk of early relapse. The Memorial Sloan-Kettering-New York-II protocol. Cancer. 1993;72(10):3120-3130. [PubMed 8221579]
  123. Stock W, Luger SM, Advani AS, et al. A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403. Blood. 2019;133(14):1548-1559. doi:10.1182/blood-2018-10-881961 [PubMed 30658992]
  124. Stone RM, Berg DT, George SL, et al. Postremission Therapy in Older Patients With de novo Acute Myeloid Leukemia: A Randomized Trial Comparing Mitoxantrone and Intermediate-Dose Cytarabine With Standard-Dose Cytarabine. Blood. 2001;98(3):548-553. [PubMed 11468148]
  125. Storring JM, Minden MD, Kao S, et al. Treatment of Adults With BCR-ABL Negative Acute Lymphoblastic Leukaemia With a Modified Paediatric Regimen. Br J Haematol. 2009;146(1):76-85. [PubMed 19438471]
  126. Termuhlen AM, Smith LM, Perkins SL, et al. Disseminated lymphoblastic lymphoma in children and adolescents: results of the COG A5971 trial: a report from the Children's Oncology Group. Br J Haematol. 2013;162(6):792-801. [PubMed 23889312]
  127. Thomas DA, Faderl S, O'Brien S, et al. Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia. Cancer. 2006;106(7):1569-1580. doi:10.1002/cncr.21776 [PubMed 16502413]
  128. Tilly H, Castaigne S, Bordessoule D, et al. Low-dose cytarabine versus intensive chemotherapy in the treatment of acute nonlymphocytic leukemia in the elderly. J Clin Oncol. 1990;8(2):272-279. doi:10.1200/JCO.1990.8.2.272 [PubMed 2299370]
  129. Trissel LA, King KM, Zhang Y, et al. Physical and Chemical Stability of Methotrexate, Cytarabine, and Hydrocortisone in Elliott's B Solution for Intrathecal Use. J Oncol Pharm Pract. 2002;8(1):27-32.
  130. Truica CI, Frankel SR. Acute rhabdomyolysis as a complication of cytarabine chemotherapy for acute myeloid leukemia: case report and review of literature. Am J Hematol. 2002;70(4):320-323. [PubMed 12210815]
  131. Tsimberidou AM, Wierda WG, Plunkett W, et al. Phase I-II Study of Oxaliplatin, Fludarabine, Cytarabine, and Rituximab Combination Therapy in Patients With Richter's Syndrome or Fludarabine-Refractory Chronic Lymphocytic Leukemia. J Clin Oncol. 2008;26(2):196-203. [PubMed 18182662]
  132. US Department of Health and Human Services; Centers for Disease Control and Prevention; National Institute for Occupational Safety and Health. NIOSH list of antineoplastic and other hazardous drugs in healthcare settings 2016. http://www.cdc.gov/niosh/topics/antineoplastic/pdf/hazardous-drugs-list_2016-161.pdf. Updated September 2016. Accessed September 28, 2020.
  133. van Imhoff GW, van der Holt B, MacKenzie MA, et al. Short Intensive Sequential Therapy Followed by Autologous Stem Cell Transplantation in Adult Burkitt, Burkitt-Like and Lymphoblastic Lymphoma. Leukemia. 2005;19(6):945-952. [PubMed 15800666]
  134. Velasquez WS, Cabanillas F, Salvador P, et al. Effective Salvage Therapy for Lymphoma With Cisplatin in Combination With High-Dose Ara-C and Dexamethasone (DHAP). Blood. 1988;71(1):117-122. [PubMed 3334893 ]
  135. Velasquez WF, McLaughlin P, Tucker S, et al. ESHAP - An Effective Chemotherapy Regimen in Refractory and Relapsing Lymphoma: A 4-Year Follow-up Study. J Clin Oncol. 1994;12(6):1169-1176. [PubMed 8201379 ]
  136. Visco C, Chiappella A, Nassi L, et al. Rituximab, bendamustine, and low-dose cytarabine as induction therapy in elderly patients with mantle cell lymphoma: a multicentre, phase 2 trial from Fondazione Italiana Linfomi. Lancet Haematol. 2017;4(1):e15-e23. doi:10.1016/S2352-3026(16)30185-5 [PubMed 27927586]
  137. Visco C, Finotto S, Zambello R, et al. Combination of rituximab, bendamustine, and cytarabine for patients with mantle-cell non-Hodgkin lymphoma ineligible for intensive regimens or autologous transplantation. J Clin Oncol. 2013;31(11):1442-1449. doi:10.1200/JCO.2012.45.9842 [PubMed 23401442]
  138. Vogler WR, Velez-Garcia E, Weiner RS, et al. A phase III trial comparing idarubicin and daunorubicin in combination with cytarabine in acute myelogenous leukemia: a Southeastern Cancer Study Group Study. J Clin Oncol. 1992;10(7):1103-1111. [PubMed 1607916]
  139. Vrooman LM, Blonquist TM, Harris MH, et al. Refining risk classification in childhood B acute lymphoblastic leukemia: results of DFCI ALL Consortium Protocol 05-001. Blood Adv. 2018;2(12):1449-1458. doi:10.1182/bloodadvances.2018016584 [PubMed 29941458]
  140. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137‐2145. doi:10.1182/blood.2020004856 [PubMed 32219442]
  141. Weiss MA, Aliff TB, Tallman MS, et al. A Single, High Dose of Idarubicin Combined With Cytarabine as Induction Therapy for Adult Patients With Recurrent or Refractory Acute Lymphoblastic Leukemia. Cancer. 2002;95(3):581-587. [PubMed 12209751]
  142. Wenzell CM, Gallagher EM, Earl M, et al. Outcomes in obese and overweight acute myeloid leukemia patients receiving chemotherapy dosed according to actual body weight. Am J Hematol. 2013;88(10):906-909. doi:10.1002/ajh.23530 [PubMed 23828018]
  143. Wierzbowska A, Robak T, Pluta A, et al. Cladribine Combined With High Doses of Arabinoside Cytosine, Mitoxantrone, and G-CSF (CLAG-M) is a Highly Effective Salvage Regimen in Patients With Refractory and Relapsed Acute Myeloid Leukemia of the Poor Risk: A Final Report of the Polish Adult Leukemia Group. Eur J Haematol. 2008;80(2):115-126. [PubMed 18076637]
  144. Wiernik PH, Banks P, Case Jr DC, et al. Cytarabine Plus Idarubicin or Daunorubicin as Induction and Consolidation Therapy for Previously Untreated Adult Patients With Acute Myeloid Leukemia. Blood. 1992;79(2):313-319. [PubMed 1730080]
  145. Willemze R, Suciu S, Meloni G, et al. High-dose cytarabine in induction treatment improves the outcome of adult patients younger than age 46 years with acute myeloid leukemia: results of the EORTC-GIMEMA AML-12 trial. J Clin Oncol. 2014;32(3):219-228. doi:10.1200/JCO.2013.51.8571 [PubMed 24297940]
  146. Winick NJ, Smith SD, Shuster J, et al. Treatment of CNS relapse in children with acute lymphoblastic leukemia: A Pediatric Oncology Group study. J Clin Oncol. 1993;11(2):271-278. [PubMed 8426204]
  147. Wittbrodt ET, Aviles VM. Pancreatitis and Fatal ARDS Following High-Dose Cytarabine Induction for Acute Myelogenous Leukemia. J Oncol Pharm Pract. 1997;3(2):110-115.
  148. Woods T, Vidarsson B, Mosher D, Stein JH. Transient effusive-constrictive pericarditis due to chemotherapy. Clin Cardiol. 1999;22(4):316-318. doi:10.1002/clc.4960220414 [PubMed 10198745]
  149. Woods WG, Ruymann FB, Lampkin BC, et al. The Role of Timing of High-Dose Cytosine Arabinoside Intensification and of Maintenance Therapy in the Treatment of Children With Acute Nonlymphocytic Leukemia. Cancer. 1990;66(6):1106-1113. [PubMed 2205352]
  150. Wrzesień-Kuś A, Robak T, Lech-Marańda E, et al. A Multicenter, Open, Non-Comparative Phase II Study of the Combination of Cladribine (2-Chlorodeoxyadenosine), Cytarabine, and G-CSF as Induction Therapy in Refractory Acute Myeloid Leukemia – A Report of the Polish Adult Leukemia Group (PALG). Eur J Haematol. 2003;71(3):155-162. [PubMed 12930315]
  151. Yang QJ, Zheng J, Dang FT, Wan YM, Yang J. Acute pancreatitis induced by combination chemotherapy used for the treatment of acute myeloid leukemia: a case report. Medicine (Baltimore). 2020;99(35):e21848. doi:10.1097/MD.0000000000021848 [PubMed 32871908]
Topic 9314 Version 321.0