Your activity: 4 p.v.

Cyclophosphamide: Drug information

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

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
Brand Names: Canada
  • Procytox
Pharmacologic Category
  • Antineoplastic Agent, Alkylating Agent;
  • Antineoplastic Agent, Alkylating Agent (Nitrogen Mustard);
  • Antirheumatic, Miscellaneous;
  • Immunosuppressant Agent
Dosing: Adult

Cyclophosphamide is associated with a moderate to high emetic potential (depending on dose, regimen, or administration route); antiemetics are recommended to prevent nausea and vomiting (ASCO [Hesketh 2020]; MASCC [Roila 2016]). Increased hydration and frequent voiding are recommended to help prevent cystitis; some protocols utilize mesna to prevent bladder toxicity/hemorrhagic cystitis (refer to protocol). May require dose reduction or treatment interruption based on clinical response and hematologic toxicity (eg, ANC <1,500 mm3). Antimicrobial prophylaxis may be considered in appropriate patients. Consider growth factors (primary or secondary prophylaxis) in patients at increased risk for complications due to neutropenia. Patient should be under the care of a clinician experienced with using cyclophosphamide.

Acute lymphoblastic leukemia

Acute lymphoblastic leukemia (off-label dosing): Multiple-agent regimens:

CALGB 10403 regimen: Patients <40 years of age: IV: Remission consolidation phase (course 2): 1,000 mg/m2 on days 1 and 29; Delayed intensification phase: 1,000 mg/m2 on day 29; phases are part of combination chemotherapy; refer to protocol for details (Stock 2019).

Hyper-CVAD regimen: IV: 300 mg/m2 over 3 hours every 12 hours for 6 doses on days 1, 2, and 3 (in combination with mesna, vincristine, doxorubicin, and dexamethasone) during odd-numbered cycles (cycles 1, 3, 5, 7) of an 8-cycle phase (Kantarjian 2000); plus a BCR-ABL tyrosine kinase inhibitor (for Philadelphia chromosome-positive disease) (Ravandi 2010; Thomas 2004).

CALGB8811 regimen:

Adults <60 years of age: IV: Induction phase: 1,200 mg/m2 on day 1 of a 4-week cycle; Early intensification phase: 1,000 mg/m2 on day 1 of a 4-week cycle (repeat once); Late intensification phase: 1,000 mg/m2 on day 29 of an 8-week cycle (Larson 1995).

Adults ≥60 years of age: IV: Induction phase: 800 mg/m2 on day 1 of a 4-week cycle; Early intensification phase: 1,000 mg/m2 on day 1 of a 4-week cycle (repeat once); Late intensification phase: 1,000 mg/m2 on day 29 of an 8-week cycle (Larson 1995).

GRAALL 2003 regimen: Patients <60 years of age: IV: Induction phase: 750 mg/m2 on day 1; then 750 mg/m2 on day 15 (in good early responders) or 500 mg/m2 every 12 hours for 4 doses on days 15 and 16 (in poor early responders); Consolidation phase: 500 mg/m2 on days 29 and 30 of consolidation blocks 3, 6, and 9; Late intensification phase: 500 mg/m2 every 12 hours on day 15; phases are part of combination chemotherapy; refer to protocol for further information (Huguet 2009).

GRAALL 2005 regimen: Patients <60 years of age: IV: Induction phase: 750 mg/m2 on day 1; then 750 mg/m2 on day 15 or 300 mg/m2 every 12 hours for 6 doses on days 15 to 17; First, second, and third consolidation phases (block 3, block 6, and block 9, respectively): 500 mg/m2 on days 29 and 30; Late intensification phase (if complete response after first course): 750 mg/m2 on day 1; then 750 mg/m2 on day 15 or 300 mg/m2 every 12 hours for 6 doses on days 15 to 17; phases are part of combination chemotherapy; refer to protocol for further information (Huguet 2018).

MRC UKALL XII/ECOG E2993 regimen: Patients <60 years of age: IV: Induction (phase 2): 650 mg/m2 on days 1, 15, and 29; Consolidation phase (cycle 3): 650 mg/m2 on day 29; phases are part of combination chemotherapy; refer to protocol for further information (Rowe 2005).

Adult T-cell leukemia/lymphoma

Adult T-cell leukemia/lymphoma (off-label dosing): IV: 350 mg/m2 on day 1 every 28 days (as part of the VCAP-AMP-VECP multiagent chemotherapy regimen) for 6 cycles (Tsukasaki 2007).

Anti-glomerular basement membrane disease

Anti-glomerular basement membrane disease (anti-GBM or Goodpasture disease) (off-label use):

Note: For use in combination with glucocorticoids and plasmapheresis in patients with kidney involvement who do not require immediate dialysis, or in patients with pulmonary hemorrhage regardless of kidney involvement (Kaplan 2022; KDIGO 2012).

Oral: 2 mg/kg once daily (Huart 2016; Kaplan 2022); maximum dose has not been established; some experts do not exceed 200 mg/day (Kaplan 2022). On plasmapheresis days, administer cyclophosphamide after plasmapheresis. Discontinue therapy after 2 to 3 months; some experts continue therapy for up to 6 months if anti-GBM antibodies are not substantially reduced within 3 months (Huart 2016; Kaplan 2022; KDIGO 2012).

Breast cancer

Breast cancer (off-label dosing):

AC regimen: IV: 600 mg/m2 on day 1 every 21 days (in combination with doxorubicin) for 4 cycles (Fisher 1990).

CEF regimen: Oral: 75 mg/m2/day days 1 to 14 every 28 days (in combination with epirubicin and fluorouracil) for 6 cycles (Levine 1998).

CMF regimen: Oral: 100 mg/m2/day days 1 to 14 every 28 days (in combination with methotrexate and fluorouracil) for 6 cycles (Levine 1998) or IV: 600 mg/m2 on day 1 every 21 days (in combination with methotrexate and fluorouracil) (Goldhirsch 1998).

Castleman disease, idiopathic, multicentric

Castleman disease, idiopathic, multicentric (off-label use; based on limited data): Note: Cyclophosphamide-containing combination chemotherapy regimens (± rituximab) may benefit certain patients with severe idiopathic multicentric Castleman disease (van Rhee 2018).

CHOP regimen: IV: 750 mg/m2 on day 1 every 3 weeks for 4 cycles (in combination with doxorubicin, vincristine, and prednisone ± rituximab) (Seo 2009).

TCP regimen: Oral: 300 mg/m2 once weekly on days 1, 8, 15, and 22 every 4 weeks (in combination with thalidomide and prednisone). TCP was administered for 2 years or until treatment failure; cyclophosphamide and prednisone were administered for 1 year, while thalidomide was administered for 2 years (Zhang 2019).

Chronic lymphocytic leukemia

Chronic lymphocytic leukemia (off-label dosing): IV: R-FC regimen: 250 mg/m2/day for 3 days every 28 days (in combination with rituximab and fludarabine) for 6 cycles (Robak 2010).

Dermatomyositis/Polymyositis, severe, life-threatening or refractory

Dermatomyositis/polymyositis, severe, life-threatening or refractory (adjunctive agent) (off-label use):

Note: For use as an adjunct to glucocorticoids and other immunosuppressive agents in patients with severe disease that is refractory to other preferred therapies, or as part of initial combination therapy in patients with impending respiratory failure (Dellaripa 2021; Targoff 2021).

IV: 500 to 750 mg/m2 every 4 weeks; maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021).

Oral: 1.5 to 2 mg/kg/day (Marie 2011); maximum dose has not been established; some experts do not exceed 200 mg/day (McCune 2021).

Duration of therapy: Continue therapy for up to 6 months, then transition to an alternative immunosuppressive agent to maintain remission (Dellaripa 2021).

Eosinophilic granulomatosis with polyangiitis

Eosinophilic granulomatosis with polyangiitis (Churg-Strauss) (off-label use):

Note: For use in combination with glucocorticoids to initiate disease remission in patients with severe, multiorgan disease or severe cardiovascular or CNS involvement (ACR/VF [Chung 2021]; EULAR [Mukhtyar 2009]; King 2021).

IV: 600 mg/m2 once every month (Cohen 2007; Gayraud 1997; Guillevin 1995); maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021).

Oral: 2 mg/kg once daily (maximum: 200 mg/day) (EULAR [Mukhtyar 2009]; Gayraud 1997; Guillevin 1991).

Duration of therapy: Continue therapy for 6 to 12 months, then transition to an alternative immunosuppressive agent to maintain remission (Cohen 2007; EULAR [Mukhtyar 2009]; Guillevin 1991; King 2021).

Ewing sarcoma

Ewing sarcoma (off-label use): VAC/IE regimen: VAC: IV: 1,200 mg/m2 (plus mesna) on day 1 of a 21-day treatment cycle (in combination with vincristine and doxorubicin [then dactinomycin when maximum doxorubicin dose reached]), alternates with IE (ifosfamide and etoposide) for a total of 17 cycles (Grier 2003).

Gestational trophoblastic neoplasia, high-risk

Gestational trophoblastic neoplasia, high-risk (off-label use): EMA/CO regimen: IV: 600 mg/m2 on day 8 of 2-week treatment cycle (in combination with etoposide, methotrexate, leucovorin, dactinomycin, and vincristine), continue for at least 2 treatment cycles after a normal hCG level (Escobar 2003; Lurain 2006).

Graft-vs-host disease, acute and chronic, prophylaxis

Graft-vs-host disease, acute and chronic, prophylaxis (off-label use): IV: 50 mg/kg on days +3 and +4 following allogeneic blood or marrow transplantation (in combination with mesna) (Kanakry 2014a; Kanakry 2014b; Luzkin 2010).

Granulomatosis with polyangiitis and microscopic polyangiitis, organ- or life-threatening

Granulomatosis with polyangiitis and microscopic polyangiitis, organ- or life-threatening (off-label use):

IV: 15 mg/kg (maximum: 1,200 mg) once every 2 weeks for 3 doses, followed by 15 mg/kg (maximum: 1,200 mg) once every 3 weeks for 3 to 6 months. Use in combination with glucocorticoids (de Groot 2009; Harper 2012).

Oral: 1.5 to 2 mg/kg/day (maximum: 200 mg/day) in combination with glucocorticoids until remission is induced (usually within 3 to 6 months) (de Groot 2009; Jayne 2003; Stone 2010).

Duration of therapy: Transition to an alternative immunosuppressive agent to maintain remission; consider alternative induction therapy if remission is not attained after 3 to 6 months of cyclophosphamide therapy (ACR/VF [Chung 2021]; EULAR [Mukhtyar 2009]; Falk 2021a).

Hematopoietic stem cell or marrow transplant

Hematopoietic stem cell or marrow transplant (off-label use):

Conditioning regimens:

Nonmyeloablative transplant (allogeneic): IV: 750 mg/m2/day for 3 days beginning 5 days prior to transplant (in combination with fludarabine) (Khouri 2008).

Myeloablative transplant: IV:

100 mg/kg (based on ideal body weight [IBW], unless actual weight <95% of IBW) as a single dose 2 days prior to transplant (in combination with total body irradiation and etoposide) (Thompson 2008).

50 mg/kg/day for 4 days beginning 5 days before transplant (with or without antithymocyte globulin [equine]) (Champlin 2007).

50 mg/kg/day for 4 days beginning 5 days prior to transplant (in combination with busulfan) (Cassileth 1993; Cassileth 1998).

60 mg/kg/day for 2 days (in combination with busulfan and total body irradiation) (Anderson 1996).

60 mg/kg/day for 2 days beginning 4 days prior to transplant (in combination with busulfan) (Tutschka 1987).

1,800 mg/m2/day for 4 days beginning 7 days prior to transplant (in combination with etoposide and carmustine) (Reece 1991).

Reduced-intensity conditioning regimens: IV:

30 mg/kg/day for 2 days beginning 4 days prior to transplant (in combination with thiotepa and fludarabine) (Corradini 2002).

50 mg/kg/day for 4 days beginning 5 days prior to transplant (in combination with antithymocyte globulin [equine]) (Storb 1997).

Hematopoietic stem cell mobilization: IV: 1,500 mg/m2/day to 2,000 mg/m2/day for 2 days on days 1 and 2 (in combination with mesna and G-CSF [filgrastim]) (Hamadani 2012).

Hodgkin lymphoma

Hodgkin lymphoma (off-label dosing):

BEACOPP regimen: IV: 650 mg/m2 on day 1 every 3 weeks (in combination with bleomycin, etoposide, doxorubicin, vincristine, procarbazine, and prednisone) for 8 cycles (Diehl 2003).

BEACOPP escalated regimen: IV: 1,200 mg/m2 on day 1 every 3 weeks (in combination with bleomycin, etoposide, doxorubicin, vincristine, procarbazine, and prednisone) for 8 cycles (Diehl 2003).

Interstitial pneumonia, nonspecific, refractory or rapidly progressive

Interstitial pneumonia, nonspecific, refractory or rapidly progressive (off-label use):

IV (preferred route): 600 mg/m2 once every month (Corte 2009); maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021). Use in combination with glucocorticoids (Corte 2009; Flaherty 2022; McCune 2021).

Oral: 1 to 2 mg/kg once daily (Kondoh 2005); maximum dose has not been established; some experts do not exceed 200 mg/day (McCune 2021). Use in combination with glucocorticoids (Kondoh 2005; McCune 2021).

Duration of therapy: Continue therapy for at least 3 to 6 months to determine efficacy; discontinue therapy or transition to an alternative immunosuppressive agent after 6 to 12 months (Corte 2009; Flaherty 2022; Kondoh 2005).

Lupus nephritis, focal or diffuse

Lupus nephritis, focal or diffuse (off-label use):

IV (shorter, low-dose regimen; preferred regimen): 500 mg once every 2 weeks for 6 doses, then transition to an alternative immunosuppressive agent (ACR [Hahn 2012]; EULAR/ERA-EDTA [Fanouriakis 2020]).

IV (longer, high-dose regimen): 500 to 1,000 mg/m2 once every month for 6 doses, then transition to an alternative immunosuppressive agent (ACR [Hahn 2012]; EULAR/ERA-EDTA [Fanouriakis 2020]). Maximum dose has not been established; some experts do not exceed 1,000 mg/dose (Falk 2021b).

Oral: Initial: 1 to 1.5 mg/kg once daily; some experts increase by 0.5 mg/kg/day every week up to 2 mg/kg once daily if needed based on response, and do not exceed 150 mg/day (Almaani 2017; Falk 2021b). Continue therapy for 2 to 4 months once the dose is stabilized, then transition to an alternative immunosuppressive agent (Almaani 2017).

Lymphodepleting therapy prior to chimeric antigen receptor T-cell immunotherapy

Lymphodepleting therapy prior to chimeric antigen receptor T-cell immunotherapy (off-label use):

Prior to axicabtagene ciloleucel: IV: 500 mg/m2/day for 3 days (in combination with fludarabine) beginning 5 days (on days −5, −4, and −3) prior to chimeric antigen receptor (CAR) T-cell infusion on day 0 (Locke 2019; Locke 2022; Neelapu 2017).

Prior to brexucabtagene autoleucel: IV: 500 mg/m2/day for 3 days (in combination with fludarabine; for relapsed/refractory mantle cell lymphoma) beginning 5 days (on days −5, −4, and −3) prior to CAR T-cell infusion on day 0 (Wang 2020) or 900 mg/m2 once beginning 2 days (on day −2) prior to CAR T-cell infusion on day 0 (in combination with fludarabine; for relapsed/refractory B-cell precursor acute lymphoblastic leukemia) (Shah 2021).

Prior to ciltacabtagene autoleucel: IV: 300 mg/m2/day for 3 days (in combination with fludarabine) followed 2 to 4 days later by CAR T-cell infusion (Berdeja 2021).

Prior to idecabtagene vicleucel: IV: 300 mg/m2/day for 3 days (in combination with fludarabine) beginning 5 days (on days -5, -4, and -3) prior to CAR T-cell infusion on day 0 (Munshi 2021).

Prior to lisocabtagene maraleucel: IV: 300 mg/m2/day for 3 days (in combination with fludarabine) followed 2 to 7 days later by CAR T-cell infusion (Abramson 2020).

Prior to tisagenlecleucel : IV: 250 mg/m2/day for 3 days (in combination with fludarabine; for relapsed/refractory diffuse large B-cell lymphoma) followed 2 to 11 days later by CAR T-cell infusion (Schuster 2019) or 500 mg/m2/day for 2 days (in combination with fludarabine; for relapsed/refractory B-cell acute lymphoblastic leukemia) followed 2 to 14 days later by CAR T-cell infusion (Maude 2018).

Membranous nephropathy

Membranous nephropathy (off-label use):

Note: For use in combination with glucocorticoids to initiate disease remission in patients at high or very high risk of disease progression, regardless of the presence of lupus (KDIGO [Floege 2019]). Some experts also use cyclophosphamide (in combination with glucocorticoids) as an alternative to other immunosuppressive therapies in patients at moderate risk of disease progression (De Vriese 2022).

Oral, cyclical (preferred): 2 mg/kg once daily during months 2, 4, and 6 (alternating with glucocorticoid therapy during months 1, 3, and 5) (Fernández-Juárez 2021; Jha 2007); maximum dose has not been established; some experts do not exceed 200 mg/day (De Vriese 2022).

Oral, continuous: 1.5 to 2 mg/kg once daily for 12 months (du Buf-Vereijken 2004); maximum dose has not been established; some experts do not exceed 200 mg/day, and also limit the total cumulative dose to 25 g to reduce the risk of malignancy (De Vriese 2022).

Merkel cell carcinoma, advanced or recurrent

Merkel cell carcinoma, advanced or recurrent (off-label use; based on limited data): IV: CAV regimen: 1,000 mg/m2 on day 1 every 21 days (in combination with doxorubicin and vincristine) (Fenig 1997).

Minimal change disease

Minimal change disease (off-label use):

Note: For use in patients with frequently relapsing or glucocorticoid-dependent disease, regardless of the presence of lupus nephritis. Initiate cyclophosphamide after remission is induced by glucocorticoids (KDIGO [Rovin 2019]).

Oral: 2 to 2.5 mg/kg once daily (Mak 1996; Ponticelli 1993); some experts continue therapy for 12 weeks, and do not exceed 200 mg/day (Meyrier 2022; Waldman 2007). Gradually taper and discontinue glucocorticoids during cyclophosphamide therapy (Mak 1996; Ponticelli 1993). Note: Avoid prolonged (>12 weeks) or repeat courses of cyclophosphamide to reduce the risk of adverse effects (Meyrier 2022).

Mixed cryoglobulinemia syndrome, moderate to severe

Mixed cryoglobulinemia syndrome, moderate to severe (alternative agent) (off-label use):

Note: For use as an alternative to rituximab, in combination with glucocorticoids, to induce disease remission (Fervenza 2021).

IV: 750 mg/m2 once every month for 3 months (Cakir 2005; Fervenza 2021); maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021).

Oral: 2 mg/kg once daily for 2 to 4 months (Frankel 1992; KDIGO 2012); maximum dose has not been established; some experts do not exceed 200 mg/day (McCune 2021).

Multiple myeloma

Multiple myeloma (off-label dosing):

CyBorD regimen: Oral: 300 mg/m2 on days 1, 8, 15, and 22 every 4 weeks (in combination with bortezomib and dexamethasone) for 4 cycles; may continue beyond 4 cycles (Khan 2012) or 500 mg/m2 on days 1, 8, and 15 every 3 weeks (in combination with bortezomib and dexamethasone) for 8 cycles (Kumar 2012a).

VDT-PACE regimen: IV: 400 mg/m2/day administered as a continuous infusion on days 1 to 4 of each cycle; repeat every 4 to 6 weeks (in combination with bortezomib, dexamethasone, thalidomide, cisplatin, doxorubicin, and etoposide) (Lee 2003; Pineda-Roman 2008).

Non-Hodgkin lymphomas

Non-Hodgkin lymphomas (off-label dosing):

Burkitt lymphoma:

CALGB 10002 regimen (cycles 1, 3, 5, and 7): IV: 200 mg/m2/day on days 1 to 5 every 3 weeks (in combination with vincristine, prednisone, ifosfamide, dexamethasone, methotrexate, leucovorin, cytarabine, etoposide, rituximab, doxorubicin, intrathecal therapy, and filgrastim); refer to protocol for further information (Rizzieri 2014).

CODOX-M/IVAC: Cycles 1 and 3 (CODOX-M): IV: 800 mg/m2 on day 1, followed by 200 mg/m2/day on days 2 to 5 (Magrath 1996) or 800 mg/m2/day on days 1 and 2 (Lacasce 2004), in combination with vincristine, doxorubicin, and methotrexate; CODOX-M alternates with IVAC (etoposide, ifosfamide, mesna, and cytarabine) for a total of 4 cycles.

R-Hyper-CVAD: IV: 300 mg/m2 every 12 hours on days 1 to 3 (total of 6 doses) of courses 1, 3, 5, and 7 (in combination with rituximab, mesna, vincristine, doxorubicin, and dexamethasone) and alternates with even courses 2, 4, 6, and 8 (rituximab, methotrexate and cytarabine) (Thomas 2006).

Diffuse large B-cell lymphoma:

CHOP/R-CHOP regimen: IV: 750 mg/m2 on day 1 every 3 weeks (in combination with rituximab, doxorubicin, vincristine, and prednisone) for 8 cycles (Coiffier 2002).

Dose-adjusted EPOCH/EPOCH-R regimen: IV: 750 mg/m2 on day 5 every 3 weeks (in combination with rituximab, etoposide, prednisone, vincristine, and doxorubicin) for 6 to 8 cycles (Garcia-Suarez 2007). Refer to protocol for dosage adjustments.

R-CEOP regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle for 3 to 6 cycles (in combination with rituximab, vincristine, etoposide, and prednisone) (Moccia 2009).

Follicular lymphoma:

CVP regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle (in combination with vincristine and prednisone [± rituximab or obinutuzumab]) for 8 cycles (Marcus 2005; Marcus 2017).

R-CHOP regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle for 6 to 8 cycles (in combination with rituximab, vincristine, doxorubicin, and prednisone) (Hiddemann 2005).

Peripheral T-cell lymphoma:

BV-CHP (or A-CHP) regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle (in combination with brentuximab vedotin, doxorubicin, and prednisone) for 6 to 8 cycles (Horwitz 2019).

CHOEP regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle (in combination with vincristine, doxorubicin, etoposide, and prednisone) for 6 to 8 cycles (Pfreundschuh 2004; Schmitz 2010).

CHOP regimen: IV: 750 mg/m2 on day 1 of a 21-day treatment cycle (in combination with vincristine, doxorubicin, and prednisone) for 6 to 8 cycles (Schmitz 2010).

Primary mediastinal B-cell lymphoma: DA-EPOCH-R regimen: IV: 750 mg/m2 as a 2-hour infusion on day 5, dose-adjusted for subsequent cycles based on neutrophil and platelet counts during nadir (in combination with etoposide, prednisone, vincristine, doxorubicin, rituximab, and filgrastim); repeat cycle every 3 weeks for a total of 6 to 8 cycles (Dunleavy 2013). Refer to protocol for dosage adjustments.

Other non- Hodgkin lymphoma regimens:

CEPP(B) regimen: IV: Initial: 600 mg/m2 on days 1 and 8; may increase dose with subsequent cycles (refer to protocol for details) every 28 days (in combination with etoposide, procarbazine, prednisone, ± bleomycin) (Chao 1990).

PEP-C regimen: Oral: 50 mg (flat dose) once daily after lunch (length of induction cycle depends on blood counts; frequency may vary based on tolerance in maintenance cycle [refer to protocol for details]; in combination with prednisone, etoposide, and procarbazine) (Coleman 2008).

Osteosarcoma, relapsed/refractory

Osteosarcoma, relapsed/refractory (off-label use): IV: 500 mg/m2/day on days 1 to 5 every 21 to 28 days (in combination with mesna, etoposide, and growth factor support) (Rodriguez-Galindo 2002) or 4,000 mg/m2 as a 3-hour infusion on day 1 (in combination with mesna and etoposide) every 21 to 28 days for 2 cycles; refer to protocol for further information (Berger 2009).

Ovarian germ cell tumors, malignant

Ovarian germ cell tumors, malignant (off-label use): IV: 150 mg/m2 on days 1 to 5 every 28 days (in combination with dactinomycin and vincristine) for at least 10 cycles (Slayton 1985).

Pheochromocytoma, malignant

Pheochromocytoma, malignant (off-label use): IV: 750 mg/m2 on day 1 every 3 or 4 weeks (in combination with dacarbazine and vincristine) (Huang 2008).

Polyarteritis nodosa, moderate to severe

Polyarteritis nodosa, moderate to severe (off-label use):

Note: Some experts prefer the regimen identified as “preferred” below based on clinical experience and data in patients with antineutrophil cytoplasmic antibody-associated vasculitis; this regimen has not been studied in polyarteritis nodosa (Merkel 2021a).

IV (preferred regimen): 15 mg/kg (maximum: 1,200 mg) once every 2 weeks for 3 doses, followed by 15 mg/kg (maximum: 1,200 mg) once every 3 weeks for 3 to 6 months (in combination with glucocorticoids) (de Groot 2009; Harper 2012; Merkel 2021a).

IV: 600 mg/m2 once every 2 weeks for 3 doses, then once every 4 weeks thereafter (in combination with glucocorticoids) (Gayraud 1997; Guillevin 1995). Maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021).

Oral: 2 mg/kg once daily in combination with glucocorticoids (Gayraud 1997; Guillevin 1991). Maximum dose has not been established; some experts do not exceed 200 mg/day (McCune 2021).

Duration of therapy : Continue therapy for ≥4 months and until disease remission is achieved (maximum duration: 6 months); some experts limit duration to 4 months if remission is achieved by then. Once remission is achieved, transition to an alternative immunosuppressive agent to maintain remission (EULAR [Mukhtyar 2009]; Guillevin 1991; Guillevin 1995; Merkel 2021a).

Primary CNS lymphoma

Primary CNS lymphoma (off-label use): TBC conditioning regimen: IV: 60 mg/kg/day for 2 days beginning 3 days prior to transplant (in combination with thiotepa and busulfan), followed by autologous stem cell transplant (DeFilipp 2017; Soussain 2001; Soussain 2008).

Pure red cell aplasia

Pure red cell aplasia (off-label use):

Note: For use in patients with acquired pure red cell aplasia (PRCA) that is not self-limiting and is refractory to other preferred agents, or as initial therapy in patients with PRCA due to anti-erythropoiesis-stimulating agent antibodies (Berns 2021; Means 2022).

Oral: 50 to 100 mg once daily (in combination with glucocorticoids); initial improvement may be observed after as early as 8 weeks, and complete response after a median of 3 to 6 months (Bennett 2005; Verhelst 2004; Yamada 1997).

Duration of therapy:

Acquired PRCA: Continue therapy for up to 6 months, then transition to an alternative immunosuppressive agent to maintain remission (Sawada 2008).

PRCA due to anti-erythropoiesis-stimulating agent antibodies: Continue treatment until antibody levels become undetectable; switch to alternative therapy if there is no response within 3 to 4 months of treatment initiation (Berns 2021).

Rhabdomyosarcoma

Rhabdomyosarcoma (off-label use): Adults <50 years: VAC regimen:

Low risk: IV: 1,200 mg/m2 every 21 days (in combination with mesna, vincristine, and dactinomycin) for 4 cycles (Walterhouse 2014).

Intermediate risk: IV: 2,200 mg/m2 every 21 days (in combination with mesna, vincristine, and dactinomycin) for 14 cycles (Arndt 2009).

Small cell lung cancer, refractory

Small cell lung cancer, refractory (off-label use): IV: 1,000 mg/m2 (maximum: 2,000 mg) on day 1 every 3 weeks (in combination with doxorubicin and vincristine) until disease progression or unacceptable toxicity (von Pawel 1999).

Systemic light chain amyloidosis

Systemic light chain amyloidosis (off-label use):

CRd regimens: IV or Oral: 300 mg/m2 orally on days 1, 8, and 15 of a 28-day cycle (in combination with lenalidomide and dexamethasone); cyclophosphamide was administered for a maximum of 12 cycles; refer to protocol for further information (Kumar 2012b) or 300 mg/m2 IV on days 1 and 8 every 28 days (in combination with lenalidomide and dexamethasone) for 6 cycles, followed by 300 mg/m2 IV on day 1 only every 28 days (in combination with lenalidomide and dexamethasone) for 6 additional cycles (Cibeira 2015).

CyBorD regimen: Oral: 300 mg/m2 once weekly (in combination with bortezomib and dexamethasone) (Mikhael 2012; Palladini 2015) or 350 mg/m2 on days 1, 8, and 15 (in combination with bortezomib and dexamethasone) for up to 8 cycles (Venner 2012).

Systemic sclerosis–related interstitial lung disease

Systemic sclerosis–related interstitial lung disease (alternative agent) (off-label use):

Note: For use in patients with features suggesting high risk for progression (eg, declining pulmonary function) or with symptomatic interstitial lung disease. Generally administered in combination with low-dose glucocorticoids (Varga 2021).

IV (preferred route): 600 mg/m2 once every 4 weeks (Hoyles 2006); maximum dose has not been established; some experts do not exceed 1,200 mg/dose (McCune 2021).

Oral: Initial: 1 mg/kg/day (rounded to the nearest 25 mg); may increase daily dose based on response and tolerability by 25 mg once monthly up to 2 mg/kg/day (Tashkin 2006); maximum dose has not been established; some experts do not exceed 200 mg/day (McCune 2021), or 50 to 100 mg daily initially (50 mg daily for patients weighing <81 kg and 100 mg daily for patients weighing ≥81 kg), followed by increases in 25 to 50 mg increments each month up to 2 mg/kg/day (maximum: 200 mg/day) (Tashkin 2016).

Duration of therapy: Limit duration of IV therapy to 6 months or oral therapy to 12 months, then transition to an alternative immunosuppressive agent (Tashkin 2006; Tashkin 2016; Varga 2021; Hoyles 2006).

Thymomas, advanced or metastatic

Thymomas, advanced or metastatic (off-label use):

ADOC regimen: IV: 700 mg/m2 on day 4 every 3 weeks (in combination with doxorubicin, cisplatin, and vincristine) (Fornasiero 1991).

CAP regimen: IV: 500 mg/m2 on day 1 every 3 weeks for up to 8 cycles (in combination with cisplatin and doxorubicin) (Loehrer 1994).

Waldenström macroglobulinemia

Waldenström macroglobulinemia (off-label use):

DRC regimen: Oral: 100 mg/m2 twice daily on days 1 to 5 every 21 days (in combination with dexamethasone and rituximab) for 6 cycles (Dimopoulos 2007).

FCR regimen: IV: 250 mg/m2 once daily on days 2 to 4 every 28 days (in combination with fludarabine and rituximab) for up to 6 cycles (Tedeschi 2012).

Warm autoimmune hemolytic anemia, relapsed or refractory

Warm autoimmune hemolytic anemia, relapsed or refractory (alternative agent) (off-label use):

Low-dose therapy:

Oral: 1 to 2 mg/kg once daily (maximum dose is not established) or 50 to 150 mg once daily, with or without concomitant glucocorticoids; response typically achieved within 2 to 4 weeks (Go 2017; Salama 2015).

High-dose therapy:

Note: Generally reserved for patients with refractory disease or severe anemia due to greater potential for toxicity (Salama 2015).

IV: 1,000 mg once every 4 weeks for 4 doses (Go 2017; Thabet 2014).

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

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Note: Specific recommendations for kidney dose adjustment have not been established. A dose-effect relationship is difficult to determine due to interpatient variability in pharmacokinetics and pharmacodynamics, and the complex metabolism and precise mechanism of action are not completely understood (Haubitz 2002).

The following general and indication-specific dosage adjustments have been recommended. Use with caution, especially with more severe impairment; consider indication, goals of therapy, and risks versus benefits when selecting a dose, and monitor for signs and symptoms of toxicity; also refer to institution-specific protocols (when available):

General dosage adjustment recommendations: IV, Oral:

Altered kidney function:

CrCl ≥30 mL/minute: No dosage adjustment necessary.

CrCl 10 to 29 mL/minute: Administer 75% (Krens 2019) or 100% (Aronoff 2007; Kintzel 1995) of normal dose.

CrCl <10 mL/minute: Administer 50% (Krens 2019), 75% (Aronoff 2007), or 100% (Kintzel 1995) of normal dose.

Hemodialysis, intermittent (thrice weekly): Moderately dialyzable (20% to 50% removal based on limited data with low-flux dialyzers [Haubitz 2002; Nemecek 2019; Wang 1981]).

Administer 50% (Krens 2019) or 75% (Janus 2010; Pedrazzoli 2017) of the normal dose. On dialysis days, administer after hemodialysis, allowing at least 12 hours before the next hemodialysis session (Haubitz 2002).

Peritoneal dialysis: Administer 75% of the normal dose (Aronoff 2007). If possible, allow at least 12 hours before next peritoneal dialysis exchange (expert opinion).

CRRT: Administer 100% of the normal dose (Aronoff 2007).

Indication-specific dosage adjustments:

Granulomatosis with polyangiitis or microscopic polyangiitis:

Cyclophosphamide Oral Initial Dosage Adjustment for Granulomatosis With Polyangiitis or Microscopic Polyangiitisa

CrCl

Cyclophosphamide oral dose (mg/kg/day)b

a Regan 2001; Merkel 2021b.

b Maximum dose: 200 mg/dose (de Groot 2009).

≥100 mL/minute

2 mg/kg/day

50 to <100 mL/minute

1.5 mg/kg/day

25 to <50 mL/minute

1.2 mg/kg/day

15 to <25 mL/minute

1 mg/kg/day

<15 mL/minute or on dialysis

0.8 mg/kg/day

Cyclophosphamide IV Dosage Adjustment for Granulomatosis With Polyangiitis or Microscopic Polyangiitisa

Age

CrCl >30 mL/minuteb,d,e

CrCl <30 mL/minuteb,c,d,e

a de Groot 2009.

b The original protocol reduced doses when SCr was above 3.4 mg/dL (300 mcmol/L), but in clinical practice this has often been translated as a CrCl <30 mL/minute (expert opinion).

c Some experts use a reduced dose of 7.5 mg/kg in all patients with CrCl <30 mL/minute regardless of age (Merkel 2021b).

d Maximum dose: 1,200 mg/dose (de Groot 2009).

e Doses are administered every 2 weeks for 3 doses, followed by maintenance pulses every 3 weeks for 3 months after remission achieved.

<60 years

15 mg/kg

12.5 mg/kg

60 to 70 years

12.5 mg/kg

10 mg/kg

>70 years

10 mg/kg

7.5 mg/kg

Hematopoietic stem cell transplantation: There are no specific dosage adjustments recommended; a reduced dose may be considered in moderate to severe impairment (Bodge 2014). Refer to "General Dosage Adjustment Recommendations" for patients with kidney impairment.

Lupus nephritis:

IV: Shorter, low-dose regimen (500 mg IV once every 2 weeks for 6 doses): No dosage adjustment necessary (ACCESS Trial 2014; Euro-Lupus Nephritis Trial [Houssiau 2002]).

IV: Longer, high-dose regimen (500 to 1,000 mg/m2 IV pulses):

CrCl >30 mL/minute: No dosage adjustment necessary.

CrCl <30 mL/minute: Reduce initial dose to 500 mg/m2 (Austin 1986; Gourley 1996).

Oral regimen: Refer to “General Dosage Adjustment Recommendations” for patients with kidney impairment.

Multiple myeloma: Oral:

Mild to severe impairment: No dosage adjustment necessary (IMWG [Dimopoulos 2016]).

Hemodialysis: No dosage adjustment necessary (IMWG [Dimopoulos 2016]).

Dosing: Hepatic Impairment: Adult

The conversion between cyclophosphamide to the active metabolite may be reduced in patients with severe hepatic impairment, potentially reducing efficacy. Some dosage forms may contain ethanol; consider alcohol content of the product when administering to patients with hepatic impairment.

There are no dosage adjustments provided in the manufacturer's labeling.

The following adjustments have been recommended:

Floyd 2006:

Serum bilirubin 3.1 to 5 mg/dL or transaminases >3 times ULN: Administer 75% of dose.

Serum bilirubin >5 mg/dL: Avoid use.

Krens 2019:

Mild or moderate impairment: Dosage adjustment is not likely needed.

Severe impairment: Use is not recommended due to risk of reduced efficacy.

Dosing: Pediatric

(For additional information see "Cyclophosphamide: Pediatric drug information")

For oncology uses, all dosing presented based on clinically recognized trials; consult specific regimens concerning dosing in combination regimens. Doses IV ≥1,000 mg/m2 are associated with a high emetic potential and IV doses <1,000 mg/m2 and all oral doses are associated with a moderate emetic risk; antiemetics are recommended to prevent nausea and vomiting (Dupuis 2011; Dupuis 2013; Paw Cho Sing 2019). In pediatric patients, dosing may be based on either BSA (mg/m2) or weight (mg/kg); use extra precaution to verify dosing parameters during calculations. To lessen potential toxicity with doses for oncologic uses, hydration, and follow-up mesna therapy are typically administered; refer to specific protocol.

Oncologic uses:

Acute lymphoblastic leukemia, high-risk

Acute lymphoblastic leukemia, high-risk: Limited data available (Steinherz 1993): NYII Protocol:

Induction: Children and Adolescents: IV: 1,200 mg/m2 on Day 2 (in combination with steroid, daunorubicin, vincristine, and asparaginase).

Maintenance I: Children and Adolescents: IV: 600 mg/m2on Day 4 (in combination with steroid, mercaptopurine, vincristine, daunorubicin, methotrexate, cytarabine, and thioguanine).

Acute lymphoblastic leukemia, relapsed or refractory

Acute lymphoblastic leukemia, relapsed or refractory: Limited data available:

Hijiya 2011: Children and Adolescents: IV: 440 mg/m2 over 30 to 60 minutes in combination with clofarabine and etoposide; number of doses dependent on phase of protocol: Induction: Days 1 to 5, and Consolidation: Days 1 to 4.

Parker 2010: UK-ALL R3 protocol: Children and Adolescents:

Phase 2 Consolidation (weeks 5 to 8): IV: 440 mg/m2 on Days 15 to 19 in combination with steroid, vincristine, methotrexate, PEG-asparaginase, and etoposide.

Phase 5, before Continuation (weeks 14 to 29): IV: 300 mg/m2 on Days 42, 49, 99, and 106 in combination with steroid, mercaptopurine, vincristine, methotrexate, etoposide, and cytarabine.

CNS tumors, malignant

CNS tumors, malignant (medulloblastoma, PNET, ependymoma, brainstem glioma): Limited data available (Chi 2004; Mason 1998): Head Start II Protocol: Infants and Children <10 years: IV: 65 mg/kg on Days 2 and 3 with mesna every 21 days for 5 cycles (in combination with cisplatin, vincristine, etoposide, and high-dose methotrexate; then followed by an auto-transplantation).

Desmoplastic small round cell tumor

Desmoplastic small round cell tumor (DSRCT): Limited data available (Kushner 1996): High-dose cyclophosphamide: HD-CAV/IE regimen: P6 Protocol:

Children <10 years: IV: 70 mg/kg over 6 hours with mesna and hydration on Days 1 and 2 of a 21-day treatment cycle in combination with vincristine and doxorubicin for courses 1, 2, 4, and 6, alternate with ifosfamide and etoposide for courses 4, 5, and 7.

Children 10 years and Adolescents: IV: 2,100 mg/m2over 6 hours with mesna and hydration on Day 1 and 2 of a 21-day treatment cycle in combination with vincristine and doxorubicin for courses 1, 2, 4, and 6, alternate with ifosfamide and etoposide for courses 4, 5, and 7.

Ewing sarcoma

Ewing sarcoma:

VAC/IE regimen: Limited data available (Grier 2003): Children and Adolescents: IV: 1,200 mg/m2 with mesna on Day 1 of a 21-day treatment cycle in combination with vincristine and doxorubicin (then dactinomycin when maximum doxorubicin dose reached), alternate with IE (ifosfamide and etoposide) for a total of 17 cycles.

High-dose cyclophosphamide: HD- CAV/IE regimen: P6 Protocol: Limited data available (Kushner 1996):

Children <10 years: IV: 70 mg/kg over 6 hours with mesna and hydration on Days 1 and 2 of a 21-day treatment cycle in combination with vincristine and doxorubicin for courses 1, 2, 4, and 6, alternate with ifosfamide and etoposide for courses 4, 5, and 7.

Children ≥10 years and Adolescents: IV: 2,100 mg/m2 over 6 hours with mesna and hydration on Days 1 and 2 of a 21-day treatment cycle in combination with vincristine and doxorubicin for courses 1, 2, 4, and 6, alternate with ifosfamide and etoposide for courses 4, 5, and 7.

Hodgkin lymphoma

Hodgkin lymphoma: Limited data available:

BEACOPP (high-risk): Children and Adolescents: IV: 1,200 mg/m2 on Day 0 of a 21-day treatment cycle for 4 cycles in combination with bleomycin, etoposide, doxorubicin, vincristine, prednisone, and procarbazine (Kelly 2011).

ABVE-PC (intermediate-risk): Children and Adolescents: IV: 800 mg/m2 over 1 hour on the first day of a 21-day cycle for up to 4 cycles (depending upon response) in combination with doxorubicin, vincristine, etoposide, prednisone, and bleomycin (Dharmarajan 2015; Friedman 2014; Schwartz 2009).

HSCT conditioning; myeloablative transplant

HSCT conditioning; myeloablative transplant: Limited data available: Infants, Children, and Adolescents: IV: 50 mg/kg/day for 4 days beginning 5 days before transplant (Champlin 2007); other regimens have used 60 mg/kg/day for 2 days following busulfan (Locatelli 2005; Mårtensson 2013).

Dosing adjustment in obese patients (ASBMT [Bubalo 2014]):

Cy200 (cyclophosphamide total dose of 200 mg/kg): Use the lesser of ideal body weight (IBW) or actual body weight (ABW).

Cy120 (cyclophosphamide total dose of 120 mg/kg): Use IBW or ABW until >120% IBW; then use ABW25 for patients >120% IBW.

ABW25: Adjusted weight (kg) = Ideal body weight (kg) + 0.25 [actual body weight (kg) - ideal body weight (kg)].

Neuroblastoma during infancy

Neuroblastoma during infancy (unresectable): Limited data available (Rubie 2011): INES 99.1 regimen courses 1 and 2:

Infants:

<10 kg: IV: 3.5 mg/kg on Days 1 to 5 administered at 2-week intervals for 2 cycles initially, in combination with vincristine.

≥10 kg: IV: 5 mg/kg on Days 1 to 5 administered at 2-week intervals for 2 cycles initially, in combination with vincristine.

Neuroblastoma; high-risk, newly diagnosed

Neuroblastoma ; high-risk, newly diagnosed: Limited data available (Park 2011): Infants, Children, and Adolescents:

Induction cycles 1 and 2 (21-day cycles):

Patient weight ≤12 kg: IV: 13.3 mg/kg over 30 minutes on Days 1 to 5 (in combination with topotecan) for 2 cycles.

Patient weight >12 kg: IV: 400 mg/m2over 30 minutes on Days 1 to 5 (in combination with topotecan) for 2 cycles.

Induction cycles 4 and 6 (21-day cycles):

Patient weight ≤12 kg: IV: 70 mg/kg over 6 hours with mesna on Days 1 and 2 (in combination with doxorubicin and vincristine).

Patient weight >12 kg: IV: 2,100 mg/m2 over 6 hours with mesna on Days 1 and 2 (in combination with doxorubicin and vincristine).

Neuroblastoma, relapsed or refractory

Neuroblastoma, relapsed or refractory: Limited data available (Kushner 2010; Kushner 2011): HD-CCV or HD-CTV regimen: Infants and Children ≤10 years: IV: 70 mg/kg on Days 1 and 2 (in combination with irinotecan and vincristine or in combination with topotecan and vincristine).

Non-Hodgkin lymphoma

Non-Hodgkin lymphoma: Limited data available:

COP regimen: Reduction: Infants ≥6 months, Children, and Adolescents: IV: 300 mg/m2over 15 minutes on Day 1 (in combination with vincristine and prednisone) (Cairo 2007; Goldman 2013; Goldman 2014).

COPADM 1 regimen: Induction 1: Infants ≥6 months, Children, and Adolescents: IV: 250 mg/m2/dose over 15 minutes every 12 hours on Days 2 to 4 (6 doses) (in combination with doxorubicin, vincristine, prednisone, methotrexate) (Cairo 2007; Goldman 2013; Goldman 2014; Patte 2007).

COPADM 2 regimen: Induction 2: Infants ≥6 months, Children, and Adolescents: IV: 500 mg/m2/dose over 15 minutes every 12 hours on Days 2 to 4 (6 doses) (in combination with doxorubicin, vincristine, prednisone, methotrexate, +/- Rituximab) (Cairo 2007; Goldman 2013; Goldman 2014).

COPADM regimen: Maintenance 1: High-risk patients: Infants ≥6 months, Children, and Adolescents: IV: 500 mg/m2 over 15 minutes on Days 2 and 3 (2 doses) (in combination with doxorubicin, vincristine, prednisone, methotrexate) (Cairo 2007; Goldman 2013; Goldman 2014).

COPA regimen: Maintenance course 3: High-risk patients: Infants ≥6 months, Children, and Adolescents: IV: 500 mg/m2/dose over 30 minutes on the first 2 days of the cycle (2 doses) (in combination with doxorubicin, vincristine, prednisone) (Cairo 2007; Goldman 2013; Goldman 2014).

Non-Hodgkin lymphoma, T-cell or anaplastic large cell lymphoma

Non-Hodgkin lymphoma, T-cell or anaplastic large cell lymphoma (ALCL): Limited data available (Reiter 1994; Seideman 2001): NHL-BFM90 protocol: Infants, Children, and Adolescents: IV: 200 mg/m2over 1 hour; number of doses, days of administration, and other chemotherapy combinations are dependent on protocol specific phase.

Palliative intent chemotherapy

Palliative intent chemotherapy (metronomic therapy): Limited data available: Infants, Children, and Adolescents: Oral: 2.5 mg/kg once daily, maximum dose: 100 mg/dose for 21 days, alternating with a 21-day cycle of etoposide in combination with continuous thalidomide, celecoxib, and fenofibrate therapy; in the trial, the youngest patient reported was 191 days old (around 6 months of age) (Robison 2014).

Post-transplant lymphoproliferative disease

Post-transplant lymphoproliferative disease (PTLD): Limited data available: Infants, Children, and Adolescents: IV: 600 mg/m2 on Day 1 every 21 days for 6 cycles in combination with prednisone (all 6 cycles) and rituximab (first 2 cycles only); the youngest patient included in the reported experience was 0.8 years of age (Gross 2012).

Retinoblastoma, extraocular

Retinoblastoma, extraocular: Limited data available (Chantada 2003): Protocol 94 regimen: Children: IV: 65 mg/kg over 1 hour on Day 1 with mesna of a 21-day treatment cycle (in combination with idarubicin and vincristine) alternating cycles with carboplatin and etoposide for a total of 8 cycles.

Rhabdomyosarcoma

Rhabdomyosarcoma:

Low risk: Limited data available (Walterhouse 2014): VAC regimen: Doses were administered with hydration and mesna every 21 days for 4 doses of a 22-week cycle in combination with vincristine and dactinomycin.

Infants and Children <3 years: IV: 40 mg/kg.

Children ≥3 years and Adolescents: 1,200 mg/m2.

Intermediate risk: Limited data available (Arndt 2009): VAC regimen: Doses were administered with hydration and mesna every 21 days for 14 cycles in combination with vincristine and dactinomycin.

Infants: IV: 36 mg/kg.

Children 1 to 3 years: IV: 73 mg/kg.

Children >3 years: 2,200 mg/m2.

Wilms tumor; anaplastic

Wilms tumor; anaplastic (stage II to IV): Limited data available (Green 1994): National Wilms’ Tumor Study (NWTS) protocol 4 regimen J: Children and Adolescents <16 years: IV: 10 mg/kg on Days 1 to 3 (3 doses) every 6 weeks (in combination with vincristine, dactinomycin, and doxorubicin).

Wilms tumor, relapsed or refractory

Wilms tumor, relapsed or refractory: Limited data available: National Wilms’ Tumor Study (NWTS) protocol-5 regimen:

Infants and Children weighing ≤30 kg: IV: 14.7 mg/kg with mesna; reported frequency and combination chemotherapy variable, refer to specific protocols. One regimen alternated the frequency based on week of therapy: Weeks 3, 9, 15, and 21: Doses administered once daily for 5 days (in combination with etoposide); Weeks 6, 12, 18, and 24 administered once daily for 3 days (in combination with doxorubicin and vincristine) (Green 2007); another protocol administered for 5 days, on Weeks 0 and 3 in combination with etoposide; alternate with carboplatin and etoposide on Weeks 6 and 9 and continued for 90 weeks (Malogolowkin 2008).

Children >30 kg and Adolescents <16 years at diagnosis: IV: 440 mg/m2 with mesna; reported frequency and combination chemotherapy variable, refer to specific protocols. One regimen alternated the frequency based on week of therapy: Weeks 3, 9, 15, and 21: Doses administered once daily for 5 days (in combination with etoposide); Weeks 6, 12, 18, and 24 administered once daily for 3 days (in combination with doxorubicin and vincristine) (Green 2007).

Non-Oncologic uses:

Aplastic anemia, severe; refractory

Aplastic anemia, severe; refractory: Limited data available: Children and Adolescents ≥2 years: High-dose therapy: IV: 45 to 50 mg/kg/day for 4 days has been used in several small trials; concurrent prophylactic antimicrobial therapy should be considered (Audino 2010; Brodsky 1996; Brodsky 2010; DeZern 2011; Jaime-Perez 2013).

Kawasaki Disease; refractory to multiple therapies

Kawasaki Disease; refractory to multiple therapies: Very limited data available: Infants and Young Children: IV: 2 mg/kg/dose once daily; some have suggested a gradual taper over 1.5 to 7 months; however, details of taper have not been described. Dosing based on a retrospective study of patients with refractory Kawasaki Disease that included two patients (ages 2.7 and 9 years) who had failed to respond to 3 doses of IVIG and required high-dose IV methylprednisolone; after initiation of cyclophosphamide, steroids were able to be successfully tapered and final ECHO showed resolution of aneurysm. While taper schedule was not described, both patients were discharged on a taper over 1.5 to 7 months (AHA [McCrindle 2017]; Wallace 2000).

Lupus nephritis; proliferative

Lupus nephritis; proliferative: Limited data available: Children and Adolescents:

Initial phase; pulse therapy:

IV:

6-month course: Usual range: 500 to 1,000 mg/m2/dose once monthly (KDIGO 2012); the following regimen has been used for dosage escalation: Initial: 500 mg/m2 then titrate as tolerated every 4 weeks in 250 mg/m2 increments up to 750 or 1,000 mg/m2 every month; maximum monthly dose: 1,500 mg/month (Bertsias 2012; Mina 2012).

3-month course: 500 mg every 2 weeks for 3 months (Bertsias 2012; KDIGO 2012).

Oral: 1 to 1.5 mg/kg/day for 2 to 4 months; maximum daily dose: 150 mg/day (KDIGO 2012); higher doses (2 to 2.5 mg/kg/day for 3 months) may be required in patients with worsening prognostic factors (eg, acute renal function deterioration) (Bertsias 2012).

Maintenance phase: IV: 500 to 1,000 mg/m2 every 3 months for a total of 1.5 to 3 years has been used; however, current guidelines recommend other oral immunosuppressive agents for maintenance therapy (KDIGO 2012; Kliegman 2011; Lehman 2000).

Nephrotic syndrome, minimal change

Nephrotic syndrome, minimal change (frequently relapsing): Infants, Children, and Adolescents: Oral: 2 mg/kg/day for 8 to 12 weeks; reported range: 2 to 3 mg/kg/day; maximum cumulative dose: 168 mg/kg; dosing based on ideal bodyweight (Gipson 2009; KDIGO 2012; KDOQI 2013). Treatment beyond 90 days may increase the potential for sterility in males.

Uveitis, severe; recalcitrant, high-risk vision loss

Uveitis, severe; recalcitrant, high-risk vision loss: Limited data available: Children and Adolescents: Oral: Initial: 2 mg/kg/day once daily; usual reported range: 1 to 3 mg/kg/day in combination with corticosteroids (which may be decreased while on cyclophosphamide) (Jabs 2000; Pujari 2010; Simonini 2010); dosing based on large, multicenter report of 215 patients with ocular inflammatory disease which included 44 patients with uveitis (age range: 11.5 to 76.4 years); after 12 months of therapy ~89% of patients had no inflammatory disease activity or only slightly active disease (Pujari 2010). Note: Some data suggests that pulse intravenous therapy may be less effective than oral cyclophosphamide (Pujari 2010).

Vasculitis, ANCA-associated

Vasculitis, ANCA-associated (eg, granulomatosis with polyangiitis [GPA], Wegener granulomatosis): Limited data available: Children and Adolescents:

IV: Initial: 15 mg/kg every 2 weeks for 3 doses, then 15 mg/kg every 3 weeks until remission or azathioprine maintenance; dosing based on experience from a pediatric case series (n=5) and a larger adult trial (n=76); in the pediatric case series, the median cumulative dose was 90 mg/kg (range: 63 to 115 mg/kg), most patients received 6 to 7 pulses of therapy; in the adult trial, therapy was continued for 3 months after remission (de Groot 2009; Krmar 2013).

Oral: 2 mg/kg/day in combination with corticosteroids until remission; a subsequent decrease in dose to 1.5 mg/kg/day for another 3 months has been reported (de Groot 2009; Kliegman 2011).

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

Dosing adjustment for toxicity: Infants, Children, and Adolescents:

Hematologic toxicity: May require dose reduction or treatment interruption.

Hemorrhagic cystitis, severe: Discontinue treatment.

Dosing: Kidney Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling; decreased renal excretion of cyclophosphamide and its metabolites may occur; monitor patients with severe impairment (CrCl 10 to 24 mL/minute) for signs and symptoms of toxicity. The following guidelines have been used by some clinicians:

Aronoff 2007: Infants, Children, and Adolescents:

CrCl ≥10 mL/minute: No dosage adjustment required.

CrCl <10 mL/minute: Administer 75% of normal dose.

Hemodialysis: Moderately dialyzable (20% to 50%); administer 50% of normal dose; administer after hemodialysis.

Continuous ambulatory peritoneal dialysis (CAPD): Administer 75% of normal dose.

Continuous renal replacement therapy (CRRT): Administer 100% of normal dose.

KDIGO 2012: Lupus nephritis: Children and Adolescents:

CrCl 25 to 50 mL/minute: Administer 80% of normal dose.

CrCl 10 to <25 mL/minute: Administer 70% of normal dose.

Dosing: Hepatic Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling; in severe hepatic impairment, conversion to an active metabolite may be reduced potentially affecting efficacy. The half-life and clearance of cyclophosphamide metabolites may be increased/decreased respectively.

The following adjustments have been recommended with oncologic uses (Floyd 2006): All patients:

Serum bilirubin 3.1 to 5 mg/dL or transaminases >3 times ULN: Administer 75% of dose.

Serum bilirubin >5 mg/mL: Avoid use.

Dosing: Older Adult

Refer to adult dosing; adjust for renal clearance.

Dosing: Obesity: Adult

American Society of Clinical Oncology guidelines for appropriate systemic therapy dosing in adults with cancer with a BMI ≥30 kg/m2 (Note: Excludes hematopoietic cell transplantation and CAR T-cell lymphodepletion dosing): Utilize patient's actual body 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 practice guideline committee position statement on chemotherapy dosing in obesity (ASBMT [Bubalo 2014]):

Cy200 (cyclophosphamide total dose of 200 mg/kg): Use the lesser of IBW or actual body weight (ABW).

Cy120 (cyclophosphamide total dose of 120 mg/kg): Use either IBW (preferred) or ABW for patients ≤120% IBW. Use ABW25 for patients >120% IBW.

ABW25: Adjusted wt (kg) = Ideal body weight (kg) + 0.25 [actual wt (kg) - ideal body weight (kg)]

Dosing: Adjustment for Toxicity: Adult

Cardiotoxicity: Symptoms may be managed with diuretics, angiotensin-converting enzyme inhibitors, beta-blockers, or inotropics (Floyd 2005).

Hematologic toxicity: May require dose reduction or treatment interruption. Initiate antibiotics for neutropenic fever; antifungal and antiviral medications may also be necessary. For oncology uses, consider growth factors (primary or secondary prophylaxis) in patients at increased risk for complications due to neutropenia.

Hemorrhagic cystitis, severe: Discontinue cyclophosphamide treatment. Severe or prolonged hemorrhagic cystitis may require medical or surgical treatment.

Infection (serious): May require dose reduction, or interruption or discontinuation of treatment.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Capsule, Oral:

Generic: 25 mg, 50 mg

Solution, Intravenous:

Generic: 500 mg/2.5 mL (2.5 mL); 1 g/5 mL (5 mL); 2 g/10 mL (10 mL)

Solution Reconstituted, Injection:

Generic: 500 mg (1 ea [DSC]); 1 g (1 ea [DSC]); 2 g (1 ea [DSC])

Solution Reconstituted, Injection [preservative free]:

Generic: 500 mg (1 ea); 1 g (1 ea); 2 g (1 ea)

Tablet, Oral:

Generic: 25 mg, 50 mg

Generic Equivalent Available: US

Yes

Dosage Forms: Canada

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

Solution Reconstituted, Injection:

Procytox: 200 mg (1 ea); 500 mg (1 ea); 1 g (50 mL); 2 g (100 mL)

Tablet, Oral:

Procytox: 25 mg, 50 mg

Administration: Adult

Cyclophosphamide is associated with a moderate or high emetic potential (depending on dose, regimen, or administration route); antiemetics may be recommended to prevent nausea and vomiting (ASCO [Hesketh 2020]; MASCC [Roila 2016]).

IV: Infusion rate may vary based on protocol (refer to specific protocol for infusion rate). Administer by direct IV injection, IVPB, or continuous IV infusion. Refer to product labeling for specific information regarding reconstitution and dilution.

Bladder toxicity: To minimize bladder toxicity, increase normal fluid intake during and for 1 to 2 days after cyclophosphamide dose. Most adult patients will require a fluid intake of at least 2 L/day. High-dose regimens should be accompanied by vigorous hydration with or without mesna therapy. Morning administration may be preferred to ensure adequate hydration throughout the day.

Hematopoietic stem cell transplant (off-label use): Approaches to reduction of hemorrhagic cystitis include infusion of 0.9% NaCl 3 L/m2/24 hours, infusion of 0.9% NaCl 3 L/m2/24 hours with continuous 0.9% NaCl bladder irrigation 300 to 1000 mL/hour, and infusion of 0.9% NaCl 1.5 to 3 L/m2/24 hours with intravenous mesna. Hydration should begin at least 4 hours before cyclophosphamide and continue at least 24 hours after completion of cyclophosphamide. The daily mesna dose (as a percentage of cyclophosphamide dose) may vary; refer to protocol and/or primary literature for mesna dose. Mesna can be administered as a continuous 24-hour intravenous infusion or be given in divided doses every 4 hours. Mesna should begin at the start of treatment, and continue at least 24 hours following the last dose of cyclophosphamide.

Oral: Swallow whole; do not crush or chew; do not open capsules. To minimize bladder toxicity, increase normal fluid intake. Morning administration may be preferred to ensure adequate hydration throughout the day; do not administer at bedtime. Avoid exposure to broken capsules and tablets; if contact occurs, wash hands immediately and thoroughly.

Administration: Pediatric

Antiemetics may be recommended to prevent nausea and vomiting; doses ≥1,200 mg/m2 IV are associated with a high emetic potential and doses of 1,000 mg/m2 IV are associated with a moderate emetic risk (POGO [Dupuis 2011]; POGO [Paw Cho Sing 2019]).

Oral: Capsules and tablets should be swallowed whole. Tablets are not scored and should not be cut, crushed, or chewed. Capsules should not be opened, crushed, or chewed. Wear gloves when handling capsules/tablets and container; avoid exposure to broken capsules. If exposure to capsule contents or crushed/cut tablets, wash hands immediately and thoroughly. Morning administration may be preferred to ensure adequate hydration throughout the day; do not administer tablets/capsules at bedtime.

Parenteral:

IV push: May administer reconstituted solution without further dilution (20 mg/mL); rate may vary based on protocols (refer to specific protocols)

IV infusion (intermittent or continuous): Infusion rate may vary based on protocol (refer to specific protocol for infusion rate); usually over 15 to 60 minutes; larger doses (>1,800 mg/m2) have been infused over 1 to 6 hours by some centers and protocols

Bladder toxicity: To minimize bladder toxicity, increase normal fluid intake during and for 1 to 2 days after cyclophosphamide dose. Most adult patients will require a fluid intake of at least 2 L/day and in pediatric patients twice maintenance (3 L/m2/day). High-dose regimens and depending upon the protocol some standard (low) cyclophosphamide doses should be accompanied by vigorous hydration with mesna therapy (refer to specific protocols and Mesna monograph for additional information). Morning administration may be preferred to ensure adequate hydration throughout the day.

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

Oncology uses: Treatment of acute lymphoblastic leukemia, acute myeloid leukemia, breast cancer, chronic lymphocytic leukemia, chronic myeloid leukemia, Hodgkin lymphoma, mycosis fungoides, multiple myeloma, neuroblastoma, non-Hodgkin lymphomas (including Burkitt lymphoma), ovarian adenocarcinoma, and retinoblastoma.

Limitations of use: Although potentially effective as a single agent in susceptible malignancies, cyclophosphamide is more frequently used in combination with other chemotherapy drugs.

Nononcology uses: Nephrotic syndrome: Treatment of minimal change nephrotic syndrome (biopsy proven) in children who are unresponsive or intolerant to corticosteroid therapy.

Limitations of use: The safety and efficacy for the treatment of nephrotic syndrome in adults or in other renal diseases has not been established.

Use: Off-Label: Adult

Anti-glomerular basement membrane disease (anti-GBM or Goodpasture disease); Castleman disease (idiopathic, multicentric); Dermatomyositis/polymyositis, severe, life-threatening or refractory; Eosinophilic granulomatosis with polyangiitis (Churg-Strauss); Ewing sarcoma; Gestational trophoblastic neoplasia, high-risk; Graft-vs-host disease (acute and chronic), prophylaxis; Granulomatosis with polyangiitis and microscopic polyangiitis, organ- or life-threatening; Hematopoietic stem cell or marrow transplant; Interstitial pneumonia, nonspecific, refractory or rapidly progressive; Lupus nephritis, focal or diffuse; Lymphodepleting therapy prior to chimeric antigen receptor T-cell immunotherapy; Membranous nephropathy; Merkel cell carcinoma (advanced or recurrent); Minimal change disease; Mixed cryoglobulinemia syndrome, moderate to severe; Osteosarcoma (relapsed/refractory); Ovarian germ cell tumors (malignant); Pheochromocytoma (malignant); Polyarteritis nodosa, moderate to severe; Primary CNS lymphoma; Pure red cell aplasia; Rhabdomyosarcoma; Small cell lung cancer (refractory); Systemic light chain amyloidosis; Systemic sclerosis–related interstitial lung disease; Thymomas (advanced or metastatic); Waldenström macroglobulinemia; Warm autoimmune hemolytic anemia, relapsed or refractory

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

Cyclophosphamide may be confused with cycloSPORINE, ifosfamide

Cytoxan may be confused with cefOXitin, Ciloxan, cytarabine, CytoGam, Cytosar, Cytosar-U, Cytotec

High alert medication:

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

Adverse Reactions (Significant): Considerations
Bone marrow suppression and infection

Leukopenia, neutropenia, thrombocytopenia, and anemia commonly occur. Bone marrow failure has been reported. Bone marrow failure and severe immunosuppression may lead to serious (and fatal) infections, including sepsis and septic shock, or reactive latent infections. Serious infections have been reported in patients receiving high-dose concurrent corticosteroids (Ref). No difference has been found between oral and intravenous cyclophosphamide (Ref). Recovery of platelet and neutrophil nadirs is expected after ~20 days.

Mechanism: Dose-related (Ref); related to pharmacologic action. Alkylating agent that induces DNA damage in all rapidly dividing cells, including those of the bone marrow (Ref).

Onset: Intermediate; platelet and neutrophil nadirs are usually at weeks 1 and 2 of treatment.

Risk factors:

• Concurrent corticosteroid use (≥20 mg/day of prednisone equivalent) (Ref)

• Concurrent/multidrug chemotherapy and/or radiation therapy

• Heavy pretreatment

Cardiotoxicity

Cardiotoxicities reported include arrhythmias (supraventricular cardiac arrhythmia and ventricular arrhythmia [some with prolonged QT interval on ECG]), heart failure, heart block, hemopericardium (secondary to hemorrhagic myocarditis and myocardial necrosis), myocarditis (including fatal hemorrhagic), pericarditis, pericardial effusion including cardiac tamponade, and tachyarrhythmias.

Mechanism: Dose-related; not clearly established. Metabolites, specifically acrolein and/or phosphoramide mustard, are believed to cause oxidative stress to the myocardium and direct endothelial capillary damage. This leads to extravasation of erythrocytes, proteins, and toxic metabolites that cause direct damage to the myocardium, manifesting as arrhythmias and/or heart failure (Ref). Capillary microthrombosis and fibrin deposition occur with cyclophosphamide-induced cardiac injury (Ref).

Onset: Varied; usually within the first 48 hours but may occur up to 10 days after initiation (Ref).

Risk factors:

• Higher doses commonly used in hematopoietic stem cell transplantation. Acute heart failure is reported in 7% to 33% of patients receiving a total dose >150 mg/kg (Ref)

• Preexisting arrhythmias, cardiac disease, diabetes, hypertension (Ref)

• Older age (>55 years) (Ref)

• Prior radiation therapy to the chest

• Prior or concurrent cardiotoxic medications

• Heavy pretreatment

Hemorrhagic cystitis

Cyclophosphamide is associated with the development of hemorrhagic cystitis, which may lead to pyelitis, ureteral disease (ureteritis), and hematuria. Hemorrhagic cystitis may rarely be severe or fatal. Bladder fibrosis, necrosis or contracture may also occur, either with or without cystitis. Occurrence of hemorrhagic cystitis increases risk of bladder cancer. While hematuria generally resolves within a few days after treatment is withheld, it may persist in some cases.

Mechanism: Dose- and duration-related; hemorrhagic cystitis is due to excretion of cyclophosphamide metabolite, acrolein, in the urine. Acrolein irritates the lining of the bladder, specifically the transitional cells, causing damage and sloughing of those cells leading to hemorrhagic cystitis (Ref). Chloroacetaldehyde may also be implicated in bladder and kidney toxicity through oxidative effects (Ref).

Onset: Varied; within hours or days of treatment with intravenous cyclophosphamide (Ref), but may occur up to a median of 2 weeks later (Ref). With oral therapy for Wegener granulomatosis, median time to development was 37 months (Ref).

Risk factors:

• Higher doses associated such as with bone marrow transplantation (Ref)

• Cumulative dose when used in rheumatic diseases (Ref)

• In bone marrow transplant, male patients (Ref)

• Active urinary tract infections, especially BK viruria, or obstructions (Ref)

• Dehydration (due to lower frequency of voiding) (Ref)

• Older patients (Ref)

Hepatotoxicity

Hepatic sinusoidal obstruction syndrome (SOS), formerly called veno-occlusive liver disease, has been reported in patients receiving chemotherapy regimens containing cyclophosphamide. Signs and symptoms of hepatic SOS include bilirubin >1.4 mg/dL, unexplained weight gain, ascites, hepatomegaly, and unexplained right upper quadrant pain (Ref). Patients may also experience cholestatic hepatitis or cytolytic hepatitis, including hepatic failure with associated liver function test abnormalities and sequelae.

Mechanism: Direct toxicity of cyclophosphamide and its metabolites (phosphoramide mustard and acrolein) to hepatic sinusoids (Ref). These metabolites induce oxidative stress by producing free radicals that inhibit the activity of antioxidant enzymes such as superoxide dismutase, glutathione, and catalase (Ref).

Onset: Varied; within 2 to 8 weeks of initiation. In the case of transplantation, onset of SOS is usually within 10 to 20 days of myeloablation (Ref).

Risk factors:

• Cytoreductive conditional transplantation regimens in combination with total body irradiation, busulfan or other hepatotoxic agents (Ref)

• Concurrent azathioprine (Ref)

• Concurrent dactinomycin (Ref)

• Long-term lower doses for immunosuppressive indications (Ref)

• Preexisting hepatic dysfunction (Ref)

• Prior radiation to the abdominal area (Ref)

• Children <3 years of age (Ref)

• Patients with CYP2B6*7 variant (Ref)

Pulmonary toxicity

Pulmonary toxicity, including pneumonitis, pulmonary fibrosis, pulmonary veno-occlusive disease, and acute respiratory distress syndrome, have been reported. Pulmonary toxicity may be difficult to identify due to confounding pulmonary conditions occurring in the same population, including infections and pulmonary malignancies. This toxicity often presents as dyspnea, fever, cough, parenchymal infiltrates, abnormal pulmonary function tests and pleural thickening. Early onset pulmonary toxicity may be reversible, but late onset is associated with pleural thickening and may persist (Ref). Late onset pneumonitis develops rarely in patients receiving prolonged treatment over several months to years with lower doses of cyclophosphamide and may be associated with increased mortality (Ref).

Mechanism: Exact mechanism unknown; metabolism of cyclophosphamide in lung to alkylating metabolites and acrolein, a reactive aldehyde, is in part responsible for pulmonary toxicity. Acrolein may cause direct injury to the pulmonary epithelium, leading to alveolitis and fibrosis (Ref). Alterations in pulmonary mixed-function oxidase activity, glutathione content, along with cyclophosphamide- and acrolein-induced alterations in the physical state of membrane lipid bilayer may be the major cause of inactivation of membrane-bound enzymes (Ref).

Onset: Varied; onset can be early (within 1 to 6 months of initiation) or late (several months to years after initiation) (Ref).

Risk factors:

• Oxygen therapy (hyperoxic conditions) may potentiate the effects (Ref)

• Concurrent use of pulmonary toxic medications, including bleomycin, busulfan, and carmustine (Ref)

• Prior or concurrent thoracic radiation (Ref)

• Concurrent therapies such as amiodarone and G-CSF or GM-CSF

• Preexisting lung disease

Second primary malignancy

Secondary primary malignant neoplasm (acute leukemia, bladder carcinoma, malignant lymphoma, malignant neoplasm of thyroid, myelodysplastic syndrome, sarcoma, and other solid tumors) have been reported with both single-agent cyclophosphamide and with combination chemotherapy regimens.

Mechanism: Dose-related; as an alkylating agent, cyclophosphamide is directly genotoxic. DNA-induced damage causes double-strand breaks, leading to genomic instability due to loss of DNA repair (Ref). Inherited genetic polymorphisms may affect the risk post-alkylator exposure (Ref). Bladder inflammation, secondary to acrolein exposure, is also associated with increased risk of bladder cancer after therapy (Ref).

Onset: Delayed; several years to decades after treatment (Ref).

Risk factors:

• Higher cumulative doses (Ref)

• Higher doses associated with stem cell transplantation (Ref)

• Concurrent use of other carcinogenic chemotherapy agents (Ref)

• Patients previously experiencing hemorrhagic cystitis (bladder cancer)

• Tobacco smoking (Ref)

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified.

Postmarketing:

Cardiovascular: Acute myocardial infarction (Yeh 2021), atrial fibrillation (Yeh 2021), atrial flutter (Yeh 2021), bradycardia, cardiac tamponade (Yeh 2021), cardiogenic shock, cardiomyopathy, chest pain, edema, flushing, heart failure (Yeh 2021), hemorrhagic myocarditis, hypertension, hypotension, myocarditis, palpitations, pericardial effusion (Yeh 2021), pericarditis (Yeh 2021), peripheral ischemia, prolonged QT interval on ECG, pulmonary embolism, supraventricular cardiac arrhythmia (Yeh 2021), tachycardia, vasculitis, venous thrombosis, ventricular arrhythmia (Yeh 2021), ventricular fibrillation, ventricular tachyarrhythmia

Dermatologic: Alopecia, changes in nails, dermatitis, erythema multiforme, erythema of skin, hyperhidrosis, palmar-plantar erythrodysesthesia, pruritus, skin abnormalities related to radiation recall, skin blister, skin rash, skin toxicity, Stevens-Johnson syndrome (Assier-Bonnet 1996), toxic epidermal necrolysis (Sasak 2016), urticaria (Thong 2002)

Endocrine & metabolic: Amenorrhea, decreased serum glucose, fluid retention, hot flash, hyponatremia (Chen 2020; Esposito 2017; Gilbar 2012), increased lactate dehydrogenase, increased serum glucose, malignant neoplasm of thyroid, nephrogenic diabetes insipidus, water intoxication

Gastrointestinal: Acute pancreatitis, ageusia, cholestasis, colitis, constipation, dysgeusia, enteritis, gastrointestinal hemorrhage, hemorrhagic colitis, nausea, neutropenic typhlitis, oral mucosa ulcer, parotitis, stomatitis, vomiting

Genitourinary: Azoospermia, bladder carcinoma (Brown 2005), bladder disease (necrosis, contracture, or atypical epithelial cells), cystitis (ulcerative), defective oogenesis, defective spermatogenesis, hematuria (Wang 2015), hemorrhagic cystitis (Wang 2015), infertility, infrequent uterine bleeding, oligospermia, ovarian failure, ovarian fibrosis, premature labor, pyelitis, testicular atrophy, toxic nephrosis, ureteral disease (ureteritis)

Hematologic & oncologic: Anemia (Malpica 2020), bone marrow depression (including bone marrow failure) (Malpica 2020), C-reactive protein increased, disseminated intravascular coagulation, febrile neutropenia, hemolytic-uremic syndrome, leukopenia (Malpica 2020), myelodysplastic syndrome (Brown 2005), neutropenia (Malpica 2020), pulmonary hemorrhage, secondary primary malignant neoplasm (including acute leukemia, malignant lymphoma, malignant neoplasm of urinary tract [pelvic, renal, and ureteric], myelatelia, sarcoma) (Brown 2005), thrombocytopenia (Malpica 2020), thrombotic microangiopathy

Hepatic: Ascites (Arndt 2004), cholestatic hepatitis, hepatic encephalopathy, hepatic failure (Arndt 2004), hepatic sinusoidal obstruction syndrome (Arndt 2004), hepatitis (including hepatic cytolysis), hepatomegaly (Arndt 2004), increased liver enzymes (Arndt 2004), increased serum bilirubin (Arndt 2004), jaundice (Arndt 2004)

Hypersensitivity: Anaphylactic shock, anaphylaxis (Garas 1995; Jones 1989), facial swelling

Immunologic: Immunosuppression (Malpica 2020)

Infection: Cytomegalovirus disease (Goldsmith 2021), infection (including reactivation of latent infection)

Local: Infusion-site reaction (including erythema, inflammation, necrosis, pain, phlebitis, swelling, and thrombosis), injection-site reaction (including erythema at injection site, inflammation at injection site, injection-site phlebitis, pain at injection site, swelling at injection site, tissue necrosis at injection site, venous thrombosis at injection site)

Nervous system: Altered sense of smell, asthenia, chills, confusion, dizziness, dysesthesia, encephalopathy, fatigue, headache, hypoesthesia, malaise, neuralgia, neurotoxicity, pain, paresthesia, peripheral neuropathy, polyneuropathy, reversible posterior leukoencephalopathy syndrome, seizure, tremor

Neuromuscular & skeletal: Arthralgia, muscle spasm, myalgia, myelopathy, rhabdomyolysis, systemic sclerosis

Ophthalmic: Conjunctivitis, lacrimation, visual impairment

Otic: Auditory impairment, deafness, tinnitus

Renal: Hemorrhagic ureteritis, renal cell carcinoma, renal failure syndrome, renal insufficiency, renal tubular disease

Respiratory: Acute respiratory distress syndrome (Malik 1996), bronchiolitis obliterans, bronchospasm, cough, dyspnea, flu-like symptoms, hypersensitivity pneumonitis, hypoxia, interstitial pulmonary disease, nasal congestion, nasal discomfort, oropharyngeal pain, pleural effusion, pneumonia, pneumonitis (can be late onset [>6 months]) (Malik 1996), pulmonary edema, pulmonary fibrosis (Malik 1996), pulmonary hypertension, pulmonary veno-occlusive disease, respiratory distress (Malik 1996), respiratory failure (Malik 1996), rhinorrhea

Miscellaneous: Fever, multi-organ failure

Contraindications

History of severe hypersensitivity to cyclophosphamide, its metabolites, or any component of the formulation; urinary outflow obstruction.

Canadian labeling: Additional contraindications (not in the US labeling): Severe myelosuppression, severe renal or hepatic impairment, active infection (especially varicella zoster), severe immunosuppression.

Warnings/Precautions

Concerns related to adverse effects:

• Hypersensitivity: Possible cross-sensitivity with other alkylating agents may occur.

Disease-related concerns:

• Hepatic impairment: Use with caution in hepatic impairment; see "Dosing: Hepatic Impairment" for additional information.

• Renal impairment: Use with caution in patients with renal impairment; see "Dosing: Altered Kidney Function" for additional information.

Dosage form specific issues:

• Cyclophosphamide injection: Some cyclophosphamide injection dosage forms may contain alcohol. The alcohol content (in some dosage forms) may affect the CNS and impair the ability to drive or operate machinery; review available dosage forms for ethanol content in order to select the appropriate product, particularly for patients who should avoid or minimize alcohol intake, including patients with hepatic impairment.

Metabolism/Transport Effects

Substrate of CYP2A6 (minor), CYP2B6 (major), CYP2C19 (minor), CYP2C9 (minor), CYP3A4 (minor); Note: Assignment of Major/Minor substrate status based on clinically relevant drug interaction potential

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

Allopurinol: May increase the serum concentration of Cyclophosphamide. Risk C: Monitor therapy

Amiodarone: Cyclophosphamide may enhance the adverse/toxic effect of Amiodarone. Specifically, the risk of pulmonary toxicity may be enhanced. Risk C: Monitor therapy

Anthracyclines: Cyclophosphamide may enhance the cardiotoxic effect of Anthracyclines. Risk C: Monitor therapy

AzaTHIOprine: May enhance the hepatotoxic effect of Cyclophosphamide. Risk C: Monitor therapy

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

Bupivacaine: Cyclophosphamide may enhance the adverse/toxic effect of Bupivacaine. Specifically, the risk of methemoglobinemia may be increased. Risk C: Monitor therapy

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: 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 Vaccines: Cyclophosphamide may diminish the therapeutic effect of COVID-19 Vaccines. Management: Adjust timing of intravenous cyclophosphamide so that administration occurs 1 week after each vaccine dose, if feasible; hold oral cyclophosphamide for 1 to 2 weeks after vaccine administration as permitted by the underlying disease. Risk D: Consider therapy modification

CycloSPORINE (Systemic): Cyclophosphamide may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

CYP2B6 Inducers (Moderate): May increase serum concentrations of the active metabolite(s) of Cyclophosphamide. 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

Etanercept: May enhance the adverse/toxic effect of Cyclophosphamide. An increased risk of solid cancer development may be present. 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

Filgrastim: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, the risk of pulmonary toxicity may be enhanced. Risk C: Monitor therapy

Fluconazole: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, serum bilirubin and serum creatinine may be increased. Fluconazole may increase the serum concentration of Cyclophosphamide. 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

Itraconazole: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, serum creatinine and serum bilirubin may be increased. Itraconazole may increase the serum concentration of Cyclophosphamide. Risk C: Monitor therapy

Ketoconazole (Systemic): May increase the serum concentration of Cyclophosphamide. Risk C: Monitor therapy

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: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, the risk of pulmonary toxicity may be enhanced. Cyclophosphamide may diminish the therapeutic effect of Lenograstim. Management: Avoid the use of lenograstim 24 hours before until 24 hours after the completion of bleomycin infusion. Monitor for enhanced pulmonary toxicity when cyclophosphamide and lenograstim are given in combination. Risk D: Consider therapy modification

Lipegfilgrastim: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, the risk of pulmonary toxicity may be increased. Cyclophosphamide may diminish the therapeutic effect of Lipegfilgrastim. Management: Lipegfilgrastim should be administered at least 24 hours after the completion of cyclophosphamide. Consider monitoring for enhanced pulmonary toxicity when cyclophosphamide and lipegfilgrastim are given in combination. Risk D: Consider therapy modification

Lumacaftor and Ivacaftor: May decrease the serum concentration of CYP2B6 Substrates (High risk with Inducers). Risk C: Monitor therapy

MiFEPRIStone: May increase the serum concentration of CYP2B6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

Mivacurium: Cyclophosphamide may increase the serum concentration of Mivacurium. Risk C: Monitor therapy

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

Protease Inhibitors: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, the incidences of neutropenia, infection, and mucositis may be increased. Protease Inhibitors may increase the serum concentration of Cyclophosphamide. 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

Sargramostim: Cyclophosphamide may enhance the adverse/toxic effect of Sargramostim. Specifically, the risk of pulmonary toxicity may be enhanced. Risk C: Monitor therapy

Schisandra: May increase the serum concentration of Cyclophosphamide. Risk C: Monitor therapy

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

Succinylcholine: Cyclophosphamide may increase the serum concentration of Succinylcholine. Management: Consider alternatives to succinylcholine in patients who have received cyclophosphamide in the past 10 days, or reduced succinylcholine doses (a serum pseudocholinesterase assay may help inform this reduction) with close monitoring. Risk D: Consider therapy modification

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

Thiazide and Thiazide-Like Diuretics: May enhance the adverse/toxic effect of Cyclophosphamide. Specifically, granulocytopenia may be enhanced. Risk C: Monitor therapy

Thiotepa: May increase the serum concentration of CYP2B6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

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

Vasopressin: Drugs Suspected of Causing SIADH may enhance the therapeutic effect of Vasopressin. Specifically, the pressor and antidiuretic effects of vasopressin may be increased. Risk C: Monitor therapy

Voclosporin: Cyclophosphamide may enhance the adverse/toxic effect of Voclosporin. 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

Evaluate pregnancy status prior to use in patients who could become pregnant.

Patients who could become pregnant should use effective contraception while receiving cyclophosphamide and for up to 1 year after completion of cyclophosphamide treatment. Patients with partners who are or could become pregnant should use a condom during and for at least 4 months after cyclophosphamide treatment.

Cyclophosphamide is used off label in the management of lupus nephritis in nonpregnant adults (ACR [Hahn 2012]; EULAR/ERA-EDTA [Fanouriakis 2020]). Females treated for rheumatic and musculoskeletal diseases should consider discontinuing cyclophosphamide 3 to 6 months prior to attempted pregnancy to allow for disease monitoring and potential change to another immunosuppressant. Cyclophosphamide should also be discontinued 12 weeks prior to attempted conception in males with rheumatic and musculoskeletal diseases who are planning to father a child (ACR [Sammaritano 2020]).

Cyclophosphamide may cause ovarian insufficiency in females, and infertility and long-term gonadal damage in males. Dose-related sterility (which may be irreversible) may occur in both males and females. Recommendations are available for fertility preservation of male and female adult patients treated with anticancer agents (ASCO [Oktay 2018]). Recommendations for preserving fertility in females and males treated with cyclophosphamide for autoimmune and systemic inflammatory diseases are available (ACR [Sammaritano 2020]).

Pregnancy Considerations

Cyclophosphamide crosses the placenta and can be detected in amniotic fluid (D'Incalci 1982).

Birth defects (including malformations of the skeleton, palate, limbs, and eyes), miscarriage, fetal growth retardation, and fetotoxic effects in the newborn (including anemia, gastroenteritis leukopenia, pancytopenia, and severe bone marrow hypoplasia) have been reported.

Cyclophosphamide, if indicated, may be administered to pregnant patients with breast cancer as part of some combination chemotherapy regimens; chemotherapy should not be administered during the first trimester, after 35 weeks' gestation, or within 3 weeks of planned delivery (Amant 2010; Loibl 2015; Shachar 2017). Use of regimens containing cyclophosphamide are generally avoided for the treatment of Hodgkin or non-Hodgkin lymphoma in pregnancy. However, use of cyclophosphamide may be considered as part of some regimens to treat patients diagnosed with aggressive non-Hodgkin lymphomas during the second or third trimester (Lishner 2016; Moshe 2017). The European Society for Medical Oncology has published guidelines for diagnosis, treatment, and follow-up of cancer during pregnancy. The guidelines recommend referral to a facility with expertise in cancer during pregnancy and encourage a multidisciplinary team (obstetrician, neonatologist, oncology team). In general, if chemotherapy is indicated, it should be avoided during in the first trimester, there should be a 3-week time period between the last chemotherapy dose and anticipated delivery, and chemotherapy should not be administered beyond week 33 of gestation (ESMO [Peccatori 2013]).

Cyclophosphamide is used off label in the management of lupus nephritis in nonpregnant adults (ACR [Hahn 2012]; EULAR/ERA-EDTA [Fanouriakis 2020). In patients with life- or organ-threatening maternal disease, cyclophosphamide may be used in the second or third trimesters only when an alternative therapy is not available (ACR [Sammaritano 2020]).

A pregnancy registry is available for all cancers diagnosed during pregnancy at Cooper Health (1-877-635-4499).

Breastfeeding Considerations

Cyclophosphamide and its metabolites are present in breast milk (Codacci-Pisanelli 2019).

Cyclophosphamide breast milk concentrations were evaluated following maternal treatment for stage IV diffuse large B-cell lymphoma at 4 months postpartum. Peak breast milk concentrations occurred within 7 days; however, cyclophosphamide was detectable in breast milk for 21 days after the first dose (Codacci-Pisanelli 2019). Leukopenia and thrombocytopenia were noted in an infant exposed to cyclophosphamide while breastfeeding. The mother was treated with one course of cyclophosphamide 6 weeks prior to delivery then cyclophosphamide IV 6 mg/kg (300 mg) once daily for 3 days beginning 20 days postpartum. CBCs were obtained in the breastfed infant on each day of therapy; WBC and platelets decreased by day 3 (Durodola 1979).

Cyclophosphamide is not recommended for use in breastfeeding mothers with autoimmune and systemic inflammatory diseases (ACR [Sammaritano 2020]). Due to the potential for serious adverse reactions in the breastfed infant, breastfeeding is not recommended by the manufacturer during therapy and for 1 week after the last cyclophosphamide dose. Others recommend breastfeeding be avoided for at least 6 weeks after the last dose of cyclophosphamide (Codacci-Pisanelli 2019).

Monitoring Parameters

CBC with differential and platelets, BUN, serum electrolytes, serum creatinine, urinalysis. Evaluate pregnancy status prior to use in patients who can become pregnant. Monitor for signs/symptoms of hemorrhagic cystitis or other urinary/renal toxicity, pulmonary toxicity (if suspect pneumonitis, consider pulmonary function testing to assess the severity of pneumonitis [Morgan 2011]), cardiac toxicity (particularly in patients with cardiac risk factors or preexisting cardiac disease), hepatic toxicity (including hepatic sinusoidal obstruction syndrome), secondary malignancies, and/or wound healing impairment. Monitor adherence (for oral dosing).

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

Cyclophosphamide is an alkylating agent that prevents cell division by cross-linking DNA strands and decreasing DNA synthesis. It is a cell cycle phase nonspecific agent. Cyclophosphamide also possesses potent immunosuppressive activity. Cyclophosphamide is a prodrug that must be metabolized to active metabolites in the liver.

Pharmacokinetics

Absorption: Oral: Well absorbed

Distribution: Vd: 30 to 50 L (approximates total body water); crosses into CSF (not in high enough concentrations to treat meningeal leukemia)

Protein binding: ~20%; some metabolites are bound at >60%

Metabolism: Hepatic to active metabolites acrolein, 4-aldophosphamide, 4-hydroperoxycyclophosphamide, and nor-nitrogen mustard

Bioavailability: >75%

Half-life elimination: IV: 3 to 12 hours; Children: 4 hours; Adults: 6 to 8 hours

Time to peak: Oral: ~1 hour; IV: Metabolites: 2 to 3 hours

Excretion: Urine (10 to 20% as unchanged drug); feces (4%)

Pharmacokinetics: Additional Considerations

Altered kidney function: Systemic exposure is increased as renal function declines. The mean (dose-corrected AUC) increased by 38% in patients with moderate renal impairment (CrCl 25 to 50 mL/minute), increased by 64% in patients with severe impairment (CrCl 10 to 24 mL/minute), and by 23% in patients undergoing hemodialysis (CrCl <10 mL/minute), when compared to a control group.

Hepatic function impairment: In patients with severe hepatic impairment, the elimination half-life is prolonged by 64%.

Pricing: US

Capsules (Cyclophosphamide Oral)

25 mg (per each): $6.00 - $9.36

50 mg (per each): $8.40 - $17.75

Solution (Cyclophosphamide Intravenous)

1 g/5 mL (per mL): $175.20 - $175.80

2 g/10 mL (per mL): $175.80

500 mg/2.5 mL (per mL): $175.20 - $175.80

Solution (reconstituted) (Cyclophosphamide Injection)

1 g (per each): $444.00 - $879.00

2 g (per each): $888.00 - $1,758.00

500 mg (per each): $222.00 - $439.50

Tablets (Cyclophosphamide Oral)

25 mg (per each): $3.54

50 mg (per each): $5.05

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
  • Alkyloxan (EG, KR, SG);
  • Alkyroxan (KR);
  • Cryofaxol (CR, DO, GT, HN, MX, NI, PA, SV);
  • Cycloblastin (AU, NZ);
  • Cycloblastine (LU);
  • Cyclonex (AU);
  • Cyclostin (DE);
  • Cyclostin N (DE);
  • Cycram (EG, VN);
  • Cyphos (LK);
  • Cytoxan (CO, HU, ID);
  • Endoksan (UA);
  • Endoxan (AT, AU, BE, BG, CL, CZ, DE, EE, EG, GR, HN, HR, HU, ID, IL, IT, JP, KR, LT, LU, LV, NL, NZ, PK, PL, PT, RO, RU, SG, SI, SK, TR, UY, VN, ZA);
  • Endoxan-Asta (AE, BH, CH, CY, FR, HK, IN, IQ, IR, JO, KW, LB, LY, MY, OM, PH, QA, SA, SY, TH, TW, YE);
  • Endoxana (GB, IE);
  • Endoxon-Asta (AU);
  • Enduxan (BR);
  • Formitex (CR, DO, GT, HN, NI, PA, SV);
  • Genoxal (BR, ES);
  • Hidrofosmin (CR, DO, GT, HN, NI, PA, SV);
  • Ledoxan (PH);
  • Ledoxina (MX);
  • Neophamid (KR);
  • Neophos (EG, LB);
  • Oncomide (LK);
  • Sendoxan (DK, FI, NO, SE);
  • Syklofosfamid (TR);
  • Uniphos (LK);
  • Xyclomed (PH)


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. Abid SH, Malhotra V, Perry MC. Radiation-induced and chemotherapy-induced pulmonary injury. Curr Opin Oncol. 2001;13(4):242-248. doi:10.1097/00001622-200107000-00006 [PubMed 11429481]
  3. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839-852. doi:10.1016/S0140-6736(20)31366-0 [PubMed 32888407]
  4. ACCESS Trial Group. Treatment of lupus nephritis with abatacept: the Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol. 2014;66(11):3096-3104. doi:10.1002/art.38790 [PubMed 25403681]
  5. Allan JM, Travis LB. Mechanisms of therapy-related carcinogenesis. Nat Rev Cancer. 2005;5(12):943-955. doi:10.1038/nrc1749 [PubMed 16294218]
  6. Almaani S, Meara A, Rovin BH. Update on lupus nephritis. Clin J Am Soc Nephrol. 2017;12(5):825-835. doi:10.2215/CJN.05780616 [PubMed 27821390]
  7. Amant F, Deckers S, Van Calsteren K, et al. Breast cancer in pregnancy: recommendations of an international consensus meeting. Eur J Cancer. 2010;46(18):3158-3168. [PubMed 20932740]
  8. American Geriatrics Society 2015 Beers Criteria Update Expert Panel. American Geriatrics Society 2015 updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2015;63(11):2227-2246. [PubMed 26446832]
  9. Anderson JE, Appelbaum FR, Schoch G, et al. Allogeneic marrow transplantation for myelodysplastic syndrome with advanced disease morphology: a phase II study of busulfan, cyclophosphamide, and total body-irradiation and analysis of prognostic factors. J Clin Oncol. 1996;14(1):220-226. [PubMed 8558201]
  10. Arndt C, Hawkins, D, Anderson JR, et al. Age is a risk factor for chemotherapy-induced hepatopathy with vincristine, dactinomycin and cyclophosphamide. J Clin Oncol. 2004;22(10):1894-1901. [PubMed 15143082]
  11. Arndt CA, Stoner JA, Hawkins DS, et al. Vincristine, actinomycin, and cyclophosphamide compared with vincristine, actinomycin, and cyclophosphamide alternating with vincristine, topotecan, and cyclophosphamide for intermediate-risk rhabdomyosarcoma: children's oncology group study D9803. J Clin Oncol. 2009;27(31):5182-5188. doi: 10.1200/JCO.2009.22.3768. [PubMed 19770373]
  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, p 97, 170.
  13. Assier-Bonnet H, Aractingi S, Cadranel J, Wechsler J, Mayaud C, Saiag P. Stevens-Johnson syndrome induced by cyclophosphamide: report of two cases. Br J Dermatol. 1996;135(5):864-866. doi:10.1111/j.1365-2133.1996.tb03915.x [PubMed 8977704]
  14. Audino AN, Blatt J, Carcamo B, et.al. High-dose cyclophosphamide treatment for refractory severe aplastic anemia in children. Pediatr Blood Cancer. 2010;54:269-272. [PubMed 19827142]
  15. Austin HA 3rd, Klipel JH, Balow JE, et al. Therapy of lupus nephritis. Controlled trial of prednisone and cytotoxic drugs N Engl J Med. 1986;314(10):614-619. [PubMed 3511372]
  16. Banerjee S, Biehl A, Ghaderi-Yeganeh M, Manna Z, Hasni S. Low incidence of opportunistic infections in lupus patients treated with cyclophosphamide and steroids in a tertiary care setting. Med Res Arch. 2017;5(3):10.18103/mra.v5i3.1084. doi:10.18103/mra.v5i3.1084 [PubMed 29276769]
  17. Bennett CL, Cournoyer D, Carson KR, et al. Long-term outcome of individuals with pure red cell aplasia and antierythropoietin antibodies in patients treated with recombinant epoetin: a follow-up report from the Research on Adverse Drug Events and Reports (RADAR) Project. Blood. 2005;106(10):3343-3347. [PubMed 16099877]
  18. Berdeja JG, Madduri D, Usmani SZ, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet. 2021;398(10297):314-324. doi:10.1016/S0140-6736(21)00933-8 [PubMed 34175021]
  19. Berger M, Grignani G, Ferrari S, et al. Phase 2 trial of two courses of cyclophosphamide and etoposide for relapsed high-risk osteosarcoma patients. Cancer. 2009;115(13):2980-2987. doi:10.1002/cncr.24368 [PubMed 19452540]
  20. Berns JS. Pure red cell aplasia (PRCA) due to anti-erythropoiesis-stimulating agent antibodies. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2021.
  21. Bertsias GK, Tektonidou M, Amoura Z, et al. Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis. Ann Rheum Dis. 2012;71(11):1771-1782. [PubMed 22851469]
  22. Bodge MN, Reddy S, Thompson MS, Savani BN. Preparative regimen dosing for hematopoietic stem cell transplantation in patients with chronic kidney disease: analysis of the literature and recommendations. Biol Blood Marrow Transplant. 2014;20(7):908-919. doi:10.1016/j.bbmt.2014.02.013 [PubMed 24565993]
  23. Braverman AC, Antin JH, Plappert MT, Cook EF, Lee RT. Cyclophosphamide cardiotoxicity in bone marrow transplantation: a prospective evaluation of new dosing regimens. J Clin Oncol. 1991;9(7):1215-1223. doi:10.1200/JCO.1991.9.7.1215 [PubMed 2045862]
  24. Brodsky RA, Chen AR, Dorr D, et.al. High-dose cyclophosphamide for severe aplastic anemia: long-term follow-up. Blood. 2010;115:2136-2141. [PubMed 20018919]
  25. Brodsky RA, Sensenbrenner LL, Jones RJ. Complete remission in severe aplastic anemia after high-dose cyclophosphamide without bone marrow transplantation. Blood. 1996;87(2): 491-494. [PubMed 8555470]
  26. Brown JR, Yeckes H, Friedberg JW, et al. Increasing incidence of late second malignancies after conditioning with cyclophosphamide and total-body irradiation and autologous bone marrow transplantation for non-Hodgkin's lymphoma. J Clin Oncol. 2005;23(10):2208-2214. doi:10.1200/JCO.2005.05.158 [PubMed 15753460]
  27. 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. [PubMed 24462742]
  28. 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]
  29. Cakir O, Ayyildiz O, Isikdogan A. Type III mixed cryoglobulinemia associated with digital necrotic ulcer successfully treated with intermittent intravenous pulse cyclophosphamide--a case report. Angiology. 2005;56(4):489-492. doi:10.1177/000331970505600418 [PubMed 16079934]
  30. Cassileth PA, Andersen J, Lazarus HM, et al. Autologous Bone Marrow Transplant in Acute Myeloid Leukemia in First Remission. J Clin Oncol. 1993;11(2):314-319. [PubMed 8426209]
  31. 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. doi:10.1056/NEJM199812033392301 [PubMed 9834301]
  32. Champlin RE, Perez WS, Passweg JR, et al. Bone marrow transplantation for severe aplastic anemia: a randomized controlled study of conditioning regimens. Blood. 2007;109(10):4582-4585. [PubMed 17272503]
  33. Chantada G, Fandiño A, Casak S, Manzitti J, Raslawski E, Schvartzman E. Treatment of overt extraocular retinoblastoma. Med Pediatr Oncol. 2003;40(3):158-161. [PubMed 12518344]
  34. Chao NJ, Rosenberg SA, Horning SJ. CEPP(B): an effective and well-tolerated regimen in poor-risk, aggressive non-Hodgkin's lymphoma. Blood. 1990;76(7):1293-1298. [PubMed 2207307]
  35. Chen J, Jin Y, Li C, Li Z. Symptomatic hyponatremia induced by low-dose cyclophosphamide in patient with systemic lupus erythematosus: a case report. Medicine (Baltimore). 2020;99(48):e22498. doi:10.1097/MD.0000000000022498 [PubMed 33235058]
  36. Chi SN, Gardner SL, Levy AS, et al. Feasibility and response to induction chemotherapy intensified with high-dose methotrexate for young children with newly diagnosed high-risk disseminated medulloblastoma. J Clin Oncol. 2004;22(24):4881-4887. [PubMed 15611503]
  37. Chou WH, McGregor B, Schmidt A, et al. Cyclophosphamide-associated bladder cancers and considerations for survivorship care: a systematic review. Urol Oncol. 2021;39(10):678-685. doi:10.1016/j.urolonc.2021.05.017 [PubMed 34134927]
  38. Chung SA, Langford CA, Maz M, et al. 2021 American College of Rheumatology/Vasculitis Foundation guideline for the management of antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheumatol. 2021;73(8):1366-1383. doi:10.1002/art.41773 [PubMed 34235894]
  39. Cibeira MT, Oriol A, Lahuerta JJ, et al; PETHEMA cooperative study group. A phase II trial of lenalidomide, dexamethasone and cyclophosphamide for newly diagnosed patients with systemic immunoglobulin light chain amyloidosis. Br J Haematol. 2015;170(6):804-813. doi:10.1111/bjh.13500 [PubMed 25974382]
  40. Codacci-Pisanelli G, Honeywell RJ, Asselin N, et al. Breastfeeding during R-CHOP chemotherapy: please abstain! Eur J Cancer. 2019;119:107-111. doi:0.1016/j.ejca.2019.07.012 [PubMed 31437753]
  41. Cohen P, Pagnoux C, Mahr A, et al; French Vasculitis Study Group. Churg-Strauss syndrome with poor-prognosis factors: A prospective multicenter trial comparing glucocorticoids and six or twelve cyclophosphamide pulses in forty-eight patients. Arthritis Rheum. 2007;57(4):686-693. doi:10.1002/art.22679 [PubMed 17471546]
  42. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346(4):235-242. [PubMed 11807147]
  43. Coleman M, Martin P, Ruan J, et al. Prednisone, etoposide, procarbazine, and cyclophosphamide (PEP-C) oral combination chemotherapy regimen for recurring/refractory lymphoma: low-dose metronomic, multidrug therapy. Cancer. 2008;112(10):2228-2232. doi:10.1002/cncr.23422 [PubMed 18338745]
  44. Copper JA Jr. Drug-induced lung disease. Adv Intern Med. 1997;42:231-268. [PubMed 9048121]
  45. Corradini P, Tarella C, Olivieri A, et al. Reduced-intensity conditioning followed by allografting of hematopoietic cells can produce clinical and molecular remissions in patients with poor-risk hematologic malignancies. Blood. 2002;99(1):75-82. doi:10.1182/blood.v99.1.75 [PubMed 11756155]
  46. Corte TJ, Ellis R, Renzoni EA, et al. Use of intravenous cyclophosphamide in known or suspected, advanced non-specific interstitial pneumonia. Sarcoidosis Vasc Diffuse Lung Dis. 2009;26(2):132-138. [PubMed 20560293]
  47. Cyclophosphamide. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases; 2017. [PubMed 31643390]
  48. Cyclophosphamide capsules [prescribing information]. Baudette, MN: ANI Pharmaceuticals Inc; March 2020.
  49. Cyclophosphamide injection, multi-dose vial [prescribing information]. Orlando, FL: Ingenus Pharmaceuticals; September 2021.
  50. Cyclophosphamide injection, single-dose vial [prescribing information]. E. Windsor, NJ: AuroMedics Pharma LLC; August 2021.
  51. Cyclophosphamide injection [prescribing information]. BluePoint Laboratories; October 2020.
  52. Cyclophosphamide tablets [prescribing information]. Baxter Healthcare Corporation; August 2020.
  53. Dal Pra A, Berlin A, Kumareswaran R, et al. Liver failure after abdominal irradiation: identifying the right suspects. J Clin Oncol. 2016;34(9):e80-e83. doi:10.1200/JCO.2013.50.9547 [PubMed 25071139]
  54. de Groot K, Harper L, Jayne DR, et al. Pulse versus daily oral cyclophosphamide for induction of remission in antineutrophil cytoplasmic antibody-associated vasculitis: a randomized trial. Ann Intern Med. 2009;150(10):670-680. [PubMed 19451574]
  55. De Vriese AS, Wetzels JFM, Cattran DC. Membranous nephropathy: treatment and prognosis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 28, 2022.
  56. DeFilipp Z, Li S, El-Jawahri A, et al. High-dose chemotherapy with thiotepa, busulfan, and cyclophosphamide and autologous stem cell transplantation for patients with primary central nervous system lymphoma in first complete remission. Cancer. 2017;123(16):3073-3079. doi:10.1002/cncr.30695 [PubMed 28369839]
  57. Dellaripa PF, Danoff SK. Interstitial lung disease in dermatomyositis and polymyositis: treatment. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 16, 2021.
  58. DeZern AE, Petri M, Drachman DB, et al. High-dose cyclophosphamide without stem cell rescue in 207 patients with aplastic anemia and other autoimmune diseases. Medicine. 2011;90:89-98. [PubMed 21358440]
  59. Dharmarajan KV, Friedman DL, Schwartz CL, et al. Patterns of relapse from a phase 3 Study of response-based therapy for intermediate-risk Hodgkin lymphoma (AHOD0031): a report from the Children's Oncology Group. Int J Radiat Oncol Biol Phys. 2015;92(1):60-66. [PubMed 25542311]
  60. Dhesi S, Chu MP, Blevins G, et al. Cyclophosphamide-induced cardiomyopathy: a case report, review, and recommendations for management. J Investig Med High Impact Case Rep. 2013;1(1):2324709613480346. doi:10.1177/2324709613480346 [PubMed 26425570]
  61. Diehl V, Franklin J, Pfreundschuh M, et al. Standard and Increased-Dose BEACOPP Chemotherapy Compared With COPP-ABVD for Advanced Hodgkin's disease. N Engl J Med. 2003;348(24):2386-2395. [PubMed 12802024]
  62. Dimopoulos MA, Anagnostopoulos A, Kyrtsonis MC, et al. Primary treatment of Waldenström macroglobulinemia with dexamethasone, rituximab, and cyclophosphamide. J Clin Oncol. 2007;25(22):3344-3349. doi: 10.1200/JCO.2007.10.9926. [PubMed 17577016]
  63. Dimopoulos MA, Sonneveld P, Leung N, et al. International Myeloma Working Group recommendations for the diagnosis and management of myeloma-related renal impairment. J Clin Oncol. 2016;34(13):1544-1557. [PubMed 26976420]10.1200/JCO.2015.65.0044
  64. D'Incalci M, Sessa C, Colombo N, et al. Transplacental passage of cyclophosphamide. Cancer Treat Rep. 1982;66(8):1681-1682. [PubMed 7105061]
  65. Dine T, Lebegue S, Benaji B, et al. Stability and compatibility studies of four cytostatic agents (fluorouracil, dacarbazine, cyclophosphamide and ifosfamide) with PVC infusion bags. Pharm Sci Communications. 1994;4:97-101.
  66. du Buf-Vereijken PW, Branten AJ, Wetzels JF; Membranous Nephropathy Study Group. Cytotoxic therapy for membranous nephropathy and renal insufficiency: improved renal survival but high relapse rate. Nephrol Dial Transplant. 2004;19(5):1142-1148. doi:10.1093/ndt/gfh036 [PubMed 14993502]
  67. Dunleavy K, Pittaluga S, Maeda LS, et al. Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J Med. 2013;368(15):1408-1416. doi:10.1056/NEJMoa1214561 [PubMed 23574119]
  68. Dupuis LL, Boodhan S, Holdsworth M, et al; Pediatric Oncology Group of Ontario. 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]
  69. Dupuis LL, Boodhan S, Sung L. 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]
  70. Durodola JI. Administration of cyclophosphamide during late pregnancy and early lactation: a case report. J Natl Med Assoc. 1979;71(2):165-166. [PubMed 423292]
  71. Eiriksson L, Dean E, Sebastianelli A, et al. Guideline No. 408: management of gestational trophoblastic diseases. J Obstet Gynaecol Can. 2021;43(1):91-105.e1. doi:10.1016/j.jogc.2020.03.001 [PubMed 33384141]
  72. Escobar PF, Lurain JR, Singh DK, et al. Treatment of High-Risk Gestational Trophoblastic Neoplasia With Etoposide, Methotrexate, Actinomycin D, Cyclophosphamide, and Vincristine Chemotherapy. Gynecol Oncol. 2003;91(3):552-557. [PubMed 14675675]
  73. Esposito P, Domenech MV, Serpieri N, et al. Severe cyclophosphamide-related hyponatremia in a patient with acute glomerulonephritis. World J Nephrol. 2017;6(4):217-220. doi:10.5527/wjn.v6.i4.217 [PubMed 28729970]
  74. Fajgenbaum DC. HHV-8-negative/idiopathic multicentric Castleman disease. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed December 16, 2020.
  75. Fanouriakis A, Kostopoulou M, Cheema K, et al. 2019 update of the joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of lupus nephritis. Ann Rheum Dis. 2020;79(6):713-723. doi:10.1136/annrheumdis-2020-216924 [PubMed 32220834]
  76. Falk RJ, Dall'Era M, Appel GB. Lupus nephritis: initial and subsequent therapy for focal or diffuse lupus nephritis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2021b.
  77. Falk RJ, Merkel PA. Granulomatosis with polyangiitis and microscopic polyangiitis: management of disease resistant to initial therapy. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 29, 2021a.
  78. Fenig E, Brenner B, Katz A, et al. The role of radiation therapy and chemotherapy in the treatment of Merkel cell carcinoma. Cancer. 1997;80(5):881-885. doi:10.1002/(sici)1097-0142(19970901)80:5<881::aid-cncr8>3.0.co;2-o [PubMed 9307187]
  79. Fenske TS, McMahon C, Edwin D, et al. Identification of candidate alkylator-induced cancer susceptibility genes by whole genome scanning in mice. Cancer Res. 2006;66(10):5029-5038. doi:10.1158/0008-5472.CAN-05-3404 [PubMed 16707424]
  80. Fernández-Codina A, Walker KM, Pope JE; Scleroderma Algorithm Group. Treatment algorithms for systemic sclerosis according to experts. Arthritis Rheumatol. 2018;70(11):1820-1828. doi:10.1002/art.40560 [PubMed 29781586]
  81. Fernández-Juárez G, Rojas-Rivera J, Logt AV, et al; STARMEN Investigators. The STARMEN trial indicates that alternating treatment with corticosteroids and cyclophosphamide is superior to sequential treatment with tacrolimus and rituximab in primary membranous nephropathy. Kidney Int. 2021;99(4):986-998. doi:10.1016/j.kint.2020.10.014 [PubMed 33166580]
  82. Fervenza FC, Leise MD, Roccatello D. Mixed cryoglobulinemia syndrome: treatment and prognosis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2021.
  83. Fisher B, Brown AM, Dimitrov NV, et al. Two Months of Doxorubicin-Cyclophosphamide With and Without Interval Reinduction Therapy Compared With 6 Months of Cyclophosphamide, Methotrexate, and Fluorouracil in Positive-Node Breast Cancer Patients With Tamoxifen-Nonresponsive Tumors: Results From the National Surgical Adjuvant Breast and Bowel Project B-15. J Clin Oncol. 1990;8(9):1483-1496. [PubMed 2202791]
  84. Flaherty KR. Treatment and prognosis of nonspecific interstitial pneumonia. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 25, 2022.
  85. Floege J, Barbour SJ, Cattran DC, et al; Conference Participants. Management and treatment of glomerular diseases (part 1): conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2019;95(2):268-280. doi:10.1016/j.kint.2018.10.018 [PubMed 30665568]
  86. Floyd J, Mirza I, Sachs B, Perry MC. Hepatotoxicity of chemotherapy. Semin Oncol. 2006;33(1):50-67. doi:10.1053/j.seminoncol.2005.11.002 [PubMed 16473644]
  87. Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, Perry MC. Cardiotoxicity of Cancer Therapy. J Clin Oncol. 2005;23(30):7685-7696. doi:10.1200/JCO.2005.08.789 [PubMed 16234530]
  88. Fornasiero A, Daniele O, Ghiotto C, et al. Chemotherapy for invasive thymoma. A 13-year experience. Cancer. 1991;68(1):30-33. doi:10.1002/1097-0142(19910701)68:1<30::aid-cncr2820680106>3.0.co;2-4 [PubMed 2049749]
  89. Frankel AH, Singer DR, Winearls CG, Evans DJ, Rees AJ, Pusey CD. Type II essential mixed cryoglobulinaemia: presentation, treatment and outcome in 13 patients. Q J Med. 1992;82(298):101-124. [PubMed 1620812]
  90. Friedman DL, Chen L, Wolden S, et al. Dose-intensive response-based chemotherapy and radiation therapy for children and adolescents with newly diagnosed intermediate-risk hodgkin lymphoma: a report from the Children's Oncology Group Study AHOD0031. J Clin Oncol. 2014;32(32):3651-3658. [PubMed 25311218]
  91. Garas G, Crawford GP, Cain M. Anaphylactic reaction to intravenous cyclophosphamide. Aust N Z J Med. 1995;25(1):59. doi:10.1111/j.1445-5994.1995.tb00585.x [PubMed 7786253]
  92. García-Suárez J, Bañas H, Arribas I, et al. Dose-adjusted EPOCH plus rituximab is an effective regimen in patients with poor-prognostic untreated diffuse large B-cell lymphoma: results from a prospective observational study. Br J Haematol. 2007;136(2):276-285. [PubMed 17233819]
  93. Gayraud M, Guillevin L, Cohen P, et al. Treatment of good-prognosis polyarteritis nodosa and Churg-Strauss syndrome: comparison of steroids and oral or pulse cyclophosphamide in 25 patients. French Cooperative Study Group for Vasculitides. Br J Rheumatol. 1997;36(12):1290-1297. doi:10.1093/rheumatology/36.12.1290 [PubMed 9448590]
  94. Gilbar PJ, Richmond J, Wood J, Sullivan A. Syndrome of inappropriate antidiuretic hormone secretion induced by a single dose of oral cyclophosphamide. Ann Pharmacother. 2012;46(9):e23. doi:10.1345/aph.1R296 [PubMed 22911342]
  95. Gipson DS, Massengill SF, Yao L, et al. Management of childhood onset nephrotic syndrome. Pediatrics. 2009. 124:747-757. [PubMed 19651590]
  96. Go RS, Li CY, Tefferi A, Phyliky RL. Acquired pure red cell aplasia associated with lymphoproliferative disease of granular T lymphocytes. Blood. 2001;98(2):483-485. doi:10.1182/blood.v98.2.483 [PubMed 11435321]
  97. Go RS, Winters JL, Kay NE. How I treat autoimmune hemolytic anemia. Blood. 2017;129(22):2971-2979. doi: 10.1182/blood-2016-11-693689. [PubMed 28360039]
  98. Goldhirsch A, Colleoni M, Coates AS, et al. Adding Adjuvant CMF Chemotherapy to Either Radiotherapy or Tamoxifen: Are All CMFs Alike? The International Breast Cancer Study Group (IBCSG). Ann Oncol. 1998;9(5):489-493. [PubMed 9653488]
  99. 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]
  100. Goldman S, Smith L, Galardy P, et al. Rituximab with chemotherapy in children and adolescents with central nervous system and/or bone marrow-positive Burkitt lymphoma/leukaemia: a Children's Oncology Group Report. Br J Haematol. 2014;167(3):394-401. [PubMed 25066629]
  101. Goldsmith SR, Abid MB, Auletta JJ, et al. Posttransplant cyclophosphamide is associated with increased cytomegalovirus infection: a CIBMTR analysis. Blood. 2021;137(23):3291-3305. doi:10.1182/blood.2020009362 [PubMed 33657221]
  102. Gottdiener JS, Appelbaum FR, Ferrans VJ, Deisseroth A, Ziegler J. Cardiotoxicity associated with high-dose cyclophosphamide therapy. Arch Intern Med. 1981;141(6):758-763. [PubMed 7235784]
  103. Gourley MF, Austin HA 3rd, Scott D, et al. Methylprednisolone and cyclophosphamide, alone or in combination, in patients with lupus nephritis. A randomized, controlled trial. Ann Intern Med. 1996;125(7):549-557. [PubMed 8815753]
  104. Green DM, Beckwith JB, Breslow NE, et al. Treatment of children with stages II to IV anaplastic Wilms' tumor: a report from the National Wilms' Tumor Study Group. J Clin Oncol. 1994;12(10):2126-2131. [PubMed 7931483]
  105. Green DM, Cotton CA, Malogolowkin M, et al. Treatment of Wilms Tumor Relapsing After Initial Treatment With Vincristine and Actinomycin D: A Report From the National Wilms Tumor Study Group. Pediatr Blood Cancer. 2007;48(5):493-499. [PubMed 16547940]
  106. Grier HE, Krailo MD, Tarbell NJ, et al. Addition of Ifosfamide and Etoposide to Standard Chemotherapy for Ewing's Sarcoma and Primitive Neuroectodermal Tumor of Bone. N Engl J Med. 2003;348(8):694-701. [PubMed 12594313]
  107. 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]
  108. Gross TG, Orjuela MA, Perkins SL, et al. Low-dose chemotherapy and rituximab for posttransplant lymphoproliferative disease (PTLD): a Children's Oncology Group Report. Am J Transplant. 2012;12(11):3069-3075. [PubMed 22883417]
  109. Guillevin L, Jarrousse B, Lok C, et al. Longterm followup after treatment of polyarteritis nodosa and Churg-Strauss angiitis with comparison of steroids, plasma exchange and cyclophosphamide to steroids and plasma exchange. A prospective randomized trial of 71 patients. The Cooperative Study Group for Polyarteritis Nodosa. J Rheumatol. 1991;18(4):567-574. [PubMed 1676753]
  110. Guillevin L, Lhote F, Cohen P, et al. Corticosteroids plus pulse cyclophosphamide and plasma exchanges versus corticosteroids plus pulse cyclophosphamide alone in the treatment of polyarteritis nodosa and Churg-Strauss syndrome patients with factors predicting poor prognosis. A prospective, randomized trial in sixty-two patients. Arthritis Rheum. 1995;38(11):1638-1645. doi:10.1002/art.1780381116 [PubMed 7488285]
  111. Gupta D, Zachariah A, Roppelt H, Patel AM, Gruber BL. Prophylactic antibiotic usage for Pneumocystis jirovecii pneumonia in patients with systemic lupus erythematosus on cyclophosphamide: a survey of US rheumatologists and the review of literature. J Clin Rheumatol. 2008;14(5):267-272. doi:10.1097/RHU.0b013e31817a7e30 [PubMed 18679133]
  112. Hahn BH, McMahon MA, Wilkinson A, et al. American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res (Hoboken). 2012;64(6):797-808. [PubMed 22556106]
  113. Hahn KM, Johnson PH, Gordon N, et al. Treatment of Pregnant Breast Cancer Patients and Outcomes of Children Exposed to Chemotherapy In utero. Cancer. 2006;107(6):1219-1226. [PubMed 16894524]
  114. Hamada K, Nagai S, Kitaichi M, et al. Cyclophosphamide-induced late-onset lung disease. Intern Med. 2003;42(1):82-87. doi:10.2169/internalmedicine.42.82 [PubMed 12583625]
  115. Hamadani M, Kochuparambil ST, Osman S, et al. Intermediate-dose versus low-dose cyclophosphamide and granulocyte colony-stimulating factor for peripheral blood stem cell mobilization in patients with multiple myeloma treated with novel induction therapies. Biol Blood Marrow Transplant. 2012;18(7):1128-1135. doi:10.1016/j.bbmt.2012.01.005 [PubMed 22248715]
  116. Hamzeh M, Hosseinimehr SJ, Khalatbary AR, Mohammadi HR, Dashti A, Amiri FT. Atorvastatin mitigates cyclophosphamide-induced hepatotoxicity via suppression of oxidative stress and apoptosis in rat model. Res Pharm Sci. 2018;13(5):440-449. doi:10.4103/1735-5362.236837 [PubMed 30271446]
  117. Harper L, Morgan MD, Walsh M, et al. Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis: long-term follow-up. Ann Rheum Dis. 2012;71(6):955-960. [PubMed 22128076]
  118. Haubitz M, Bohnenstengel F, Brunkhorst R, Schwab M, Hofmann U, Busse D. Cyclophosphamide pharmacokinetics and dose requirements in patients with renal insufficiency. Kidney Int. 2002;61(4):1495-1501. doi:10.1046/j.1523-1755.2002.00279.x [PubMed 11918757]
  119. 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]
  120. Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2005;106(12):3725-3732. doi:10.1182/blood-2005-01-0016 [PubMed 16123223]
  121. Hijiya N, Thomson B, Isakoff MS, et al. Phase 2 trial of clofarabine in combination with etoposide and cyclophosphamide in pediatric patients with refractory or relapsed acute lymphoblastic leukemia. Blood. 2011;118(23):6043-6049. [PubMed 21967976]
  122. Horwitz S, O'Connor OA, Pro B, et al; ECHELON-2 Study Group. Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): a global, double-blind, randomised, phase 3 trial. Lancet. 2019;393(10168):229-240. doi:10.1016/S0140-6736(18)32984-2 [PubMed 30522922]
  123. Houssiau FA, Vasconcelos C, D'Cruz D, et al. Immunosuppressive therapy in lupus nephritis: the Euro-Lupus Nephritis Trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis Rheum. 2002;46(8):2121-2131. doi:10.1002/art.10461 [PubMed 12209517]
  124. Hoyles RK, Ellis RW, Wellsbury J, et al. A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma. Arthritis Rheum. 2006;54(12):3962-3970. doi: 10.1002/art.22204. [PubMed 17133610]
  125. Huang H, Abraham J, Hung E, et al. Treatment of malignant pheochromocytoma/paraganglioma with cyclophosphamide, vincristine, and dacarbazine: recommendation from a 22-year follow-up of 18 patients. Cancer. 2008;113(8):2020-2028. [PubMed 18780317]
  126. Huart A, Josse AG, Chauveau D, et al; French Society of Hemapheresis. Outcomes of patients with Goodpasture syndrome: a nationwide cohort-based study from the French Society of Hemapheresis. J Autoimmun. 2016;73:24-29. doi:10.1016/j.jaut.2016.05.015 [PubMed 27267459]
  127. Huguet F, Chevret S, Leguay T, et al; Group of Research on Adult ALL (GRAALL). Intensified therapy of acute lymphoblastic leukemia in adults: report of the randomized GRAALL-2005 clinical trial. J Clin Oncol. 2018;36(24):2514-2523. doi:10.1200/JCO.2017.76.8192 [PubMed 29863974]
  128. Huguet F, Leguay T, Raffoux E, et al. Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study. J Clin Oncol. 2009;27(6):911-918. doi:10.1200/JCO.2008.18.6916 [PubMed 19124805]
  129. 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]
  130. Iqubal A, Iqubal MK, Sharma S, et al. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: old drug with a new vision. Life Sci. 2019;218:112-131. doi:10.1016/j.lfs.2018.12.018 [PubMed 30552952]
  131. Ishida S, Doki N, Shingai N, et al. The clinical features of fatal cyclophosphamide-induced cardiotoxicity in a conditioning regimen for allogeneic hematopoietic stem cell transplantation (allo-HSCT). Ann Hematol. 2016;95(7):1145-1150. doi:10.1007/s00277-016-2654-6 [PubMed 27079957]
  132. Iwaki N, Fajgenbaum DC, Nabel CS, et al. Clinicopathologic analysis of TAFRO syndrome demonstrates a distinct subtype of HHV-8-negative multicentric Castleman disease. Am J Hematol. 2016;91(2):220-226. doi:10.1002/ajh.24242 [PubMed 26805758]
  133. Jabs DA, Rosenbaum JT, Foster CS, et al. Guidelines for the use of immunosuppressive drugs in patients with ocular inflammatory disorders: recommendations of an expert panel. Am J Ophthalmol. 2000;130(4):492-513. [PubMed 11024423]
  134. Jacobson C, Chavez JC, Sehgal AR, et al. Primary analysis of zuma-5: a phase 2 study of axicabtagene ciloleucel (Axi-Cel) in patients with relapsed/refractory (R/R) indolent non-hodgkin lymphoma (iNHL). Blood. 2020;136(suppl 1):40-41. [Abstract 623 from ASH 2020 annual meeting]. doi:10.1182/blood-2020-136834
  135. Jaime-Perez JC, Gonzalez-Llano O, Cantu-Rodriguez OG, et al. Long-term remission and bone marrow findings in children with severe aplastic anemia immunosuppressed with high doses of cyclophosphamide. Journal of Bone Marrow Research. 2013;1(1):1-4.
  136. Janus N, Thariat J, Boulanger H, Deray G, Launay-Vacher V. Proposal for Dosage Adjustment and Timing of Chemotherapy in Hemodialyzed Patients. Ann Oncol. 2010;21(7):1395-1403. doi: 10.1093/annonc/mdp598 [PubMed 20118214]
  137. Jayne D, Rasmussen N, Andrassy K, et al. A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med. 2003;349(1):36-44. [PubMed 12840090]
  138. Jha V, Ganguli A, Saha TK, et al. A randomized, controlled trial of steroids and cyclophosphamide in adults with nephrotic syndrome caused by idiopathic membranous nephropathy. J Am Soc Nephrol. 2007;18(6):1899-1904. doi:10.1681/ASN.2007020166 [PubMed 17494881]
  139. Jones JB, Purdy CY, Bailey RT Jr. Cyclophosphamide anaphylaxis. DICP. 1989;23(1):88-89. doi:10.1177/106002808902300125 [PubMed 2718490]
  140. Jones S, Holmes FA, O'Shaughnessy J, et al. Docetaxel with cyclophosphamide is associated with an overall survival benefit compared with doxorubicin and cyclophosphamide: 7-year follow-up of US Oncology Research Trial 9735. J Clin Oncol. 2009;27(8):1177-1183. doi:10.1200/JCO.2008.18.4028 [PubMed 19204201]
  141. Juma FD. Effect of liver failure on the pharmacokinetics of cyclophosphamide. Eur J Clin Pharmacol. 1984;26(5):591-593. doi:10.1007/BF00543491 [PubMed 6468474]
  142. Kanakry CG, O'Donnell PV, Furlong T, et al. Multi-institutional study of post-transplantation cyclophosphamide as single-agent graft-versus-host disease prophylaxis after allogeneic bone marrow transplantation using myeloablative busulfan and fludarabine conditioning. J Clin Oncol. 2014a;32(31):3497-505. doi:10.1200/JCO.2013.54.0625 [PubMed 25267759]
  143. Kanakry CG, Tsai HL, Bolaños-Meade J, et al. Single-agent GVHD prophylaxis with posttransplantation cyclophosphamide after myeloablative, HLA-matched BMT for AML, ALL, and MDS. Blood. 2014b;124(25):3817-3827. doi:10.1182/blood-2014-07-587477 [PubMed 25316679]
  144. 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]
  145. Kaplan AA, Appel GB, Pusey CD. Anti-GBM (Goodpasture) disease: treatment and prognosis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2021.
  146. KDIGO. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for glomerulonephritis. Am J Kidney Dis. 2013;62(3):403-441. [PubMed 23871408]
  147. Kelly KM, Sposto R, Hutchinson R, et al. BEACOPP Chemotherapy is a Highly Effective Regimen in Children and Adolescents With High-Risk Hodgkin lymphoma: A Report from the Children's Oncology Group. Blood. 2011;117(9):2596-2603. [PubMed 21079154]
  148. Kennedy R, Groepper D, Tagen M, et al. Stability of Cyclophosphamide in Extemporaneous Oral Suspensions. Ann Pharmacother. 2010, 44(2):295-301. [PubMed 20103616]
  149. Khan ML, Reeder CB, Kumar SK, et al. A Comparison of Lenalidomide/Dexamethasone versus Cyclophosphamide/Lenalidomide/Dexamethasone versus Cyclophosphamide/Bortezomib/Dexamethasone in Newly Diagnosed Multiple Myeloma. Br J Haematol. 2012;156(3):326-333. [PubMed 22107129]
  150. Khouri IF, McLaughlin P, Saliba RM, et al. Eight-year experience with allogeneic stem cell transplantation for relapsed follicular lymphoma after nonmyeloablative conditioning with fludarabine, cyclophosphamide, and rituximab. Blood. 2008;111(12):5530-5536. doi:10.1182/blood-2008-01-136242 [PubMed 18411419]
  151. Kidney Disease: Improving Global Outcomes (KDIGO). KDIGO clinical practice guideline for glomerulonephritis. Kidney Int Suppl. 2012;2(2):139-274. https://kdigo.org/wp-content/uploads/2017/02/KDIGO-2012-GN-Guideline-English.pdf
  152. King TE Jr. Treatment and prognosis of eosinophilic granulomatosis with polyangiitis (Churg-Strauss). Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 22, 2021.
  153. Kintzel PE, Dorr RT. Anticancer drug renal toxicity and elimination: dosing guidelines for altered renal function. Cancer Treat Rev. 1995;21(1):33-64. doi:10.1016/0305-7372(95)90010-1 [PubMed 7859226]
  154. Kliegman RM, Stanton BF, St. Gemell JW, et al, eds. Nelson Textbook of Pediatrics. 19th ed. Philadelphia, PA: Saunders Elsevier; 2011.
  155. Kondoh Y, Taniguchi H, Yokoi T, et al. Cyclophosphamide and low-dose prednisolone in idiopathic pulmonary fibrosis and fibrosing nonspecific interstitial pneumonia. Eur Respir J. 2005;25(3):528-533. doi:10.1183/09031936.05.00071004 [PubMed 15738299]
  156. Kowal-Bielecka O, Fransen J, Avouac J, et al; EUSTAR Coauthors. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76(8):1327-1339. doi:10.1136/annrheumdis-2016-209909 [PubMed 27941129]
  157. 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]
  158. Krmar RT, Kagebrand M, Hansson MEA, et al. Renal-limited vasculitis in children: a single-center retrospective long-term follow-up analysis. Clinical Nephrology. 2013;80:388-394. [PubMed 22541686]
  159. Kumar S, Flinn I, Richardson PG, et al. Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood. 2012a;119(19):4375-4382. [PubMed 22422823]
  160. Kumar SK, Hayman SR, Buadi FK, et al. Lenalidomide, cyclophosphamide, and dexamethasone (CRd) for light-chain amyloidosis: long-term results from a phase 2 trial. Blood. 2012b;119(21):4860-4867. doi:10.1182/blood-2012-01-407791 [PubMed 22504925]
  161. Kushner BH, Kramer K, Modak S, Qin LX, Cheung NK. Differential impact of high-dose cyclophosphamide, topotecan, and vincristine in clinical subsets of patients with chemoresistant neuroblastoma. Cancer. 2010;116(12):3054-3060. [PubMed 20564411]
  162. Kushner BH, Kramer K, Modak S, Yataghene K, Cheung NK. High-dose cyclophosphamide-irinotecan-vincristine for primary refractory neuroblastoma. Eur J Cancer. 2011;47(1):84-89 [PubMed 20934323]
  163. Kushner BH, LaQuaglia MP, Bonilla MA, et al. Highly Effective Induction Therapy for Stage 4 Neuroblastoma in Children Over 1 Year of Age. J Clin Oncol. 1994;12(12):2607-2613. [PubMed 7527454]
  164. Kushner BH, LaQuaglia MP, Wollner N, et al. Desmoplastic small round-cell tumor: prolonged progression-free survival with aggressive multimodality therapy. J Clin Oncol. 1996;14(5):1526-1531. [PubMed 8622067]
  165. Lacasce A, Howard O, Fisher D, et al. Modified Magrath regimens for adults with Burkitt and Burkitt-like lymphomas: preserved efficacy with decreased toxicity. Leuk Lymphoma. 2004;45(4):761-767. [PubMed 15160953]
  166. Lacy MQ, Kurtin PJ, Tefferi A. Pure red cell aplasia: association with large granular lymphocyte leukemia and the prognostic value of cytogenetic abnormalities. Blood. 1996;87(7):3000-3006. [PubMed 8639922]
  167. 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]
  168. Leblond V, Kastritis E, Advani R, et al. Treatment recommendations from the Eighth International Workshop on Waldenström's Macroglobulinemia. Blood. 2016;128(10):1321-1328. doi:10.1182/blood-2016-04-711234 [PubMed 27432877]
  169. Lee CK, Barlogie B, Munshi N, et al. DTPACE: an effective, novel combination chemotherapy with thalidomide for previously treated patients with myeloma. J Clin Oncol. 2003;21(14):2732-2739. doi:10.1200/JCO.2003.01.055 [PubMed 12860952]
  170. Lee S, Lee J, Khimji C, et al. Repeated low-dose acrolein triggers irreversible lamina propria edema in urinary bladder, transient voiding behavior and widening of eyes to mechanical stimuli. Cells. 2021;10(12):3477. doi:10.3390/cells10123477 [PubMed 34943985]
  171. Lehman TJ, Onel K. Intermittent Intravenous Cyclophosphamide Arrests Progression of the Renal Chronicity Index in Childhood Systemic Lupus Erythematosus. J Pediatr. 2000;136(2):243-247. [PubMed 10657833]
  172. Levine MN, Bramwell VH, Pritchard KI, et al. Randomized Trial of Intensive Cyclophosphamide, Epirubicin, and Fluorouracil Chemotherapy Compared With Cyclophosphamide, Methotrexate, and Fluorouracil in Premenopausal Women With Node-Positive Breast Cancer. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 1998;16(8):2651-2658. [PubMed 9704715]
  173. Levy JB, Turner AN, Rees AJ, Pusey CD. Long-term outcome of anti-glomerular basement membrane antibody disease treated with plasma exchange and immunosuppression. Ann Intern Med. 2001;134(11):1033-1042. doi:10.7326/0003-4819-134-11-200106050-00009 [PubMed 11388816]
  174. Lishner M, Avivi I, Apperley JF, et al. Hematologic malignancies in pregnancy: management guidelines from an international consensus meeting. J Clin Oncol. 2016;34(5):501-508. doi:10.1200/JCO.2015.62.4445 [PubMed 26628463]
  175. Liu AY, Nabel CS, Finkelman BS, et al. Idiopathic multicentric Castleman's disease: a systematic literature review. Lancet Haematol. 2016;3(4):e163-e175. doi:10.1016/S2352-3026(16)00006-5 [PubMed 27063975]
  176. Locatelli F, Nöllke P, Zecca M, et al. Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): results of the EWOG-MDS/EBMT trial. Blood. 2005;105(1):410-419. [PubMed 15353481]
  177. Locke FL, Ghobadi A, Jacobson CA, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20(1):31-42. doi:10.1016/S1470-2045(18)30864-7 [PubMed 30518502]
  178. Locke FL, Miklos DB, Jacobson CA, et al; All ZUMA-7 Investigators and Contributing Kite Members. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med. 2022;386(7):640-654. doi:10.1056/NEJMoa2116133 [PubMed 34891224]
  179. Loehrer PJ Sr, Kim K, Aisner SC, et al; The Eastern Cooperative Oncology Group, Southwest Oncology Group, and Southeastern Cancer Study Group. Cisplatin plus doxorubicin plus cyclophosphamide in metastatic or recurrent thymoma: final results of an intergroup trial. J Clin Oncol. 1994;12(6):1164-1168. doi:10.1200/JCO.1994.12.6.1164 [PubMed 8201378]
  180. Loibl S, Schmidt A, Gentilini O, et al. Breast cancer diagnosed during pregnancy: adapting recent advances in breast cancer care for pregnant patients. JAMA Oncol. 2015;1(8):1145-1153. doi: 10.1001/jamaoncol.2015.2413 [PubMed 26247818]
  181. Loibl S, von Minckwitz G, Gwyn K, et al. Breast carcinoma during pregnancy. International recommendations from an expert meeting. Cancer. 2006;106(2):237-246. [PubMed 16342247]
  182. Lunde LE, Dasaraju S, Cao Q, et al. Hemorrhagic cystitis after allogeneic hematopoietic cell transplantation: risk factors, graft source and survival. Bone Marrow Transplant. 2015;50(11):1432-1437. doi:10.1038/bmt.2015.162 [PubMed 26168069]
  183. Lurain JR, Singh DK, Schink JC. Primary treatment of metastatic high-risk gestational trophoblastic neoplasia with EMA-CO chemotherapy. J Reprod Med. 2006;51(10):767-772. [PubMed 17086804]
  184. Luznik L, Bolaños-Meade J, Zahurak M, et al. High-dose cyclophosphamide as single-agent, short-course prophylaxis of graft-versus-host disease. Blood. 2010;115(16):3224-3230. doi:10.1182/blood-2009-11-251595 [PubMed 20124511]
  185. 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. [PubMed 8622041]
  186. Mac S, Ngo D, Yang D, et al. Use of high-dose mesna and hyperhydration leads to lower incidence of hemorrhagic cystitis after posttransplant cyclophosphamide-based allogeneic transplantation. Bone Marrow Transplant. 2021;56(10):2464-2470. doi:10.1038/s41409-021-01364-0 [PubMed 34108676]
  187. Mak SK, Short CD, Mallick NP. Long-term outcome of adult-onset minimal-change nephropathy. Nephrol Dial Transplant. 1996;11(11):2192-2201. doi:10.1093/oxfordjournals.ndt.a027136 [PubMed 8941578]
  188. Majhail NS, Brazauskas R, Rizzo JD, et al. Secondary solid cancers after allogeneic hematopoietic cell transplantation using busulfan-cyclophosphamide conditioning. Blood. 2011;117(1):316-322. doi:10.1182/blood-2010-07-294629 [PubMed 20926773]
  189. Malik SW, Myers JL, DeRemee RA, et al. Lung toxicity associated with cyclophosphamide use. Two distinct patterns. Am J Respir Crit Care Med. 1996;154(6, pt 1):1851-1856. [PubMed 8970380]
  190. Malogolowkin M, Cotton CA, Green DM, et al. Treatment of Wilms tumor relapsing after initial treatment with vincristine, actinomycin D, and doxorubicin. A report from the National Wilms Tumor Study Group. Pediatr Blood Cancer. 2008;50(2):236-241. [PubMed 17539021]
  191. Malpica L, Moll S. Practical approach to monitoring and prevention of infectious complications associated with systemic corticosteroids, antimetabolites, cyclosporine, and cyclophosphamide in nonmalignant hematologic diseases. Hematology Am Soc Hematol Educ Program. 2020;2020(1):319-327. doi:10.1182/hematology.2020000116 [PubMed 33275674]
  192. Marcus R, Davies A, Ando K, et al. Obinutuzumab for the first-line treatment of follicular lymphoma. N Engl J Med. 2017;377(14):1331-1344. doi:10.1056/NEJMoa1614598 [PubMed 28976863]
  193. Marcus R, Imrie K, Belch A, et al. CVP chemotherapy plus rituximab compared with CVP as first-line treatment for advanced follicular lymphoma. Blood. 2005;105(4):1417-1423. doi:10.1182/blood-2004-08-3175 [PubMed 15494430]
  194. Marie I, Mouthon L. Therapy of polymyositis and dermatomyositis. Autoimmun Rev. 2011;11(1):6-13. doi:10.1016/j.autrev.2011.06.007 [PubMed 21740984]
  195. Mårtensson T1, Priftakis P, Casswall T, et al. Increased risk of gastrointestinal acute GVHD following the addition of melphalan to busulfan/cyclophosphamide conditioning. Pediatr Transplant. 2013;17(3):285-293. [PubMed 23489519]
  196. Martínez-Gabarrón M, Enríquez R, Sirvent AE, García-Sepulcre M, Millán I, Amorós F. Hepatotoxicity following cyclophosphamide treatment in a patient with MPO-ANCA vasculitis. Nefrologia. 2011;31(4):496-498. doi:10.3265/Nefrologia.pre2011.May.10917 [PubMed 21738257]
  197. Mason WP, Grovas A, Halpern S, et al. Intensive chemotherapy and bone marrow rescue for young children with newly diagnosed malignant brain tumors. J Clin Oncol. 1998;16(1):210-221. [PubMed 9440745]
  198. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. doi:10.1056/NEJMoa1709866 [PubMed 29385370]
  199. McCrindle BW, Rowley AH, Newburger JW, et al. Diagnosis, treatment, and long-term management of Kawasaki Disease: a scientific statement for health professionals from the American Heart Association. Circulation. 2017;135(17):e927-e999. [PubMed 28356445]
  200. McCune WJ, Clowse MB. General principles of the use of cyclophosphamide in rheumatic diseases. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 22, 2021.
  201. McDonald GB, Slattery JT, Bouvier ME, et al. Cyclophosphamide metabolism, liver toxicity, and mortality following hematopoietic stem cell transplantation. Blood. 2003;101(5):2043-2048. doi:10.1182/blood-2002-06-1860 [PubMed 12406916]
  202. Means RT Jr. Acquired pure red cell aplasia in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 20, 2022.
  203. Mecoli CA, Saylor D, Gelber AC, Christopher-Stine L. Pneumocystis jiroveci pneumonia in rheumatic disease: a 20-year single-centre experience. Clin Exp Rheumatol. 2017;35(4):671-673. [PubMed 28134084]
  204. Merkel PA. Treatment and prognosis of polyarteritis nodosa. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 17, 2021a.
  205. Merkel PA, Kaplan AA, Falk RJ. Granulomatosis with polyangiitis and microscopic polyangiitis: induction and maintenance therapy. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 6, 2021b.
  206. Meyrier A, Radhakrishnan J. Minimal change disease: treatment in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 20, 2022.
  207. Mikhael JR, Schuster SR, Jimenez-Zepeda VH, et al. Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis. Blood. 2012;119(19):4391-4394. doi:10.1182/blood-2011-11-390930 [PubMed 22331188]
  208. Mills KA, Chess-Williams R, McDermott C. Novel insights into the mechanism of cyclophosphamide-induced bladder toxicity: chloroacetaldehyde's contribution to urothelial dysfunction in vitro. Arch Toxicol. 2019;93(11):3291-3303. doi:10.1007/s00204-019-02589-1 [PubMed 31598736]
  209. Mina R, Scheven E, Ardoin SP, et al. Consensus treatment plans for induction therapy of newly diagnosed proliferative lupus nephritis in juvenile systemic lupus erythematosus. Arthritis Care Res (Hoboken). 2012;64(3):375-383. [PubMed 22162255]
  210. Ming Z, Yongqiang Z, Zijin Z, Yan X, Di C, Xiaoxin T. Severe and prolonged cyclophosphamide-induced hepatotoxicity in a breast cancer patient carrying a CYP2B6*7 variant. Pharmacogenomics. 2019;20(16):1119-1124. doi:10.2217/pgs-2019-0093 [PubMed 31686598]
  211. Moccia AA, Schaff K, Hoskins P, et al. R-CHOP with etoposide substituted for doxorubicin (R-CEOP): excellent outcome in diffuse large B cell lymphoma for patients with a contraindication to anthracyclines. Blood. 2009;114(22).
  212. Mok CC, Lau CS, Wong RW. Risk Factors for Ovarian Failure in Patients With Systemic Lupus Erythematosus Receiving Cyclophosphamide Therapy. Arthritis Rheum. 1998;41(5):831-837. [PubMed 9588734]
  213. Mok CC, To CH, Szeto ML. Successful treatment of dermatomyositis-related rapidly progressive interstitial pneumonitis with sequential oral cyclophosphamide and azathioprine. Scand J Rheumatol. 2003;32(3):181-183. doi:10.1080/03009740310002542 [PubMed 12892257]
  214. Moore MJ. Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet. 1991;20(3):194-208. doi:10.2165/00003088-199120030-00002 [PubMed 2025981]
  215. Morgan C, Tillett T, Braybrooke J, Ajithkumar T. Management of Uncommon Chemotherapy-Induced Emergencies. Lancet Oncol. 2011;12(8):806-814. doi:10.1016/S1470-2045(10)70208-4 [PubMed 21276754]
  216. Moshe Y, Bentur OS, Lishner M, Avivi I. The management of hodgkin lymphomas in pregnancies. Eur J Haematol. 2017;99(5):385-391. doi:10.1111/ejh.12956 [PubMed 28850718]
  217. Moyo VM, Smith D, Brodsky I, Crilley P, Jones RJ, Brodsky RA. High-dose cyclophosphamide for refractory autoimmune hemolytic anemia. Blood. 2002;100(2):704-706. doi: 10.1182/blood-2002-01-0087. [PubMed 12091370]
  218. Mukhtyar C, Guillevin L, Cid MC, et al; European Vasculitis Study Group. EULAR recommendations for the management of primary small and medium vessel vasculitis. Ann Rheum Dis. 2009;68(3):310-317. doi:10.1136/ard.2008.088096 [PubMed 18413444]
  219. Munshi NC, Anderson LD Jr, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 2021;384(8):705-716. doi:10.1056/NEJMoa2024850 [PubMed 33626253]
  220. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544. doi:10.1056/NEJMoa1707447 [PubMed 29226797]
  221. Nemecek BD, Hammond DA, eds. Demystifying Drug Dosing in Renal Dysfunction. American Society of Health-System Pharmacists; 2019.
  222. Oktay K, Harvey BE, Partridge AH, et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018;36(19):1994-2001. doi:10.1200/JCO.2018.78.1914 [PubMed 29620997]
  223. Ostermann-Kraljevic S, Berger M, Voeffray M, Pannatier A. Stability and compatibility studies of antineoplastic agents in glass and polyethylene containers. EHP. 1999;5(4):162-168.
  224. Page RL 2nd, O'Bryant CL, Cheng D, et al; American Heart Association Clinical Pharmacology and Heart Failure and Transplantation Committees of the Council on Clinical Cardiology; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular and Stroke Nursing; and Council on Quality of Care and Outcomes Research. Drugs That May Cause or Exacerbate Heart Failure: A Scientific Statement From the American Heart Association [published correction appears in Circulation. 2016;134(12):e261]. Circulation. 2016;134(6):e32-e69. [PubMed 27400984]
  225. Palladini G, Sachchithanantham S, Milani P, et al. A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis. Blood. 2015;126(5):612-615. doi:10.1182/blood-2015-01-620302 [PubMed 25987656]
  226. Park JR, Scott JR, Stewart CF, et al. Pilot induction regimen incorporating pharmacokinetically guided topotecan for treatment of newly diagnosed high-risk neuroblastoma: a Children's Oncology Group study. J Clin Oncol. 2011;29(33):4351-4357. [PubMed 22010014]
  227. Parker C, Waters R, Leighton C, et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet. 2010;376(9757):2009-2017. [PubMed 21131038]
  228. Patel JM. Metabolism and pulmonary toxicity of cyclophosphamide. Pharmacol Ther. 1990;47(1):137-146. doi:10.1016/0163-7258(90)90049-8 [PubMed 2195554]
  229. 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]
  230. Pavlov-Dolijanovic S, Vujasinovic Stupar N, Zugic V, et al. Long-term effects of immunosuppressive therapy on lung function in scleroderma patients. Clin Rheumatol. 2018;37(11):3043-3050. doi:10.1007/s10067-018-4266-0 [PubMed 30143960]
  231. 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]
  232. Peccatori FA, Azim HA Jr, Orecchia R, et al. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013;24(suppl 6):vi160-vi167. [PubMed 23813932]
  233. Pedrazzoli P, Silvestris N, Santoro A, et al. Management of patients with end-stage renal disease undergoing chemotherapy: recommendations of the Associazione Italiana di Oncologia Medica (AIOM) and the Società Italiana di Nefrologia (SIN). ESMO Open. 2017;2(3):e000167. doi:10.1136/esmoopen-2017-000167 [PubMed 29209521]
  234. Pfreundschuh M, Trümper L, Kloess M, et al; German High-Grade Non-Hodgkin's Lymphoma Study Group. Two-weekly or 3-weekly CHOP chemotherapy with or without etoposide for the treatment of young patients with good-prognosis (normal LDH) aggressive lymphomas: results of the NHL-B1 trial of the DSHNHL. Blood. 2004;104(3):626-633. doi:10.1182/blood-2003-06-2094 [PubMed 14982884]
  235. Pineda-Roman M, Zangari M, Haessler J, et al. Sustained complete remissions in multiple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2. Br J Haematol. 2008;140(6):625-634. doi:10.1111/j.1365-2141.2007.06921.x [PubMed 18302711]
  236. Ponticelli C, Edefonti A, Ghio L, et al. Cyclosporin versus cyclophosphamide for patients with steroid-dependent and frequently relapsing idiopathic nephrotic syndrome: a multicentre randomized controlled trial. Nephrol Dial Transplant. 1993;8(12):1326-1332. [PubMed 8159300]
  237. Procytox (cyclophosphamide) [product monograph]. Mississauga, Ontario, Canada: Baxter; September 2012.
  238. Pryor BD, Bologna SG, Kahl LE. Risk factors for serious infection during treatment with cyclophosphamide and high-dose corticosteroids for systemic lupus erythematosus. Arthritis Rheum. 1996;39(9):1475-1482. doi:10.1002/art.1780390906 [PubMed 8814058]
  239. Pujari SS, Kempen JH, Newcomb CW, et al. Cyclophosphamide for ocular inflammatory diseases. Ophthalmology. 2010;117(2):356-365. [PubMed 19969366]
  240. Ravandi F, O'Brien S, Thomas D, et al. First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia. Blood. 2010;116(12):2070-2077. doi:10.1182/blood-2009-12-261586 [PubMed 20466853]
  241. Reece DE, Barnett MJ, Connors JM, et al. Intensive chemotherapy with cyclophosphamide, carmustine, and etoposide followed by autologous bone marrow transplantation for relapsed Hodgkin's disease. J Clin Oncol. 1991;9(10):1871-1879. [PubMed 1919637]
  242. Reinhold-Keller E, Beuge N, Latza U, et al. An interdisciplinary approach to the care of patients with Wegener's granulomatosis: long-term outcome in 155 patients. Arthritis Rheum. 2000;43(5):1021-1032. doi:10.1002/1529-0131(200005)43:5<1021::AID-ANR10>3.0.CO;2-J [PubMed 10817555]
  243. Reiter A, Schrappe M, Tiemann M, et al. Successful treatment strategy for Ki-1 anaplastic large-cell lymphoma of childhood: a prospective analysis of 62 patients enrolled in three consecutive Berlin-Frankfurt-Munster group studies. J Clin Oncol. 1994;12(5):899-908. [PubMed 8164040]
  244. Regan MJ, Hellmann DB, Stone JH. Treatment of Wegener's granulomatosis. Rheum Dis Clin North Am. 2001;27(4):863-886, viii. doi:10.1016/s0889-857x(05)70240-4 [PubMed 11723769]
  245. Rizzieri DA, Johnson JL, Byrd JC, et al; Alliance for Clinical Trials In Oncology (ACTION). Improved efficacy using rituximab and brief duration, high intensity chemotherapy with filgrastim support for Burkitt or aggressive lymphomas: cancer and Leukemia Group B study 10 002. Br J Haematol. 2014;165(1):102-111. doi:10.1111/bjh.12736 [PubMed 24428673]
  246. Robak T, Dmoszynska A, Solal-Céligny P, et al. Rituximab Plus Fludarabine and Cyclophosphamide Prolongs Progression-Free Survival Compared With Fludarabine and Cyclophosphamide Alone in Previously Treated Chronic Lymphocytic Leukemia. J Clin Oncol. 2010;28(10):1756-1765. [PubMed 20194844]
  247. Robison NJ, Campigotto F, Chi SN, et al. A phase II trial of a multi-agent oral antiangiogenic (metronomic) regimen in children with recurrent or progressive cancer. Pediatr Blood Cancer. 2014;61(4):636-642. [PubMed 24123865]
  248. Rodríguez-Galindo C, Daw NC, Kaste SC, et al. Treatment of refractory osteosarcoma with fractionated cyclophosphamide and etoposide. J Pediatr Hematol Oncol. 2002;24(4):250-255. doi:10.1097/00043426-200205000-00006 [PubMed 11972091]
  249. 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]
  250. Rossi SE, Erasmus JJ, McAdams HP, Sporn TA, Goodman PC. Pulmonary drug toxicity: radiologic and pathologic manifestations. Radiographics. 2000;20(5):1245-1259. doi:10.1148/radiographics.20.5.g00se081245 [PubMed 10992015]
  251. Roumier M, Loustau V, Guillaud C, et al. Characteristics and outcome of warm autoimmune hemolytic anemia in adults: New insights based on a single-center experience with 60 patients. Am J Hematol. 2014;89(9):E150-E155. doi: 10.1002/ajh.23767. [PubMed 24847759]
  252. Rovin BH, Caster DJ, Cattran DC, et al; Conference Participants. Management and treatment of glomerular diseases (part 2): conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2019;95(2):281-295. doi:10.1016/j.kint.2018.11.008 [PubMed 30665569]
  253. Rowe JM, Buck G, Burnett AK, et al; MRC/NCRI Adult Leukemia Working Party. Induction therapy for adults with acute lymphoblastic leukemia: results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993. Blood. 2005;106(12):3760-3767. doi:10.1182/blood-2005-04-1623 [PubMed 16105981]
  254. Rubie H, De Bernardi B, Gerrard M, et al. Excellent outcome with reduced treatment in infants with nonmetastatic and unresectable neuroblastoma without MYCN amplification: results of the prospective INES 99.1. J Clin Oncol. 2011;29(4):449-455. [PubMed 21172879]
  255. Saba, Khan S, Parvez S, et al. Ellagic acid attenuates bleomycin and cyclophosphamide-induced pulmonary toxicity in Wistar rats. Food Chem Toxicol. 2013;58:210-219. doi:10.1016/j.fct.2013.03.046 [PubMed 23603381]
  256. Salama A. Treatment options for primary autoimmune hemolytic anemia: a short comprehensive review. Transfus Med Hemother. 2015;42(5):294-301. doi:10.1159/000438731 [PubMed 26696797]
  257. Sammaritano LR, Bermas BL, Chakravarty EE, et al. 2020 American College of Rheumatology guideline for the management of reproductive health in rheumatic and musculoskeletal diseases. Arthritis Rheumatol. 2020;72(4):529‐556. doi:10.1002/art.41191 [PubMed 32090480]
  258. Sampath S, Schultheiss TE, Wong J. Dose response and factors related to interstitial pneumonitis after bone marrow transplant. Int J Radiat Oncol Biol Phys. 2005;63(3):876-884. doi:10.1016/j.ijrobp.2005.02.032 [PubMed 16199317]
  259. Sasak G, Zemheri E, Ozkok A. Toxic epidermal necrolysis due to cyclophosphamide. Clin Exp Nephrol. 2016;20(4):660-661. doi:10.1007/s10157-015-1181-4 [PubMed 26475490]
  260. Sawada K, Fujishima N, Hirokawa M. Acquired pure red cell aplasia: updated review of treatment. Br J Haematol. 2008;142(4):505-514. doi:10.1111/j.1365-2141.2008.07216.x [PubMed 18510682]
  261. Schmitz N, Trümper L, Ziepert M, et al. Treatment and prognosis of mature T-cell and NK-cell lymphoma: an analysis of patients with T-cell lymphoma treated in studies of the German High-Grade Non-Hodgkin Lymphoma Study Group. Blood. 2010;116(18):3418-3425. doi:10.1182/blood-2010-02-270785 [PubMed 20660290]
  262. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45-56. doi:10.1056/NEJMoa1804980 [PubMed 30501490]
  263. Schwartz CL, Constine LS, Villaluna D, et al. A risk-adapted, response-based approach using ABVE-PC for children and adolescents with intermediate- and high-risk Hodgkin lymphoma: the results of P9425. Blood. 2009;114(10):2051-2059. [PubMed 19584400]
  264. Segura A, Yuste A, Cercos A, et al. Pulmonary fibrosis induced by cyclophosphamide. Ann Pharmacother. 2001;35(7-8):894-897. doi:10.1345/aph.10297 [PubMed 11485142]
  265. Seidemann K, Tiemann M, Schrappe M, et al. Short-pulse B-non-Hodgkin lymphoma-type chemotherapy is efficacious treatment for pediatric anaplastic large cell lymphoma: a report of the Berlin-Frankfurt-Münster Group Trial NHL-BFM 90. Blood. 2001;97(12):3699-3706. [PubMed 11389005]
  266. Seo HY, Kim EB, Kim JW, Shin BK, Kim SJ, Kim BS. Complete remission in a patient with human herpes virus-8 negative multicentric Castleman disease using CHOP chemotherapy. Cancer Res Treat. 2009;41(2):104-107. doi:10.4143/crt.2009.41.2.104 [PubMed 19707509]
  267. Shachar SS, Gallagher K, McGuire K, et al. Multidisciplinary management of breast cancer during pregnancy. Oncologist. 2017;22(3):324-334. doi:10.1634/theoncologist.2016-0208 [PubMed 28232597]
  268. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021;398(10299):491-502. doi:10.1016/S0140-6736(21)01222-8 [PubMed 34097852]
  269. Shaunak S, Munro JM, Weinbren K, Walport MJ, Cox TM. Cyclophosphamide-induced liver necrosis: a possible interaction with azathioprine. Q J Med. 1988;67(252):309-317. [PubMed 3060893]
  270. Simonini G, Cantarini L, Bresci C, Lorusso M, Galeazzi M, Cimaz R. Current therapeutic approaches to autoimmune chronic uveitis in children. Autoimmun Rev. 2010;9(10):674-683 [PubMed 20553975]
  271. Slayton RE, Park RC, Silverberg SG, et al. Vincristine, Dactinomycin, and Cyclophosphamide in the Treatment of Malignant Germ Cell Tumors of the Ovary. A Gynecologic Oncology Group Study (A Final Report). Cancer. 1985;56(2):243-248. [PubMed 2988740]
  272. Soussain C, Hoang-Xuan K, Taillandier L, et al; Société Française de Greffe de Moëlle Osseuse-Thérapie Cellulaire. Intensive chemotherapy followed by hematopoietic stem-cell rescue for refractory and recurrent primary CNS and intraocular lymphoma: Société Française de Greffe de Moëlle Osseuse-Thérapie Cellulaire. J Clin Oncol. 2008;26(15):2512-2518. doi:10.1200/JCO.2007.13.5533 [PubMed 18413641]
  273. Soussain C, Suzan F, Hoang-Xuan K, et al. Results of intensive chemotherapy followed by hematopoietic stem-cell rescue in 22 patients with refractory or recurrent primary CNS lymphoma or intraocular lymphoma. J Clin Oncol. 2001;19(3):742-749. doi:10.1200/JCO.2001.19.3.742 [PubMed 11157026]
  274. 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]
  275. Stentoft J. Progressive pulmonary fibrosis complicating cyclophosphamide therapy. Acta Med Scand. 1987;221(4):403-407. doi:10.1111/j.0954-6820.1987.tb03363.x [PubMed 3604756]
  276. 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]
  277. Stone JH, Merkel PA, Spiera R, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010;363(3):221-232. [PubMed 20647199]
  278. Storb R, Leisenring W, Anasetti C, et al. Long-term follow-up of allogeneic marrow transplants in patients with aplastic anemia conditioned by cyclophosphamide combined with antithymocyte globulin. Blood. 1997;89(10):3890-3891. [PubMed 9160700]
  279. Subramaniam SR, Cader RA, Mohd R, Yen KW, Ghafor HA. Low-dose cyclophosphamide-induced acute hepatotoxicity. Am J Case Rep. 2013;14:345-349. doi:10.12659/AJCR.889401 [PubMed 24023976]
  280. Tai PT, Yu E, Winquist E, et al. Chemotherapy in neuroendocrine/Merkel cell carcinoma of the skin: case series and review of 204 cases. J Clin Oncol. 2000;18(12):2493-2499. doi:10.1200/JCO.2000.18.12.2493 [PubMed 10856110]
  281. Talar-Williams C, Hijazi YM, Walther MM, et al. Cyclophosphamide-induced cystitis and bladder cancer in patients with Wegener granulomatosis. Ann Intern Med. 1996;124(5):477-484. doi:10.7326/0003-4819-124-5-199603010-00003 [PubMed 8602705]
  282. Tanaka F, Origuchi T, Migita K, et al. Successful combined therapy of cyclophosphamide and cyclosporine for acute exacerbated interstitial pneumonia associated with dermatomyositis. Intern Med. 2000;39(5):428-430. doi:10.2169/internalmedicine.39.428 [PubMed 10830189]
  283. Targoff IN. Treatment of recurrent and resistant dermatomyositis and polymyositis in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 22, 2021.
  284. Tashkin DP, Elashoff R, Clements PJ, et al; Scleroderma Lung Study Research Group. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med. 2006;354(25):2655-2666. doi:10.1056/NEJMoa055120 [PubMed 16790698]
  285. Tashkin DP, Roth MD, Clements PJ, et al; Sclerodema Lung Study II Investigators. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial. Lancet Respir Med. 2016;4(9):708-719. doi:10.1016/S2213-2600(16)30152-7 [PubMed 27469583]
  286. Tedeschi A, Benevolo G, Varettoni M, et al. Fludarabine plus cyclophosphamide and rituximab in Waldenstrom macroglobulinemia: an effective but myelosuppressive regimen to be offered to patients with advanced disease. Cancer. 2012;118(2):434-443. doi: 10.1002/cncr.26303. [PubMed 21732338]
  287. Teepen JC, van Leeuwen FE, Tissing WJ, et al. Long-term risk of subsequent malignant neoplasms after treatment of childhood cancer in the DCOG LATER study cohort: role of chemotherapy. J Clin Oncol. 2017;35(20):2288-2298. doi:10.1200/JCO.2016.71.6902 [PubMed 28530852]
  288. Thabet AF, Faisal M. Pulse cyclophosphamide therapy in refractory warm autoimmune hemolytic anemia: a new perspective. Indian J Hematol Blood Transfus. 2014;30(4):313-318. doi:10.1007/s12288-013-0290-z [PubMed 25435734]
  289. Thomas DA, Faderl S, Cortes J, et al. Treatment of Philadelphia chromosome-positive acute lymphocytic leukemia with hyper-CVAD and imatinib mesylate. Blood. 2004;103(12):4396-4407. doi:10.1182/blood-2003-08-2958 [PubMed 14551133]
  290. 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]
  291. Thompson JA, Fisher RI, Leblanc M, et al. Total body irradiation, etoposide, cyclophosphamide, and autologous peripheral blood stem-cell transplantation followed by randomization to therapy with interleukin-2 versus observation for patients with non-Hodgkin lymphoma: results of a phase 3 randomized trial by the Southwest Oncology Group (SWOG 9438). Blood. 2008;111(8):4048-4054. [PubMed 18256325]
  292. Thong BY, Leong KP, Thumboo J, Koh ET, Tang CY. Cyclophosphamide type I hypersensitivity in systemic lupus erythematosus. Lupus. 2002;11(2):127-129. doi:10.1191/0961203302lu140cr [PubMed 11958577]
  293. Travis LB, Gospodarowicz M, Curtis RE, et al. Lung cancer following chemotherapy and radiotherapy for Hodgkin's disease. J Natl Cancer Inst. 2002;94(3):182-192. doi:10.1093/jnci/94.3.182 [PubMed 11830608]
  294. Tsukasaki K, Utsunomiya A, Fukuda H, et al; Japan Clinical Oncology Group Study JCOG9801. VCAP-AMP-VECP compared with biweekly CHOP for adult T-cell leukemia-lymphoma: Japan Clinical Oncology Group Study JCOG9801. J Clin Oncol. 2007;25(34):5458-5464. doi:10.1200/JCO.2007.11.9958 [PubMed 17968021]
  295. Tutschka PJ, Copelan EA, Klein JP. Bone marrow transplantation for leukemia following a new busulfan and cyclophosphamide regimen. Blood. 1987;70(5):1382-1388. [PubMed 3311203]
  296. 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 October 5, 2016.
  297. van Rhee F, Voorhees P, Dispenzieri A, et al. International, evidence-based consensus treatment guidelines for idiopathic multicentric Castleman disease. Blood. 2018;132(20):2115-2124. doi:10.1182/blood-2018-07-862334 [PubMed 30181172]
  298. Varga J, Montesi S. Treatment and prognosis of interstitial lung disease in systemic sclerosis (scleroderma). Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed July 19, 2021.
  299. Venner CP, Lane T, Foard D, et al. Cyclophosphamide, bortezomib, and dexamethasone therapy in AL amyloidosis is associated with high clonal response rates and prolonged progression-free survival. Blood. 2012;119(19):4387-4390. doi:10.1182/blood-2011-10-388462 [PubMed 22331187]
  300. Verhelst D, Rossert J, Casadevall N, Krüger A, Eckardt KU, Macdougall IC. Treatment of erythropoietin-induced pure red cell aplasia: a retrospective study. Lancet. 2004;363(9423):1768-1771. doi:10.1016/S0140-6736(04)16302-2 [PubMed 15172775]
  301. von Pawel J, Schiller JH, Shepherd FA, et al. Topotecan versus cyclophosphamide, doxorubicin, and vincristine for the treatment of recurrent small-cell lung cancer. J Clin Oncol. 1999;17(2):658-667. [PubMed 10080612]
  302. Waldman M, Crew RJ, Valeri A, et al. Adult minimal-change disease: clinical characteristics, treatment, and outcomes. Clin J Am Soc Nephrol. 2007;2(3):445-453. doi:10.2215/CJN.03531006 [PubMed 17699450]
  303. Wallace CA, French JW, Kahn SJ, Sherry DD. Initial intravenous gammaglobulin treatment failure in Kawasaki disease. Pediatrics. 2000;105(6):E78. [PubMed 10835091]
  304. Walterhouse DO, Meza JL, Breneman JC, et al. Local control and outcome in children with localized vaginal rhabdomyosarcoma: a report from the Soft Tissue Sarcoma committee of the Children's Oncology Group. Pediatr Blood Cancer. 2011;57(1):76-83. doi: 10.1002/pbc.22928. [PubMed 21298768]
  305. Walterhouse DO, Pappo AS, Meza JL, et al. Shorter-duration therapy using vincristine, dactinomycin, and lower-dose cyclophosphamide with or without radiotherapy for patients with newly diagnosed low-risk rhabdomyosarcoma: a report from the Soft Tissue Sarcoma Committee of the Children's Oncology Group. J Clin Oncol. 2014;32(31):3547-3552. [PubMed 25267746]
  306. Wang CC, Weng TI, Lu MY, et al. Hemorrhagic cystitis in children treated with alkylating agent cyclophosphamide: the experience of a medical center in Taiwan. J Formos Med Assoc. 2015;114(8):691-697. doi:10.1016/j.jfma.2013.06.004 [PubMed 23880225]
  307. Wang LH, Lee CS, Majeske BL, Marbury TC. Clearance and recovery calculations in hemodialysis: application to plasma, red blood cells, and dialysate measurements for cyclophosphamide. Clin Pharmacol Ther. 1981;29(3):365-372. doi:10.1038/clpt.1981.50 [PubMed 7471607]
  308. Wang M, Munoz J, Goy A, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2020;382(14):1331-1342. doi:10.1056/NEJMoa1914347 [PubMed 32242358]
  309. Wynn RF, Cross MA, Hatton C, et al. Accelerated telomere shortening in young recipients of allogeneic bone-marrow transplants. Lancet. 1998;351(9097):178-181. doi:10.1016/S0140-6736(97)08256-1 [PubMed 9449873]
  310. Xie H, Griskevicius L, Stahle L, et al. Pharmacogenetics of Cyclophosphamide in Patients With Hematologic Malignancies. Eur J Pharm Sci. 2006;27(1):54-61. [PubMed 16183265]
  311. Xu Y, Wang H, Zhou S, et al. Risk of second malignant neoplasms after cyclophosphamide-based chemotherapy with or without radiotherapy for non-Hodgkin lymphoma. Leuk Lymphoma. 2013;54(7):1396-1404. doi:10.3109/10428194.2012.743657 [PubMed 23101661]
  312. Yamada O, Mizoguchi H, Oshimi K. Cyclophosphamide therapy for pure red cell aplasia associated with granular lymphocyte-proliferative disorders. Br J Haematol. 1997;97(2):392-399. doi:10.1046/j.1365-2141.1997.282672.x [PubMed 9163606]
  313. Yamasaki Y, Yamada H, Yamasaki M, et al. Intravenous cyclophosphamide therapy for progressive interstitial pneumonia in patients with polymyositis/dermatomyositis. Rheumatology (Oxford). 2007;46(1):124-130. doi:10.1093/rheumatology/kel112 [PubMed 16754626]
  314. Yeh J, Whited L, Saliba RM, et al. Cardiac toxicity after matched allogeneic hematopoietic cell transplant in the posttransplant cyclophosphamide era. Blood Adv. 2021;5(24):5599-5607. doi:10.1182/bloodadvances.2021004846 [PubMed 34592759]
  315. Yilmaz N, Emmungil H, Gucenmez S, et al. Incidence of cyclophosphamide-induced urotoxicity and protective effect of Mesna in rheumatic diseases. J Rheumatol. 2015;42(9):1661-1666. doi:10.3899/jrheum.150065 [PubMed 26178288]
  316. Zhang L, Zhao AL, Duan MH, et al. Phase 2 study using oral thalidomide-cyclophosphamide-prednisone for idiopathic multicentric Castleman disease. Blood. 2019;133(16):1720-1728. doi:10.1182/blood-2018-11-884577 [PubMed 30760451]
Topic 9308 Version 455.0