Page last updated: 2024-11-07

clofarabine

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Clofarabine is a nucleoside analog that is used as a chemotherapy agent for the treatment of acute lymphoblastic leukemia. Its synthesis involves the modification of the purine base guanine. Clofarabine is a prodrug that is converted to its active form, 2'-deoxycoformycin, which inhibits adenosine deaminase. This inhibition leads to the accumulation of deoxyadenosine, which is toxic to leukemia cells. Clofarabine is studied due to its potent anti-leukemic activity and its ability to overcome resistance to other chemotherapy drugs. Its importance lies in its ability to improve survival rates and reduce the risk of relapse in patients with acute lymphoblastic leukemia.'

Cross-References

ID SourceID
PubMed CID119182
CHEMBL ID1750
CHEBI ID681569
CHEBI ID47311
SCHEMBL ID9040
MeSH IDM0186928

Synonyms (100)

Synonym
AC-274
HY-A0005
AB00430247-03
AB00430247-04
clofarabine ,
123318-82-1
clolar
evoltra
2-cl-2'-f-araa
cafda
clofarex
sar-393590
9h-purin-6-amine, 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-
clofarabine [usan]
c1-f-ara-a
2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-9h-purin-6-amine
2-chloro-9-(2-deoxy-2-fluoro-b-d-arabinofuranosyl)-9h-purin-6-amine
2-chloro-9-(2-deoxy-2-fluoro-b -d-arabinofuranosyl)-9h-purin-6-amine
clolar (tn)
D03546
clofarabine (jan/usan/inn)
2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)adenine
2-chloro-9-(2'-deoxy-2'-fluoro-beta-d-arabinofuranosyl)adenine
DB00631
(2r,3r,4s,5r)-5-(6-amino-2-chloropurin-9-yl)-4-fluoro-2-(hydroxymethyl)oxolan-3-ol
cl-f-ara-a
HMS2090A07
clofarabinum
CHEBI:681569 ,
clofarabina
CHEMBL1750
nsc-759857
AKOS005063562
2-chloro-2'-fluoroarabino-2'-deoxyadenosine
2-chloro-9-(2-deoxy-2-fluoro-beta-d-arbinofuranosyl)adenine
2-chloro-2'-arabino-fluoro-2'-deoxyadenosine
2-chloro-9-(2-deoxy-2-fluoroarabinofuranosyl)adenine
dtxsid5046437 ,
dtxcid3026437
tox21_112182
cas-123318-82-1
BCP9000540
nsc 759857
clofarabine [usan:inn:ban]
762rdy0y2h ,
unii-762rdy0y2h
AKOS015919355
c10h11clfn5o3
smr002530055
MLS003915615
clofarabine,clolar, evoltra
BCP0726000280
2-chloro-9-(2-deoxy-2-fluoro-.beta.-d-arabinofuranosyl)-9h-purin-6-amine
clofarabine [orange book]
9h-purin-6-amine, 2-chloro-9-(2-deoxy-2-fluoro-.beta.-d-arabinofuranosyl)
clofarabine [ema epar]
clofarabine [mi]
clofarabine [who-dd]
clofarabine [inn]
clofarabine [jan]
clofarabine [vandf]
clofarabine [mart.]
CS-0373
S1218
gtpl6802
cl-f-araa
SCHEMBL9040
tox21_112182_1
NCGC00164553-02
(2r,3r,4s,5r)-5-(6-amino-2-chloro-purin-9-yl)-4-fluoro-2-(hydroxymethyl)tetrahydrofuran-3-ol
2-chloro-9-(2-deoxy-2-fluoro-?-d-arabinofuranosyl)-9h-purin-6-amine
(2r,3r,4s,5r)-5-(6-amino-2-chloro-9h-purin-9-yl)-4-fluoro-2-(hydroxymethyl)tetrahydrofuran-3-ol
AB00430247_06
(2r,3r,4s,5r)-5-(6-amino-2-chloro-9h-purin-9-yl)-4-fluoro-2-(hydroxymethyl)oxolan-3-ol
clofarabine, >=98% (hplc)
SR-01000930565-3
sr-01000930565
clofarabine; clofarex; clolar; evoltra; 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-9h-purin-6-amine; 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)adenine
9h-purine-6-amine,2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-
WDDPHFBMKLOVOX-AYQXTPAHSA-N
SW218080-2
bdbm50247921
Q5134875
Z1551900487
HMS3677J15
2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-adenine
AS-12958
BCP23422
HMS3413J15
CCG-264865
2 chloro 2' fluoroarabino 2' deoxyadenosine
2 chloro 2' arabino fluoro 2' deoxyadenosine
(2r,3r,4s,5r)-5-(6-amino-2-chloropurin-9-yl)-4-fluoro-2-(hydroxymethyl)oxolan-3-ol 2-chloro-2''-arabino-fluoro-2''-deoxyadenosine
NCGC00164553-05
clofarabin
clofarabine (mart.)
9h-purin-6-amine, 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)
chebi:47311
l01bb06
EN300-123437

Research Excerpts

Overview

Clofarabine (CLO) is a nucleoside analog with efficacy in relapsed/refractory acute lymphoblastic leukemia (ALL) ClofarABine is an active antileukemic drug for subgroups of patients with acute myeloid leukemia (AML) Clo is an immunosuppressive purine analog that may have better anti-leukemic activity than fludarabines.

ExcerptReferenceRelevance
"Clofarabine (CLO) is a nucleoside analog with efficacy in relapsed/refractory acute lymphoblastic leukemia (ALL). "( Clofarabine added to intensive treatment in adult patients with newly diagnosed ALL: the HOVON-100 trial.
Bakkus, M; Beel, K; Bellido, M; Beverloo, HB; Biemond, BJ; Boersma, RS; Breems, DA; Cornelissen, JJ; de Haas, V; de Weerdt, O; Deeren, D; Fijnheer, R; Gadisseur, A; Halkes, CJM; Havelange, V; Homburg, CHE; Jaspers, A; Legdeur, MC; Legrand, O; Maertens, JA; Mazure, D; Rijneveld, AW; Selleslag, D; Sinnige, HAM; Terpstra, WE; Tick, LW; Triffet, A; van de Loosdrecht, AA; van der Holt, B; van der Velden, VHJ; van der Velden, WJFM; van der Wagen, LE; van Gelder, M; van Lammeren-Venema, D; van Sluis, GL, 2022
)
3.61
"Clofarabine is an active antileukemic drug for subgroups of patients with acute myeloid leukemia (AML). "( The added value of multi-state modelling in a randomized controlled trial: The HOVON 102 study re-analyzed.
Bakunina, K; Breems, DA; Cloos, J; Cornelissen, JJ; de Wreede, LC; Gjertsen, BT; Koster, EAS; Löwenberg, B; Manz, MG; Ossenkoppele, GJ; Pabst, T; Passweg, J; Putter, H; Valk, PJM; van der Holt, B; Versluis, J, 2022
)
2.16
"Clofarabine (Clo) is an immunosuppressive purine analog that may have better anti-leukemic activity than fludarabine (Flu). "( Clofarabine and total body irradiation (TBI) as conditioning regimen for allogeneic stem cell transplantation in high-risk acute leukemia patients: a two-center retrospective cohort study.
Abou Dalle, I; Atoui, A; Bazarbachi, A; Bidaoui, G; Dulery, R; El Cheikh, J; Mohty, M; Moukalled, N; Sharrouf, L; Terro, K; Zahreddine, A, 2023
)
3.8
"Clofarabine is a purine nucleoside analog with mechanistic overlap with fludarabine; however, toxicity is high in the upfront leukemia setting, and thus use as an LD pre-CAR should be pursued with caution."( Rational Alternatives to Fludarabine and Cyclophosphamide-Based Pre-CAR Lymphodepleting Regimens in the Pediatric and Young Adult B-ALL Setting.
Green, S; Schultz, L, 2023
)
1.63
"Clofarabine is a nucleoside analog used to treat relapsed or refractory ALL."( Testicular disposition of clofarabine in rats is dependent on equilibrative nucleoside transporters.
Cherrington, NJ; Galligan, JJ; Hau, RK; Jennings, EQ; Jilek, JL; McGrath, ME; Miller, SR; Wright, SH, 2021
)
1.64
"Clofarabine is a second-generation deoxyadenosine analog rationally synthesized to improve stability and reduce toxicity."( Clofarabine exerts antileukemic activity against cytarabine-resistant B-cell precursor acute lymphoblastic leukemia with low deoxycytidine kinase expression.
Abe, M; Akahane, K; Goi, K; Goto, H; Huang, M; Inukai, T; Iwamoto, S; Kagami, K; Minegishi, M; Miyake, K; Oshiro, H; Shinohara, T; Somazu, S; Sugihara, E; Sugita, K; Tanaka, Y; Watanabe, A, 2018
)
2.64
"Clofarabine is a second generation nucleoside analogue which has shown promising activity in elderly patients with AML."( Clofarabine in the treatment of elderly patients with acute myeloid leukemia.
Aleem, A; Algahtani, F; Almomen, A; Alsaleh, K; Anjum, F; Iqbal, Z, 2013
)
2.55
"Clofarabine is a second generation nucleoside analogue with activity in adults with acute myeloid leukemia (AML). "( Clofarabine, idarubicin, and cytarabine (CIA) as frontline therapy for patients ≤60 years with newly diagnosed acute myeloid leukemia.
Andreeff, M; Borthakur, G; Brandt, M; Choi, S; Cortes, J; Daver, N; Du, M; Estrov, Z; Faderl, S; Ferrajoli, A; Garcia-Manero, G; Huang, X; Jabbour, E; Kadia, T; Kantarjian, H; Konopleva, M; Kornblau, S; Nazha, A; Pemmaraju, N; Ravandi, F, 2013
)
3.28
"Clofarabine is a novel purine nucleoside analogue with immunosuppressive and anti-leukemic activity in acute lymphoblastic and myeloid leukemia (AML, ALL). "( Feasibility of clofarabine cytoreduction followed by haploidentical hematopoietic stem cell transplantation in patients with relapsed or refractory advanced acute leukemia.
Bücklein, V; Engel, N; Fritsch, S; Hausmann, A; Hill, W; Hubmann, M; Ledderose, G; Prevalsek, D; Schulz, C; Stemmler, HJ; Tischer, J; Zoellner, AK, 2013
)
2.19
"Clofarabine is a purine nucleoside analog indicated for treatment of relapsed or refractory acute lymphoblastic leukaemia in children. "( Clofarabine-induced kidney toxicity.
Allen, SL; Chidella, S; Fishbane, S; Jhaveri, KD, 2014
)
3.29
"Clofarabine is a second-generation purine nucleoside analog approved in 2004 for the treatment of pediatric patients with relapsed or refractory acute lymphocytic leukemia (ALL) following failure of at least two prior regimens. "( Clofarabine in the treatment of myelodysplastic syndromes.
Bryan, J; Jabbour, E; Kantarjian, H; Prescott, H, 2014
)
3.29
"Clofarabine is a nucleoside analogue with activity in children with acute lymphoblastic leukemia (ALL). "( Phase I and extension study of clofarabine plus cyclophosphamide in patients with relapsed/refractory acute lymphoblastic leukemia.
Balakrishnan, K; Borthakur, G; Burger, J; Cortes, J; Faderl, S; Feliu, J; Ferrajoli, A; Gandhi, V; Kadia, T; Kantarjian, HM; O'Brien, S; Plunkett, W; Ravandi, F; Thomas, DA, 2014
)
2.13
"Clofarabine triphosphate is an intracellular active metabolite of clofarabine. "( A sensitive LC-MS/MS method for quantifying clofarabine triphosphate concentrations in human peripheral blood mononuclear cells.
Chen, X; Lu, Y; Tu, X; Zhang, Y; Zhong, D, 2014
)
2.11
"Clofarabine is an effective drug in relapsed leukemia and lymphoma that has some adverse effects which can be fatal like capillary leak syndrome (CLS). "( Clofarabine associated capillary leak syndrome in a child with lymphoma successfully treated with intravenous immunoglobulin.
Atas, E; Babacan, O; Kesik, V; Korkmazer, N,
)
3.02
"The clofarabine-based regimen is a promising strategy to induce disease remission in r/rALL and bridge to HSCT."( Refractory acute lymphoblastic leukemia in Chinese children: bridging to stem cell transplantation with clofarabine, cyclophosphamide and etoposide.
Chiang, AK; Ha, SY; Lee, V; Li, CK; Liu, AP, 2016
)
1.13
"Clofarabine is an FDA approved ribonucleotide reductase inhibitor, which has shown potent antiretroviral activity in transformed cell lines."( Dual anti-HIV mechanism of clofarabine.
Bonnac, L; Clouser, CL; Daly, MB; Kim, B; Maldonado, JO; Mansky, LM; Patterson, SE; Roth, ME; Xie, J, 2016
)
1.45
"Clofarabine is a potent HIV-1 inhibitor in both activated CD4(+) T cells and macrophages. "( Dual anti-HIV mechanism of clofarabine.
Bonnac, L; Clouser, CL; Daly, MB; Kim, B; Maldonado, JO; Mansky, LM; Patterson, SE; Roth, ME; Xie, J, 2016
)
2.17
"Clofarabine is a nucleoside analog with activity in adult AML."( Clofarabine combinations as acute myeloid leukemia salvage therapy.
Borthakur, G; Estrov, Z; Faderl, S; Ferrajoli, A; Huang, X; Kantarjian, HM; Kwari, M; Ravandi, F; Verstovsek, S; Wierda, W, 2008
)
2.51
"Clofarabine is a next generation nucleoside analog which is under clinical investigation."( Bone marrow CFU-GM and human tumor xenograft efficacy of three antitumor nucleoside analogs.
Bagley, RG; Crawford, J; Krumbholz, R; Kurtzberg, LS; Lovett, D; Roth, S; Rouleau, C; Schmid, S; Teicher, BA; Yao, M, 2009
)
1.07
"Clofarabine is a second-generation nucleoside analogue. "( Comparison of clofarabine activity in childhood and adult acute leukemia: individual tumor response study.
Badowska, W; Balcerska, A; Balwierz, W; Derwich, K; Gil, L; Komarnicki, M; Kowalczyk, J; Krawczuk-Rybak, M; Matysiak, M; Sobol, G; Sonta-Jakimczyk, D; Stefaniak, J; Styczynski, J; Urasinski, T; Wachowiak, J; Wieczorek, M; Wysocki, M, 2009
)
2.16
"Clofarabine is a rationally designed, second-generation deoxyadenosine analog that incorporates characteristics of two other purine analogs, fludarabine and cladribine. "( Clofarabine: a new treatment option for patients with acute myeloid leukemia.
Larson, ML; Venugopal, P, 2009
)
3.24
"Clofarabine is a second generation of purine nucleoside analogues designed to combine the most favorable pharmacokinetic properties of fludarabine and cladribine. "( Clofarabine as a novel nucleoside analogue approved to treat patients with haematological malignancies: mechanism of action and clinical activity.
Korycka, A; Lech-Maranda, E; Robak, T, 2009
)
3.24
"Clofarabine is a second-generation purine nucleoside analogue. "( Clofarabine: emerging role in leukemias.
Borthakur, G; Kantarjian, H; Sampat, K, 2009
)
3.24
"Clofarabine is a second generation nucleoside analogue. "( Clofarabine in the treatment of poor risk acute myeloid leukaemia.
Ansar, N; Hayad, A; Hayden, P; Krawczyk, J; MacDonagh, B; Meenaghan, T; Murphy, T; Murray, M; O'Dwyer, M; Swords, R, 2010
)
3.25
"Clofarabine is a novel purine analog approved in 2005 for treating children in second or greater ALL relapse."( Recent progress in the treatment of acute lymphoblastic leukemia: clofarabine.
Jeha, S, 2009
)
1.31
"Clofarabine is an active agent with acceptable toxicity in patients age 60 years or older with untreated AML who have at least one unfavorable prognostic factor. "( Phase II study of clofarabine monotherapy in previously untreated older adults with acute myeloid leukemia and unfavorable prognostic factors.
Abichandani, R; Arellano, M; Claxton, D; Craig, M; Douer, D; Eckert, S; Erba, HP; Faderl, S; Gabrilove, J; Kantarjian, HM; Kovascovics, T; Lyons, RM; Maris, M; Petersdorf, S; Shami, PJ; Yeager, AM, 2010
)
2.14
"Clofarabine is an effective therapy of pediatric patients with relapsed acute lymphoblastic leukemia (ALL). "( Fatal neurotoxicity in a patient with down syndrome treated with chemotherapy, irradiation, stem cell transplant, and clofarabine.
Bains, T; Halton, J; Johnston, DL; Klaassen, R; Mandel, K, 2010
)
2.01
"Clofarabine for injection is a second-generation nucleoside analog approved in the United States (Clolar(®)) and Europe (Evoltra(®)) for the treatment of pediatric relapsed or refractory acute lymphoblastic leukemia. "( Population pharmacokinetics of clofarabine and its metabolite 6-ketoclofarabine in adult and pediatric patients with cancer.
Bonate, PL; Cunningham, CC; Gaynon, P; Jeha, S; Kadota, R; Lam, GN; Razzouk, B; Rytting, M; Steinherz, P; Weitman, S, 2011
)
2.1
"Clofarabine is a second-generation purine nucleoside analogue that has been synthesized to overcome the limitations and incorporate the best qualities of fludarabine and cladribine. "( Clofarabine in leukemia.
Faderl, S; Ghandhi, V; Ghanem, H; Jabbour, E; Kantarjian, H; Plunkett, W, 2010
)
3.25
"Clofarabine is a purine nucleoside analog that works primarily via inhibition of DNA biosynthesis and the ribonucleotide reductase enzyme with recent evidence suggesting that at low doses it may affect DNA methylation."( Clofarabine for myelodysplastic syndromes.
Sekeres, MA; Tiu, RV; Traina, F, 2011
)
2.53
"Clofarabine is a second-generation purine nucleoside analogue, which has shown promising activity in relapsed and refractory paediatric acute lymphoblastic leukaemia (ALL). "( Early UK experience in the use of clofarabine in the treatment of relapsed and refractory paediatric acute lymphoblastic leukaemia.
Caswell, M; Connor, P; Cummins, M; Kearns, PR; Mitchell, C; Motwani, J; O'Connor, D; Sibson, K; Taj, M; Vora, A; Wynn, R, 2011
)
2.09
"Clofarabine is a nucleoside analog with activity in myeloid malignancies. "( A randomized study of 2 dose levels of intravenous clofarabine in the treatment of patients with higher-risk myelodysplastic syndrome.
Borthakur, G; Byrd, AL; Cortes, J; Estrov, Z; Faderl, S; Gandhi, V; Garcia-Manero, G; Jabbour, E; Kantarjian, HM; Kwari, M; Ravandi, F, 2012
)
2.07
"Clofarabine is a novel purine nucleoside analog with immunosuppressive and antileukemia activity. "( A phase I study in adults of clofarabine combined with high-dose melphalan as reduced-intensity conditioning for allogeneic transplantation.
Chen, R; Conrad, J; Dagis, A; Delioukina, M; Forman, SJ; Frankel, P; Kirschbaum, MH; Lacey, SF; Nademanee, A; Nakamura, R; Popplewell, L; Snyder, D; Stein, AS, 2012
)
2.11
"Clofarabine is a promising new chemotherapeutic agent that is active in the treatment of pediatric acute leukemia. "( Clofarabine, cyclophosphamide and etoposide for the treatment of relapsed or resistant acute leukemia in pediatric patients.
Barisone, E; Bertorello, N; Calvillo, M; De Rossi, G; Dufour, C; Lo Nigro, L; Luciani, M; Messina, C; Miano, M; Micalizzi, C; Parasole, R; Petruzziello, F; Pistorio, A; Putti, MC; Varotto, S; Ziino, O, 2012
)
3.26
"Clofarabine is a second-generation purine nucleoside analog and has significant anti-leukemic activity as a single agent. "( Clofarabine in pediatric acute leukemia: current findings and issues.
Arceci, RJ; Barry, E; Hijiya, N, 2012
)
3.26
"Clofarabine (ClF) is a drug used in the treatment of leukemia. "( Clofarabine targets the large subunit (α) of human ribonucleotide reductase in live cells by assembly into persistent hexamers.
Asturias, FJ; Aye, Y; Brignole, EJ; Chittuluru, J; Drennan, CL; Long, MJ; Stubbe, J, 2012
)
3.26
"Clofarabine is a second-generation purine nucleoside analog that has been synthesized to overcome the limitations and incorporate the best qualities of fludarabine and cladribine. "( The role of clofarabine in acute myeloid leukemia.
Ghanem, H; Jabbour, E; Kantarjian, H; Ohanian, M, 2013
)
2.21
"Clofarabine is a purine nucleoside antimetabolite under development by Bioenvision (under license from the Southern Research Institute) and ILEX for the potential treatment of solid tumors, acute myelogenous leukemia, non-Hodgkin's lymphoma, and acute lymphoblastic and chronic lymphocytic leukemia. "( Clofarabine. Bioenvision/ILEX.
Sternberg, A, 2003
)
3.2
"Clofarabine [Clofarex] is a purine nucleoside in development with Bio-envision, the Southern Research Institute and ILEX Oncology as an anticancer agent. "( Clofarabine.
, 2004
)
3.21
"As clofarabine is a potent inhibitor of ribonucleotide reductase (RnR), we hypothesized that clofarabine will modulate ara-c triphosphate accumulation and increase the antileukemic activity of cytarabine (ara-C)."( Results of a phase 1-2 study of clofarabine in combination with cytarabine (ara-C) in relapsed and refractory acute leukemias.
Beran, M; Bonate, P; Cortes, J; Du, M; Estey, E; Estrov, Z; Faderl, S; Ferrajoli, A; Gandhi, V; Garcia-Manero, G; Giles, FJ; Kantarjian, H; Keating, M; Kwari, M; O'Brien, S; Plunkett, W; Wierda, W, 2005
)
1.13
"Clofarabine is a deoxyadenosine analog synthesized with the intention of retaining the favorable mechanistic properties of fludarabine and cladribine while eliminating their undesirable characteristics. "( Clofarabine in adult acute leukemias: clinical success and pharmacokinetics.
Cooper, T; Gandhi, V; Kantarjian, H; Plunkett, W, 2004
)
3.21
"Clofarabine is a new-generation nucleoside analog that has been synthesized to combine the most favorable pharmacokinetic properties of its congeners fludarabine and cladribine. "( The role of clofarabine in hematologic and solid malignancies--development of a next-generation nucleoside analog.
Faderl, S; Gandhi, V; Jeha, S; Kantarjian, HM; Keating, MJ; Plunkett, W, 2005
)
2.15
"Clofarabine is a purine nucleoside analog that inhibits DNA synthesis and repair. "( Clofarabine: in pediatric patients with acute lymphoblastic leukemia.
Curran, MP; Perry, CM, 2005
)
3.21
"Clofarabine is a new nucleoside analog that has demonstrated clinical benefit in phase I-II studies, and is currently being studied in children and adults with leukemias and has been approved for the treatment of children with relapsed or refractory acute lymphocytic leukemia."( Clofarabine induced durable complete remission in heavily pretreated adolescents with relapsed and refractory leukemia.
Jeha, S; Meyers, PA; Steinherz, LJ; Steinherz, PG, 2007
)
2.5
"Clofarabine is a good generation purine nucleoside analogue designed to overcome the limitations and to incorporate the best qualities of both cladribine and fludarabine. "( Clofarabine: past, present, and future.
Faderl, S; Gandhi, V; Jeha, S; Kantarjian, HM; Wess, M, 2007
)
3.23
"Clofarabine is a novel nucleoside analog that has been associated with significant clinical activity in relapsed pediatric B-ALL."( Efficacy of low dose clofarabine in refractory precursor T- acute lymphoblastic leukemia.
Choi, J; Foss, F, 2006
)
1.37
"Clofarabine acts as a powerful radiosensitizer both in vitro and in vivo by interfering with the DNA damage response."( Clofarabine acts as radiosensitizer in vitro and in vivo by interfering with DNA damage response.
Cariveau, MJ; Cui, XL; Secrist, JA; Stackhouse, M; Tiwari, K; Waud, W; Xu, B, 2008
)
3.23

Effects

Clofarabine has shown activity and tolerability in older patients with acute myeloid leukemia (AML) The drug has potent antileukemia activity and its inclusion in reduced-intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation for acute leukemia could potentially improve outcomes.

ExcerptReferenceRelevance
"Clofarabine has been shown to effectively induce remission in children with refractory leukemia. "( The combination of clofarabine, etoposide, and cyclophosphamide shows limited efficacy as a bridge to transplant for children with refractory acute leukemia: results of a monitored prospective study.
Dykes, J; Håkansson, Y; Król, L; Pronk, C; Toporski, J; Turkiewicz, D, 2021
)
2.39
"Clofarabine has recently been evaluated as part of the conditioning regimen for allogeneic hematopoietic stem cell transplantation (HCT) in children. "( Population pharmacokinetics of clofarabine for allogeneic hematopoietic cell transplantation in paediatric patients.
Bierings, M; Boelens, JJ; Huitema, ADR; Lindemans, CA; Nierkens, S; Nijstad, AL; van der Elst, KCM; Versluys, AB, 2021
)
2.35
"Clofarabine has antileukemic activity with an acceptable toxicity profile."( Haploidentical Donor Transplantation Using a Novel Clofarabine-containing Conditioning Regimen for Very High-risk Hematologic Malignant Neoplasms.
Cross, SJ; Geiger, T; Inaba, H; Jeha, S; Kang, G; Sharma, A; Sunkara, A; Triplett, B, 2018
)
1.45
"Clofarabine has activity against LCH, JXG, and RDD in heavily pretreated patients, but prospective multi-center trials are warranted to determine long-term efficacy, optimal dosing, and late toxicity of clofarabine in this population."( Clofarabine salvage therapy in refractory multifocal histiocytic disorders, including Langerhans cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman disease.
Allen, CE; Beaupin, LK; Bilgi, M; Coulter, D; Garrington, T; Hicks, MJ; Jones, J; McCavit, TL; McClain, KL; Moore, C; Rivera-Ortegón, F; Shaffer, L; Simko, SJ; Stork, L; Tran, HD; Turcotte, L; Welsh, EC, 2014
)
3.29
"Clofarabine has shown activity and tolerability in older patients with acute myeloid leukemia (AML). "( Phase I study of oral clofarabine consolidation in adults aged 60 and older with acute myeloid leukemia.
Abboud, CN; Bernabe, N; Cashen, A; Dipersio, JF; Jacoby, MA; Luo, J; Martin, MG; Reineck, T; Stockerl-Goldstein, K; Uy, GL; Vij, R; Westervelt, P, 2014
)
2.16
"Clofarabine has potent antileukemia activity and its inclusion in reduced-intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation (HSCT) for acute leukemia could potentially improve outcomes. "( Phase II Trial of Reduced-Intensity Busulfan/Clofarabine Conditioning with Allogeneic Hematopoietic Stem Cell Transplantation for Patients with Acute Myeloid Leukemia, Myelodysplastic Syndromes, and Acute Lymphoid Leukemia.
Alyea, E; Ballen, KK; Chen, YB; Cutler, C; Dey, BR; Driscoll, J; El-Jawahri, A; Ho, VT; Hunnewell, C; Li, S; McAfee, SL; Poliquin, C; Saylor, M; Soiffer, RJ; Spitzer, TR, 2016
)
2.14
"Clofarabine has shown efficacy in selected pediatric leukemias."( Clofarabine: emerging role in leukemias.
Borthakur, G; Kantarjian, H; Sampat, K, 2009
)
2.52
"Clofarabine has been safely and effectively combined with other agents and will probably become an integral part of induction and/or consolidation regimens in AML."( Clofarabine for the treatment of adult acute myeloid leukemia.
Cannas, G; Dombret, H; Elhamri, M; Lobe, I; Raffoux, E; Thomas, X, 2009
)
2.52
"Clofarabine has significant efficiency in children with relapsed or refractory leukemia. "( Clofarabine-induced capillary leak syndrome in a child with refractory acute lymphoblastic leukemia.
Baytan, B; Dönmez, O; Gunes, AM; Ozdemir, O, 2010
)
3.25
"Clofarabine has not been studied in patients on hemodialysis."( Effect of hemodialysis on the plasma levels of clofarabine.
Kobos, R; Latcha, S; Shukla, N; Steinherz, PG, 2010
)
1.34
"Oral clofarabine has achieved a response rate of 43% in patients with higher-risk MDS. "( Oral clofarabine in the treatment of patients with higher-risk myelodysplastic syndrome.
Borthakur, G; Byrd, A; Cortes, JE; Estrov, Z; Faderl, S; Gandhi, V; Garcia-Manero, G; Kantarjian, HM; Kwari, M; O'Brien, S; Plunkett, W; Ravandi, F, 2010
)
1.39
"Clofarabine has significant single-agent activity in patients with indolent and aggressive non-Hodgkin lymphoma and synergizes with DNA-damaging drugs. "( A phase I trial of high-dose clofarabine, etoposide, and cyclophosphamide and autologous peripheral blood stem cell transplantation in patients with primary refractory and relapsed and refractory non-Hodgkin lymphoma.
Abonour, R; Baute, J; Cox, E; Farag, SS; Jones, D; Kane, K; Nelson, RP; Robertson, MJ; Schwartz, JE; Secrest, A; Srivastava, S; Sullivan, C; Wood, LL, 2011
)
2.1
"Clofarabine has been shown to be effective in AML patients, either as single agent or, mainly, in association with intermediate dose cytarabine. "( Cytarabine and clofarabine after high-dose cytarabine in relapsed or refractory AML patients.
Angelucci, E; Biagiotti, C; Borin, L; Bosi, A; Caracciolo, F; Cutini, I; Fanci, R; Gianfaldoni, G; Longo, G; Mannelli, F; Pogliani, EM; Romani, C; Scappini, B; Simonetti, F; Susini, MC, 2012
)
2.17
"Clofarabine has proven to be effective in the treatment of adult and pediatric acute myelogenous leukemia (AML). "( Biochemical modulation of cytarabine triphosphate by clofarabine.
Ayres, M; Cooper, T; Gandhi, V; Nowak, B, 2005
)
2.02
"Clofarabine has also been shown to induce apoptosis in transformed cell lines, indicating that clofarabine results in cell death in both cycling and non-cycling cells."( Clofarabine in the treatment of acute myeloid leukaemia and acute lymphoblastic leukaemia: a review.
Kline, JP; Larson, RA, 2005
)
2.49
"Clofarabine has shown impressive response rates in patients with acute leukemias. "( A phase 1 clinical-laboratory study of clofarabine followed by cyclophosphamide for adults with refractory acute leukemias.
Balakrishnan, K; Briel, J; Carraway, H; Gandhi, V; Gore, SD; Greer, J; Karp, JE; Levis, MJ; McDevitt, MA; Ricklis, RM; Smith, BD, 2007
)
2.05
"Clofarabine has also shown promising clinical activity in pediatric patients, with an overall response rate of 30%, and some patients are able to proceed to allogeneic hematopoietic cell transplantation."( Three new drugs for acute lymphoblastic leukemia: nelarabine, clofarabine, and forodesine.
Larson, RA, 2007
)
1.3

Treatment

Pretreatment with clofarabine resulted in a slight decrease in metabolism of T-araC in RPMI-8226 myeloma cells (65% of control) and inhibited metabolism ofT-ara C in CCRF-CEM leukemia cells by 90%.

ExcerptReferenceRelevance
"Pretreatment with clofarabine resulted in a slight decrease in metabolism of T-araC in RPMI-8226 myeloma cells (65% of control) and inhibited metabolism of T-araC in CCRF-CEM leukemia cells by 90%."( Enhancement of the in vivo antitumor activity of clofarabine by 1-beta-D-[4-thio-arabinofuranosyl]-cytosine.
Gilbert, KS; Parker, WB; Shaddix, SC; Shepherd, RV; Waud, WR, 2009
)
0.93
"The treated with clofarabine 0.01 - 0.1 µmol/L for 24 h, apoptosis rate and Bcl-2 expression of NB4 cells were measured by flow cytometry and Western blot respectively."( [Effect of clofarabine on proliferation and Bcl-2 expression of NB4 cells].
He, PC; Li, CL; Liu, HB; Zhang, M, 2012
)
1.1

Toxicity

We hypothesized that combination of busulfan (Bu), fludarabine (Flu) and clofarabin (Clo) would provide superior efficacy. CloFluBu provides a less toxic alternative to conventional conditioning regimens, with adequate antileukemic activity.

ExcerptReferenceRelevance
" In search of less toxic alternatives, we hypothesized that combination of busulfan (Bu), fludarabine (Flu) and clofarabine (Clo) would provide superior efficacy."( The synergistic cytotoxicity of clofarabine, fludarabine and busulfan in AML cells involves ATM pathway activation and chromatin remodeling.
Andersson, BS; Champlin, RE; Li, Y; Murray, D; Valdez, BC, 2011
)
0.86
" We conducted a review of the literature and utilized the Food and Drug Administration Adverse Event Reporting System to identify spontaneous reporting of renal adverse events with this drug in 29 other cases."( Clofarabine-induced kidney toxicity.
Allen, SL; Chidella, S; Fishbane, S; Jhaveri, KD, 2014
)
1.85
"The occurrence of chemotherapy-related adverse cutaneous reactions in the setting of capillary leak syndrome (CLS) is quite rare."( Chemotherapy-induced skin toxicity and capillary leak syndrome.
Bridges, AG; El-Azhary, RA; Hashmi, SK; Hunjan, MK; Lehman, JS; Nowsheen, S; Ramos-Rodriguez, AJ, 2019
)
0.51
"Five patients were identified, two with a diagnosis of toxic erythema of chemotherapy (TEC) and three others with a skin diagnosis of toxic epidermal necrolysis (TEN)."( Chemotherapy-induced skin toxicity and capillary leak syndrome.
Bridges, AG; El-Azhary, RA; Hashmi, SK; Hunjan, MK; Lehman, JS; Nowsheen, S; Ramos-Rodriguez, AJ, 2019
)
0.51
"Although TEC is generally self-limited, both TEC and TEN can present with severe adverse skin manifestations during CLS secondary to toxicity from chemotherapy."( Chemotherapy-induced skin toxicity and capillary leak syndrome.
Bridges, AG; El-Azhary, RA; Hashmi, SK; Hunjan, MK; Lehman, JS; Nowsheen, S; Ramos-Rodriguez, AJ, 2019
)
0.51
" Response rates, survival outcomes, and adverse events were assessed."( Effectiveness and Safety of Clofarabine Monotherapy or Combination Treatment in Relapsed/Refractory Childhood Acute Lymphoblastic Leukemia: A Pragmatic, Non-interventional Study in Korea.
Cho, B; Cho, H; Choi, JY; Chung, NG; Hahn, SM; Han, JW; Hong, CR; Hong, KT; Im, HJ; Jang, PS; Jeon, IS; Kang, HJ; Kim, H; Kim, S; Koh, KN; Koo, HH; Kook, H; Lee, JW; Lim, YT; Lyu, CJ; Seo, JJ; Shin, HY; Yang, EJ; Yoo, KH, 2021
)
0.92
" There were 24 deaths; 14 due to treatment-emergent adverse events."( Effectiveness and Safety of Clofarabine Monotherapy or Combination Treatment in Relapsed/Refractory Childhood Acute Lymphoblastic Leukemia: A Pragmatic, Non-interventional Study in Korea.
Cho, B; Cho, H; Choi, JY; Chung, NG; Hahn, SM; Han, JW; Hong, CR; Hong, KT; Im, HJ; Jang, PS; Jeon, IS; Kang, HJ; Kim, H; Kim, S; Koh, KN; Koo, HH; Kook, H; Lee, JW; Lim, YT; Lyu, CJ; Seo, JJ; Shin, HY; Yang, EJ; Yoo, KH, 2021
)
0.92
"We prospectively studied clofarabine-fludarabine-busulfan (CloFluBu)-conditioning in allogeneic hematopoietic cell therapy (HCT) for lymphoid and myeloid malignancies and hypothesized that CloFluBu provides a less toxic alternative to conventional conditioning regimens, with adequate antileukemic activity."( Clofarabine-fludarabine-busulfan in HCT for pediatric leukemia: an effective, low toxicity, TBI-free conditioning regimen.
Bierings, M; Boelens, JJ; Bresters, D; de Koning, CCH; Kollen, WJ; Lankester, AC; Lindemans, CA; Nierkens, S; Versluijs, AB, 2022
)
2.47

Pharmacokinetics

Plasma clofarabine levels and pharmacokinetic parameters were monitored continuously for up to 8 hours after the single intravenous administration of 20 mg/kg. There was a linear, dose dependent increase in clofArabine concentration in the plasma.

ExcerptReferenceRelevance
"The purpose of our study was to investigate the pharmacology of clofarabine and its triphosphate and the pharmacodynamic actions in circulating blasts obtained from acute leukemia patients who entered a Phase I clinical trial of clofarabine."( Pharmacokinetics and pharmacodynamics of plasma clofarabine and cellular clofarabine triphosphate in patients with acute leukemias.
Ayres, M; Bonate, P; Du, M; Faderl, S; Gandhi, V; Kantarjian, H; Keating, MJ; Plunkett, W; Rios, MB, 2003
)
0.81
" Cellular pharmacokinetic studies done at the end of the first clofarabine infusion in 26 patients appeared dose proportional but showed a wide variation in the concentrations of clofarabine triphosphate."( Pharmacokinetics and pharmacodynamics of plasma clofarabine and cellular clofarabine triphosphate in patients with acute leukemias.
Ayres, M; Bonate, P; Du, M; Faderl, S; Gandhi, V; Kantarjian, H; Keating, MJ; Plunkett, W; Rios, MB, 2003
)
0.81
" A 2-compartment model with weight (scaled to a 40-kg reference patient) modeled as a power function on all pharmacokinetic parameters (0."( Population pharmacokinetics of clofarabine, a second-generation nucleoside analog, in pediatric patients with acute leukemia.
Bonate, PL; Craig, A; Gandhi, V; Gaynon, P; Jeha, S; Kadota, R; Lam, GN; Plunkett, W; Razzouk, B; Rytting, M; Steinherz, P; Weitman, S, 2004
)
0.61
" Pharmacokinetic studies in the phase I trial revealed marked heterogeneity in peak levels of clofarabine among patients at the end of infusion, however; there was a linear, dose dependent increase in clofarabine concentration in the plasma."( Clofarabine in adult acute leukemias: clinical success and pharmacokinetics.
Cooper, T; Gandhi, V; Kantarjian, H; Plunkett, W, 2004
)
1.99
"The median clofarabine clearance was 17 mL/min/kg (range, 15-20), the median plasma area under the concentration-time curve was 452 mumol/L minutes (range, 380-487), and the median terminal half-life was 105 minutes (range, 78-138)."( Plasma and cerebrospinal fluid pharmacokinetics of clofarabine in nonhuman primates.
Berg, SL; Bernacky, B; Blaney, SM; Bonate, PL; Dauser, R; McGuffey, L; Nuchtern, JG, 2005
)
0.97
" The elimination half-life was dependent on all the covariates but was generally less than 7 h in all cases."( Population pharmacokinetics of clofarabine and its metabolite 6-ketoclofarabine in adult and pediatric patients with cancer.
Bonate, PL; Cunningham, CC; Gaynon, P; Jeha, S; Kadota, R; Lam, GN; Razzouk, B; Rytting, M; Steinherz, P; Weitman, S, 2011
)
0.66
" We developed a pharmacodynamic model to characterize the interaction between CLO and DEC on an AML cell line and down-regulated p53R2 protein to understand its role."( Synergism between clofarabine and decitabine through p53R2: a pharmacodynamic drug-drug interaction modeling.
Fetterly, GJ; Ghoshal, S; Haese, JP; Karpf, AR; Thudium, KE; Wetzler, M, 2012
)
0.71
" Plasma clofarabine levels and pharmacokinetic parameters were monitored continuously for up to 8 hours after the single intravenous administration of 20 mg/kg at 12:00 noon and 12:00 midnight by high performance liquid chromatography (HPLC)-UV method."( Dosing-time contributes to chronotoxicity of clofarabine in mice via means other than pharmacokinetics.
Liu, XY; Luan, JJ; Song, JG; Wang, WS; Wang, YQ; Zhang, W; Zhang, YS; Zuo, J, 2016
)
1.13
" We studied the correlation of pharmacokinetic (PK) parameters with these toxicities and the efficacy of CFB in patients with relapsed or refractory acute myeloid leukemia."( Impact of pharmacokinetics on the toxicity and efficacy of clofarabine in patients with relapsed or refractory acute myeloid leukemia.
Alakel, N; Bornhäuser, M; Büttner, B; Ehninger, G; Knoth, H; Kramer, M; Middeke, JM; Oertel, R; Platzbecker, U; Röllig, C; Schetelig, J; Seeling, A; Sockel, K; Stölzel, F; von Bonin, M, 2017
)
0.7
"We sought to examine the pharmacodynamic activation of the DNA damage response (DDR) pathway in tumors following anticancer treatment for confirmation of target engagement."( Evaluation of Pharmacodynamic Responses to Cancer Therapeutic Agents Using DNA Damage Markers.
Barrett, AM; Chen, A; Doroshow, JH; Dull, AB; Hollingshead, MG; Kinders, RJ; Kummar, S; Lawrence, SM; Parchment, RE; Wilsker, DF, 2019
)
0.51
"Because of inherent biological variability, baseline DDR biomarker levels were evaluated in a colorectal cancer microarray to establish clinically relevant thresholds for pharmacodynamic activation."( Evaluation of Pharmacodynamic Responses to Cancer Therapeutic Agents Using DNA Damage Markers.
Barrett, AM; Chen, A; Doroshow, JH; Dull, AB; Hollingshead, MG; Kinders, RJ; Kummar, S; Lawrence, SM; Parchment, RE; Wilsker, DF, 2019
)
0.51
" Pharmacokinetic (PK) exposure of different agents commonly used in conditioning regimens is strongly related to HCT outcome."( Population pharmacokinetics of clofarabine for allogeneic hematopoietic cell transplantation in paediatric patients.
Bierings, M; Boelens, JJ; Huitema, ADR; Lindemans, CA; Nierkens, S; Nijstad, AL; van der Elst, KCM; Versluys, AB, 2021
)
0.91
"5-18 years) and 1 adult patient (37 years), 805 plasma concentrations were included in pharmacokinetic analyses using nonlinear mixed effects modelling."( Population pharmacokinetics of clofarabine for allogeneic hematopoietic cell transplantation in paediatric patients.
Bierings, M; Boelens, JJ; Huitema, ADR; Lindemans, CA; Nierkens, S; Nijstad, AL; van der Elst, KCM; Versluys, AB, 2021
)
0.91

Compound-Compound Interactions

Clofarabine 5 × 40 mg/m(2) in combination with pegylated asparaginase (PEG-ASP) 1 × 2500 iu/m() in high risk ALL patients as a novel post-induction element in German Co-operative Study Group for treatment of ALL (CoALL) trial 08-09.

ExcerptReferenceRelevance
"To assess the toxicity, pharmacokinetics, and pharmacodynamics of multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in children with relapsed/refractory leukemia."( Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia.
Baker, SD; Campana, D; Christensen, R; Coustan-Smith, E; Furmanski, BD; Heym, KM; Inaba, H; Li, L; Mascara, GP; Onciu, M; Pounds, SB; Pui, CH; Ribeiro, RC; Rubnitz, JE; Shurtleff, SA, 2011
)
0.79
"Sorafenib in combination with clofarabine and cytarabine is tolerable and shows activity in relapsed/refractory pediatric AML."( Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia.
Baker, SD; Campana, D; Christensen, R; Coustan-Smith, E; Furmanski, BD; Heym, KM; Inaba, H; Li, L; Mascara, GP; Onciu, M; Pounds, SB; Pui, CH; Ribeiro, RC; Rubnitz, JE; Shurtleff, SA, 2011
)
0.87
" We conducted a phase I study to determine the maximum tolerated dose (MTD) and dose limiting toxicities (DLTs) of clofarabine when combined with GO in adult patients with relapsed or refractory AML."( A phase I dose-escalation study of clofarabine in combination with fractionated gemtuzumab ozogamicin in patients with refractory or relapsed acute myeloid leukemia.
Amin, C; Barr, ND; Chiu, WM; Foster, MC; Ivanova, A; Richards, KL; Shea, T; van Deventer, HW; Voorhees, PM; Whitman, J, 2012
)
0.87
" busulfan (Bu) in combination with clofarabine (Clo) in patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation (SCT)."( Clofarabine combined with busulfan provides excellent disease control in adult patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation.
Alousi, A; Andersson, BS; Basset, R; Champlin, RE; Ciurea, S; de Lima, M; Hosing, C; Jones, RB; Kebriaei, P; Khouri, I; Ledesma, C; Nieto, Y; Parmar, S; Popat, U; Qazilbash, M; Shpall, EJ, 2012
)
2.1
" To investigate its value in the frontline treatment of ALL we applied clofarabine 5 × 40 mg/m(2) in combination with pegylated asparaginase (PEG-ASP) 1 × 2500 iu/m(2) in high risk ALL patients as a novel post-induction element in the German Co-operative Study Group for treatment of ALL (CoALL) trial 08-09."( Clofarabine in combination with pegylated asparaginase in the frontline treatment of childhood acute lymphoblastic leukaemia: a feasibility report from the CoALL 08-09 trial.
Escherich, G; Horstmann, MA; Zimmermann, M; Zur Stadt, U, 2013
)
2.07
"This study aims to determine the maximum tolerated dose (MTD) of clofarabine combined with the EORTC-GIMEMA 3 + 10 induction regimen (idarubicin + cytosine arabinoside) in adults with untreated acute myelogenous leukemia or high-risk myelodysplastic syndrome."( Clofarabine in combination with a standard remission induction regimen (cytosine arabinoside and idarubicin) in patients with previously untreated intermediate and bad-risk acute myelogenous leukemia (AML) or high-risk myelodysplastic syndrome (HR-MDS): p
Amadori, S; de Witte, T; Halkes, CJ; Karrasch, M; Marie, JP; Meert, L; Meloni, G; Muus, P; Rapion, J; Suciu, S; Vignetti, M; Willemze, R, 2014
)
2.08
" In this phase 1/2 study, the tolerability and overall response rate of clofarabine in combination with cytarabine was investigated in children with recurrent/refractory AML."( AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia.
Alonzo, TA; Cooper, TM; Gamis, AS; Gerbing, RB; Horton, TM; Loken, MR; Meshinchi, S; Perentesis, JP; Razzouk, BI; Taub, JW; Whitlock, JA, 2014
)
0.87
" The recommended pediatric phase 2 dose of clofarabine in combination with cytarabine was 52 mg/m(2)/day for 5 days."( AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia.
Alonzo, TA; Cooper, TM; Gamis, AS; Gerbing, RB; Horton, TM; Loken, MR; Meshinchi, S; Perentesis, JP; Razzouk, BI; Taub, JW; Whitlock, JA, 2014
)
0.9
" We evaluated two new lower-intensity regimens with clofarabine (n = 119) or cladribine (n = 129) combined with low-dose cytarabine (LDAC) alternating with decitabine."( Long-term results of low-intensity chemotherapy with clofarabine or cladribine combined with low-dose cytarabine alternating with decitabine in older patients with newly diagnosed acute myeloid leukemia.
Borthakur, G; Burger, J; Daver, N; DiNardo, CD; Estrov, Z; Ferrajoli, A; Garcia-Manero, G; Huang, X; Jabbour, E; Jain, N; Kadia, TM; Kanagal-Shamanna, R; Kantarjian, H; Konopleva, M; Pemmaraju, N; Pierce, S; Popat, U; Rausch, C; Ravandi, F; Verstovsek, S; Wang, X, 2021
)
1.12

Bioavailability

ExcerptReferenceRelevance
" Administration of equivalent oral doses resulted in bioavailability estimates of approximately 50%, indicating that oral treatment regimens of CL-F-ara-A are feasible."( Pharmacokinetics of the anticancer agent 2-chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)adenine in rats.
Chu, CK; Gallo, JM; Qian, M; Shanmuganathan, K; Wang, X, 1994
)
0.29
" Clofarabine was rapidly absorbed following oral administration with a mean absorption time of less than 2 h and bioavailability of 57."( Population pharmacokinetics of clofarabine and its metabolite 6-ketoclofarabine in adult and pediatric patients with cancer.
Bonate, PL; Cunningham, CC; Gaynon, P; Jeha, S; Kadota, R; Lam, GN; Razzouk, B; Rytting, M; Steinherz, P; Weitman, S, 2011
)
1.57
" This new anticancer drug is more effective (in low doses) and indicates higher oral bioavailability in comparison to its congeners."( Clofarabine (2-chloro-2'-fluoro-2'-deoxyarabinosyladenine)--biochemical aspects of anticancer activity.
Fabianowska-Majewska, K; Kaufman-Szymczyk, A; Lubecka, K; Majda, K,
)
1.57
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51

Dosage Studied

The distribution, metabolism, and elimination of intravenous [14C]clofarabine was studied in Fischer 344 male rats under a once daily for 5 days dosing schedule of 25 or 50 mg/kg/day. The results confirm that clofArabine should continue being dosed on a per-squaremeter or per-body-weight basis. ClofARabine dosed at 52 mg/m2 was used in adult patients with refractory ALL to maximize response before allo-HSCT.

ExcerptRelevanceReference
" The results confirm that clofarabine should continue being dosed on a per-squaremeter or per-body-weight basis."( Population pharmacokinetics of clofarabine, a second-generation nucleoside analog, in pediatric patients with acute leukemia.
Bonate, PL; Craig, A; Gandhi, V; Gaynon, P; Jeha, S; Kadota, R; Lam, GN; Plunkett, W; Razzouk, B; Rytting, M; Steinherz, P; Weitman, S, 2004
)
0.91
"The distribution, metabolism, and elimination of intravenous [14C]clofarabine was studied in Fischer 344 male rats under a once daily for 5 days dosing schedule of 25 or 50 mg/kg/day."( The distribution, metabolism, and elimination of clofarabine in rats.
Arthaud, L; Bonate, PL; Press, RJ; Rose, JQ; Stuhler, J; Yerino, P, 2005
)
0.82
" Clofarabine dosed at 52 mg/m2 was used in adult patients with refractory ALL to maximize response before allo-HSCT."( The use of higher dose clofarabine in adults with relapsed acute lymphoblastic leukemia.
Brown, AW; McGregor, BA; Osswald, MB; Savona, MR, 2009
)
1.57
" Clo 40 mg/m(2) was given once daily, with each dose followed by pharmacokinetically dosed Bu infused over 3 hours daily for 4 days, followed by hematopoietic SCT 2 days later."( Clofarabine combined with busulfan provides excellent disease control in adult patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation.
Alousi, A; Andersson, BS; Basset, R; Champlin, RE; Ciurea, S; de Lima, M; Hosing, C; Jones, RB; Kebriaei, P; Khouri, I; Ledesma, C; Nieto, Y; Parmar, S; Popat, U; Qazilbash, M; Shpall, EJ, 2012
)
1.82
" These data suggest that renal-adjustment of clofarabine dosing should be considered for older and at-risk patients even when renal function is ostensibly normal."( Clofarabine-associated acute kidney injury in patients undergoing hematopoietic stem cell transplant.
Artz, AS; Cao, H; Hard, M; O'Donnell, PH; Petri, CR; Stock, W; van Besien, K; Wickrema, A, 2014
)
2.1
" High-dose cytarabine is clearly effective and there definitely is a dose-response relationship for cytarabine and remission duration."( Optimal therapy for adult patients with acute myeloid leukemia in first complete remission.
Wiernik, PH, 2014
)
0.4
" A simulation of clofarabine pharmacokinetics in mice and pediatric patients suggested that a dosage of 30 mg/kg IP in mice would give exposures comparable to that in children at a dosage of 148 mg/m(2)."( Preclinical examination of clofarabine in pediatric ependymoma: intratumoral concentrations insufficient to warrant further study.
Boulos, N; Dapper, JD; Davis, AD; Freeman, BB; Gilbertson, RJ; Jacus, MO; Mohankumar, KM; Patel, YT; Stewart, CF; Throm, SL; Vuppala, PK, 2015
)
1.05
"A clofarabine dosing algorithm based on this PK model, using body weight and eGFR, results in a more predictable exposure than BSA-based dosing."( Population pharmacokinetics of clofarabine for allogeneic hematopoietic cell transplantation in paediatric patients.
Bierings, M; Boelens, JJ; Huitema, ADR; Lindemans, CA; Nierkens, S; Nijstad, AL; van der Elst, KCM; Versluys, AB, 2021
)
1.63
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (2)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
antimetaboliteA substance which is structurally similar to a metabolite but which competes with it or replaces it, and so prevents or reduces its normal utilization.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (2)

ClassDescription
organofluorine compoundAn organofluorine compound is a compound containing at least one carbon-fluorine bond.
adenosinesAny purine ribonucleoside that is a derivative of adenosine.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Protein Targets (31)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
RAR-related orphan receptor gammaMus musculus (house mouse)Potency5.44940.006038.004119,952.5996AID1159521; AID1159523
Fumarate hydrataseHomo sapiens (human)Potency4.46680.00308.794948.0869AID1347053
TDP1 proteinHomo sapiens (human)Potency0.00210.000811.382244.6684AID686978; AID686979
GLI family zinc finger 3Homo sapiens (human)Potency0.89260.000714.592883.7951AID1259369; AID1259392
AR proteinHomo sapiens (human)Potency7.15280.000221.22318,912.5098AID1259243; AID1259247; AID743035; AID743063
estrogen receptor 2 (ER beta)Homo sapiens (human)Potency10.59090.000657.913322,387.1992AID1259378
nuclear receptor subfamily 1, group I, member 3Homo sapiens (human)Potency0.11660.001022.650876.6163AID1224838; AID1224893
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency0.79430.01237.983543.2770AID1346984
glucocorticoid receptor [Homo sapiens]Homo sapiens (human)Potency1.42380.000214.376460.0339AID720691; AID720692
retinoic acid nuclear receptor alpha variant 1Homo sapiens (human)Potency3.75780.003041.611522,387.1992AID1159552; AID1159555
estrogen-related nuclear receptor alphaHomo sapiens (human)Potency9.13860.001530.607315,848.9004AID1224841; AID1224842; AID1224848; AID1224849; AID1259401; AID1259403
farnesoid X nuclear receptorHomo sapiens (human)Potency1.67840.375827.485161.6524AID743217
pregnane X nuclear receptorHomo sapiens (human)Potency0.76540.005428.02631,258.9301AID1346982; AID1346985
estrogen nuclear receptor alphaHomo sapiens (human)Potency8.71030.000229.305416,493.5996AID1259244; AID743069; AID743075; AID743078; AID743080; AID743091
polyproteinZika virusPotency4.46680.00308.794948.0869AID1347053
peroxisome proliferator activated receptor gammaHomo sapiens (human)Potency8.41200.001019.414170.9645AID743191
cytochrome P450, family 19, subfamily A, polypeptide 1, isoform CRA_aHomo sapiens (human)Potency6.68240.001723.839378.1014AID743083
Histone H2A.xCricetulus griseus (Chinese hamster)Potency1.68470.039147.5451146.8240AID1224845; AID1224896
thyroid hormone receptor beta isoform 2Rattus norvegicus (Norway rat)Potency0.13590.000323.4451159.6830AID743065; AID743067
nuclear factor erythroid 2-related factor 2 isoform 1Homo sapiens (human)Potency1.93700.000627.21521,122.0200AID743202; AID743219
Voltage-dependent calcium channel gamma-2 subunitMus musculus (house mouse)Potency3.75780.001557.789015,848.9004AID1259244
Cellular tumor antigen p53Homo sapiens (human)Potency0.09060.002319.595674.0614AID651631; AID720552
Glutamate receptor 2Rattus norvegicus (Norway rat)Potency3.75780.001551.739315,848.9004AID1259244
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency19.95260.009610.525035.4813AID1479145
ATPase family AAA domain-containing protein 5Homo sapiens (human)Potency0.53080.011917.942071.5630AID651632
Ataxin-2Homo sapiens (human)Potency0.53080.011912.222168.7989AID651632
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
cGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)IC50 (µMol)3.12000.00001.77679.2000AID1368375
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)133.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)133.00000.20005.677410.0000AID1473741
Bile salt export pumpHomo sapiens (human)IC50 (µMol)133.00000.11007.190310.0000AID1473738
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (205)

Processvia Protein(s)Taxonomy
heart valve developmentcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
ventricular septum developmentcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
aorta developmentcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwaycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
regulation of cGMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cAMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cGMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to macrophage colony-stimulating factor stimuluscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
negative regulation of vascular permeabilitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
positive regulation of vascular permeabilitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
regulation of cAMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cGMP catabolic processcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
positive regulation of inflammatory responsecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
establishment of endothelial barriercGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to mechanical stimuluscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to cAMPcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to cGMPcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to transforming growth factor beta stimuluscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
negative regulation of adenylate cyclase-activating G protein-coupled receptor signaling pathwaycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cellular response to 2,3,7,8-tetrachlorodibenzodioxinecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
positive regulation of gene expressioncGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
negative regulation of cGMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
negative regulation of cAMP-mediated signalingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
regulation of mitochondrion organizationcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell population proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of B cell proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
nuclear DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
signal transduction in response to DNA damageATPase family AAA domain-containing protein 5Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
isotype switchingATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of isotype switching to IgG isotypesATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloadingATPase family AAA domain-containing protein 5Homo sapiens (human)
regulation of mitotic cell cycle phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of cell cycle G2/M phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
negative regulation of receptor internalizationAtaxin-2Homo sapiens (human)
regulation of translationAtaxin-2Homo sapiens (human)
RNA metabolic processAtaxin-2Homo sapiens (human)
P-body assemblyAtaxin-2Homo sapiens (human)
stress granule assemblyAtaxin-2Homo sapiens (human)
RNA transportAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (70)

Processvia Protein(s)Taxonomy
magnesium ion bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
3',5'-cyclic-AMP phosphodiesterase activitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cGMP-stimulated cyclic-nucleotide phosphodiesterase activitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
protein bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
zinc ion bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cAMP bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cGMP bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
TPR domain bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
phosphate ion bindingcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
protein homodimerization activitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
3',5'-cyclic-GMP phosphodiesterase activitycGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo sapiens (human)
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
protein bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP hydrolysis activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloader activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
RNA bindingAtaxin-2Homo sapiens (human)
epidermal growth factor receptor bindingAtaxin-2Homo sapiens (human)
protein bindingAtaxin-2Homo sapiens (human)
mRNA bindingAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (45)

Processvia Protein(s)Taxonomy
plasma membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
nucleuscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cytoplasmcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
mitochondrial outer membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
mitochondrial inner membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
endoplasmic reticulumcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
Golgi apparatuscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cytosolcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
plasma membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
presynaptic membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
perinuclear region of cytoplasmcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
nucleuscGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
mitochondrial inner membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
perinuclear region of cytoplasmcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
mitochondrial outer membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
synaptic membranecGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
cytosolcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
mitochondrial matrixcGMP-dependent 3',5'-cyclic phosphodiesteraseHomo sapiens (human)
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
plasma membraneGlutamate receptor 2Rattus norvegicus (Norway rat)
virion membraneSpike glycoproteinSevere acute respiratory syndrome-related coronavirus
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
Elg1 RFC-like complexATPase family AAA domain-containing protein 5Homo sapiens (human)
nucleusATPase family AAA domain-containing protein 5Homo sapiens (human)
cytoplasmAtaxin-2Homo sapiens (human)
Golgi apparatusAtaxin-2Homo sapiens (human)
trans-Golgi networkAtaxin-2Homo sapiens (human)
cytosolAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
membraneAtaxin-2Homo sapiens (human)
perinuclear region of cytoplasmAtaxin-2Homo sapiens (human)
ribonucleoprotein complexAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (157)

Assay IDTitleYearJournalArticle
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1347125qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347122qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347119qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347124qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347135qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347127qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347425Rhodamine-PBP qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347114qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347137qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for Daoy cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347424RapidFire Mass Spectrometry qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347118qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347139qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347115qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID1347116qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347128qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347407qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Pharmaceutical Collection2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347121qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347138qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D caspase screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347110qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for A673 cells)2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1347113qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347141qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347123qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1347111qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347140qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347117qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347136qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Orthogonal 3D viability screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347126qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347129qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347109qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347112qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1298312Effect on 5-aza-dCTP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625286Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1408357Cytotoxicity against human CCRF-CEM cells assessed as decrease in cell viability after 72 hrs by MTT assay2018European journal of medicinal chemistry, Sep-05, Volume: 157CD73 inhibition by purine cytotoxic nucleoside analogue-based diphosphonates.
AID1298258Increase in 5-aza-dCTP/dCTP ratio in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-C2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID44000Compound was tested for cytotoxicity against CCRF-CEM cell lines1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID135606Log chance in viable tumor cells population was measured after intraperitoneal administration of 25 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID445360Cytotoxicity against human PC3 cells after 5 days by SRB assay2010Journal of medicinal chemistry, Jan-14, Volume: 53, Issue:1
6-(Het)aryl-7-deazapurine ribonucleosides as novel potent cytostatic agents.
AID140653Tumor-free survivors of P388 leukemia infected mice was measured after ip administration of 200 mg/kg in 3 mice1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID625285Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic necrosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1408362Cytotoxicity against human RL cells assessed as decrease in cell viability after 72 hrs by MTT assay2018European journal of medicinal chemistry, Sep-05, Volume: 157CD73 inhibition by purine cytotoxic nucleoside analogue-based diphosphonates.
AID1298282Reduction in dTTP level in human U373-MAGI cells at 200 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298385Reduction in 5-aza-dCTP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID135604Log chance in viable tumor cells population was measured after intraperitoneal administration of 100 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID135603Log chance in viable tumor cells population was measured after intraperitoneal administration of 200 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID1298316Effect on 5-aza-dCTP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID96710Compound was tested for cytotoxicity against L1210 cell lines1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID140652Tumor-free survivors of P388 leukemia infected mice was measured after ip administration of 20 mg/kg in 6 mice1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID625282Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cirrhosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1408363Cytotoxicity against human HL60 cells assessed as decrease in cell viability after 72 hrs by MTT assay2018European journal of medicinal chemistry, Sep-05, Volume: 157CD73 inhibition by purine cytotoxic nucleoside analogue-based diphosphonates.
AID611376Cytostatic activity against human MT4 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID625292Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) combined score2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298267Selectivity index, ratio of 5-Aza-C CC50 for human U373-MAGI cells to 5-Aza-C EC50 for VSV-G pseudotyped HIV-1 NL4-3 in presence of 50 nM compound (Rvb = 2.8 No_unit)2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298286Reduction in dGTP level in human U373-MAGI cells at 50 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298272Cytotoxicity against human U373-MAGI cells at 50 nM measured at 72 hrs by Celltiter-Glo luminescent cell viability assay2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625289Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver disease2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298290Reduction in dGTP level in human U373-MAGI cells at 200 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID625279Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for bilirubinemia2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298337Increase in 5-aza-dCTP/dCTP ratio in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-dC2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298278Reduction in dTTP level in human U373-MAGI cells at 50 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625291Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver function tests abnormal2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID135605Log chance in viable tumor cells population was measured after intraperitoneal administration of 20 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1368376Inhibition of recombinant human PDE2A catalytic domain (580 to 919 residues) expressed in Escherichia coli BL21 (Codonplus) at 1 uM using [3H]cGMP as substrate after 15 mins by liquid scintillation counting method relative to control2018Bioorganic & medicinal chemistry, 01-01, Volume: 26, Issue:1
Discovery of novel purine nucleoside derivatives as phosphodiesterase 2 (PDE2) inhibitors: Structure-based virtual screening, optimization and biological evaluation.
AID1894173Inhibition of Ribonucleotide reductase (unknown origin)2021European journal of medicinal chemistry, Mar-15, Volume: 214FDA-approved pyrimidine-fused bicyclic heterocycles for cancer therapy: Synthesis and clinical application.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID611371Cytostatic activity against human PC3 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID611373Cytostatic activity against human HCT15 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID140654Tumor-free survivors of P388 leukemia infected mice was measured after ip administration of 25 mg/kg in 5 mice1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID625284Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic failure2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID625287Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatomegaly2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID611372Cytostatic activity against human HCT116 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID131310In Vivo increase in life span of dying mice implanted ip with 10E6 P388 leukemia, after ip administration of 200 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID764207Cytostatic activity against human A549 cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry, Sep-01, Volume: 21, Issue:17
Synthesis and in vitro cytostatic activity of 1,2- and 1,3-diacylglycerophosphates of clofarabine.
AID1298378Reduction in 5-aza-dCTP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298308Reduction in dATP level in human U373-MAGI cells at 200 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298304Reduction in dATP level in human U373-MAGI cells at 50 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID764206Cytostatic activity against human HL60 cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry, Sep-01, Volume: 21, Issue:17
Synthesis and in vitro cytostatic activity of 1,2- and 1,3-diacylglycerophosphates of clofarabine.
AID1408358Cytotoxicity against human Granta cells assessed as decrease in cell viability after 72 hrs by MTT assay2018European journal of medicinal chemistry, Sep-05, Volume: 157CD73 inhibition by purine cytotoxic nucleoside analogue-based diphosphonates.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID625280Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholecystitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID445359Cytotoxicity against human NCI-H23 cells after 5 days by SRB assay2010Journal of medicinal chemistry, Jan-14, Volume: 53, Issue:1
6-(Het)aryl-7-deazapurine ribonucleosides as novel potent cytostatic agents.
AID1298389Effect on dRGU-TP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID611370Cytostatic activity against human DU145 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID1298375Increase in 5-aza-dCTP/dCTP ratio in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-C2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID131311In Vivo increase in life span of dying mice implanted ip with 10E6 P388 leukemia, after ip administration of 25 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID140651Tumor-free survivors of P388 leukemia infected mice was measured after ip administration of 100 mg/kg in 5 mice1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1298393Reduction in dCTP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-dC2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID445361Cytotoxicity against human HCT15 cells after 5 days by SRB assay2010Journal of medicinal chemistry, Jan-14, Volume: 53, Issue:1
6-(Het)aryl-7-deazapurine ribonucleosides as novel potent cytostatic agents.
AID1298264Increase of 5-Aza-C-mediated cytotoxicity against human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-Aza-C addition for 2 hrs measured after 72 hrs by Celltiter-Glo luminescent cell viability assay (Rvb = 387 uM)2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID477295Octanol-water partition coefficient, log P of the compound2010European journal of medicinal chemistry, Apr, Volume: 45, Issue:4
QSPR modeling of octanol/water partition coefficient of antineoplastic agents by balance of correlations.
AID1298295Reduction in dCTP level in human U373-MAGI cells at 50 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298328Effect on dRGU-TP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625290Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver fatty2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298329Reduction in dRGU-TP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298299Reduction in dCTP level in human U373-MAGI cells at 200 nM after 6 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625281Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholelithiasis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298320Reduction in dCTP level in human U373-MAGI cells at 50 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID625283Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for elevated liver function tests2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298333Reduction in dRGU-TP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID611368Cytostatic activity against human A549 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID94831Compound was tested for cytotoxicity against K562 cell lines1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID611369Cytostatic activity against human NCI-H23 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID131309In Vivo increase in life span of dying mice implanted ip with 10E6 P388 leukemia, after ip administration of 20 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID764205Cytostatic activity against human MCF7 cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry, Sep-01, Volume: 21, Issue:17
Synthesis and in vitro cytostatic activity of 1,2- and 1,3-diacylglycerophosphates of clofarabine.
AID1298261Potentiation of 5-Aza-C-induced antiviral activity against VSV-G pseudotyped HIV-1 NL4-3 infected in human U373-MAGI cells assessed as decrease in 5-Aza-C EC50 at 50 nM preincubated for 2 hrs followed by 5-Aza-C addition for 2 hrs and subsequent viral inf2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298324Reduction in dCTP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-C addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-C2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID611375Cytostatic activity against human BT549 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID81745Compound was tested for cytotoxicity against HEp-2 cell lines1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID1368375Inhibition of recombinant human PDE2A catalytic domain (580 to 919 residues) expressed in Escherichia coli BL21 (Codonplus) using [3H]cGMP as substrate after 15 mins by liquid scintillation counting method2018Bioorganic & medicinal chemistry, 01-01, Volume: 26, Issue:1
Discovery of novel purine nucleoside derivatives as phosphodiesterase 2 (PDE2) inhibitors: Structure-based virtual screening, optimization and biological evaluation.
AID445363Cytotoxicity against human BT549 cells after 5 days by SRB assay2010Journal of medicinal chemistry, Jan-14, Volume: 53, Issue:1
6-(Het)aryl-7-deazapurine ribonucleosides as novel potent cytostatic agents.
AID764204Cytostatic activity against human HeLa cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry, Sep-01, Volume: 21, Issue:17
Synthesis and in vitro cytostatic activity of 1,2- and 1,3-diacylglycerophosphates of clofarabine.
AID1298268Antiviral activity against VSV-G pseudotyped HIV-1 NL4-3 infected in human U373-MAGI cells assessed as reduction in viral infectivity at 50 nM incubated for 4 hrs prior to viral infection measured at 72 hrs post infection by flow cytometric analysis2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1298341Increase in 5-aza-dCTP/dCTP ratio in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-dC2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID131308In Vivo increase in life span of dying mice implanted ip with 10E6 P388 leukemia, after ip administration of 100 mg/kg1992Journal of medicinal chemistry, Jan-24, Volume: 35, Issue:2
Synthesis and biologic activity of 2'-fluoro-2-halo derivatives of 9-beta-D-arabinofuranosyladenine.
AID625288Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for jaundice2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1298262Potentiation of 5-Aza-C-induced antiviral activity against VSV-G pseudotyped HIV-1 NL4-3 infected in human U373-MAGI cells assessed as 5-Aza-C EC50 at 50 nM preincubated for 2 hrs followed by 5-Aza-C addition for 2 hrs and subsequent viral infection measu2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1298397Reduction in dCTP level in human U373-MAGI cells at 200 nM preincubated for 2 hrs followed by 5-aza-dC addition measured after 4 hrs by LC-MS/MS analysis relative to 5-aza-dC2016Bioorganic & medicinal chemistry, 06-01, Volume: 24, Issue:11
Synergistic reduction of HIV-1 infectivity by 5-azacytidine and inhibitors of ribonucleotide reductase.
AID611374Cytostatic activity against human Hs578 cells after 5 days by SRB assay2011Journal of medicinal chemistry, Aug-11, Volume: 54, Issue:15
Synthesis and significant cytostatic activity of 7-hetaryl-7-deazaadenosines.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1346042Human ribonucleotide reductase catalytic subunit M1 (Nucleoside synthesis and metabolism)1991Cancer research, May-01, Volume: 51, Issue:9
Effects of 2-chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)adenine on K562 cellular metabolism and the inhibition of human ribonucleotide reductase and DNA polymerases by its 5'-triphosphate.
AID1346136Human ribonucleotide reductase regulatory subunit M2 (Nucleoside synthesis and metabolism)1991Cancer research, May-01, Volume: 51, Issue:9
Effects of 2-chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)adenine on K562 cellular metabolism and the inhibition of human ribonucleotide reductase and DNA polymerases by its 5'-triphosphate.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (415)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's14 (3.37)18.2507
2000's94 (22.65)29.6817
2010's257 (61.93)24.3611
2020's50 (12.05)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 45.49

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index45.49 (24.57)
Research Supply Index6.27 (2.92)
Research Growth Index5.42 (4.65)
Search Engine Demand Index70.70 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (45.49)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials101 (23.82%)5.53%
Reviews60 (14.15%)6.00%
Case Studies48 (11.32%)4.05%
Observational2 (0.47%)0.25%
Other213 (50.24%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (165)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Population Pharmacokinetics of the Nucleoside Analogues Clofarabine and Fludarabine in Pediatric Patients Undergoing Hematopoietic Cell Transplantation (alloHCT) [NCT03609814]30 participants (Actual)Observational2016-01-26Completed
A Pharmacokinetic Study of Oral and Intravenous Clofarabine in Patients With High Risk Myelodysplasia and Acute Leukemias - Determination of Oral Bioavailability and the Effect of Cimetidine on Clofarabine Clearance [NCT01169012]Phase 113 participants (Actual)Interventional2010-09-30Completed
A Phase I/II Dose Escalation Trial of Clofarabine, in Addition to Melphalan and Thiotepa as Myeloablative Regimen Followed by an Allogeneic Unmodified Hematopoietic Stem Cell Transplant From HLA-Compatible Related or Unrelated Donors for the Treatment of [NCT00423514]Phase 1/Phase 238 participants (Actual)Interventional2006-11-20Completed
Clofarabine/Ara-C Treatment Combined With Reduced-intensity Conditioning Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia in Primary Treatment Failure [NCT01188174]Phase 226 participants (Actual)Interventional2010-07-31Completed
Multicentric Phase II Trial, Prospective, Open, Single Group, to Discuss Induction Therapy With a Combination of Clofarabine and Low-dose Cytarabine Followed by Consolidation Therapy With Clofarabine and Low-dose Cytarabine for the Treatment of AML Patien [NCT01193400]Phase 275 participants (Anticipated)Interventional2010-09-30Terminated
A Phase I, Open-label, Multi-center Study of Clofarabine (JC0707) in Japanese Patients With Acute Myeloid Leukemia (AML) [NCT01090167]Phase 114 participants (Actual)Interventional2010-02-28Completed
A Phase II Trial of Alpha Beta T-cell and CD19 B-cell Depleted Peripheral Blood Stem Cell Transplantation Using the CliniMACS System for Patients With Non-Malignant Hematologic Disorders From Matched or Mismatched, Related or Unrelated Donors [NCT03615144]Phase 21 participants (Actual)Interventional2018-07-23Terminated(stopped due to Participant accrual low and the study was closed)
Clofarabine Salvage Therapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia [NCT01295307]Phase 286 participants (Actual)Interventional2011-03-31Completed
A Phase I, Open-label, Multi-center Study of Clofarabine in Japanese Paediatric Patients With Relapsed or Refractory Acute Lymphoblastic Leukaemia [NCT01196013]Phase 17 participants (Actual)Interventional2010-08-31Completed
Vorinostat With Gemcitabine/Clofarabine/Busulfan for Allogeneic Transplantation for Aggressive Lymphomas [NCT04220008]Phase 230 participants (Anticipated)Interventional2023-06-01Not yet recruiting
An Open-label, Phase II, Two-stage, Study of Bisantrene(Xantrene) in Combination With Fludarabine and Clofarabine as Salvage Therapy for Adult Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) [NCT04989335]Phase 229 participants (Anticipated)Interventional2021-08-02Recruiting
A Reduced Intensity Conditioning With Clofarabine Antithymocyte Globulin and Total Lymphoid Irradiation Followed by Allogeneic Hematopoietic Stem Cell Transplantation [NCT00697684]Phase 129 participants (Actual)Interventional2008-06-30Completed
Phase II Study of Clofarabine in Combination With Low Dose Cytarabine for Untreated Patients With Poor Risk Myelodysplastic Syndromes [NCT01302106]Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to For increased induction mortality in older patients due to specific combinations of drugs, other clinical factors such as patient age and comorbidities.)
A Phase II Study of Single Agent Clofarabine in Previously Untreated Older Adult Patients With Acute Myelogenous Leukemia (AML) for Whom Standard Induction Chemotherapy is Unlikely to be of Benefit [NCT00373529]Phase 2116 participants (Actual)Interventional2006-10-31Completed
AML08: A Phase II Randomized Trial of Clofarabine Plus Cytarabine Versus Conventional Induction Therapy And A Phase II Study Of Natural Killer Cell Transplantation In Patients With Newly Diagnosed Acute Myeloid Leukemia [NCT00703820]Phase 3324 participants (Actual)Interventional2008-08-04Completed
Phase II Study of Clofarabine and High-Dose Melphalan Conditioning Prior to Allogeneic Hematopoietic Cell Transplantation for Myelodysplasia or Acute Leukemia in Remission [NCT01885689]Phase 272 participants (Actual)Interventional2014-02-10Active, not recruiting
A Dose Escalation Phase I/II Study of Clofarabine Plus Cytarabine With Growth Factor Priming in Patients Who Are Not Felt to be Candidates for More Aggressive Treatment, With Int-2 and High-Risk MDS [NCT00503880]Phase 1/Phase 22 participants (Actual)Interventional2007-05-07Terminated(stopped due to Genzyme discontinued Funding)
A Phase I Feasibility Study Assessing the Role of Maintenance Oral Clofarabine in Older Adults With Acute Myeloid Leukemia [NCT01065545]Phase 10 participants (Actual)Interventional2011-06-30Withdrawn(stopped due to Due to funding issues.)
A Phase III Randomized, Double-blind, Controlled Study Comparing Clofarabine and Cytarabine Versus Cytarabine Alone in Adult Patients 55 Years and Older With Acute Myelogenous Leukemia (AML) Who Have Relapsed or Are Refractory After Receiving up to Two Pr [NCT00317642]Phase 3326 participants (Actual)Interventional2006-08-31Completed
A Phase 1/2 Trial of Uproleselan Combined With High Dose Busulfan Pre-Transplant Conditioning in Hematopoietic Stem Cell Transplantation for Patients With Chemotherapy Resistant Acute Myeloid Leukemia [NCT05569512]Phase 1/Phase 228 participants (Anticipated)Interventional2022-10-06Recruiting
Fludarabine/Clofarabine/Busulfan Combined With SAHA in Patients Receiving Allogeneic Hematopoietic Stem Cell Transplantation for Acute Leukemia [NCT02083250]Phase 170 participants (Actual)Interventional2014-03-06Completed
A Phase I Study of the Histone Deacetylase Inhibitor Entinostat (SNDX-275, NSC 706995) Plus Clofarabine for Philadelphia Chromosome-Negative, Poor Risk Acute Lymphoblastic Leukemia or Bilineage/Biphenotypic Leukemia in Newly Diagnosed Older Adults or in A [NCT01132573]Phase 127 participants (Actual)Interventional2010-04-30Completed
A Randomized Study of Once Daily Fludarabine-Clofarabine Versus Fludarabine Alone Combined With Intervenous Busulfan Followed by Allogeneic Hemopoietic Stem Cell Transplantation for Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) [NCT01471444]Phase 3256 participants (Actual)Interventional2011-11-02Completed
Phase II Study Testing Prophylaxis Feasibility of Graft Versus Host Disease With Only High Dose Cyclophosphamide Post-transplantation for Patients Eligible to a Reduced-intensity Conditioning Regiment Prior to Allogenic Transplantation With a Compatible F [NCT03263767]Phase 247 participants (Actual)Interventional2018-01-15Terminated(stopped due to Security criteria (MTD))
The Biodistribution of [18F]-Clofarabine in Cancer Patients Before and After Interventions That Increase the Activity of Deoxycytidine Kinase [NCT02888301]12 participants (Actual)Observational2015-06-12Terminated(stopped due to slow accrual)
Clofarabine-melphalan-alemtuzumab Conditioning in Patients With Advanced Hematologic Malignancies [NCT00943592]Phase 1/Phase 282 participants (Actual)Interventional2006-03-31Completed
Phase II Trial of Clofarabine With Parenteral Busulfan (Busulfex®) Followed by Allogeneic Related or Unrelated Donor Transplantation for the Treatment of Hematologic Malignancies and Diseases [NCT00852163]Phase 220 participants (Actual)Interventional2007-03-31Completed
Phase I Dose-Escalation Trial of Clofarabine Followed by Escalating Doses of Fractionated Cyclophosphamide in Children With Relapsed or Refractory Acute Leukemias [NCT00852709]Phase 135 participants (Anticipated)Interventional2007-09-01Terminated(stopped due to Lack of accrual)
Conditioning for Graft Failure After Hematopoietic Stem Cell Transplantation [NCT00617929]Phase 212 participants (Actual)Interventional2008-01-31Terminated(stopped due to Replaced by another protocol)
18F-Clofarabine (CFA) as a PET Imaging Agent to Measure Deoxycytidine Kinase (DCK) Activity in Metastatic Cancer, as a Candidate Predictive Biomarker for Response to DCK-dependent Drugs Such as Gemcitabine [NCT05065736]Early Phase 14 participants (Anticipated)Interventional2022-04-27Recruiting
A Phase I/II Open-Label Study of ECT-001-Expanded Cord Blood Transplantation in Pediatric and Young Adult (<21year) Patients With High-Risk and Very High-Risk Myeloid Malignancies [NCT04990323]Phase 1/Phase 212 participants (Anticipated)Interventional2021-12-01Recruiting
A Phase II, Open-Label Study of Clofarabine in Paediatric Patients With Refractory/Relapsed Acute Lymphoblastic Leukaemia [NCT00930098]Phase 274 participants (Actual)Interventional2003-12-31Completed
Clofarabine in Combination With Cytarabine (Ara-C) and G-CSF Priming Followed by Infusion of Ex Vivo Expanded Cord Blood Progenitors for Patients With AML [NCT01031368]Phase 129 participants (Actual)Interventional2009-12-31Completed
Total Therapy Study XVI for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia [NCT00549848]Phase 3600 participants (Actual)Interventional2007-10-29Completed
Phase I Feasibility Study of Clofarabine and Low Dose Total Body Irradiation (TBI) as a Non-myeloablative Preparative Regimen for Stem Cell Transplantation (SCT) for Hematologic Malignancies [NCT01041508]Phase 136 participants (Anticipated)Interventional2010-02-28Completed
S0910, A Phase II Study of Epratuzumab (NSC-716711) in Combination With Cytarabine and Clofarabine for Patients With Relapsed or Refractory Ph- Negative Precursor B-Cell Acute Lymphoblastic Leukemia [NCT00945815]Phase 235 participants (Actual)Interventional2010-09-30Completed
A Pilot Pharmacokinetic, Pharmacodynamic and Feasibility Study of Sorafenib in Combination With Cytarabine and Clofarabine in Patients With Refractory or Relapsed Hematologic Malignancies [NCT00908167]Phase 144 participants (Actual)Interventional2009-09-30Completed
An Open Label Phase II Trial of Clofarabine and Temsirolimus in Older Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) [NCT00775593]Phase 260 participants (Actual)Interventional2008-12-31Completed
A Phase I Study of Clofarabine Plus High Dose Melphalan as a Conditioning Regimen for Allogeneic Transplantation [NCT00641030]Phase 120 participants (Actual)Interventional2007-07-31Completed
Allogeneic Hematopoietic Cell Transplantation Using α/β+ T-lymphocyte Depleted Grafts From HLA Mismatched Donors [NCT03615105]Phase 29 participants (Actual)Interventional2018-07-25Active, not recruiting
A Phase II Trial of Clofarabine in Older Patients With Acute Myeloid Leukemia for Whom Intensive Chemotherapy is Not Considered Suitable [NCT00924443]Phase 269 participants (Actual)Interventional2004-06-30Completed
Pilot Study of Haploidentical Natural Killer Cell Infusions for Poor Prognosis Non-AML Hematologic Malignancies [NCT00697671]Phase 148 participants (Actual)Interventional2007-03-31Completed
Haploidentical Hematopoietic Stem Cell Transplantation Using A Novel Clofarabine Containing Conditioning Regimen For Patients With Refractory Hematologic Malignancies [NCT00824135]Phase 134 participants (Actual)Interventional2009-01-31Completed
A Phase II Trial of Cytarabine and Clofarabine in Patients With Relapsed or Refractory Acute Lymphoblastic Leukemia (ALL) [NCT00337168]Phase 236 participants (Actual)Interventional2006-10-31Completed
Busulfan-Fludarabine-Clofarabine With Allogeneic Stem Cell Transplantation for Advanced Refractory Acute Myeloid Leukemia, Myelodysplastic Syndrome, and Advanced, Gleevec Refractory Chronic Myeloid Leukemia. A Randomized Phase II Study. [NCT00469014]Phase 272 participants (Actual)Interventional2006-09-30Completed
Clofarabine Based Remission Induction Followed by Haploidentical Stem Cell Transplantation in Children With Refractory Hematological Malignancies [NCT01025778]Phase 27 participants (Actual)Interventional2009-12-31Completed
A Phase 1, Open-Label, Multi-Center Safety and Tolerability Pilot Combination Study of Clofarabine, Etoposide, Cyclophosphamide, PEG-asparaginase, and Vincristine in Pediatric Patients With Acute Lymphoblastic Leukemia (ALL) in First Relapse [NCT00991133]Phase 18 participants (Actual)Interventional2009-10-31Completed
Clofarabine in Combination With a Standard Remission Induction Regimen (AraC and Idarubicin) in Patients 18-60 Years Old With Previously Untreated Intermediate and Bad Risk Acute Myelogenous Leukemia (AML) or High Risk Myelodysplasia (MDS) : a Phase I-II [NCT00838240]Phase 1/Phase 2114 participants (Anticipated)Interventional2008-11-30Recruiting
A Phase II Study Evaluating Mechanisms of Resistance Following Treatment With Clofarabine and Daunorubicin in Newly Diagnosed Adult Acute Myeloid Leukemia Patients > or = to 60 Years Old [NCT00814164]Phase 221 participants (Actual)Interventional2008-12-31Terminated(stopped due to Sponsor withdrew support)
Phase I Study of Oral Clofarabine for the Treatment of Patients With Relapsed/Refractory Chronic Lymphocytic Leukemia [NCT00800566]Phase 10 participants (Actual)Interventional2008-11-30Withdrawn(stopped due to Sponsor terminated.)
A Pilot Study of IV Clofarabine for Patients With Myelodysplastic Syndrome Who Have Failed 5-azacytidine [NCT00700011]Phase 210 participants (Actual)Interventional2008-03-31Terminated(stopped due to poor accrual)
A Phase II Trial of Transplants From HLA-Compatible Related or Unrelated Donors With CD34+ Enriched, T-cell Depleted Peripheral Blood Stem Cells Isolated by the CliniMACS System in the Treatment of Patients With Hematologic Malignancies and Other Lethal H [NCT01119066]Phase 2422 participants (Actual)Interventional2010-05-03Completed
Phase I Study of Oral Clofarabine Consolidation in Adults Aged 60 and Older With Acute Myeloid Leukemia [NCT00727766]Phase 125 participants (Actual)Interventional2009-01-31Completed
A Randomized Study of Decitabine Alternating With Clofarabine Versus Decitabine Until Failure in Patients With Higher Risk Myelodysplastic Syndromes (MDS) [NCT00903760]Phase 242 participants (Actual)Interventional2010-01-31Completed
Clofarabine, Etoposide, and Mitoxantrone for Relapsed and Refractory Acute Leukemias [NCT00882076]Phase 122 participants (Actual)Interventional2009-03-31Terminated(stopped due to study closed prematurely)
A Phase I/II Study of CLOLAR® (Clofarabine, IND# 73, 789) in Combination With Cytarabine in Pediatric Patients With Refractory/Relapsed Leukemia [NCT00372619]Phase 1/Phase 274 participants (Actual)Interventional2007-03-31Completed
Clofarabine, Idarubicin, and Cytarabine Combination as Induction Therapy for Younger Patients With Acute Myeloid Leukemia (AML) [NCT01025154]Phase 263 participants (Actual)Interventional2010-01-31Completed
Phase II, Multi-center, and Pharmacokinetic Study of Efficacy, Safety and Pharmacokinetic of Clofarabine in Chinese Pediatric Patients With Refractory or Relapsed Acute Lymphoblastic Leukemia [NCT02544789]Phase 244 participants (Actual)Interventional2009-06-30Completed
Clofarabine, Cyclophosfamide, And Etoposide For The Treatment Of Relapsed Or Resistant Acute Leukemia In Pediatric Patients [NCT01385891]Phase 2/Phase 340 participants (Actual)Interventional2008-08-31Completed
Clofarabine Plus Low-Dose Cytarabine Induction Followed by Consolidation of Clofarabine Plus Low-Dose Cytarabine Alternating With Decitabine in Frontline Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS) [NCT00778375]Phase 2122 participants (Actual)Interventional2008-10-31Completed
A Randomized, Multi-Center Phase III Trial Comparing Two Conditioning Regimens (CloFluBu and BuCyMel) in Children With Acute Myeloid Leukemia Undergoing Allogeneic Stem Cell Transplantation. [NCT05477589]Phase 3170 participants (Anticipated)Interventional2022-06-07Recruiting
A Phase I Study Evaluating the Safety and Tolerability of Oral Clofarabine in Intermediate to High Risk Myelodysplastic Patients [NCT01003678]Phase 14 participants (Actual)Interventional2009-10-31Terminated(stopped due to Drug provider withdrew support)
A Phase II Open-label, Multicenter, Non Randomized Study Evaluating the Efficacy and the Safety of Clofarabine in Combination With IV Busulfan and Thymoglobulin (CBT) as a Reduced Intensity Conditioning Regimen Prior to Allogeneic Stem Cell Transplantatio [NCT00863148]Phase 230 participants (Actual)Interventional2009-10-31Completed
Clofarabine in Combination With Cytarabine and Total Body Irradiation Followed by Allogeneic Stem Cell Transplantation in Children With Acute Lymphoblastic Leukemia and Acute Non-Lymphoblastic Leukemia [NCT00529360]Phase 1/Phase 232 participants (Actual)Interventional2007-06-30Completed
Phase II Study of Clofarabine in Patients With Recurrent or Refractory Langerhans Cell Histiocytosis and LCH-related Disorders [NCT02425904]Phase 225 participants (Actual)Interventional2015-05-31Active, not recruiting
A Phase 1/2 Dose-Escalation Study of Clofarabine in Combination With Etoposide and Cyclophosphamide in Pediatric Patients With Refractory or Relapsed Acute Leukemias. [NCT00315705]Phase 1/Phase 250 participants (Actual)Interventional2006-03-31Completed
A Phase I/II Multicenter Study of IV Clofarabine in Patients With High-Risk Myelodysplastic Syndrome Who Have Failed Therapy With Azacitidine: the NIDEVOL Study [NCT01063257]Phase 1/Phase 276 participants (Actual)Interventional2010-04-30Completed
Pilot Study Of Haplo-Identical Natural Killer Cell Transplantation For Acute Myeloid Leukemia [NCT00187096]49 participants (Actual)Interventional2005-09-30Completed
Phase II Study of Oral Clofarabine in Myelodysplastic Syndrome (MDS) [NCT00299156]Phase 265 participants (Actual)Interventional2006-03-31Completed
A Phase I Trial of Bendamustine in Combination With Clofarabine and Etoposide in Pediatric Patients With Relapsed or Refractory Hematologic Malignancies [NCT01900509]Phase 116 participants (Actual)Interventional2013-08-31Completed
A Phase I and Pharmacokinetic Study of Clofarabine in Adult Patients With Advanced Solid Tumors [NCT00125840]Phase 10 participants Interventional2002-08-31Completed
A Phase II Study of Clofarabine in Patients With Relapsed or Refractory Multiple Myeloma [NCT00241371]Phase 28 participants (Actual)Interventional2005-03-31Terminated(stopped due to No response seen in patients)
Personalized NK Cell Therapy in CBT [NCT02727803]Phase 2100 participants (Anticipated)Interventional2016-05-19Recruiting
A Phase I/IIa, Open-label, Non-randomized, Study of ASC-101 in Patients With Hematologic Malignancies or Myelodysplastic Syndrome (MDS) Who Are Candidates for Dual-cord Umbilical Cord Blood Transplantation (UCBT) [NCT01983761]Phase 1/Phase 225 participants (Anticipated)Interventional2013-11-30Active, not recruiting
Phase III Randomized Trial of Clofarabine as Induction and Post-Remission Therapy vs. Standard Daunorubicin &Amp; Cytarabine Induction and Intermediate Dose Cytarabine Post-Remission Therapy, Followed by Decitabine Maintenance vs. Observation in Newly-Dia [NCT02085408]Phase 3727 participants (Actual)Interventional2011-02-04Active, not recruiting
A Phase 2 Study of Clofarabine, Idarubicin, Cytarabine, Vincristine, and Corticosteroids for Patients With Newly Diagnosed or Relapsed Mixed Phenotype Acute Leukemia [NCT02135874]Phase 218 participants (Actual)Interventional2014-10-27Completed
A Phase I, Open-Label, Dose-Finding and Food Effect Study of Oral Clofarabine in Previously Treated Adult Patients With Myelodysplastic Syndromes (MDS) [NCT00750334]Phase 12 participants (Actual)Interventional2008-09-30Terminated(stopped due to Terminated to focus on comparable trial, CLOMDS02507)
A Nonrandomized Dose-escalation Study of Clofarabine in Combination With Gemtuzumab Ozogamicin for Relapsed/Refractory Acute Myeloid Leukemia (AML) for Patients Less Than 60 Years-old [NCT00577694]Phase 121 participants (Actual)Interventional2007-11-30Terminated(stopped due to funding unavailable)
Lead-In and Phase II Study of Clofarabine, Etoposide, Cyclophosphamide [CEC], Liposomal Vincristine (VCR), Dexamethasone and Bortezomib in Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL) and Lymphoblastic Lymphoma (LL) [NCT03136146]Phase 242 participants (Anticipated)Interventional2017-08-09Recruiting
A Phase II Study of Optimally Dosed Clofarabine in Combination With Low-Dose TBI to Decrease Relapse Rates After Related or Unrelated Donor Hematopoietic Cell Transplantation in Patients With AML [NCT01252667]Phase 244 participants (Actual)Interventional2011-01-25Completed
Busulfan Plus Clofarabine Followed by Allogeneic Hematopoietic Stem Cell Transplantation for Patients With Acute Lymphoblastic Leukemia or Lymphoma, or Biphenotypic Leukemia [NCT00990249]Phase 2120 participants (Actual)Interventional2009-10-31Completed
Thiotepa-Clofarabine-Busulfan With Allogeneic Stem Cell Transplant for High Risk Malignancies [NCT00857389]Phase 260 participants (Actual)Interventional2009-03-02Completed
Phase I/II Study of Plerixafor and Clofarabine in Previously Untreated Older (>/=60 Years) Adult Patients With Acute Myelogenous Leukemia (AML) With Two or More Unfavorable Prognostic Factors for Whom Standard Induction Chemotherapy is Unlikely to be of B [NCT01160354]Phase 1/Phase 222 participants (Actual)Interventional2010-08-31Terminated(stopped due to Slow Accrual)
A Pilot Study of Clofarabine Pre-conditioning Prior to Full or Reduced Intensity Allogeneic Transplantation in the Treatment of High Risk Acute Myeloid Leukaemia and Myelodysplasia. [NCT01422603]Phase 1/Phase 29 participants (Actual)Interventional2011-02-28Completed
Randomized, Multi-centre, Phase II Trial to Compare the Event-Free Survival of Clofarabine / Ara-C (ClAraC) or of FLAMSA Treatment in Patients With High Risk AML or Advanced MDS Scheduled for Allogeneic Stem Cell Transplantation [NCT01423175]Phase 260 participants (Anticipated)Interventional2011-07-31Recruiting
A Phase II Study of Clofarabine in Patients With Aggressive Non-Hodgkin's Lymphoma [NCT00201669]Phase 26 participants (Actual)Interventional2004-10-31Completed
Phase I Trial of the Combination of Bortezomib and Clofarabine in Adults With Refractory Solid Tumors, Lymphomas, or Myelodysplastic Syndromes [NCT02211755]Phase 175 participants (Anticipated)Interventional2014-10-06Recruiting
Dose Escalation Study of Clofarabine in Patients With Relapsed or Refractory Low Grade or Intermed.Grade B-Cell/T-Cell Lymphoma [NCT00338494]Phase 116 participants (Actual)Interventional2005-10-31Completed
Phase I/II Study of Oral Clofarabine + Rituximab in Relapsed B Cell NHL [NCT00691652]Phase 1/Phase 22 participants (Actual)Interventional2008-05-31Terminated(stopped due to Funding ended when only one subject enrolled and subject withdrew early.)
Cord Blood Fucosylation to Enhance Homing and Engraftment in Patients With Hematologic Malignancies [NCT01471067]Phase 133 participants (Actual)Interventional2012-07-13Completed
The Biodistribution of 18F-Clofarabine in Patients With Autoimmune and Inflammatory Diseases [NCT02925351]Early Phase 18 participants (Actual)Interventional2016-01-25Completed
Cord Blood Ex-Vivo MSC Expansion Plus Fucosylation to Enhance Homing and Engraftment [NCT03096782]Phase 26 participants (Actual)Interventional2017-10-13Completed
A Phase II Trial Investigating Clofarabine, Cyclophosphamide and Etoposide for Minimal Residual Disease Positive Acute Leukemia [NCT01677949]Phase 20 participants (Actual)Interventional2013-12-31Withdrawn(stopped due to Slow accrual)
Phase I Study of Weekly Clofarabine for the Treatment of Relapsed/Refractory Chronic Lymphocytic Leukemia [NCT00081887]Phase 18 participants (Actual)Interventional2004-05-31Terminated(stopped due to low accrual)
Phase I Dose-Escalation Trial of Clofarabine Followed by Escalating Doses of Fractionated Cyclophosphamide in Adults and Children With Relapsed or Refractory Acute Leukemias [NCT00293410]Phase 170 participants (Anticipated)Interventional2005-11-30Completed
Phase II Clinical and Pharmacodynamic Investigation of Clofarabine in Acute Leukemias [NCT00098033]Phase 264 participants Interventional2002-09-30Completed
A Programme of Treatment Development for Older Patients With Acute Myeloid Leukemia and High Risk Myelodysplastic Syndrome [NCT00454480]Phase 2/Phase 32,000 participants (Anticipated)Interventional2006-08-31Completed
A Phase I Dose Escalation Trial of Clofarabine in Addition to Topotecan, Vinorelbine, Thiotepa, and Dexamethasone in Pediatric Patients With Relapsed or Refractory Acute Leukemia [NCT00462787]Phase 123 participants (Actual)Interventional2007-04-30Completed
Cord Blood Expansion on Mesenchymal Stem Cells [NCT00498316]Phase 198 participants (Actual)Interventional2007-07-03Completed
Treatment of High Risk, Inherited Lysosomal And Peroxisomal Disorders by Reduced Intensity Hematopoietic Stem Cell Transplantation [NCT00383448]Phase 238 participants (Actual)Interventional2006-09-30Completed
Phase I/II Study of Myeloablative Allogeneic Stem Cell Transplantation for Aggressive Hematologic Malignancies Using Clofarabine and Busulfan x 4 (Clo/BU4) Regimen [NCT00556452]Phase 1/Phase 246 participants (Actual)Interventional2007-10-31Completed
Reduced Intensity Allogeneic Transplantation For Severe Osteopetrosis Incorporating A Second Cd34 Selected Graft [NCT00638820]Phase 23 participants (Actual)Interventional2007-09-30Terminated(stopped due to Excess toxicity)
Study of Oral Clofarabine Plus Low-dose Cytarabine in Previously Treated AML and High-Risk MDS Patients at Least 60 Years of Age [NCT00839982]Phase 1/Phase 235 participants (Actual)Interventional2008-11-30Completed
Treatment of Older Adults With Acute Lymphoblastic Leukemia [NCT00973752]Phase 230 participants (Actual)Interventional2009-08-31Completed
A Pilot Study of Mitoxantrone in Combination With Clofarabine (MITCL) in Children, Adolescents and Young Adults (CAYA) With Refractory/Relapsed Acute Leukemia or High Grade Non-Hodgkin Lymphoma [NCT01842672]Phase 1/Phase 241 participants (Actual)Interventional2013-03-31Completed
A Phase I Trial of Clofarabine in Combination With High-Dose Etoposide and Cyclophosphamide and Autologous Peripheral Blood Stem Cell Transplantation for Patients With High-Risk or Refractory Non-Hodgkin's Lymphoma [NCT00477945]Phase 118 participants (Actual)Interventional2007-05-31Completed
A Phase I Trial of Myeloablative Conditioning Using Clofarabine and High-Dose Busulfan for Patients With Refractory Hematological Malignancies Undergoing Allogeneic HSCT [NCT00477542]Phase 118 participants (Actual)Interventional2007-05-31Completed
Phase I/II Study on Cytarabine and Idarubicin Combined With Escalating Doses of Clofarabine as Induction Therapy in Patients With Acute Myeloid Leukemia and High Risk for Induction Failure (AMLSG 17-10) [NCT01534702]Phase 1/Phase 260 participants (Anticipated)Interventional2012-01-31Recruiting
A Phase II Study of Clofarabine in Combination With Cytarabine (Ara-C) in Patients >/= 50 Years With Newly Diagnosed and Previously Untreated Acute Myeloid Leukemia (AML) and High-risk Myelodysplastic Syndrome (MDS) (>/= 10% Bone Marrow Blasts) [NCT00065143]Phase 260 participants (Actual)Interventional2003-06-23Completed
A Phase II Study of Oral Clofarabine in Patients With Relapsed/Refractory Non-Hodgkin Lymphoma [NCT00644189]Phase 1/Phase 250 participants (Actual)Interventional2008-06-30Completed
A Phase II, Open Label Study of Clofarabine in Pediatric Patients With Refractory or Relapsed Acute Myelogenous Leukemia [NCT00042354]Phase 240 participants Interventional2002-05-31Completed
Phase I Study of CL-F-ARA-A in Solid and Hematologic Malignancies [NCT00028418]Phase 1100 participants Interventional1999-02-28Completed
A Phase I Dose Escalation and Pharmacokinetic Study of Oral Clofarabine Administered Daily for 5 Days in Adult Patients With Refractory Solid Tumors [NCT00125827]Phase 124 participants (Actual)Interventional2003-10-31Completed
A Phase I Study of Clofarabine & Cytosine Arabinoside Therapy for Older Adults With Acute Myeloid Leukemia [NCT00081822]Phase 123 participants (Actual)Interventional2004-01-31Completed
A Phase II, Open-Label Study of Clofarabine in Adult Patients With Refractory or Relapsed Acute Myelogenous Leukemia [NCT00044889]Phase 240 participants Interventional2002-05-31Completed
Phase I/II Study of Clofarabine Plus Cyclophosphamide for Relapsed and Refractory Acute Lymphoblastic Leukemia (ALL) [NCT00412243]Phase 1/Phase 251 participants (Actual)Interventional2006-03-31Completed
Phase II Study of Clofarabine in Patients With Recurrent or Refractory Langerhans Cell Histiocytosis [NCT01796405]Phase 20 participants (Actual)InterventionalWithdrawn
Phase I/II Study of Low-dose Oral Clofarabine for the Treatment of IPSS INT-1, INT-2 or HIGH Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia (Dysplastic Type) Patients Who Have Failed Hypomethylating Therapy [NCT00708721]Phase 1/Phase 211 participants (Actual)Interventional2008-03-31Terminated(stopped due to Data analysis revealed sufficient data for safety and efficacy)
A Non-Myeloablative Conditioning Regimen for Allogeneic Transplantation With Clofarabine, Cytarabine, and Thymoglobulin for Myelodysplastic Syndrome and Acute Myeloid Leukemia [NCT00593645]Phase 27 participants (Actual)Interventional2007-11-30Terminated(stopped due to toxicities were worse than expected)
Dose Escalation Study of Clofarabine in Combination With Cytarabine (Ara-C) and G-CSF Priming for Relapsed or Refractory Acute Myeloid Leukemia (AML) Patients [NCT00602225]Phase 1/Phase 250 participants (Actual)Interventional2007-12-31Completed
Clofarabine Bone Marrow Cytoreduction : Feasibility of Induction as a Bridge to Allogeneic Stem Cell Transplantation for Patients With Relapsed or Refractory Acute Leukemias, Myelodysplastic Syndromes, and Advanced Myeloproliferative Diseases. [NCT00724009]Phase 229 participants (Actual)Interventional2007-12-31Completed
Phase I/II Study of Decitabine (DAC) Followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia [NCT01794702]Phase 1/Phase 265 participants (Actual)Interventional2013-02-20Completed
A Phase I Open-Label Study of High-Dose Cytarabine and Clofarabine in Adult Patients With Refractory or Relapsed Acute Myelogenous Leukemia or Refractory or Relapsed Acute Lymphoblastic Leukemia [NCT00295841]Phase 153 participants (Actual)Interventional2005-02-28Completed
A Phase II Study Of Pooled Unrelated Donor Umbilical Cord Blood (UCB) Transplant For Patients With Hematologic Malignancies Needing Allogeneic Stem Cell Transplant But Do Not Have A Related HLA-Matched Donor [NCT01500161]Phase 21 participants (Actual)Interventional2011-11-30Terminated(stopped due to Insufficient accruals)
Clofarabine Followed by Lenalidomide for Treatment of High Risk Myelodysplastic Syndromes and Acute Myeloid Leukemia [NCT01629082]Phase 14 participants (Actual)Interventional2012-06-06Completed
Clofarabine, Gemcitabine, and Busulfan Followed by Allogeneic Stem Cell Transplantation for Chronic Lymphocytic Leukemia (CLL) [NCT01629511]Phase 1/Phase 215 participants (Actual)Interventional2012-11-21Terminated
A Phase IIa Open-label, Dose Confirmation Study of Oral Clofarabine in Adult Patients Previously Treated for Myelodysplastic Syndromes (MDS) [NCT00531232]Phase 238 participants (Actual)Interventional2007-05-07Completed
Phase II Study of Reduced Intensity Conditioning With Busulfan/Clofarabine Followed by Allogeneic Stem Cell Transplantation [NCT01643668]Phase 234 participants (Actual)Interventional2012-07-31Completed
Total Therapy XVII for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia and Lymphoma [NCT03117751]Phase 2/Phase 3790 participants (Actual)Interventional2017-03-29Active, not recruiting
Treatment of High Risk, Inherited Lysosomal and Peroxisomal Disorders by Reduced-Intensity Hematopoietic Cell Transplantation and Low-Dose Total Body Irradiation With Marrow Boosting by Volumetric-Modulated Arc Therapy (VMAT) [NCT01626092]3 participants (Actual)Interventional2012-07-11Completed
A Phase II Study of Therapy for Pediatric Relapsed or Refractory Precursor B-Cell Acute Lymphoblastic Leukemia and Lymphoma [NCT01700946]Phase 280 participants (Actual)Interventional2013-04-15Completed
A Phase II Open-Label Study of High-Dose Cytarabine and Clofarabine in Adult Patients With Refractory or Relapsed Acute Myelogenous Leukemia or Refractory or Relapsed Acute Lymphoblastic Leukemia [NCT01656031]Phase 239 participants (Actual)Interventional2005-02-28Completed
T-Cell Replete Haploidentical Donor Hematopoietic Stem Cell Plus Natural Killer (NK) Cell Transplantation in Patients With Hematologic Malignancies Relapsed or Refractory Despite Previous Allogeneic Transplant [NCT01621477]Phase 234 participants (Actual)Interventional2012-08-31Terminated(stopped due to Study was terminated in August 2016 due to replacement by a new study.)
T2007-002 A Phase II Study of Clofarabine With Etoposide and Cyclophosphamide in Relapsed/Refractory AML (IND 104,650) [NCT00939653]Phase 26 participants (Actual)Interventional2009-07-10Terminated(stopped due to Due to insufficient research institution participation and patient enrollment)
A Phase I/II Study of Alemtuzumab and Clofarabine for Relapsed or Refractory Acute Lymphoblastic Leukemia [NCT00983528]Phase 1/Phase 26 participants (Actual)Interventional2009-09-30Terminated(stopped due to Support withdrawn due to slow accrual)
A Phase II, Open Label Study of Clofarabine in Pediatric Patients With Refractory or Relapsed Acute Lymphoblastic Leukemia [NCT00042341]Phase 260 participants Interventional2002-05-31Completed
A Phase II Study Incorporating Panobinostat, Bortezomib and Liposomal Vincristine Into Re-Induction Therapy for Relapsed Pediatric T-Cell Acute Lymphoblastic Leukemia or Lymphoma [NCT02518750]Phase 23 participants (Actual)Interventional2016-11-23Terminated(stopped due to Due to slow accrual)
A Prospective Randomized Phase I/II Study of Clofarabine (Clo) and Ara-C vs Clo and Ida vs Clo Plus Ida and Ara-C in Patients With First Relapse or First Salvage of Primary Refractory AML; and High-Grade MDS(>/= 10% Blasts); or CML in Myeloid Blasts Phase [NCT00067028]Phase 2116 participants (Actual)Interventional2003-12-31Completed
Clofarabine With Cytarabine for MRD Positive Leukemia [NCT01158885]Phase 22 participants (Actual)Interventional2010-08-31Terminated(stopped due to Study terminated for lack of accrual.)
Phase I/II Randomized Study of Clofarabine, Idarubicin, and Cytarabine (CIA) Versus Fludarabine, Idarubicin, and Cytarabine (FLAI) in Acute Myelogenous Leukemia and High-Risk Myelodysplastic Syndrome [NCT01289457]Phase 1/Phase 2282 participants (Actual)Interventional2011-02-02Completed
A Phase I/II Open-label Study of Clofarabine in Patients With Relapsed or Refractory Diffuse Large Cell B-Cell NHL [NCT00156013]Phase 1/Phase 233 participants (Actual)Interventional2005-09-30Completed
A Pilot Trial of Clofarabine Added to Standard Busulfan and Fludarabine for Conditioning Prior to Allogeneic Hematopoietic Cell Transplantation [NCT01596699]Phase 216 participants (Actual)Interventional2012-05-24Terminated(stopped due to Triggering of futility rule; Interim analysis suggested no statistical superiority over historic controls)
Clofarabine Plus Low-Dose Cytarabine for the Treatment of Patients With Higher-Risk Myelodysplastic Syndrome (MDS) Who Have Been Relapsing After, or Are Refractory to, Hypomethylator Therapy [NCT01444742]Phase 281 participants (Actual)Interventional2011-11-16Completed
Clofarabine, Cytarabine and Mitoxantrone (CLAM) for Relapsed or Refractory AML [NCT02686593]Phase 250 participants (Actual)Interventional2016-02-01Completed
Randomized Phase II Study of Clofarabine Alone Versus Clofarabine in Combination With Low-Dose Cytarabine in Previously Untreated Patients >= 60 Years With AML and High-Risk MDS [NCT00088218]Phase 295 participants (Actual)Interventional2004-07-31Completed
Phase II Trial of Clofarabine and Cytarabine in Relapsed Standard-Risk AML and Untreated High-Risk MDS in Adult Patients, and Untreated AML in Selected Elderly Patients at High Risk of Anthracycline Toxicity [NCT00334074]Phase 230 participants (Actual)Interventional2005-08-31Completed
Phase 2 Study Of Clofarabine With High Dose Cytarabine And G-CSF Priming In Adult Patients Less Than Age 65 With Newly Diagnosed Acute Myeloid Leukemia Or Advanced Myelodysplastic Syndrome and/or Advanced Myeloproliferative Neoplasm [NCT01101880]Phase 250 participants (Actual)Interventional2010-08-31Completed
A Phase II Study of Clofarabine and Cytarabine for Patients With Newly Diagnosed Acute Myeloid Leukemia Who Have Persistent Disease After Treatment With an Anthracycline and Cytarabine [NCT01960387]Phase 22 participants (Actual)Interventional2013-10-31Terminated(stopped due to lack of accrual)
Gemcitabine/Clofarabine/Busulfan and Allogeneic Transplantation for Aggressive Lymphomas [NCT01701986]Phase 1/Phase 280 participants (Anticipated)Interventional2012-10-25Active, not recruiting
NHL16: Study For Newly Diagnosed Patients With Acute Lymphoblastic Lymphoma [NCT01451515]Phase 223 participants (Actual)Interventional2012-05-25Completed
Allogeneic Hematopoietic Stem Cell Transplantation as Initial Salvage Therapy for Patients With Primary Induction Failure Acute Myeloid Leukemia Refractory to High-Dose Cytarabine-Based Induction Chemotherapy [NCT02441803]Phase 211 participants (Actual)Interventional2015-09-14Active, not recruiting
A Randomized Multi-Center Treatment Study (COALL 08-09) to Improve the Survival of Children With Acute Lymphoblastic Leukemia on Behalf of the German Society of Pediatric Hematology and Oncology [NCT01228331]Phase 2/Phase 3745 participants (Actual)Interventional2010-10-31Active, not recruiting
A Phase I/II Study of Clofarabine in Patients With Relapsed T-Cell and NK-Cell Lymphomas [NCT00416351]Phase 1/Phase 229 participants (Actual)Interventional2006-06-27Completed
FLUCLORIC: Randomized Multicentric Phase III Study Comparing the Efficacy of 2 Reduced Intensity Conditioning Regimens (Clofarabine/Busulfan vs Fludarabine/Busulfan) in Adults With AML and Eligible to Allogeneic Stem Cell Transplantation [NCT05917405]Phase 2302 participants (Anticipated)Interventional2023-09-14Recruiting
Multi-center Single Arm Phase II Study of Myeloablative Allogeneic Stem Cell Transplantation for Non-remission Acute Myeloblastic Leukemia (AML) Using Clofarabine and Busulfan x 4 (CloBu4) Regimen [NCT01457885]Phase 275 participants (Actual)Interventional2011-11-30Completed
"A Phase II Study With a Sequential Clofarabine-cyclophosphamide Combination Schedule as Salvage Therapy for Refractory and Relapsed Acute Lymphoblastic Leukemia (ALL) in Adult Patients" [NCT01462253]Phase 235 participants (Actual)Interventional2012-10-31Completed
Phase II Randomized Study of Two Different Schedules of Intravenous Clofarabine in Myelodysplastic Syndrome (MDS) [NCT00422032]Phase 260 participants (Actual)Interventional2006-01-31Completed
A Phase II Randomized Study to Assess Outcomes With Treosulfan-Based Versus Clofarabine-Based Conditioning in Patients With Myelodysplastic Syndromes With Excess Blasts (MDS-EB), or Acute Myeloid Leukemia (AML) Undergoing Allogeneic Hematopoietic Cell Tra [NCT04994808]Phase 280 participants (Anticipated)Interventional2023-08-11Recruiting
A Phase III Randomized Trial for Newly Diagnosed High Risk B-Lymphoblastic Leukemia (B-ALL) Including a Stratum Evaluating Dasatinib (NSC#732517) in Patients With Ph-like Tyrosine Kinase Inhibitor (TKI) Sensitive Mutations [NCT02883049]Phase 35,937 participants (Actual)Interventional2012-02-29Active, not recruiting
Clofarabine and Non-Myeloablative Allogeneic Hematopoietic Transplantation [NCT00626626]Phase 1/Phase 28 participants (Actual)Interventional2007-05-31Terminated(stopped due to Investigator decision)
A Phase I Dose Escalation Study of Clofarabine Given in Combination With Multi-agent Therapy for Remission Induction in Pediatric Patients With Acute Lymphoblastic Leukemia in First Relapse or Refractory to First Line Therapy - [NCT01279096]Phase 120 participants (Actual)Interventional2010-01-31Completed
A Phase II Trial of Clofarabine and Cytarabine to Treat Minimal Residual Disease (MRD) in Acute Myeloid Leukemia [NCT00863434]Phase 22 participants (Actual)Interventional2009-02-28Terminated
Cord Blood Transplantation in Children and Young Adults With Hematologic Malignancies and Non-malignant Disorders [NCT04644016]Phase 231 participants (Anticipated)Interventional2020-11-20Recruiting
Clofarabine Pre-conditioning Followed by Hematopoietic Stem Cell Transplant With Post-Transplant Cyclophosphamide for Non-remission Acute Myeloid Leukemia [NCT04002115]Phase 22 participants (Actual)Interventional2020-06-03Terminated(stopped due to terminated due to low accrual)
Bridging Study: A Phase 2 Study Investigating Clofarabine, Cyclophosphamide and Etoposide for Children, Adolescents, and Young Adults (AYA) With Acute Leukemia and Minimal Residual Disease [NCT02349178]Phase 26 participants (Actual)Interventional2014-12-08Terminated(stopped due to Low accrual)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00067028 (2) [back to overview]Overall Response Rate (ORR)
NCT00067028 (2) [back to overview]Participants With a Response
NCT00088218 (1) [back to overview]Number of Participants With Response
NCT00156013 (1) [back to overview]Phase I Maximum Tolerated Dose
NCT00187096 (10) [back to overview]Percent of Detectable Donor NK Cells at Day 28
NCT00187096 (10) [back to overview]Number of Participants With Evidence of NK Cells Lysing a Target Cell Line (K562)
NCT00187096 (10) [back to overview]Day That Maximum NK Cell Engraftment Was Reached
NCT00187096 (10) [back to overview]Duration of Engraftment of Natural Killer (NK) Cells
NCT00187096 (10) [back to overview]Number of Patients Experiencing Grade 3 or 4 Toxicities During Conditioning and up to 100 Days Post-transplant
NCT00187096 (10) [back to overview]Overall Survival
NCT00187096 (10) [back to overview]Percent of Peak NK Cell Chimerism
NCT00187096 (10) [back to overview]Proportion of Patients Experiencing Grade 3 or 4 Toxicities During Conditioning and up to 100 Days Post-transplant
NCT00187096 (10) [back to overview]Relapse-free Survival
NCT00187096 (10) [back to overview]Number of KIR-mismatched NK Cells
NCT00299156 (1) [back to overview]Participants With a Complete Remission (CR)
NCT00315705 (16) [back to overview]Percentage of Participants Achieving A Response Over the First Two Treatment Cycles in Phase 2
NCT00315705 (16) [back to overview]Summary of Participants With Adverse Events (AEs) in Phase 1
NCT00315705 (16) [back to overview]Summary of Participants With Adverse Events (AEs) in Phase 2
NCT00315705 (16) [back to overview]Percentage of Participants Achieving A Response Over the First Two Treatment Cycles in Phase 1
NCT00315705 (16) [back to overview]Kaplan Meier Estimate of Duration of Remission (DOR) for Participants Who Achieved Overall Remission (OR) in Phase 1
NCT00315705 (16) [back to overview]Time to Remission for Participants Who Had a Response in Phase 2
NCT00315705 (16) [back to overview]Time to Remission for Participants Who Had a Response in Phase 1
NCT00315705 (16) [back to overview]Participants With Dose Limiting Toxicity in Phase 1
NCT00315705 (16) [back to overview]Number of Participants With 4-month Event Free Survival in Phase 2
NCT00315705 (16) [back to overview]Number of Participants With 4-month Event Free Survival in Phase 1
NCT00315705 (16) [back to overview]Maximum Tolerated Dose (MTD) in Phase 1
NCT00315705 (16) [back to overview]Kaplan Meier Estimates of Overall Survival (OS) for Participants in Phase 2
NCT00315705 (16) [back to overview]Kaplan Meier Estimates of Overall Survival (OS) for Participants in Phase 1
NCT00315705 (16) [back to overview]Kaplan Meier Estimates of Event-free Survival (EFS) for Participants in Phase 2
NCT00315705 (16) [back to overview]Kaplan Meier Estimates of Event-free Survival (EFS) for Participants in Phase 1
NCT00315705 (16) [back to overview]Kaplan Meier Estimate of Duration of Remission (DOR) for Participants Who Achieved Overall Remission (OR) in Phase 2
NCT00317642 (12) [back to overview]Four-Month Event-free Survival Per IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Four-Month Event-free Survival Per IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)
NCT00317642 (12) [back to overview]Overall Survival - Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Overall Survival - Overall and by Randomized Strata (CSR 9-July-12)
NCT00317642 (12) [back to overview]Best Response Per Independent Response Review Panel (IRRP) Assessment - Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Participants With Adverse Events (CSR 7-April-11)
NCT00317642 (12) [back to overview]Disease-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)
NCT00317642 (12) [back to overview]Disease-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Duration of Remission (DOR) Per IRRP Assessment-Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Duration of Remission (DOR) Per IRRP Assessment-Overall and by Randomized Strata (CSR 9-July-12)
NCT00317642 (12) [back to overview]Event-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)
NCT00317642 (12) [back to overview]Event-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)
NCT00334074 (2) [back to overview]Response Rate (Complete Response [CR] Plus Partial Response [PR]) of Clofarabine Plus Cytarabine in Patients With Relapsed/Refractory AML, Untreated MDS, CML in Blast Phase, or in Selected Untreated Patients With High Risk of Anthracycline Toxicity
NCT00334074 (2) [back to overview]Number of Participants Who Had an Adverse Event While on Treatment With Clofarabine Plus Cytarabine
NCT00337168 (4) [back to overview]Expression of Nucleoside Transporters
NCT00337168 (4) [back to overview]Toxicity
NCT00337168 (4) [back to overview]Number of Patients With Complete Remission
NCT00337168 (4) [back to overview]Number of Patients With Very Poor Risk Cytogenetics
NCT00372619 (2) [back to overview]Safety and Tolerability as Measured by CTCAE v3.0
NCT00372619 (2) [back to overview]Overall Response (CR for ALL Patients), (CR + CRp for AML Patients)
NCT00373529 (7) [back to overview]Kaplan Meier Estimate for Duration of Remission (DOR)
NCT00373529 (7) [back to overview]Kaplan Meier Estimates for Overall Survival (OS)
NCT00373529 (7) [back to overview]Percentage of Participants Who Died Within Thirty Days of Treatment (30-day Mortality Rate)
NCT00373529 (7) [back to overview]Number of Participants Achieving Overall Remission After A Maximum of Two Cycles by Subgroup of Baseline Prognostic Factors
NCT00373529 (7) [back to overview]Overall Participant Counts Summarizing Adverse Events (AEs) During the Treatment and Follow-up Periods
NCT00373529 (7) [back to overview]Percentage of Participants Achieving Overall Remission (OR) After No More Than Two Cycles (Approximately Month 2)
NCT00373529 (7) [back to overview]Kaplan Meier Estimate for Disease-free Survival (DFS)
NCT00383448 (4) [back to overview]Number of Patients Whose Death Was Related to the Transplant
NCT00383448 (4) [back to overview]Number of Patients With Acute Graft Versus Host Disease (GVHD)
NCT00383448 (4) [back to overview]Number of Patients With Chronic Graft Versus Host Disease (GVHD)
NCT00383448 (4) [back to overview]Number of Patients With Donor Cell Engraftment
NCT00412243 (1) [back to overview]Maximum Tolerated Dose for Cyclophosphamide (MTD)
NCT00416351 (2) [back to overview]Maximum Tolerated Dose
NCT00416351 (2) [back to overview]Participants Evaluated for Toxicity
NCT00422032 (1) [back to overview]Number of Participants With Response for Two Dose Schedules of Clofarabine
NCT00423514 (1) [back to overview]Participants Evaluated for Early Post-transplant Regimen-related Severe Morbidity (Grade III to IV Nonhematologic Toxicity) and Mortality as Measured by the NCI Cancer Therapy Evaluation Program CTCAE v 3.0
NCT00469014 (1) [back to overview]Treatment Related Mortality
NCT00531232 (14) [back to overview]Number of Participants Who Reported Death Within 30 Days of First Dose
NCT00531232 (14) [back to overview]Number of Participants Who Achieved Hematologic Improvement (HI)
NCT00531232 (14) [back to overview]Maximum Tolerated Dose (MTD) of Oral Clofarabine
NCT00531232 (14) [back to overview]Duration of Response (DoR)
NCT00531232 (14) [back to overview]Number of Participants With Unacceptable Drug-related Toxicities During Cycle 1
NCT00531232 (14) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00531232 (14) [back to overview]Time to Acute Myeloid Leukemia (AML) Transformation
NCT00531232 (14) [back to overview]PK Parameter: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Clofarabine
NCT00531232 (14) [back to overview]PK Parameter: Area Under the Concentration-time Curve From Time 0 to Time of Last Measurable Plasma Concentration (AUC0-last) of Clofarabine
NCT00531232 (14) [back to overview]Pharmacokinetics (PK) Parameter: Maximum Observed Plasma Concentration (Cmax) of Clofarabine
NCT00531232 (14) [back to overview]Percentage of Participants With Overall Response
NCT00531232 (14) [back to overview]Percentage of Participants Achieving Overall Remission (OR)
NCT00531232 (14) [back to overview]Overall Survival (OS)
NCT00531232 (14) [back to overview]Number of Participants With Febrile Neutropenia
NCT00549848 (6) [back to overview]Probability of Event-free Survival
NCT00549848 (6) [back to overview]Proportion of Participants With Minimal Residual Disease (MRD) on the 15th Day of Remission Induction ≥ 5%
NCT00549848 (6) [back to overview]Proportion of Participants With Minimal Residual Disease (MRD) at End of Remission Induction ≥ 0.01%
NCT00549848 (6) [back to overview]Probability of Overall Survival
NCT00549848 (6) [back to overview]Probability of CNS Relapse
NCT00549848 (6) [back to overview]Percentage of Participants With Continuous Complete Remission of Patients Receiving High-dose and Conventional Dose PEG-asparaginase.
NCT00556452 (4) [back to overview]One-year Overall Survival Rate for AML
NCT00556452 (4) [back to overview]Two-year Overall Survival for All Cases.
NCT00556452 (4) [back to overview]Regimen Related Toxicities
NCT00556452 (4) [back to overview]Five Year Overall Survival for All Cases
NCT00593645 (8) [back to overview]Engraftment as Measured by Percent Donor Chimerism
NCT00593645 (8) [back to overview]Engraftment as Measured by Percent Donor Chimerism
NCT00593645 (8) [back to overview]Disease-free Survival
NCT00593645 (8) [back to overview]Median Time to Progression
NCT00593645 (8) [back to overview]Engraftment as Measured by Percent Donor Chimerism
NCT00593645 (8) [back to overview]Overall Survival
NCT00593645 (8) [back to overview]Rate of Acute Graft-versus-host Disease (GVHD)
NCT00593645 (8) [back to overview]Rate of Chronic Graft-versus-host Disease (GVHD)
NCT00602225 (9) [back to overview]Response Rates by Cytogenetic Risk Category
NCT00602225 (9) [back to overview]Disease-free Survival
NCT00602225 (9) [back to overview]Maximum Tolerated Dose of Clofarabine
NCT00602225 (9) [back to overview]Dose-limiting Toxicity as Assessed by NCI CTCAE v3.0
NCT00602225 (9) [back to overview]Efficacy
NCT00602225 (9) [back to overview]Overall Survival
NCT00602225 (9) [back to overview]Response Rates by Cytogenetic Risk Category and Clofarabine Dose
NCT00602225 (9) [back to overview]Response Rates by Duration First Complete Remission (CR1)
NCT00602225 (9) [back to overview]Response Rates by Salvage Number
NCT00617929 (7) [back to overview]Treatment-related Death
NCT00617929 (7) [back to overview]Chimerism
NCT00617929 (7) [back to overview]Acute Graft-vs-host Disease
NCT00617929 (7) [back to overview]Rate of Sustained Donor Engraftment
NCT00617929 (7) [back to overview]Survival
NCT00617929 (7) [back to overview]Survival at 100 Days Post Transplant
NCT00617929 (7) [back to overview]Time to Primary Neutrophil Engraftment
NCT00626626 (1) [back to overview]Engraftment of Allogeneic Blood Cells.
NCT00638820 (3) [back to overview]Number of Patients With Transplant Related Toxicity
NCT00638820 (3) [back to overview]Number of Patients With Transplant Related Death
NCT00638820 (3) [back to overview]Number of Patients Achieving Donor Cell Engraftment
NCT00644189 (9) [back to overview]Phase I Participants: Progression-free Survival (PFS)
NCT00644189 (9) [back to overview]Phase I Participants Only: Overall Response Rate (ORR)
NCT00644189 (9) [back to overview]All Phase I-II Participants: Overall Survival (OS)
NCT00644189 (9) [back to overview]All Phase I-II Participants: Overall Response Rate (ORR)
NCT00644189 (9) [back to overview]All Phase I-II Participants: Progression-free Survival (PFS)
NCT00644189 (9) [back to overview]All Phase I Participants: Overall Survival (OS)
NCT00644189 (9) [back to overview]All Phase I-II Participants: Safety
NCT00644189 (9) [back to overview]Phase I Participants: Safety
NCT00644189 (9) [back to overview]Phase I-II Participants Treated at the RP2D (3mg): Overall Response Rate (ORR)
NCT00700011 (4) [back to overview]To Determine the Non-hematologic Toxicity Profile of This Dose Schedule (Grade 2 and Above)
NCT00700011 (4) [back to overview]Improvement in Peripheral Blood Count and Reduction in Number of Transfusions
NCT00700011 (4) [back to overview]Determine Frequency and Duration of Bone Marrow Responses to IV Clofarabine
NCT00700011 (4) [back to overview]Number of Participants With DNA Hypomethylation During the Study
NCT00703820 (2) [back to overview]Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Alone.
NCT00703820 (2) [back to overview]Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Followed by Natural Killer Cell Transplantation.
NCT00708721 (7) [back to overview]The Overall Response Rate in Response to Low Dose Daily Oral Clofarabine in Patients With High Risk Myelodysplastic Syndrome or Chronic Myelomonocytic Leukemia (Dysplastic Type).
NCT00708721 (7) [back to overview]The Effect of Low Dose Daily Oral Clofarabine on miRNA and mRNA Expression Patterns in Patients With MDS
NCT00708721 (7) [back to overview]The Effect of Low Dose Daily Oral Clofarabine on Global Methylation in Patients With MDS.
NCT00708721 (7) [back to overview]Time to Progression to Acute Myeloid Leukemia (AML)
NCT00708721 (7) [back to overview]Number of Participants Who Experienced a Dose-Limiting Toxicity
NCT00708721 (7) [back to overview]Number of Participants With Adverse Events
NCT00708721 (7) [back to overview]Response of MDS Patients Treated With Low Dose Daily Oral Clofarabine.
NCT00724009 (8) [back to overview]Number of Participants With Hepatic (SGOT) Adverse Events
NCT00724009 (8) [back to overview]Number of Participants With Cardiac Adverse Events
NCT00724009 (8) [back to overview]Leukemia Free Survival
NCT00724009 (8) [back to overview]Cytoreductive Response
NCT00724009 (8) [back to overview]Number of Participants Infection Adverse Events
NCT00724009 (8) [back to overview]Number of Participants With Renal Adverse Events
NCT00724009 (8) [back to overview]Number of Participants With Skin Adverse Events
NCT00724009 (8) [back to overview]Number of Participants With Hepatic (Total Bilirubin) Adverse Events
NCT00775593 (4) [back to overview]Number of Serious Adverse Events Within 2 Years
NCT00775593 (4) [back to overview]Duration of Survival
NCT00775593 (4) [back to overview]Duration of Response
NCT00775593 (4) [back to overview]Complete Response Rate
NCT00778375 (6) [back to overview]Disease-free (DFS) or Relapse-free Survival (RFS) Time
NCT00778375 (6) [back to overview]Complete Remission (CR) Rate for First 60 Participants
NCT00778375 (6) [back to overview]Event Free Survival (EFS)
NCT00778375 (6) [back to overview]Overall Response Rate (CR, CRp/CRi and PR)
NCT00778375 (6) [back to overview]Median Overall Survival (OS)
NCT00778375 (6) [back to overview]Number of Participants With Complete Remission [Complete Response (CR), Complete Response With Platelet Recover (CRp) or Complete Response With Incomplete Marrow Recovery (CRi)]
NCT00814164 (3) [back to overview]Overall Survival
NCT00814164 (3) [back to overview]Disease-free Survival
NCT00814164 (3) [back to overview]Complete Remission (CR)
NCT00839982 (4) [back to overview]Disease Free Survival
NCT00839982 (4) [back to overview]Maximum Tolerated Dose
NCT00839982 (4) [back to overview]Number of Patients With Dose Limiting Toxicity
NCT00839982 (4) [back to overview]Overall Survival
NCT00857389 (7) [back to overview]Relapse Rate of Participants Treated With Thiotepa, Busulfan, and Clofarabine
NCT00857389 (7) [back to overview]Overall Survival Rate
NCT00857389 (7) [back to overview]Number of Participants With Survival Rate at 100 Days Post-transplant
NCT00857389 (7) [back to overview]Number of Participants With Serious Adverse Events
NCT00857389 (7) [back to overview]Number of Participants With Disease Free Survival
NCT00857389 (7) [back to overview]Graft vs Host Disease (GVHD)
NCT00857389 (7) [back to overview]Engraftment
NCT00863434 (3) [back to overview]Overall Survival
NCT00863434 (3) [back to overview]Disease-free Survival
NCT00863434 (3) [back to overview]Minimal Residual Disease as Assessed by Bone Marrow Flow Cytometry
NCT00903760 (2) [back to overview]Event Free Survival (EFS) at 1 Year
NCT00903760 (2) [back to overview]Participant Response
NCT00924443 (5) [back to overview]Duration of Overall Response
NCT00924443 (5) [back to overview]Duration of Complete Remission
NCT00924443 (5) [back to overview]Rate of Response (Complete, Complete With Incomplete Blood Count Recovery, Partial)
NCT00924443 (5) [back to overview]Overall Survival
NCT00924443 (5) [back to overview]Overall Response Rate (ORR)
NCT00939653 (2) [back to overview]Death
NCT00939653 (2) [back to overview]Achievement of Complete Remission (CR) at Reinduction
NCT00943592 (8) [back to overview]Number of Participants With Skin Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation
NCT00943592 (8) [back to overview]Number of Participants With Renal Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation
NCT00943592 (8) [back to overview]Number of Participants With Hepatic Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation
NCT00943592 (8) [back to overview]Treatment-related Mortality (TRM)
NCT00943592 (8) [back to overview]Relapse Rate
NCT00943592 (8) [back to overview]Progression-free Survival (PFS)
NCT00943592 (8) [back to overview]Overall Survival (OS)
NCT00943592 (8) [back to overview]Number of Participants With Other Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation
NCT00945815 (2) [back to overview]Number of Patients With Grade 3 Through 5 Treatment-Related Adverse Events
NCT00945815 (2) [back to overview]Complete Remission
NCT00973752 (1) [back to overview]Overall Survival at One Year
NCT00983528 (1) [back to overview]Number of Adverse Events
NCT00990249 (1) [back to overview]Treatment-Related Mortality (TRM) Defined as Non Relapse Mortality (NRM)
NCT01025154 (2) [back to overview]Median Event-Free Survival (EFS)
NCT01025154 (2) [back to overview]Overall Response: Number of Participants With Complete Remission or Complete Remission Without Platelet Recovery
NCT01101880 (5) [back to overview]Overall Survival
NCT01101880 (5) [back to overview]Event Free Survival
NCT01101880 (5) [back to overview]Duration of Remission
NCT01101880 (5) [back to overview]Time to Progression
NCT01101880 (5) [back to overview]Rates of Complete Remission and Complete Remission With Incomplete Recovery of Counts
NCT01119066 (6) [back to overview]Survival and Disease-free Survival (DFS)
NCT01119066 (6) [back to overview]Survival and Disease-free Survival (DFS)
NCT01119066 (6) [back to overview]Survival and Disease-free Survival (DFS)
NCT01119066 (6) [back to overview]Number of Participants With Acute and Chronic GVHD Following T-cell Depleted, CD34+ Progenitor Cell Enriched Transplants Fractionated by the CliniMACS System.
NCT01119066 (6) [back to overview]Incidence of Non-relapse Mortality (Transplant-related Mortality) Following Each Cytoreduction Regimen and a Transplant Fractionated by the CliniMACS System.
NCT01119066 (6) [back to overview]The Incidence of Durable Hematopoietic Engraftment for T-cell Depleted Transplants Fractionated by the CliniMACS System Administered After Each of the Four Disease Targeted Cytoreduction Regimens.
NCT01158885 (1) [back to overview]Number of Patients With Dose-Limiting Toxicity (DLT)
NCT01160354 (1) [back to overview]Number of Participants in Phase I With First Cycle Dose Limiting Toxicities (DLT) Observed
NCT01252667 (9) [back to overview]Part 2: Number of Participants With Relapsed Disease
NCT01252667 (9) [back to overview]Pharmacokinetics (PK) of Clofarabine
NCT01252667 (9) [back to overview]Pharmacokinetics (PK) of Clofarabine
NCT01252667 (9) [back to overview]Number of Participants With Minimal Residual/Recurring Disease (MRD) Post-transplant
NCT01252667 (9) [back to overview]Number of Participants Surviving Overall
NCT01252667 (9) [back to overview]Part 1: Number of Participants With Dose Limiting Toxicities (DLT)
NCT01252667 (9) [back to overview]Number of Participants Surviving Progression-free.
NCT01252667 (9) [back to overview]Number of Participants Who Graft Rejected.
NCT01252667 (9) [back to overview]Number of Non-Relapse Mortalities (NRM)
NCT01289457 (4) [back to overview]Overall Survival
NCT01289457 (4) [back to overview]Response Rates of Clofarabine, Idarubicin, and Cytarabine (CIA) Versus Fludarabine, Idarubicin, and Cytarabine (FLAI)
NCT01289457 (4) [back to overview]Event-Free Survival (EFS) at 2 Years
NCT01289457 (4) [back to overview]Maximum Tolerated Dose (MTD) of Clofarabine, Idarubicin, and Cytarabine
NCT01444742 (2) [back to overview]Overall Survival (OS)
NCT01444742 (2) [back to overview]Number of Participants With Complete Response (CR)
NCT01451515 (4) [back to overview]Minimal Residual Disease (MRD)
NCT01451515 (4) [back to overview]Probability of Event-free Survival (EFS)
NCT01451515 (4) [back to overview]Minimal Disseminated Disease (MDD)
NCT01451515 (4) [back to overview]Probability of Overall Survival (OS)
NCT01457885 (3) [back to overview]The Percentage of Patients Alive at 1 Year
NCT01457885 (3) [back to overview]Incidence of Relapse
NCT01457885 (3) [back to overview]Cumulative Incidence of Non Relapse Mortality (NRM)
NCT01462253 (6) [back to overview]Cumulative Incidence of Relapse (CIR)
NCT01462253 (6) [back to overview]Number of Participants With Toxicity of Grade 2 or Greater
NCT01462253 (6) [back to overview]The Primary End-point is the Number of Patients in CR After Induction Therapy.
NCT01462253 (6) [back to overview]Overall Survival (OS)
NCT01462253 (6) [back to overview]Number of Participants With Minimal Residual Disease (MRD) Response in Remission.
NCT01462253 (6) [back to overview]Disease-free Survival (DFS)
NCT01471444 (4) [back to overview]Number of Participants in the Study Who Are With no Grade 3 or 4 Acute Graft-versus-host Disease at Any Time During the First 100 Days Post Transplant.
NCT01471444 (4) [back to overview]Overall Survival (OS) Post Transplant at 1, 3 and 5 Years
NCT01471444 (4) [back to overview]Progression-Free Survival (PFS)
NCT01471444 (4) [back to overview]Number of Participants With Non Relapse Mortality at 100 Day Post Transplant
NCT01596699 (3) [back to overview]Number of Participants With Treatment-Related Adverse Events as a Measure of Safety and Tolerability
NCT01596699 (3) [back to overview]Engraftment Rate of Patients With Non-malignant Diseases (Stratum A)
NCT01596699 (3) [back to overview]Mixed-donor Chimerism Rate of Patients With High-risk Myeloid Malignancies (Stratum B)
NCT01621477 (7) [back to overview]Incidence and Severity of Chronic Graft Versus Host Disease (GVHD)
NCT01621477 (7) [back to overview]Incidence and Severity of Acute Graft Versus Host Disease (GVHD)
NCT01621477 (7) [back to overview]Number of Participants With Transplant Related Mortality (TRM)
NCT01621477 (7) [back to overview]One-year Survival (OS)
NCT01621477 (7) [back to overview]Incidence of Malignant Relapse
NCT01621477 (7) [back to overview]Event-Free Survival (EFS)
NCT01621477 (7) [back to overview]Disease-Free Survival (DFS)
NCT01626092 (2) [back to overview]Donor (Allogeneic) Hematopoietic Engraftment
NCT01626092 (2) [back to overview]Transplant-Related Mortality
NCT01629511 (3) [back to overview]100 Day Treatment Related Mortality (TRM)
NCT01629511 (3) [back to overview]Time-to-engraftment
NCT01629511 (3) [back to overview]Overall Survival
NCT01643668 (9) [back to overview]Incidence of Hepatic Veno-occlusive Disease
NCT01643668 (9) [back to overview]Infection-related Complications
NCT01643668 (9) [back to overview]Assessment of Donor Stem Cell Engraftment: ANC Count
NCT01643668 (9) [back to overview]Cumulative Incidence and Severity of Acute GVHD Within 100 Days Post Transplant
NCT01643668 (9) [back to overview]Cumulative Incidence of Non-relapse Mortality
NCT01643668 (9) [back to overview]Donor Stem Cell Engraftment: Platelet Count
NCT01643668 (9) [back to overview]Progression-Free and Overall Survival
NCT01643668 (9) [back to overview]Grade 3 or 4 Toxicities
NCT01643668 (9) [back to overview]Cumulative Incidence of Chronic GVHD at One Year
NCT01656031 (1) [back to overview]Measure Patient Response to High-dose Cytarabine Followed by Clofarabine in Adult Patients With Relapsed or Refractory AML
NCT01700946 (4) [back to overview]Median CD20 Expression Levels
NCT01700946 (4) [back to overview]3-year Event-free Survival Rates in Patients With Relapsed ALL
NCT01700946 (4) [back to overview]Mean of CD20 Expression Levels
NCT01700946 (4) [back to overview]3-year Overall Survival Rate of Patients With Relapsed ALL
NCT01794702 (4) [back to overview]Overall Survival
NCT01794702 (4) [back to overview]Number of Participants With a Response
NCT01794702 (4) [back to overview]Maximum Tolerated Dose (MTD) of Clofarabine
NCT01794702 (4) [back to overview]To Determine the Disease-free Survival (DFS).
NCT01885689 (2) [back to overview]Overall Survival at 2 Years
NCT01885689 (2) [back to overview]Progression-free Survival at 2 Years
NCT01960387 (1) [back to overview]Complete Clinical Response
NCT02085408 (4) [back to overview]Proportion of Patients With Complete Remission
NCT02085408 (4) [back to overview]Overall Survival by Donor Status
NCT02085408 (4) [back to overview]Overall Survival
NCT02085408 (4) [back to overview]Disease-free Survival for Maintenance
NCT02349178 (1) [back to overview]Minimal Residual Disease
NCT02425904 (5) [back to overview]Response Rate (OR) of LCH Cohort
NCT02425904 (5) [back to overview]Number of Participants With at Least One Grade 3 or Higher Treatment-Related Toxicity
NCT02425904 (5) [back to overview]1-year Progression Free Survival (PFS)
NCT02425904 (5) [back to overview]1-year Overall Survival (OS)
NCT02425904 (5) [back to overview]Response Rate of LCH-related Disorders Cohort
NCT03096782 (3) [back to overview]Time to Engraftment
NCT03096782 (3) [back to overview]Disease-free Survival
NCT03096782 (3) [back to overview]Overall Survival
NCT04002115 (7) [back to overview]Complete Remission (CR) Rate at Day 30 Post HSCT
NCT04002115 (7) [back to overview]Neutrophil Engraftment
NCT04002115 (7) [back to overview]Non-relapse Related Mortality
NCT04002115 (7) [back to overview]Rate of Acute Graft-versus-host Disease (GVHD)
NCT04002115 (7) [back to overview]Rate of Chronic GVHD
NCT04002115 (7) [back to overview]Severity of Acute Graft-versus-host Disease (GVHD)
NCT04002115 (7) [back to overview]Severity of Chronic GVHD

Overall Response Rate (ORR)

Percentage of participants with complete response following treatment out of all participants in that particular treatment group. Response assessed by blood test or bone marrow aspirate following day 21 of induction and then every 2 weeks thereafter until remission or non-response. Complete remission (CR): Disappearance all clinical and/or radiologic evidence of disease; Neutrophil count > 1.0 x10^9/L and platelet count >100x10^9/L, and normal bone marrow differential (< 5% blasts). Complete remission without platelet recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of > 20 x 10^9/L and < 100 x 10^9/L. Partial remission (PR): Peripheral blood count recovery as for CR, but with decrease in marrow blasts of > 50% and not more than 6-25% abnormal cells in the marrow. All other responses considered as failures. (NCT00067028)
Timeframe: Up to 6 years

InterventionPercentage of Participants (Number)
Clofarabine + Ara-C36
Clofarabine + Idarubicin44
Clofarabine + Idarubicin + Ara-C24

[back to top]

Participants With a Response

Response assessed by blood test or bone marrow aspirate following day 21 of induction and then every 2 weeks thereafter until remission or non-response. Complete remission (CR): Disappearance all clinical and/or radiologic evidence of disease; Neutrophil count > 1.0 x10^9/L and platelet count >100x10^9/L, and normal bone marrow differential (< 5% blasts). Complete remission without platelet recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of > 20 x 10^9/L and < 100 x 10^9/L. Partial remission (PR): Peripheral blood count recovery as for CR, but with decrease in marrow blasts of > 50% and not more than 6-25% abnormal cells in the marrow. All other responses considered as failures. (NCT00067028)
Timeframe: Up to 6 years

,,
Interventionparticipants (Number)
CRCRpPR
Clofarabine + Ara-C420
Clofarabine + Idarubicin951
Clofarabine + Idarubicin + Ara-C1671

[back to top]

Number of Participants With Response

"Participant responses are categorized as 'Complete Remission,' Complete Remission, No Platelet Recovery,' 'No Response.'~Complete Remission: Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 109/L and platelet count > 100 x 109/L, and normal bone marrow differential (< 5% blasts); Complete Remission, No Platelet Recovery: Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 109/L.~Blood draws once a week until remission then every 2 to 8 weeks during therapy." (NCT00088218)
Timeframe: Every 2 to 8 weeks

,
InterventionParticipants (Number)
Complete RemissionComplete Remission, No Platelet RecoveryNo Response
Clofarabine5011
Clofarabine Plus Ara-C49426

[back to top]

Phase I Maximum Tolerated Dose

Maximum Tolerated Dose for Clofarabine. Cohorts of 3 patients each will receive doses of clofarabine increased in increments as follows: 4, 6, 8, 10, 12,…etc mg/m2/day for 5 days. The dose level immediately below the MTD will be used to treat patients in the Phase II part of the study. Starting dose of 4 mg/m2. (NCT00156013)
Timeframe: days 1 -28, maximum 6 cycles

Interventionmg/m^2 (Number)
Clofarabine6

[back to top]

Percent of Detectable Donor NK Cells at Day 28

The percent of detectable donor NK cells in recipients at 28 days after NK cell infusion. Three of 10 participants had detectable donor cells at week 4. The results report the percent of detectable cells in the 3 participants. (NCT00187096)
Timeframe: At 28 days

Interventionpercent of donor NK cells (Median)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine29

[back to top]

Number of Participants With Evidence of NK Cells Lysing a Target Cell Line (K562)

NK cells in recipient achieving ability to lyse target cell line (K562) within normal range established by donor NK cells. (NCT00187096)
Timeframe: Days 2, 7, 14, 21, and 28 after NK cell transplantation

Interventionparticipants (Number)
Lysed within normal rangeDid not Lyse within normal range
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine100

[back to top]

Day That Maximum NK Cell Engraftment Was Reached

The time elapsed after transplantation in days until peak KIR-mismatched donor NK cell expansion was reached in recipients (NCT00187096)
Timeframe: Day 0 through Day 28 post NK cell transplantation

Interventionnumber of days (Median)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine14

[back to top]

Duration of Engraftment of Natural Killer (NK) Cells

NK cell engraftment defined as NK cell chimerism in recipients. (NCT00187096)
Timeframe: Measured at days 2, 7, 14, 21 and 28 after NK cell transplantation, and up to 189 days post transplant as clinically indicated

InterventionDays (Median)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine10

[back to top]

Number of Patients Experiencing Grade 3 or 4 Toxicities During Conditioning and up to 100 Days Post-transplant

Document the number of patients experiencing grade 3 or 4 toxicities during conditioning and up to 100 days post-transplant. Toxicities were identified using Common Toxicity Criteria V 3.0 criteria. (NCT00187096)
Timeframe: Beginning at on therapy through 100 days post-transplant

Interventionparticipants (Number)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine2
Arm 2a: Conditioning Regimen: Cyclophosphamide/Fludarabine3
Arm 2b: Conditioning: Clofarabine/Etoposide/Cyclophosphamide11

[back to top]

Overall Survival

"Overall survival is defined as the time relapse from on study date to death with those alive at last follow up date censored. The Kaplan-Meier method was used to compute survival probability estimates and confidence interval was determined by binomial distribution (for no events or all events) or by log hazard method. The binomial interval is based on the number of patients at risk.~The confidence intervals for Arm 1 and Arm 2a were determined by binomial distribution.~The confidence interval for Arm 2b was determined by log hazard method." (NCT00187096)
Timeframe: Up to 2 years post NK cell transplantation

InterventionPercent probability (Number)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine100
Arm 2a: Conditioning Regimen: Cyclophosphamide/Fludarabine0
Arm 2b: Conditioning: Clofarabine/Etoposide/Cyclophosphamide45.0

[back to top]

Percent of Peak NK Cell Chimerism

The maximum percent of donor NK cell in recipients during a four-week period after NK cell infusion. (NCT00187096)
Timeframe: Days 2, 7, 14, 21 and 28 after NK cell transplantation

Interventionpercent of NK cells (Median)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine7

[back to top]

Proportion of Patients Experiencing Grade 3 or 4 Toxicities During Conditioning and up to 100 Days Post-transplant

Document the proportion of patients experiencing grade 3 or 4 toxicities during conditioning and up to 100 days post-transplant. Toxicities were identified using Common Toxicity Criteria V 3.0 criteria. (NCT00187096)
Timeframe: Beginning at on therapy through 100 days post-transplant

Interventionproportion of patients (Number)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine0.20
Arm 2a: Conditioning Regimen: Cyclophosphamide/Fludarabine1.00
Arm 2b: Conditioning: Clofarabine/Etoposide/Cyclophosphamide0.917

[back to top]

Relapse-free Survival

For Arm 1, the efficacy of NK cell transplantation will be reported as the proportion of participants who achieve complete or partial remission. Kaplan-Meier estimates of relapse-free survival and confidence interval was determined by binomial distribution because no events were observed. The binomial interval is based on the number of patients at risk. (NCT00187096)
Timeframe: Up to 2 years post NK cell transplantation

InterventionPercent probability (Number)
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine100

[back to top]

Number of KIR-mismatched NK Cells

Number of KIR-mismatched donor NK cells in recipients' blood at day 2 and day 14 post NK cell infusion. (NCT00187096)
Timeframe: Day 2 and day 14 post NK cell transplantation

Interventioncells/µl (Median)
Day 2Day 14
Arm 1: Conditioning Regimen: Cyclophosphamide/Fludarabine2105,800

[back to top]

Participants With a Complete Remission (CR)

"Complete Remission (CR): Normalization of the peripheral blood and bone marrow with <5% bone marrow blasts, a peripheral blood granulocyte count > (1.0 x 109/ L, and a platelet count > 100 x 109/L).~Partial Remission: as above except for the presence of 6-15% marrow blasts, or 50% reduction if <15% at start of treatment.~Hematologic Improvement: meets all criteria for CR except for platelet recovery to >100 x 109/L.~Clinical Benefit: Platelets increase by 50% and to above 30 x 109/L untransfused (if lower than that pretherapy); or granulocytes increase by 100% and to above 109/L (if lower than that pretherapy); or hemoglobin increase by 2 g/dl; or transfusion independent; or splenomegaly reduction by > 50%; or monocytosis reduction by > 50% if pretreatment > 5 x 109/L." (NCT00299156)
Timeframe: After 3 courses of treatment, up to 24 weeks.

,,
InterventionParticipants (Number)
Complete RemissionPartial RemissionHematologic ImprovementClinical Benefit
Oral Clofarabine8033
Randomized, Oral Clofarabine 10mg3010
Randomized, Oral Clofarabine 20mg0030

[back to top]

Percentage of Participants Achieving A Response Over the First Two Treatment Cycles in Phase 2

Response categories 1) complete remission (CR): without circulating blasts or extramedullary disease, bone marrow (BM) with <5% blasts, and platelet (plt)/ANC recovery: ≥75/ ≥0.75 [x 10^9/L] 2) CR in absence of plt recovery (CRp): plt ≥20 to <75 x 10^9/L 3) partial remission (PR): no circulating blasts, appearance of normal hematopoietic progenitors, and either a BM with ≥5% and ≤25% blasts with recovery of plts/ANC or a BM with <5% blasts not meeting CR/CRp definition 4) Overall remission (OR): CR+CRp 5) Any response: CR+CRp+PR. (NCT00315705)
Timeframe: Approximately 28-56 days (Phase 2 portion of study)

Interventionpercentage of total participants (Number)
Complete remission (CR)Complete remission/absence total platelet recoveryPartial remission (PR)Overall remission (OR)Any response (CR+CRp+PR)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide2816124456

[back to top]

Summary of Participants With Adverse Events (AEs) in Phase 1

Number of participants with AEs that occurred during treatment and follow-up period (45 days after last cycle). Drug-related AEs and SAEs were followed until resolved or mutually agreed by the investigator and Genzyme to discontinue reporting. AEs were classified by the investigator according to severity (graded using National Cancer Institute [NCI] Common Terminology Criteria for Adverse Events [CTCAE] version 3.0) and relationship to study drug. The severity scale is:> Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Life-threatening or disabling, Grade 5=Death related to AE (NCT00315705)
Timeframe: Up to 9.5 months (Phase 1 portion of study)

,,,,
Interventionparticipants (Number)
At least one AEAt least one AE related to clofarabineAt least one serious AEAt least one serious AE related to clofarabineDiscontinued study due to AEDiedAE with the worst grade of: 1AE with the worst grade of: 2AE with the worst grade of: 3AE with the worst grade of: 4AE with the worst grade of: 5
Phase 1 - Cohort 133330300210
Phase 1 - Cohort 233320100111
Phase 1 - Cohort 333330300120
Phase 1 - Cohort 41010990900352
Phase 1 - Cohort 566650500321

[back to top]

Summary of Participants With Adverse Events (AEs) in Phase 2

Number of participants with AEs that occurred during treatment and follow-up period (45 days after last cycle). Drug-related AEs and SAEs were followed until resolved or mutually agreed by the investigator and Genzyme to discontinue reporting. AEs were classified by the investigator according to severity (graded using National Cancer Institute [NCI] Common Terminology Criteria for Adverse Events [CTCAE] version 3.0) and relationship to study drug. The severity scale is:> Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Life-threatening or disabling, Grade 5=Death related to AE (NCT00315705)
Timeframe: Up to 9.5 months (Phase 2 portion of study)

Interventionparticipants (Number)
At least one AEAt least one AE related to clofarabineAt least one serious AEAt least one serious AE related to clofarabineDiscontinued study due to AEDiedAE with the worst grade of: 1AE with the worst grade of: 2AE with the worst grade of: 3AE with the worst grade of: 4AE with the worst grade of: 5
Phase 2: Clofarabine, Etoposide, Cyclophosphamide25252120116001168

[back to top]

Percentage of Participants Achieving A Response Over the First Two Treatment Cycles in Phase 1

Response categories 1) complete remission (CR): without circulating blasts or extramedullary disease, bone marrow (BM) with <5% blasts, and platelet (plt)/ANC recovery: ALL ≥75/ ≥0.75 [x 10^9/L]; AML ≥100/ ≥1.0 [x 10^9/L] 2) CR in absence of plt recovery (CRp): ALL plt ≥20 to <75 x 10^9/L; AML plt ≥20 to <100 x 10^9/L 3) partial remission (PR): no circulating blasts, appearance of normal hematopoietic progenitors, and either a BM with ≥5% and ≤25% blasts with recovery of plts/ANC or a BM with <5% blasts not meeting CR/CRp definition 4) Overall remission (OR): CR+CRp 5) Any response: CR+CRp+PR. (NCT00315705)
Timeframe: Approximately 2 months (Phase 1 portion of study)

Interventionpercentage of total participants (Number)
Complete remission (CR)Complete remission/absence total platelet recoveryPartial remission (PR)Overall remission (OR)Any response (CR+CRp+PR)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide402406464

[back to top]

Kaplan Meier Estimate of Duration of Remission (DOR) for Participants Who Achieved Overall Remission (OR) in Phase 1

Duration of response is the time from the first objective measurement of complete response (CR) or complete response with the absence of total platelet recovery (CRp) to the date of first objective documentation of disease relapse or death due to any cause, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 1 portion of study)

Interventionweeks (Median)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide18.2

[back to top]

Time to Remission for Participants Who Had a Response in Phase 2

The weeks between start of intervention and remission as assessed by the investigator in Phase 2. Participants who had a complete remission (CR) or complete remission with the absence of total platelet recovery (CRp) are included. (NCT00315705)
Timeframe: up to 8 weeks (Phase 2 portion of study)

Interventionweeks (Mean)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide4.84

[back to top]

Time to Remission for Participants Who Had a Response in Phase 1

The weeks between start of intervention and remission as assessed by the investigator in Phase 1. Participants who had a complete remission (CR) or complete remission with the absence of total platelet recovery (CRp) are included. (NCT00315705)
Timeframe: up to 8 weeks (Phase 1 portion of study)

Interventionweeks (Mean)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide4.96

[back to top]

Participants With Dose Limiting Toxicity in Phase 1

The number of participants in each cohort that had dose limiting toxicity is summarized. Toxicities were reviewed by an independent Data Safety Monitoring Board (DSMB) who determined if additional participants should be added to the cohort and the criteria for escalating to the next cohort. (NCT00315705)
Timeframe: Up to Day 42 (Phase 1 portion of study)

Interventionparticipants (Number)
Phase 1 - Cohort 10
Phase 1 - Cohort 20
Phase 1 - Cohort 30
Phase 1 - Cohort 41
Phase 1 - Cohort 51

[back to top]

Number of Participants With 4-month Event Free Survival in Phase 2

Number of participants with event-free survival at four months post first dose of therapy. A participant is considered event-free if at month 4 they have not died or had a response assessment confirming a relapse. (NCT00315705)
Timeframe: 4 months (Phase 2 portion of study)

Interventionparticipants (Number)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide11

[back to top]

Number of Participants With 4-month Event Free Survival in Phase 1

Number of participants with event-free survival at four months post first dose of therapy. A participant is considered event-free if at month 4 they have not died or had a response assessment confirming a relapse. (NCT00315705)
Timeframe: 4 months (Phase I portion of study)

Interventionparticipants (Number)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide13

[back to top]

Maximum Tolerated Dose (MTD) in Phase 1

"The MTD was to be the highest dose level of clofarabine in combination with etoposide and cyclophosphamide that caused <= 1 of 6 participants to experience a dose limiting toxicity (DLT) with the next higher dose level having at least 2 of 3 or 2 of 6 participants experiencing a DLT. The MTD would be used as the recommended phase 2 dose (RP2D). If the MTD could not be determined, then the target dose of clofarabine 40 mg/m^2, etoposide 100 mg/m^2 and cyclophosphamide 440 mg/m^2 as taken by Cohort 5 was to become the RP2D.~The rating scale used is 0 = not the MTD, 1 = the MTD." (NCT00315705)
Timeframe: Up to Day 42 (Phase 1 portion of study)

Interventionunits on a scale (Number)
Phase 1 - Cohort 10
Phase 1 - Cohort 20
Phase 1 - Cohort 30
Phase 1 - Cohort 40
Phase 1 - Cohort 50

[back to top]

Kaplan Meier Estimates of Overall Survival (OS) for Participants in Phase 2

Overall survival is defined as the time from date of first administration of study interventions until date of death, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 2 portion of study)

Interventionweeks (Median)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide10.7

[back to top]

Kaplan Meier Estimates of Overall Survival (OS) for Participants in Phase 1

Overall survival is defined as the time from date of first administration of study interventions until date of death, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 1 portion of study)

Interventionweeks (Median)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide27.1

[back to top]

Kaplan Meier Estimates of Event-free Survival (EFS) for Participants in Phase 2

Event-free survival (EFS) is defined as the time from date of first administration of study interventions until the earliest of the following: date of death or date of first response assessment confirming relapse or date of final response assessment which fails to confirm response, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 2 portion of study)

Interventionweeks (Median)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide10.7

[back to top]

Kaplan Meier Estimates of Event-free Survival (EFS) for Participants in Phase 1

Event-free survival (EFS) is defined as the time from date of first administration of study interventions until the earliest of the following: date of death or date of first response assessment confirming relapse or date of final response assessment which fails to confirm response, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 1 portion of study)

Interventionweeks (Median)
Phase 1: Clofarabine, Etoposide, Cyclophosphamide19.3

[back to top]

Kaplan Meier Estimate of Duration of Remission (DOR) for Participants Who Achieved Overall Remission (OR) in Phase 2

Duration of response is the time from the first objective measurement of complete response (CR) or complete response with the absence of total platelet recovery (CRp) to the date of first objective documentation of disease relapse or death due to any cause, plus one day. For summary purposes, results are presented as weeks. (NCT00315705)
Timeframe: Up to 2 years (Phase 2 portion of study)

Interventionweeks (Median)
Phase 2: Clofarabine, Etoposide, Cyclophosphamide67.3

[back to top]

Four-Month Event-free Survival Per IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Four-month event-free survival (EFS) was defined as achieving an EFS of at least 122 days, where EFS is defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) to Day 122

Interventionpercentage of participants (Number)
Clofarabine (IV Formulation) and Cytarabine37.7
Placebo and Cytarabine16.6
Clofarabine and Cytarabine - In Stratum < 6 Months35.2
Placebo and Cytarabine - In Stratum < 6 Months16.9
Clofarabine and Cytarabine In Stratum >= 6 Months40.5
Placebo and Cytarabine In Stratum >= 6 Months16.2

[back to top]

Four-Month Event-free Survival Per IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Four-month event-free survival (EFS) was defined as achieving an EFS of at least 122 days, where EFS is defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) to Day 122

Interventionpercentage of participants (Number)
Clofarabine (IV Formulation) and Cytarabine38.9
Placebo and Cytarabine17.1
Clofarabine and Cytarabine - In Stratum < 6 Months31.4
Placebo and Cytarabine - In Stratum < 6 Months17.9
Clofarabine and Cytarabine In Stratum >= 6 Months47.4
Placebo and Cytarabine In Stratum >= 6 Months16.2

[back to top]

Overall Survival - Overall and by Calculated Strata (CSR 7-April-11)

Overall survival (OS) for the Full Analysis Set (FAS) and for the 2 calculated strata. OS was defined as the number of months from date of randomization until date of death due to any cause. (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine (FAS)6.6
Placebo and Cytarabine (FAS)6.4
Clofarabine and Cytarabine - In Stratum < 6 Months5.1
Placebo and Cytarabine - In Stratum < 6 Months5.5
Clofarabine and Cytarabine In Stratum >= 6 Months8.7
Placebo and Cytarabine In Stratum >= 6 Months7.2

[back to top]

Overall Survival - Overall and by Randomized Strata (CSR 9-July-12)

Overall survival (OS) for the Full Analysis Set (FAS) and for the 2 randomized strata. OS was defined as the number of months from date of randomization until date of death due to any cause. (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine (FAS)6.6
Placebo and Cytarabine (FAS)6.3
Clofarabine and Cytarabine - In Stratum < 6 Months4.8
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months9.7
Placebo and Cytarabine In Stratum >= 6 Months6.6

[back to top]

Best Response Per Independent Response Review Panel (IRRP) Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Percentage of participants whose best response was assessed by the IRRP as complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) using the revised International Working Group for Response Criteria (Cheson 2003).~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 up to approximately 6 months

,,,,,
Interventionpercentage of participants (Number)
Overall Remission (CR + CRi)Complete Remission (CR)CR with incomplete blood count recovery (CRi)
Clofarabine (IV Formulation) and Cytarabine46.935.211.7
Clofarabine and Cytarabine - In Stratum < 6 Months45.533.012.5
Clofarabine and Cytarabine In Stratum >= 6 Months48.637.810.8
Placebo and Cytarabine22.917.85.1
Placebo and Cytarabine - In Stratum < 6 Months22.918.14.8
Placebo and Cytarabine In Stratum >= 6 Months23.017.65.4

[back to top]

Participants With Adverse Events (CSR 7-April-11)

"Number of participants with treatment emergent adverse events (TEAEs) or death due to related AE. Related AEs for the combination arm can be related to either clofarabine or cytarabine.~Grade 1 = Mild AE, Grade 2 = Moderate AE, Grade 3 = Severe AE, Grade 4 = Life Threatening AE, Grade 5 = Death" (NCT00317642)
Timeframe: Day 1 up to a maximum of 4 years (includes up to a maximum of 3 cycles of therapy plus 45 days follow up. Related AEs are followed to resolution.)

,
Interventionparticipants (Number)
Any Treatment Emergent AEAny Related Treatment Emergent AEAny Treatment Emergent Grade >=3 AEAny Related Treatment Related Grade >=3 AEDiscontinue of study medication due to AEDiscontinue of study medication due to related AE
Clofarabine (IV Formulation) and Cytarabine1611571571271714
Placebo and Cytarabine1551331338353

[back to top]

Disease-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Disease-free survival was defined as the time from first complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) until the date of first objective documentation of disease recurrence or death due to any cause, whichever occurred first.~See Outcome #3 for definition of CR and CRi.~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine9.5
Placebo and Cytarabine7.0
Clofarabine and Cytarabine - In Stratum < 6 Months6.7
Placebo and Cytarabine - In Stratum < 6 Months6.7
Clofarabine and Cytarabine In Stratum >= 6 Months15.4
Placebo and Cytarabine In Stratum >= 6 Months9.2

[back to top]

Disease-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Disease-free survival was defined as the time from first complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) until the date of first objective documentation of disease recurrence or death due to any cause, whichever occurred first.~See Outcome #3 for definition of CR and CRi.~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine8.1
Placebo and Cytarabine7.0
Clofarabine and Cytarabine - In Stratum < 6 Months5.7
Placebo and Cytarabine - In Stratum < 6 Months6.7
Clofarabine and Cytarabine In Stratum >= 6 Months10.3
Placebo and Cytarabine In Stratum >= 6 Months9.1

[back to top]

Duration of Remission (DOR) Per IRRP Assessment-Overall and by Calculated Strata (CSR 7-April-11)

"DOR was defined as the time from first CR or CRi to the date of first objective documentation of disease recurrence, initiation of alternative antileukemic therapy [including hematopoietic stem cell transplant] while in remission, or death due to any cause, whichever occurred first.~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine7.6
Placebo and Cytarabine3.8
Clofarabine and Cytarabine - In Stratum < 6 Months5.7
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months11.5
Placebo and Cytarabine In Stratum >= 6 Months3.8

[back to top]

Duration of Remission (DOR) Per IRRP Assessment-Overall and by Randomized Strata (CSR 9-July-12)

"DOR was defined as the time from first CR or CRi to the date of first objective documentation of disease recurrence, initiation of alternative antileukemic therapy [including hematopoietic stem cell transplant] while in remission, or death due to any cause, whichever occurred first.~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine7.7
Placebo and Cytarabine3.8
Clofarabine and Cytarabine - In Stratum < 6 Months6.7
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months10.2
Placebo and Cytarabine In Stratum >= 6 Months3.8

[back to top]

Event-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Event-free survival (EFS) was defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine1.9
Placebo and Cytarabine1.0
Clofarabine and Cytarabine - In Stratum < 6 Months1.4
Placebo and Cytarabine - In Stratum < 6 Months1.0
Clofarabine and Cytarabine In Stratum >= 6 Months2.0
Placebo and Cytarabine In Stratum >= 6 Months1.0

[back to top]

Event-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Event-free survival (EFS) was defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine1.9
Placebo and Cytarabine1.0
Clofarabine and Cytarabine - In Stratum < 6 Months1.1
Placebo and Cytarabine - In Stratum < 6 Months1.0
Clofarabine and Cytarabine In Stratum >= 6 Months2.8
Placebo and Cytarabine In Stratum >= 6 Months1.0

[back to top]

Response Rate (Complete Response [CR] Plus Partial Response [PR]) of Clofarabine Plus Cytarabine in Patients With Relapsed/Refractory AML, Untreated MDS, CML in Blast Phase, or in Selected Untreated Patients With High Risk of Anthracycline Toxicity

"Based on International working group for diagnosis, standardization of response criteria, and treatment outcomes for reporting standards for therapeutic trials in Acute myeloid Leukemia:~Complete Response (CR) was defined as normalization of marrow blasts (< 5%), recovery of normal heamtopoiesis (absolute neutrophil count >1 X 10^9/l, platelet count ≥100 X10^9/l, and absence of peripheral blood blasts, independent of transfusions and growth factor support.~Partial response was defined as blood count recovery as for complete response with the exception of leukemic marrow blasts in the range of 6%-25% or a ≥50% decrease in bone marrow blasts.~Treatment failure was defined as a <25% change in marrow blasts within 30 days of starting therapy" (NCT00334074)
Timeframe: Proportion of confirmed responses was estimated by the number of patients who achieved a CR or PR, defined as two consecutive evaluations at least 4 weeks apart, divided by the number of eligible participants in the study.

Interventionparticipants (Number)
Overall Response RateComplete ResponsePartial ResponseNot RespondingNot evaluable
Clofarabine Plus Cytarabine1614286

[back to top]

Number of Participants Who Had an Adverse Event While on Treatment With Clofarabine Plus Cytarabine

Patients will be monitored clinically and diagnostically using measures including blood test, bone marrow aspiration and MUGA. Toxicity assessment every week using the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0 will be performed. (NCT00334074)
Timeframe: Up to five months (includes follow up period of 30 days) from the day patient received their first dose of study drug

Interventionparticipants; with adverse events (Number)
Clofarabine and Cytarabine30

[back to top]

Expression of Nucleoside Transporters

Expression was examined in paraffin-embedded tissue by immunohistochemistry. Intensities were scored on a 0-2+ scale. High expression was a score of 2+. (NCT00337168)
Timeframe: On average, two weeks before treatment started

Interventionparticipants (Number)
High expression of hENT1High expression of hCNT3High expression of dCK cytoplasmicHigh expression of dCK nuclear
Induction7644

[back to top]

Toxicity

Number of patients with Grade 3-5 adverse events that are related to study drug by given type of adverse event (NCT00337168)
Timeframe: Patients were assess for adverse events after each induction cycle (up to two cycles) and after the one consolidation cycle

InterventionParticipants with a given type of AE (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOT (serum glut oxaloacetic transaminase)Albumin, serum-low (hypoalbuminemia)AnorexiaAscites (non-malignant)Bilirubin (hyperbilirubinemia)Calcium, serum-low (hypocalcemia)ColitisColitis, infectious (e.g., Clostridium difficile)ConfusionCreatinineDIC (disseminated intravascular coagulation)Death not assoc with CTCAE term-Multi-organ failDermatology/Skin-Other (Specify)DiarrheaDyspnea (shortness of breath)Edema: limbFatigue (asthenia, lethargy, malaise)Febrile neutropeniaGlucose, serum-high (hyperglycemia)HemoglobinHypotensionHypoxiaINRInfec with Gr 34 neutrophils - Bladder (urinIInfec with Gr 34 neutrophils - BloodInfec with Gr 34 neutrophils - Catheter-relaInfec with Gr 34 neutrophils - ColonInfec with Gr 34 neutrophils - ConjunctivaInfec with Gr 34 neutrophils - LarynxInfec with Gr 34 neutrophils - Lung (pneumonInfec with Gr 34 neutrophils - Skin (celluliInfec with Gr 34 neutrophils - Urinary tractInfec with Gr 34 neutrophils - WoundInfection with unknown ANC - Lung (pneumonia)Infection-Other (Specify)Leukocytes (total WBC)Liver dysfunction/failure (clinical)LymphopeniaMental statusNeutrophils/granulocytes (ANC/AGC)PTT (Partial thromboplastin time)Pain - Abdomen NOSPain - BackPain - BonePlateletsPleural effusion (non-malignant)Pneumonitis/pulmonary infiltratesPotassium, serum-high (hyperkalemia)Potassium, serum-low (hypokalemia)Pruritus/itchingRenal failureRestrictive cardiomyopathySodium, serum-high (hypernatremia)Sodium, serum-low (hyponatremia)Supraventricular and nodal arrhythmiaTumor lysis syndromeTyphlitis (cecal inflammation)Uric acid, serum-high (hyperuricemia)
Induction5731231121411121121411331119121121111111171181111201114131121111

[back to top]

Number of Patients With Complete Remission

Complete remission is defined as: less than 5% bone marrow blasts, neutrophils greater or equal to 1,000 per microliter, platelets greater than 100,000 per microliter, no blasts in the peripheral blood, and no extramedullary disease (NCT00337168)
Timeframe: Between day 28 and day 35 inclusive

Interventionparticipants (Number)
Induction3

[back to top]

Number of Patients With Very Poor Risk Cytogenetics

(NCT00337168)
Timeframe: On average, 2 weeks before treatment started

Interventionparticipants (Number)
Induction10

[back to top]

Safety and Tolerability as Measured by CTCAE v3.0

Number of participants with at least one grade 3 or higher adverse event during therapy. (NCT00372619)
Timeframe: End of therapy

Interventionnumber participants (Number)
Clofarabine 40 mg/m² to Assess Feasibility in ALL Patients.7
Clofarabine 40 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in ALL Patients.3
Clofarabine 52 mg/m² to Assess Efficacy in ALL Patients.10
Clofarabine 52 mg/m² to Assess Feasibility in AML Patients.7
Clofarabine 52 mg/m² to Assess Efficacy in AML Patients35
Clofarabine 52 mg/m² to Assess Efficacy - Ambiguous Lineage pt2

[back to top]

Overall Response (CR for ALL Patients), (CR + CRp for AML Patients)

"Overall response for ALL patients: CR - complete remission (attainment of an M1 bone marrow (< 5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral counts (absolute neutrophil count (ANC) > 750/μL and platelet count > 75,000/μL).~Overall response for AML patients: (CR + CRp), defined as:~CR - complete remission (attainment of an M1 bone marrow (<5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral blood counts (absolute neutrophil count (ANC) > 1000/uL and platelet count > 100,000/uL)) or CRp - remission without platelet recovery (Attainment of an M1 bone marrow (<5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of absolute neutrophil count (ANC) > 1000/uL and platelet transfusion independence (defined as: no platelet transfusions x 1 week))." (NCT00372619)
Timeframe: 2 cycles or up to 84 days

Interventionparticipants (Number)
Clofarabine 40 mg/m² to Assess Feasibility in ALL Patients.1
Clofarabine 40 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in ALL Patients.0
Clofarabine 52 mg/m² to Assess Efficacy in ALL Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Efficacy in AML Patients19
Clofarabine 52 mg/m² to Assess Efficacy - Ambiguous Lineage pt2

[back to top]

Kaplan Meier Estimate for Duration of Remission (DOR)

DOR was defined as the number of days from achievement of OR as assessed by the Independent Response Review Panel (IRRP) until IRRP-determined disease recurrence or death (any cause), plus 1 day. Participants who initiated alternative antileukemic treatment while in remission were censored on the date the therapy was initiated or on the date of last follow-up. (NCT00373529)
Timeframe: Up to 2 years

Interventionweeks (Median)
Clofarabine55.6

[back to top]

Kaplan Meier Estimates for Overall Survival (OS)

OS was defined as the number of days from first dose of clofarabine until death for all participants, plus 1 day. (NCT00373529)
Timeframe: Up to 2 years

Interventionweeks (Median)
Clofarabine40.7

[back to top]

Percentage of Participants Who Died Within Thirty Days of Treatment (30-day Mortality Rate)

Percentage of participants who died within 30 days of the first dose of study drug, regardless of cause. (NCT00373529)
Timeframe: up to Day 30

Interventionpercentage of participants (Number)
Clofarabine9.8

[back to top]

Number of Participants Achieving Overall Remission After A Maximum of Two Cycles by Subgroup of Baseline Prognostic Factors

The number of participants within each subgroup of baseline prognostic factors of the full analysis set who achieved a best response of either a complete response (CR) or a complete response in the absence of platelet recovery (CRp) as determined by the Independent Response Review Panel following a maximum of two cycles of treatment. (NCT00373529)
Timeframe: approximately Month 2

Interventionparticipants (Number)
Age >=70 (n=69)Age <70 (n=43)Antecedent hematologic disorder - Yes (n=41)Antecedent hematologic disorder - No (n=67)Antecedent hematologic disorder-Not reported (n=4)ECOG Performance Status = 0-1 (n=87)ECOG Performance Status = 2 (n=25)Karyotype = Intermediate (n=46)Karyotype = Unfavorable (n=62)Karyotype = Not Reported (n=4)
Clofarabine27242129143825260

[back to top]

Overall Participant Counts Summarizing Adverse Events (AEs) During the Treatment and Follow-up Periods

"Participants with AEs that occurred during the treatment and follow-up periods. AEs were classified according to severity (graded using National Cancer Institute [NCI] Common Terminology Criteria for Adverse Events [CTCAE] version 3.0) and relationship to study drug. Treatment emergent is defined as any event that either first presents after baseline or worsens in severity after baseline.~NCI Common Terminology Criteria for Severity:~Grade 1= Mild AE, Grade 2= Moderate AE, Grade 3= Severe AE, Grade 4= Life-threatening or disabling AE, Grade 5= Death related to AE" (NCT00373529)
Timeframe: Up to 2 years

Interventionparticipants (Number)
Treatment-emergent AEs (TEAE)Treatment-emergent AEs related to study drugTreatment-emergent serious AEsTreatment-emergent serious AEs related to drugDiscontinued due to AEsDied w/i treatment period-w/i 45 days of last doseDied due to drug-related AEsDied within 30 days of first doseDied w/i 30 days of first dose due to related AEsGrade 1: maximum severity rating for any TEAEGrade 2: maximum severity rating for any TEAEGrade 3: maximum severity rating for any TEAEGrade 4: maximum severity rating for any TEAEGrade 5: maximum severity rating for any TEAE
Clofarabine1121087641722411326463424

[back to top]

Percentage of Participants Achieving Overall Remission (OR) After No More Than Two Cycles (Approximately Month 2)

Best response was assessed by the Independent Response Review Panel(IRRP) after two cycles of treatment. Overall remission(OR) is the sum of complete remission(CR) and complete remission in the absence of platelet recovery(CRp). CR includes normal values for peripheral blood cell counts (absolute neutrophil and platelet) and leukemic blast cells from bone marrow biopsy or aspirate, and absence of extramedullary disease. Partial remission(PR) includes recovery of peripheral blood cells with improved but still abnormal values in leukemic blast cells. (NCT00373529)
Timeframe: approximately Month 2

Interventionpercentage of participants (Number)
Overall Remission (OR=CR+CRp)Complete Remission (CR)Complete Remission w/o platelet recovery (CRp)Partial Remission (PR)Treatment Failure (TF)
Clofarabine45.537.58.03.650.9

[back to top]

Kaplan Meier Estimate for Disease-free Survival (DFS)

DFS was defined as the number of days from achievement of IRRP-determined overall response until IRRP-determined disease recurrence or death (any cause), regardless of intervening alternative antileukemic treatment, plus 1 day. (NCT00373529)
Timeframe: Up to 2 years

Interventionweeks (Median)
Clofarabine43.8

[back to top] [back to top]

Number of Patients With Acute Graft Versus Host Disease (GVHD)

Graft-Versus-Host Disease is a severe short-term complication created by infusion of donor cells into a foreign host. Acute GVHD can occur once the donor's cells have engrafted in the transplant recipient. The symptoms typically appear within weeks after transplant. (NCT00383448)
Timeframe: Day 100

InterventionParticipants (Count of Participants)
Treated Patients3

[back to top]

Number of Patients With Chronic Graft Versus Host Disease (GVHD)

Graft-Versus-Host Disease is a severe complication created by infusion of donor cells into a foreign host. Chronic GVHD can appear at any time after allogeneic transplant or several years after transplant. (NCT00383448)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Treated Patients2

[back to top]

Number of Patients With Donor Cell Engraftment

Donor Cell Engraftment is defined as the process of transplanted stem cells reproducing new cells. (NCT00383448)
Timeframe: Day 100

InterventionParticipants (Count of Participants)
Treated Patients26

[back to top]

Maximum Tolerated Dose for Cyclophosphamide (MTD)

MTD is dose at which there are no dose limiting toxicity (DLT) defined as any =/> grade 3 drug-related non-hematologic toxicity that occurs within the first 14 days after start of treatment. Evaluation using continual reassessment method; 3-5 Day Cycle (NCT00412243)
Timeframe: First 14 days of each cycle

Interventionmg/m^2 (Number)
Clofarabine + Cyclophosphamide200

[back to top]

Maximum Tolerated Dose

(NCT00416351)
Timeframe: 2 years

Interventionmg/m2 of clofarabine (Number)
Clofarabine20

[back to top]

Participants Evaluated for Toxicity

Toxicity as defined by NCI Common Terminology Criteria for Adverse Events v 3.0 (NCT00416351)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
4 mg/m2/Day Clofarabine1
8 mg/m2/Day Clofarabine3
13.2 mg/m2/Day Clofarabine4
20 mg/m2/Day Clofarabine17
28 mg/m2/Day Clofarabine4

[back to top]

Number of Participants With Response for Two Dose Schedules of Clofarabine

Response defined as Complete Remission (CR): Normalization of blood counts with neutrophils >/= 1 * 10^9/L and platelet counts >/= 100 * 10^9/L, and marrow blasts /= 100 * 10^9/L. Repeat bone marrow samples collected every 1-3 cycles (4-8 week cycle). (NCT00422032)
Timeframe: 4 weeks (minimum 1 cycle) up to 24 weeks (maximum 3 cycles of 8 weeks)

,
InterventionParticipants (Number)
Complete ResponseHematologic Improvement
15 mg/m^2 Clofarabine105
30 mg/m^2 Clofarabine51

[back to top]

Participants Evaluated for Early Post-transplant Regimen-related Severe Morbidity (Grade III to IV Nonhematologic Toxicity) and Mortality as Measured by the NCI Cancer Therapy Evaluation Program CTCAE v 3.0

(NCT00423514)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Dose Level 1: Clofarabine at 20 mg/m^2/Dose x 5 + THIO-MEL31
Dose Level 2: Clofarabine at 30 mg/m^2/Dose x 5 + THIO-MEL7

[back to top] [back to top]

Number of Participants Who Reported Death Within 30 Days of First Dose

(NCT00531232)
Timeframe: Within 30 days of first dose administered on Day 1 of Cycle 1

InterventionParticipants (Count of Participants)
Clofarabine 55 mg/Day2
Clofarabine 35 mg/Day1
Clofarabine 25 mg/Day1

[back to top]

Number of Participants Who Achieved Hematologic Improvement (HI)

Per IWG criteria, HI was defined as meeting any of the erythroid, platelet,and/or neutrophil independence categories for at least 8 consecutive weeks. Erythroid response (pre-treatment,<11 grams per deciliter [g/dL]) was defined as Hgb increased by >=1.5 g/dL;relevant reduction of units of red blood cell (RBC) transfusion by an absolute number of at least 4 RBC transfusion/8 weeks compared with the pretreatment transfusion number in the previous 8 weeks; only RBC transfusions given for Hgb of <=9.0 g/dL pre-treatment counted in the RBC transfusion response evaluation. Platelet response(pretreatment, <100*109/L) was defined as absolute increase of >=30*109/L for participants starting with >20*109/L platelets, increase from <20*109/L to >20*109/L and by at least 100%. Neutrophil response (pre-treatment, <1.0*109/L) was defined by at least 100% increase and an absolute increase >0.5*109/L. Number of participants who achieved HI for MDS participants only was reported in outcome measure. (NCT00531232)
Timeframe: From date of randomization until disease recurrence or death, whichever occurred first (maximum study duration: up to 4 years)

InterventionParticipants (Count of Participants)
Clofarabine 35 mg/Day0
Clofarabine 25 mg/Day3

[back to top]

Maximum Tolerated Dose (MTD) of Oral Clofarabine

The MTD was the highest dose level of Clofarabine that caused less than (<) 2 participants to experience unacceptable drug-related toxicities after receiving 1 or more of the initial 5 daily doses of Clofarabine. Unacceptable drug-related toxicities included: drug-related prolonged myelosuppression, which was defined as hypocellular bone marrow with <5 percent (%) cellularity at 6 weeks after starting treatment, which required a 1-dose level reduction for subsequent cycles; Common Terminology Criteria for Adverse Events (CTCAE) Grade 4 drug-related non-hematologic toxicity; CTCAE Grade 3 drug-related non-hematologic toxicity that either recovered to Baseline grade but was recurrent to Grade 3 upon re-treatment during the first cycle or did not recover to Grade 1 or Baseline within 3 weeks when the drug was held or dose reduced; drug-related non-hematologic toxicity that resulted in non-completion of at least 4 daily doses of oral Clofarabine at the original assigned dose level. (NCT00531232)
Timeframe: Cycle 1 (28 days)

Interventionmg/day (Number)
Clofarabine 55 mg/Day25
Clofarabine 35 mg/Day25
Clofarabine 25 mg/Day25

[back to top]

Duration of Response (DoR)

DoR: time (in months) from 1st documentation of response to date of 1st documentation of disease relapse, progression or death due to any cause, whichever occured first. Response defined as CR, marrow CR, or PR (MDS participants) and CR/CRi (sAML participants). Per IWG criteria, relapse defined as: return to pretreatment bone marrow blast %; decrease of >=50% from maximum remission levels; reduction in Hgb by >=1.5g/dL.CR:<= 5%myeloblasts in bone marrow; persistent dysplasia; peripheral blood showing Hgb>=11g/dL. Marrow CR:<=5%myeloblasts (bone marrow) and decreased by >=50% over pretreatment; any HI response in peripheral blood. CRi:meeting all criteria for CR except for residual thrombocytopenia/neutropenia. PR: all CR criteria if abnormal before treatment except that marrow blasts should have decreased by >=50%. Progression: at least 50% decrease from maximum remission/response in granulocytes/platelets; reduction in Hgb by >=2g/dL; transfusion dependence. Analyzed by Kaplan-Meier. (NCT00531232)
Timeframe: From first documentation of response to date of documentation of disease relapse, progression or death due to any cause, whichever occurs first (maximum study duration: up to 4 years)

Interventionmonths (Median)
Clofarabine 35 mg/Day12.5
Clofarabine 25 mg/Day3.2

[back to top] [back to top]

Number of Participants With Adverse Events (AEs)

Adverse Event (AE): any undesirable physical, psychological/behavioral effect experienced by participant during their participation in clinical study, with study drug usage, whether or not product related.Treatment Emergent AEs (TEAEs): AEs that developed, worsened, or became serious during treatment period (from signature of informed consent form up to 45 days post last dose). Serious AE (SAE):any untoward medical occurrence that at any dose resulted in death, was life-threatening, required inpatient hospitalization/prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity, was a congenital anomaly/birth defect, medically important event. Per National Cancer Institute (NCI)-CTCAE v3.0 severity for each AE were graded as: Grade 1=Mild AE; Grade 2=Moderate AE; Grade 3=Severe AE, Grade 4=Life-threatening/disabling AE and Grade 5=Death related to AE. Participants with TEAEs, SAEs, death, discontinuations, Grade 4 and 5 toxicities were reported. (NCT00531232)
Timeframe: From Baseline up to 45 days post last dose of study drug (maximum duration: up to 4 years)

,,
InterventionParticipants (Count of Participants)
Any TEAEAny treatment emergent SAEAny TEAE leading to discontinuationAny TEAE leading to deathDeaths within 45 days of last doseGrade 4 ToxicitiesGrade 5 Toxicities
Clofarabine 25 mg/Day24192143184
Clofarabine 35 mg/Day8627344
Clofarabine 55 mg/Day4414313

[back to top]

Time to Acute Myeloid Leukemia (AML) Transformation

Time to AML transformation was defined as time (in months) from date of the first dose of oral Clofarabine to the date of AML transformation, (i.e., the earliest date when participants experienced bone marrow or peripheral blasts >30%). The analysis was performed by Kaplan-Meier method. (NCT00531232)
Timeframe: From date of randomization until disease recurrence or death, whichever occurred first (maximum study duration: up to 4 years)

Interventionmonths (Median)
Clofarabine 35 mg/Day4.2
Clofarabine 25 mg/DayNA

[back to top]

PK Parameter: Time to Reach Maximum Observed Plasma Concentration (Tmax) of Clofarabine

Tmax was defined as the time to reach Cmax (maximum observed plasma concentration). (NCT00531232)
Timeframe: Pre-dose and at 1, 3, and 5 hours Post-dose on Day 1 of Cycle 1

Interventionhours (Median)
Clofarabine 55 mg/Day2.02
Clofarabine 35 mg/Day1
Clofarabine 25 mg/Day2

[back to top]

PK Parameter: Area Under the Concentration-time Curve From Time 0 to Time of Last Measurable Plasma Concentration (AUC0-last) of Clofarabine

AUC0-last was defined as area under the concentration-time curve from time 0 to time of last measurable plasma concentration. (NCT00531232)
Timeframe: Pre-dose and at 1, 3, and 5 hours Post-dose on Day 1 of Cycle 1

Interventionnanograms*hours per milliliter (Mean)
Clofarabine 55 mg/Day590.188
Clofarabine 35 mg/Day194.084
Clofarabine 25 mg/Day144.604

[back to top]

Pharmacokinetics (PK) Parameter: Maximum Observed Plasma Concentration (Cmax) of Clofarabine

Cmax was defined as maximum observed plasma concentration of study drug. (NCT00531232)
Timeframe: Pre-dose and at 1, 3, and 5 hours Post-dose on Day 1 of Cycle 1

Interventionnanograms per milliliter (Mean)
Clofarabine 55 mg/Day158.25
Clofarabine 35 mg/Day79.613
Clofarabine 25 mg/Day47.813

[back to top]

Percentage of Participants With Overall Response

Overall Response: complete remission(CR) or marrow CR or partial remission (PR), or hematologic improvement (HI) in participants with myelodysplastic syndromes (MDS);CR or CR with incomplete count recovery(CRi),or PR in participants with secondary acute myeloid leukemia (sAML). Per International Working Group (IWG) criteria, CR:<= 5% myeloblasts in bone marrow; persistent dysplasia had to be noted; peripheral blood showing hemoglobin (Hgb)>=11g/dL, platelets >=100*10^9/L,neutrophils >=1*10^9/L, blasts 0%. Marrow CR:<=5%myeloblasts in bone marrow and decreased by >=50% over pretreatment; any HI response in peripheral blood. CRi: meeting all criteria for CR except for residual thrombocytopenia (platelet <100*10^9/L) or neutropenia (ANC <1.0*10^9/L). PR: all CR criteria if abnormal before treatment except that marrow blasts should have decreased by >=50% over pretreatment but still >5%. HI: meeting any of the erythroid, platelet, or neutrophil improvement categories for at least 8 weeks. (NCT00531232)
Timeframe: From date of randomization until disease recurrence or death, whichever occurred first (maximum study duration: up to 4 years)

Interventionpercentage of participants (Number)
Clofarabine 35 mg/Day13
Clofarabine 25 mg/Day25

[back to top]

Percentage of Participants Achieving Overall Remission (OR)

OR was defined as a best response of CR, marrow CR, or PR in participants with MDS or a best response of CR or CRi in participants with sAML. As per IWG criteria, CR was defined as the following: bone marrow <=5% myeloblasts with normal maturation of all cell lines; persistent dysplasia had to be noted; peripheral blood showing hemoglobin >=11 g/dL, platelets >=100*10^9/L, neutrophils >=1*10^9/L, blasts 0%. Marrow CR was defined as: bone marrow <=5% myeloblasts and decreased by >=50% over pretreatment; any HI response in peripheral blood had to be noted. CRi was defined as meeting all criteria for CR except for residual thrombocytopenia (platelet count <100*10^9/L) or neutropenia (ANC <1.0*10^9/L). PR was defined as all CR criteria if abnormal before treatment except that marrow blasts should have decreased by >=50% over pre-treatment but still >5%. HI was defined as meeting any of the erythroid, platelet, or neutrophil improvement categories for at least 8 weeks. (NCT00531232)
Timeframe: From date of randomization until disease recurrence or death, whichever occurred first (maximum study duration: up to 4 years)

Interventionpercentage of participants (Number)
Clofarabine 35 mg/Day13
Clofarabine 25 mg/Day25

[back to top]

Overall Survival (OS)

OS defined as date of the first dose of oral Clofarabine until date of death due to any cause. If death was not observed, the participant was censored at the earliest of the last date the participant was known to be alive or the study cut-off date. The analysis was performed by Kaplan-Meier method. (NCT00531232)
Timeframe: From date of first dose of study drug until date of death due to any cause (maximum study duration: up to 4 years)

Interventionmonths (Median)
Clofarabine 35 mg/Day4.8
Clofarabine 25 mg/Day7.3

[back to top]

Number of Participants With Febrile Neutropenia

Febrile neutropenia was defined as fever (e.g., greater than or equal to (>=) 38.5 Celsius (°C) on a single occasion, or greater than (>) 38°C on 2 occasions within 12 hours) in the setting of neutropenia (defined as Absolute Neutrophil Count <1.0*10^9/liter [L]). (NCT00531232)
Timeframe: From Baseline up to 45 days post last dose of study drug (maximum study duration: up to 4 years)

InterventionParticipants (Count of Participants)
Clofarabine 55 mg/Day1
Clofarabine 35 mg/Day1
Clofarabine 25 mg/Day13

[back to top]

Probability of Event-free Survival

"To estimate the event-free survival of children with ALL who are treated with risk-directed therapy and to compare the EFS results of TOTXVI with that of TOTXV (NCT00137111).~EFS will be measured from the date of complete response to the date of initial failure for patients who fail. Failure includes the traditional endpoints of failure to achieve a complete remission, relapse in any site, secondary malignancy, and death during induction or remission. EFS time will be measured to the date of last contact for patients who are failure free at the time of analysis. The EFS time is defined to be zero (0) for patients who die during induction therapy or fail to achieve a complete remission." (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 350092.4
TOTXVI PEG 250091.1
TOTXVI Not Randomized86.3
All Eligible Patients in TOTXV87.1

[back to top]

Proportion of Participants With Minimal Residual Disease (MRD) on the 15th Day of Remission Induction ≥ 5%

To study whether intensifying induction, including fractionated cyclophosphamide and thioguanine, in patients with day 15 MRD ≥ 5% will result in improved leukemia cytoreduction in this subgroup compared to therapy followed in the TOTXV protocol. (NCT00549848)
Timeframe: Middle of remission induction, Day 15 in Total XVI and Day 19 in Total XV

InterventionParticipants (Count of Participants)
TOTXVI PEG 350022
TOTXVI PEG 250026
TOTXVI Not Randomized31
All Eligible Patients in TOTXV55

[back to top]

Proportion of Participants With Minimal Residual Disease (MRD) at End of Remission Induction ≥ 0.01%

To study whether intensifying induction, including fractionated cyclophosphamide and thioguanine, in patients with day 15 MRD ≥ 5% will result in improved leukemia cytoreduction in this subgroup compared to therapy followed in the TOTXV protocol. (NCT00549848)
Timeframe: End of remission induction; day 42 in Total XVI and day 46 in Total XV

InterventionParticipants (Count of Participants)
TOTXVI PEG 35007
TOTXVI PEG 250012
TOTXVI Not Randomized20
All Eligible Patients in TOTXV44

[back to top]

Probability of Overall Survival

To estimate the overall survival of children with ALL who are treated with risk-directed therapy and to compare the EFS results of TOTXVI with that of TOTXV. (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 350097.5
TOTXVI PEG 250095.6
TOTVI Not Randomized90.8
All Eligible Patients in TOTXV93.5

[back to top]

Probability of CNS Relapse

To assess whether intensification of CNS-directed intrathecal and systemic chemotherapy will improve outcome in patients at high-risk of CNS relapse. (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 35000.8
TOTXVI PEG 25001.8
TOTXVI Not Randomized2.7
All Eligible Patients in TOTXV5.7

[back to top]

Percentage of Participants With Continuous Complete Remission of Patients Receiving High-dose and Conventional Dose PEG-asparaginase.

"The primary objective of this study is to compare the distributions of continuous complete remission of patients randomized on the first day of the continuation phase to receive a higher dose of PEG-asparaginase or to receive the conventional dose (2,500 units/m2).~The randomization will occur on the starting day of the continuation phase, at which time all information necessary for performing the randomization should be available. In the rare case that immunophenotype and/or Day-15 MRD is not available, we will make the following assumptions: If immunophenotype is unknown at the time the randomization is to be executed, then it will be assumed B-lineage. If Day-15 MRD is unknown at the time of randomization, then it will be assumed negative (<0.01%). Past experience indicate that few patients will fall into these unknown categories." (NCT00549848)
Timeframe: 3.5 years after the last enrollment up to 12.5 years

InterventionPercentage of participants (Number)
PEG 3500 Units/m^291.6
PEG 2500 Units/m^290.7

[back to top]

One-year Overall Survival Rate for AML

Percent Overall Survival (OS) for at one year for subjects with Acute Myeloid Leukemia (AML). (NCT00556452)
Timeframe: 1 year

Interventionpercent overall survival (Number)
Clo/BU448

[back to top]

Two-year Overall Survival for All Cases.

Percent Overall Survival (OS) at two years for all patients. (NCT00556452)
Timeframe: 2 years

Interventionpercent overall survival (Number)
Clo/BU428

[back to top] [back to top]

Five Year Overall Survival for All Cases

The number of patients alive at 5 years (NCT00556452)
Timeframe: five years

InterventionParticipants (Count of Participants)
Clo/BU46

[back to top]

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +30

Interventionparticipants (Number)
Not done50% donor (bone marrow)100% donor (bone marrow)23.5% donor (FISH)
Arm 1: Non-myeloablative Conditioning Regimen2111

[back to top]

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +80-+90

Interventionparticipants (Number)
0% donor33% donor - myeloid (peripheral blood)67% donor (bone marrow)100% donor (bone marrow)
Arm 1: Non-myeloablative Conditioning Regimen1111

[back to top]

Disease-free Survival

Disease-free survival is defined as the length of time after treatment ends that the participant survives without any signs or symptoms of that cancer. (NCT00593645)
Timeframe: 5 years from time of restaging

Interventionparticipants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

[back to top]

Median Time to Progression

Time to progression is defined as the length of time from the start of treatment until the disease starts to get worse or spread to other parts of the body. (NCT00593645)
Timeframe: 5 years from time of restaging

Interventiondays (Median)
Arm 1: Non-myeloablative Conditioning Regimen152

[back to top]

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +40-+60

Interventionparticipants (Number)
60% donor (bone marrow)70% donor (bone marrow)75% donor (bone marrow)100% donor (peripheral blood)
Arm 1: Non-myeloablative Conditioning Regimen1111

[back to top]

Overall Survival

(NCT00593645)
Timeframe: 5 years from time of restaging

Interventiondays (Median)
Arm 1: Non-myeloablative Conditioning Regimen237

[back to top]

Rate of Acute Graft-versus-host Disease (GVHD)

Acute GVHD occurs within 100 days of transplant. (NCT00593645)
Timeframe: Up to 100 days after transplant

Interventionpercentage of participants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

[back to top]

Rate of Chronic Graft-versus-host Disease (GVHD)

(NCT00593645)
Timeframe: 100 days-1 year after transplant

Interventionpercentage of participants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

[back to top]

Response Rates by Cytogenetic Risk Category

Number of participants who achieved Complete Remission (less than 5% blasts in the marrow and count recovery of Absolute Neutrophil Count to 1,000/microL and Platelet Count to 100,000/microL) under each cytogenetic risk category. (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Favorable risk Complete RemissionIntermediate risk Complete remissionUnfavorable risk Complete remission
Arm I3109

[back to top]

Disease-free Survival

Number of participants who survived and were disease-free at 5 years (NCT00602225)
Timeframe: At five years after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)11

[back to top]

Maximum Tolerated Dose of Clofarabine

(NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

Interventionmg/m^2 of clofarabine (Number)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)25

[back to top]

Dose-limiting Toxicity as Assessed by NCI CTCAE v3.0

(NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)2

[back to top]

Efficacy

Number of Patients Surviving at Five Years (NCT00602225)
Timeframe: At five years after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)12

[back to top]

Overall Survival

(NCT00602225)
Timeframe: At five years after the last dose of clofarabine

Interventionmonths (Median)
Arm I9

[back to top]

Response Rates by Cytogenetic Risk Category and Clofarabine Dose

Number of participants under each Cytogenetic Risk Category and Clofarabine dose who achieve CR (Complete Remission = less than 5% blasts in the marrow and count recovery of Absolute Neutrophil Count to 1,000/microL and Platelet Count to 100,000/microL) or CRp (Complete Remission, but with a platelet count of less than 100,000/microL). (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Favorable Risk + 25 mg/m^2 achieve CRIntermediate Risk + 15 mg/m^2 achieve CRIntermediate Risk + 20 mg/m^2 achieve CRIntermediate Risk + 25 mg/m^2 achieve CRIntermediate Risk + 25 mg/m^2 achieve CRpUnfavorable Risk + 15 mg/m^2 achieve CRUnfavorable Risk + 20 mg/m^2 achieve CRUnfavorable Risk + 25 mg/m^2 achieve CRUnfavorable Risk + 25 mg/mg^2 achieve CRp
Arm I223542163

[back to top]

Response Rates by Duration First Complete Remission (CR1)

Number of participants whose first Complete Remission lasted 0, 1-6, 6-12, or greater than 12 months. Only those participant who had a first CR are included in this data. (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Duration CR1 (months): 0Duration CR1 (months): 1-6Duration CR1 (months): 6-12Duration CR1 (months): greater than 12
Arm I12423

[back to top]

Response Rates by Salvage Number

Number of participants in each Salvage number category who achieved a Complete Remission. Salvage number refers to whether treatment with GCLAC on this study was the pariticipant's first salvage regimen (1), second salvage regimen (2), or third or greater salvage regimen (3 or greater). (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Salvage number 1Salvage number 2Salvage number 3 or greater
Arm I1650

[back to top] [back to top]

Chimerism

Occurrence of genetically distinct cell types in a single organism (NCT00617929)
Timeframe: Day 28 post transplantation

Interventionpercentage of donor cells (Median)
Conditioning for Graft Failure After Transplant95

[back to top]

Acute Graft-vs-host Disease

"Percent of patients with Acute Graft-vs-host Disease - a process where T-cells present in the donor's bone marrow at the time of transplant identify the transplant patient as non-self' and attack the patient's skin, liver, stomach, and/or intestines." (NCT00617929)
Timeframe: Day 30-100

Interventionpercentage of participants (Number)
Conditioning for Graft Failure After Transplant18

[back to top]

Rate of Sustained Donor Engraftment

Rate of Sustained Donor Engraftment is defined as the percent of paticipants with an absolute neutrophile count (ANC) of 500 or more without a subsequent graft rejection. (NCT00617929)
Timeframe: Day 42 post transplantation

Interventionpercentage of participants (Number)
Conditioning for Graft Failure After Transplant73

[back to top]

Survival

Percent of patients alive from beginning of study to one year post transplantation (NCT00617929)
Timeframe: One year post transplantation

Interventionpercentage of participants (Number)
Conditioning for Graft Failure After Transplant36

[back to top]

Survival at 100 Days Post Transplant

Percent of patients alive from beginning of study to Day 100 post transplantation (NCT00617929)
Timeframe: Day 100 post transplantation

Interventionpercentage of participants (Number)
Conditioning for Graft Failure After Transplant73

[back to top]

Time to Primary Neutrophil Engraftment

Time to primary neutrophil engraftment is defined as the percent of patients with an absolute neutrophil count (ANC) of 500 or more neutrophils in a cubic millimeter of blood. (NCT00617929)
Timeframe: Day 42 post transplantation

Interventionpercentage of participants (Number)
Conditioning for Graft Failure After Transplant73

[back to top]

Engraftment of Allogeneic Blood Cells.

"Establish the safety of Clofarabine and cyclophosphamide preceding allogeneic hematopoietic engraftment. Assess the efficacy of Clofarabine and cyclophosphamide as conditioning for promoting allogeneic hematopoietic engraftment.~Adequacy of engraftment will be assessed via assessment of chimerism (percent donor engraftment). Less than 20% engraftment by day 30 is then failure of engraftment.~Safety is defined per common toxicity criteria - Non-Hematological and non renal toxicities of ≥grade 3 or ≥grade 4 up to day 30 are scored as toxicity." (NCT00626626)
Timeframe: two years

,
InterventionParticipants (Count of Participants)
Adequate EngraftmentSafety
Clofarabine, Cyclophosphamide & Alemtuzumab (Phase I )25
Clofarabine, Cyclophosphamide & Alemtuzumab (Phase II)13

[back to top] [back to top] [back to top]

Number of Patients Achieving Donor Cell Engraftment

Number of patients with persistent presence of donor-derived cells at Day 100 (NCT00638820)
Timeframe: Day 100

InterventionParticipants (Number)
Patients With Osteopetrosis Who Received Transplant2

[back to top]

Phase I Participants: Progression-free Survival (PFS)

"Determine the progression-free survival rate among all phase I trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg).~Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions." (NCT00644189)
Timeframe: at 17 months

Interventionpercentage of participants (Mean)
Phase I34

[back to top]

Phase I Participants Only: Overall Response Rate (ORR)

"Determine the efficacy of oral clofarabine (any of the 4 dose levels: 1mg, 2mg, 4mg, and 3mg) in all phase I trial patients with relapsed/refractory non-Hodgkin lymphomas.~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT00644189)
Timeframe: after at most 6 28-day cycles

Interventionpercentage of participants (Mean)
Phase I47

[back to top]

All Phase I-II Participants: Overall Survival (OS)

Determine the overall survival rate among all phase I-II trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg) (NCT00644189)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Clofarabine58

[back to top]

All Phase I-II Participants: Overall Response Rate (ORR)

"Determine the efficacy of oral clofarabine (any of the 4 dose levels: 1mg, 2mg, 4mg, and 3mg) in all phase I-II trial patients with relapsed/refractory non-Hodgkin lymphomas.~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT00644189)
Timeframe: after at most 6 28-day cycles

Interventionpercentage of participants (Number)
Phase I-II28

[back to top]

All Phase I-II Participants: Progression-free Survival (PFS)

"Determine the progression-free survival rate among all phase I-II trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg).~Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions." (NCT00644189)
Timeframe: at 1 and 2 years

Interventionpercentage of patients (Number)
1-year PFS2-year PFS
Clofarabine3216

[back to top]

All Phase I Participants: Overall Survival (OS)

Determine the overall survival rate among all phase I trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg) (NCT00644189)
Timeframe: at 17 months

Interventionpercentage of participants (Mean)
Phase I75

[back to top]

All Phase I-II Participants: Safety

Grade 3-4 toxicities among all phase I-II trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg) (NCT00644189)
Timeframe: during 6 28-day cycles and 90 days out

InterventionParticipants (Count of Participants)
Any grade 3-4 toxicityLeukopenia and neutropeniaThrombocytopeniaAnemiaFatigue
Clofarabine29241573

[back to top]

Phase I Participants: Safety

Grade 2-4 toxicities and grade 3-4 infections among all phase I trial participants who are treated with any of the 4 dose levels of oral clofarabine (1mg, 2mg, 4mg, or 3mg) (NCT00644189)
Timeframe: during 6 28-day cycles and 90 days out

InterventionParticipants (Count of Participants)
AnemiaNeutropeniaThrombocytopeniaFatigueNeutropenic feverGrade 3-4 infectionsAny grade 3-4 toxicity
Phase I1918981320

[back to top]

Phase I-II Participants Treated at the RP2D (3mg): Overall Response Rate (ORR)

"To determine the efficacy of oral clofarabine (3mg) in patients with relapsed/refractory non-Hodgkin lymphoma. The 3mg dose was declared the recommended phase 2 dose (RP2D) from phase I.~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT00644189)
Timeframe: after at most 6 28-day cycles

Interventionpercentage of participants (Number)
Clofarabine28

[back to top]

To Determine the Non-hematologic Toxicity Profile of This Dose Schedule (Grade 2 and Above)

Assess for adverse events in all the patients receiving the Clofarabine at the dose schedules described in the protocol (CTCAE 3.0 used). (NCT00700011)
Timeframe: biweekly for duration of treatment , an average of 3 months

,
Interventionparticipants (Number)
FatigueMucositisVomitingNauseaHyperbilirubinemiaDehydrationRashALT elevationAST elevationDyspneaDizzinessHeadachePulmonary edema
10 mg/m2 Group4122111110110
5 mg/m2 Group1111110001001

[back to top]

Improvement in Peripheral Blood Count and Reduction in Number of Transfusions

Hematologic improvement will be an increased Hemoglobin of 1.5 g/dL or a reduction in the need for PRBC transfusions by at least 4 units over an 8 week period, at least 100% increase and an ANC of >0.5 x 10^9/L and an absolut platelet count increase of >30 x 10^9/L for patients who start at > 20 x 10^9/L, or increase from <20 x 10^9/L to >20 x 10^9/L and by at least 100%. (NCT00700011)
Timeframe: 2-3 months

Interventionparticipants (Number)
10 mg/m2 Group3
5 mg/m2 Group1

[back to top]

Determine Frequency and Duration of Bone Marrow Responses to IV Clofarabine

The International Working Group response criteria was used. Complete remission is defined as <5 % marrow blasts without evidence of dysplasia and normalization of the peripheral blood counts, including hemoglobin >11 g/dL, neutrophil count of >1 x 10^9/L. and platelet count of >100 x 10^9/L. Patients must also be transfusion-independent and not require any recombinant erythropoietin. Partial remission (PR) is defined as: satisfying complete remission criteria if abnormal before treatment, except that blasts are reduced by 50% or more compared to pretreatment levels, but still >5 %. Stable disease is defined as: failure to achieve at least a PR but without evidence of disease progression for at least 8 weeks.Progression of disease is defined as: disease progression with worsening cytopenias. Best response of these patients is used in the determination for this outcome below. (NCT00700011)
Timeframe: 2-3 months

,
Interventionparticipants (Number)
complete remissionpartial remissionstable diseaseprogressive disease
10 mg/m2 Group1142
5 mg/m2 Group0010

[back to top]

Number of Participants With DNA Hypomethylation During the Study

Since we previously observed decreases in DNA methylation in tumor cells after in vitro treatment with Clofarabine, we compared the long interspersednuclear element-1 methylation of genomic DNA obtained from CD3-depletedperipheral blood mononuclear cells between day 1 and day 5 of each cycle of Clofarabine. (NCT00700011)
Timeframe: assessed twice per cycle

Interventionparticipants (Number)
10 mg/m2 Group2
5 mg/m2 Group0

[back to top]

Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Alone.

Kaplan-Meier estimate of the probability of being alive and free of relapse or second malignancy three years after protocol enrollment (NCT00703820)
Timeframe: 3 years after completion of therapy

InterventionPercentage of participants (Number)
Cytarabine+Daunorubicin+Etoposide55.6
Clofarabine+Cytarabine54.3

[back to top]

Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Followed by Natural Killer Cell Transplantation.

Kaplan-Meier estimate of the probability of being alive and free of relapse or second malignancy three years after protocol enrollment (NCT00703820)
Timeframe: 3 years after completion of therapy

InterventionPercentage of participants (Number)
Cytarabine+Daunorubicin+Etoposide55.6
Clofarabine+Cytarabine77.8

[back to top]

The Overall Response Rate in Response to Low Dose Daily Oral Clofarabine in Patients With High Risk Myelodysplastic Syndrome or Chronic Myelomonocytic Leukemia (Dysplastic Type).

Response rate was measured as complete, partial and hematologic improvement by modified IWG criteria. For complete remission the following must be present for four weeks in a bone marrow aspirate and biopsy: <5% myeloblasts, normal maturation of all cell lines, persisted dysplasia. Peripheral blood counts need to be hemoglobin >11 g/dL, neutrophils >1000/mm3, platelets>100000/mm3, blasts 0%. Partial remission requires all criteria for complete remission except blasts decreased by >50% over pretreatment. Stable disease is defined as failure to achieve at least partial remission, but no evidence of progression for > 8 weeks. Failure is defined as death during tre3atment or disease progression characterized by worsening of cytopenias, increase in percentage of bone marrow blasts, or progression to a more advanced MDS FAB subtype than pretreatment. (NCT00708721)
Timeframe: 4 weeks

Interventionparticipants who experienced a response (Number)
All Evaluable Patients3

[back to top]

The Effect of Low Dose Daily Oral Clofarabine on miRNA and mRNA Expression Patterns in Patients With MDS

Assessment of potential change in miRNA and mRNA genetic expression patterns in patients following administration of low dose daily clofarabine. (NCT00708721)
Timeframe: through end of treatment

Intervention ()
All Patients0

[back to top]

The Effect of Low Dose Daily Oral Clofarabine on Global Methylation in Patients With MDS.

Potential genomic changes following low dose daily oral clofarabine administration will be assessed. (NCT00708721)
Timeframe: through end of treatment

Intervention ()
All Patients0

[back to top]

Time to Progression to Acute Myeloid Leukemia (AML)

Longest documented duration of time until progression to AML. (NCT00708721)
Timeframe: approximately 4 years

Interventioncycles (Number)
All Evaluable Patients51

[back to top]

Number of Participants Who Experienced a Dose-Limiting Toxicity

Dose-limiting toxicity was defined as any nonhematologic toxicity grade 3 or greater except for alopecia or nausea (which may be of grade 4 severity), or any grade 4 hematologic toxicity lasting more than 28 days after the last day of therapy. (NCT00708721)
Timeframe: 28 days after the first admistration of oral clofarabine

Interventionparticipants who experienced a DLT (Number)
Cohort 1: 10 mg/Day for 10 Days2
Cohort 2: 1 mg/Day for 10 Days4
Cohort 3: 1 mg/Day for 7 Days0
Not Evaluable0

[back to top]

Number of Participants With Adverse Events

NCI CTCAE version 3.0 will be used to assess adverse events. The number of participants experiencing adverse events and the number of adverse events per patient will be documented through the course of study. (NCT00708721)
Timeframe: To 30 days after end of treatment or until full resolution.

Interventionparticipants experiencing adverse events (Number)
All Patients11

[back to top]

Response of MDS Patients Treated With Low Dose Daily Oral Clofarabine.

Stable disease is defined as failure to achieve at least PR, but no evidence of progression for > 8 weeks. (NCT00708721)
Timeframe: approximately 4 years

InterventionParticipants (Count of Participants)
All Evaluable Patients2

[back to top]

Number of Participants With Hepatic (SGOT) Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine149

[back to top]

Number of Participants With Cardiac Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine20

[back to top]

Leukemia Free Survival

Time to event analysis used the day of transplant as day 0. (NCT00724009)
Timeframe: 2 years

Interventiondays (Median)
Clofarabine211

[back to top]

Cytoreductive Response

Percent of patients achieving cytoreductive response of marrow cellularity <20% and blasts < 10% (NCT00724009)
Timeframe: Day 12

Interventionpercentage of participants (Number)
Clofarabine52

[back to top]

Number of Participants Infection Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 3-4Grade 5
Clofarabine111

[back to top]

Number of Participants With Renal Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine91

[back to top]

Number of Participants With Skin Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine10

[back to top]

Number of Participants With Hepatic (Total Bilirubin) Adverse Events

Treatment-related toxicity was calculated according to Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00724009)
Timeframe: Day 12

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine143

[back to top]

Number of Serious Adverse Events Within 2 Years

(NCT00775593)
Timeframe: At 2 years from study entry

Interventionserious adverse events (Number)
Study Group22

[back to top]

Duration of Survival

(NCT00775593)
Timeframe: At 2 years from study entry

Interventionmonths (Median)
Study Group10.9

[back to top]

Duration of Response

Participants who responded to treatment (NCT00775593)
Timeframe: At 2 years from study entry

Interventionmonths (Mean)
Study Group3.5

[back to top]

Complete Response Rate

(NCT00775593)
Timeframe: At 2 years from study entry

Interventionparticipants (Number)
Study Group4

[back to top]

Disease-free (DFS) or Relapse-free Survival (RFS) Time

Disease (DFS) or Relapse-free survival (RFS): Time from date of treatment start until the date of first objective documentation of disease-relapse; Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy and then every 2 weeks (+/- 7 days) as required by leukemia evolution until remission or non-response. Among participants who achieved CR or CRp, RFS was defined as the time interval between the date of response (ie CR or CRp) and the date of relapse or date of death, whichever occurs first. CR or CRp participants who were alive and relapse-free were censored at the off-study date. Full range reflects time to disease progression only, therefore does not reflect a lesser survival time due to other reasons than disease progression/relapse. (NCT00778375)
Timeframe: Evaluated from treatment date until date of disease progression/relapse, followed for 5 years/60 months.

InterventionMonths (Median)
Clofarabine + Cytarabine + Decitabine15.9

[back to top]

Complete Remission (CR) Rate for First 60 Participants

All responses were defined as per IWG criteria (2003) where CR Rate defined as number of participants with CR out of total treated participants. Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy. (NCT00778375)
Timeframe: Evaluation following two 10 day cycles on day 21 of therapy, continuing up to 210 days

InterventionPercentage of Participants (Number)
Clofarabine + Cytarabine + Decitabine58

[back to top]

Event Free Survival (EFS)

EFS is defined as length of time after primary treatment for a cancer ends that the participant remains free of certain complications or events that the treatment was intended to prevent or delay, for example but not exclusive of hematologic and non-hematologic toxicities, cumulative toxicities with consolidation courses, or emergence of resistance to the chemotherapy component of treatment. (NCT00778375)
Timeframe: Follow up up to 5 years/60 months.

InterventionMonths (Median)
Clofarabine + Cytarabine + Decitabine7.7

[back to top]

Overall Response Rate (CR, CRp/CRi and PR)

IWG Response criteria (2003): Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Complete Remission without Platelet Recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 10^9/L or Complete remission with incomplete marrow recovery (CRi), defined as CR above, but without normal blood counts; Partial Remission (PR): Blood count recovery as for CR, but with both a decrease in marrow blasts of at least 50% and not more than 6 to 25% abnormal cells in the marrow. Participants not achieving a complete remission following first induction course, can receive a second induction course at least 28 days following first to optimize response if possible. (NCT00778375)
Timeframe: Beginning assessment following two 10-day induction cycles through an additional re-induction cycle, up to 40 days

InterventionPercentage of Participants (Number)
Clofarabine + Cytarabine + Decitabine73

[back to top]

Median Overall Survival (OS)

Overall survival (OS): Time from date of treatment start until date of death due to any cause. (NCT00778375)
Timeframe: Evaluated from treatment date until date of death, followed for 5 years/60 months.

InterventionMonths (Median)
Overall SurvivalOverall Survival among Responders ((n=86)
Clofarabine + Cytarabine + Decitabine11.121.1

[back to top]

Number of Participants With Complete Remission [Complete Response (CR), Complete Response With Platelet Recover (CRp) or Complete Response With Incomplete Marrow Recovery (CRi)]

All responses were defined as per IWG criteria (2003) where CR Rate defined as number of participants with CR out of total treated participants. Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Complete response with incomplete marrow recovery (CRi), defined as CR above, but without normal blood counts. Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy. (NCT00778375)
Timeframe: Beginning assessment following two 10-day induction cycles through an additional re-induction cycle, up to 40 days

,
InterventionParticipants (Number)
CRCRp/CRi
Induction Clofarabine + Cytarabine + Decitabine609
Re-Induction124

[back to top]

Overall Survival

Overall survival was defined as time from date of treatment initiation until date of death due to any cause. (NCT00814164)
Timeframe: 4 years

Interventionmonths (Median)
Clorafarbine With Daunorubicin11.2

[back to top]

Disease-free Survival

Disease-free survival was defined as time from first objective documentation of CR or CRp until the date of first objective documentation of disease relapse or death due to any cause, whichever occurs first. (NCT00814164)
Timeframe: 5 years

Interventionmonths (Median)
Clorafarbine With Daunorubicin6.8

[back to top]

Complete Remission (CR)

Complete Response/Remission (CR) was defined on morphologic criteria at a single response assessment as follows: A bone marrow aspirate or biopsy of < 5% blasts, with evidence of normal hematopoiesis; Absence of Auer rods in the blast that are present; Absence of extramedullary disease [imaging required only if obtained pretreatment for known site(s) of disease]; If applicable and available, absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping; Recovery of peripheral counts (platelets ≥100x109/L, and ANC ≥1.0x109/L). Peripheral count recovery must be documented no earlier than 7 days prior to, and no later than 14 days following, the bone marrow assessment that provides evidence of the CR. Complete Response/Remission without platelet recovery (CRp) was defined as all criteria for CR except for thrombocytopenia (platelet count ≥75x109/L). (NCT00814164)
Timeframe: 2 years

Interventionpercentage of participant (Number)
Clorafarbine With Daunorubicin38.1

[back to top]

Disease Free Survival

Median disease-free survival (NCT00839982)
Timeframe: Up to 5 years

Interventionmedian months (Median)
Treatment (Chemotherapy)7.4

[back to top]

Maximum Tolerated Dose

We identified 20 mg/d for 5 d as the maximum tolerated dose (MTD) of oral clofarabine. (NCT00839982)
Timeframe: up to 5 years

Interventionmg/day (Number)
Treatment (Chemotherapy)20

[back to top]

Number of Patients With Dose Limiting Toxicity

Dose limiting toxicity (DLT) consists of grade 3-4 non-hematologic toxicity at least possibly related to study drug. Exceptions include neutropenic fever; drug-related fever; alopecia; anorexia; inadequately treated nausea, vomiting and/or diarrhea; and grade 3/4 increase in ALT, AST, or bilirubin recovering to < grade 2 by 7 days. Prolonged grade 2 myelosuppression lasting longer than 49 days in patients who don't proceed to additional cytotoxic therapy is considered a DLT. The MTD or recommended phase II dose is the highest dose level at which no more than 1 patient out of 6 experiences DLT. (NCT00839982)
Timeframe: Outcomes by day 30

InterventionParticipants (Count of Participants)
Dose Level 10
Dose Level 24
Dose Level 32
Dose Level 42

[back to top]

Overall Survival

Median overall survival (NCT00839982)
Timeframe: Up to 5 years

Interventionmedian months (Median)
Treatment (Chemotherapy)6.8

[back to top]

Relapse Rate of Participants Treated With Thiotepa, Busulfan, and Clofarabine

Relapse Rate will be estimated using the Kaplan-Meier method. (NCT00857389)
Timeframe: Up to 2 years post transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine26

[back to top]

Overall Survival Rate

Overall Survival Rate will be estimated using the Kaplan-Meier method. (NCT00857389)
Timeframe: Up to 3 years post transplant

Interventiondays (Median)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine320

[back to top]

Number of Participants With Survival Rate at 100 Days Post-transplant

The toxicities will be monitored and scored on a daily basis following the methods of Simon R. Practical Bayesian Guideline for Phase IIB Clinical Trials. A Bayesian stopping rule will be used to stop the trial if there is a 90% chance that the true toxicity rate exceeds the target toxicity rate of 0.25. (NCT00857389)
Timeframe: 100 days post-transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine45

[back to top]

Number of Participants With Serious Adverse Events

Severity of toxicities graded according to the NCI Common Toxicity Criteria Adverse Effects (CTCAE) v3.0. Standard reporting guidelines followed for adverse events. Safety data summarized by category, severity and frequency. (NCT00857389)
Timeframe: up to 30 days post transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine52

[back to top]

Number of Participants With Disease Free Survival

Kaplan-Meier product limit method to estimate the disease free survival. (NCT00857389)
Timeframe: Up to 2 years post transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine32

[back to top]

Graft vs Host Disease (GVHD)

Severity of toxicities graded according to the NCI Common Toxicity Criteria Adverse Effects (CTCAE) v3.0. Standard reporting guidelines followed for adverse events. Safety data summarized by category, severity and frequency. (NCT00857389)
Timeframe: Up to 30 days post transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine34

[back to top]

Engraftment

Engraftment is most commonly defined as the first of three consecutive days of achieving a sustained peripheral blood neutrophil count of >500 × 10^6/L . (NCT00857389)
Timeframe: up to 100 days post transplant

InterventionParticipants (Count of Participants)
Conditioning Reg- Thiotepa, Busulfan, and Clofarabine52

[back to top]

Overall Survival

(NCT00863434)
Timeframe: Every 3 months for 2 years, and then annually for 3 years

Interventionmonths (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)9.73

[back to top]

Disease-free Survival

(NCT00863434)
Timeframe: Every 3 months for 2 years, and then annually for 3 years

Interventionmonths (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)6.43

[back to top]

Minimal Residual Disease as Assessed by Bone Marrow Flow Cytometry

Percent of white blood cells that are blasts in the bone marrow post-treatment. (NCT00863434)
Timeframe: Post-treatment

Interventionpercent of white blood cells (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)1.8

[back to top]

Event Free Survival (EFS) at 1 Year

Percentage of participants with event free survival at 1 year. Event free survival (EFS) where event is defined as either death or transformation to acute myeloid leukemia (AML) (marrow and/or blood blasts >/= 20%) (NCT00903760)
Timeframe: Assessed at 12 months/1 year

Interventionpercentage of participants (Number)
Decitabine + Clofarabine26
Decitabine65

[back to top]

Participant Response

Responses: Complete Remission (CR): Normalization peripheral blood & bone marrow /= 1.0 x 10^9/L, & platelet>/= 100 x 10^9/L. Partial Remission: all CR criteria if abnormal before treatment except marrow blasts decrease =/> 50% compared to pretreatment or a less-advanced MDS disease classification than prior to treatment. Hematologic Improvement: Response maintained 8+ weeks: Hemoglobin (pretreatment < 11 g/dL): improves 1.5 g/dL or reduced by 4 units of red blood cell (RBC) transfusions in 8 weeks compared with pretreatment transfusions in 8 weeks; or Platelet (pretreatment < 100 x 10^9/L): absolute increase >/= 30 x 10^9/L, starting platelet > 20 x 10^9/L OR increase < 20 x 10^9/L to > 20 x 10^9/L and =/> 100%. Neutrophil (pretreatment < 1 x 10^9/L): increase 100% & absolute increase > 0.5 x 10^9/L. (NCT00903760)
Timeframe: Up to 6 months

,
Interventionparticipants (Number)
Complete Remission (CR)Partial Remission (PR)Hematologic Improvement (HI)
Decitabine904
Decitabine + Clofarabine805

[back to top]

Duration of Overall Response

Duration was calculated by Kaplan-Meier estimates (NCT00924443)
Timeframe: From 20 months up to 48 months

Interventiondays (Median)
Clofarabine62

[back to top]

Duration of Complete Remission

Duration was calculated by Kaplan- Meier estimates (NCT00924443)
Timeframe: From 20 months up to 48 months

Interventiondays (Median)
Clofarabine63

[back to top]

Rate of Response (Complete, Complete With Incomplete Blood Count Recovery, Partial)

"Response was determined by assessment of morphology and blast count from bone marrow aspirates and peripheral blood performed prior to first dose and at the end of clofarabine treatment.~Response was determined at the end of each cycle of clofarabine, and assessed using the participant's best response to clofarabine treatment." (NCT00924443)
Timeframe: At month 20

Interventionpercent of participants (Number)
Complete responseComplete with incomplete blood count recoveryPartial response
Clofarabine21235

[back to top]

Overall Survival

Calculated by Kaplan-Meier estimates (NCT00924443)
Timeframe: From 20 months up to 48 months

Interventiondays (Median)
Clofarabine173

[back to top]

Overall Response Rate (ORR)

"ORR rate was defined as the sum of the number of participants in the study population with complete remission (CR), complete remission with incomplete blood count recovery (CRi), or partial remission (PR) divided by the total number of participants in the study population.~ORR rate was determined by assessment of morphology and blast count from bone marrow aspirates and peripheral blood performed prior to first dose and at the end of clofarabine treatment. The ORR was determined at the end of each cycle of clofarabine, and assessed using the participant's best response to clofarabine treatment." (NCT00924443)
Timeframe: At month 20

Interventionpercentage of participants (Number)
Clofarabine48

[back to top]

Death

Number of participants who died. (NCT00939653)
Timeframe: From the first dose of study therapy until 30 days after last therapy dose

InterventionParticipants (Count of Participants)
Enrolled Patients4

[back to top]

Achievement of Complete Remission (CR) at Reinduction

Disease response assessed after chemotherapy from bone marrow aspirates/biopsies and complete blood count. (NCT00939653)
Timeframe: Between Days 22-36 or on Day 43 and weekly thereafter if peripheral counts haven't recovered

InterventionParticipants (Count of Participants)
Enrolled Patients1

[back to top]

Number of Participants With Skin Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation

Toxicity was scored according to NCI/CTC version 3 (NCT00943592)
Timeframe: Day 7 until Day 30

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine, Melphalan, and Alemtuzumab68

[back to top]

Number of Participants With Renal Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation

Toxicity was scored according to NCI/CTC version 3 (NCT00943592)
Timeframe: Day 7 until Day 30

Interventionparticipants (Number)
Grade 1-2Grade 3-4Grade 5
Clofarabine, Melphalan, and Alemtuzumab26133

[back to top]

Number of Participants With Hepatic Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation

Toxicity was scored according to NCI/CTC version 3 (NCT00943592)
Timeframe: Day 7 until Day 30

Interventionparticipants (Number)
Grade 1-2Grade 3-4
Clofarabine, Melphalan, and Alemtuzumab4830

[back to top] [back to top]

Relapse Rate

(NCT00943592)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Clofarabine, Melphalan, and Alemtuzumab29

[back to top]

Progression-free Survival (PFS)

Progression is defined from stem cell infusion to disease relapse, i.e., recurrence of hematologic malignancy and/or need for treatment after transplant for disease or death from any cause, whichever occurred first. (NCT00943592)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Clofarabine, Melphalan, and Alemtuzumab45

[back to top]

Overall Survival (OS)

(NCT00943592)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Clofarabine, Melphalan, and Alemtuzumab59

[back to top]

Number of Participants With Other Adverse Events During the Conditioning Regimen Prior to Stem Cell Transplantation

Toxicity was scored according to NCI/CTC version 3 (NCT00943592)
Timeframe: Day 7 until Day 30

Interventionparticipants (Number)
Grade 1-2Grade 3-4Grade 5
Clofarabine, Melphalan, and Alemtuzumab1277

[back to top] [back to top]

Complete Remission

Complete remission (CR) is defined as: <5% marrow aspirate blasts. Blasts can be >=5% if the blasts are found to be myeloid and there is no evidence of lymphoblasts by flow cytometry or immunostaining. Neutrophils >= 1000/mcl; platelets >100,000/mcl; and no blasts in the peripheral blood. C1 Extramedullary disease status as defined in the protocol. Complete remission with incomplete platelet recovery (CRi) is same as CR but platelet count may be <=100,000/mcl and/or ANC may be <1,000/mcl. (NCT00945815)
Timeframe: After induction therapy was completed (1 or 2 months)

Interventionpercentage of participants (Number)
Ara-C + Clofarabine + Epratuzumab52

[back to top]

Overall Survival at One Year

The number of participants alive one year after baseline. (NCT00973752)
Timeframe: 1 years

InterventionParticipants (Count of Participants)
Experimental19

[back to top]

Number of Adverse Events

(NCT00983528)
Timeframe: 2 years

Interventionadverse events (Number)
Nose BleedDiarrheaNauseaVomitingDyspesiaFeverConstipationPainInsomniaItchingAnxietyRashHivesDry EyesDeaths
Alemtuzumab and Clofarabine156614566461114

[back to top] [back to top]

Median Event-Free Survival (EFS)

Event-free survival (EFS) defined as time from start of treatment to first documentation of disease relapse or death. Bayesian time-to-event model will be used to monitor progression free survival. (NCT01025154)
Timeframe: 2 years

InterventionMonths (Median)
Clofarabine, Cytarabine + Idarubicin13.5

[back to top]

Overall Response: Number of Participants With Complete Remission or Complete Remission Without Platelet Recovery

Overall Response (CR+CRp) defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 10^9/L and platelet count > 100 x 10^9/L, and normal bone marrow differential (< 5% blasts); and, Complete Remission without Platelet Recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 10^9/L. Response evaluated within 8 weeks after induction therapy. (NCT01025154)
Timeframe: 8 weeks after Induction therapy (induction cycle 4-6 weeks)

Interventionparticipants (Number)
Complete RemissionComplete Remission without Platelet Recovery
Clofarabine, Cytarabine + Idarubicin423

[back to top]

Overall Survival

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. (NCT01101880)
Timeframe: Up to 5 years

Interventionmonths (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)24.3

[back to top]

Event Free Survival

Number of patients in remission at a median follow up of 15 months. (NCT01101880)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy and Colony Stimulating Factor)21

[back to top]

Duration of Remission

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. Remission is defined as less than 5% blasts in the bone marrow, no appearance of blasts in the peripheral blood, and no extramedullary disease (appearance of leukemic cells in other tissues). (NCT01101880)
Timeframe: Up to 5 years

Interventionweeks (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)7

[back to top]

Time to Progression

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. (NCT01101880)
Timeframe: Up to 5 years

Interventionweeks (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)7

[back to top]

Rates of Complete Remission and Complete Remission With Incomplete Recovery of Counts

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. Complete remission is defined as less than 5% blast cells present in the bone marrow and count recovery (absolute neutrophil count greater than 1000/microL and platelet count greater than 100,000/microL). Complete remission with incomplete recovery of counts is defined as less than 5% blast cells present in the bone marrow without compete count recovery (absolute neutrophil count less than 1000/microL and platelet count less than 100,000/microL). (NCT01101880)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
CR achievedCR achieved with first course of inductionCR + CRp achievedCR achieved with no AHDCR achieved with AHDCR + CRp achieved with AHDCR achieved with FLT3 positiveCR achieved with favorable risk cytogeneticsCR achieved with intermediate risk cytogeneticsCR + CRp achieved with intermediate risk cyto.CR achieved with unfavorable risk cytogeneticsCR + CRp achieved with unfavorable risk cyto.
Treatment (Chemotherapy and Colony Stimulating Factor)383341231518742627810

[back to top]

Survival and Disease-free Survival (DFS)

(NCT01119066)
Timeframe: 2 years post transplant

,,,
Intervention% of pts at 2 years (Number)
SurvivalDisease Free Survival
Busulfan, Melphalan and Fludarabine72.162.1
Clofarabine, Melphalan and Thiotepa63.859.6
Melphalan, Fludarabine and Thiotepa71.662.3
Total Body Irradiation, Thiotepa and Cyclophosphamide68.758.9

[back to top]

Survival and Disease-free Survival (DFS)

(NCT01119066)
Timeframe: at 6 months post transplant

,,,
Interventionpercentage of participants (Number)
SurvivalDisease Free Survival
Busulfan, Melphalan and Fludarabine92.489.0
Clofarabine, Melphalan and Thiotepa87.285.1
Melphalan, Fludarabine and Thiotepa90.990.9
Total Body Irradiation, Thiotepa and Cyclophosphamide93.382.5

[back to top]

Survival and Disease-free Survival (DFS)

(NCT01119066)
Timeframe: 1 year post transplant

,,,
Intervention% of pts at 1 year post transplant (Number)
SurvivalDisease free survival
Busulfan, Melphalan and Fludarabine83.873.7
Clofarabine, Melphalan and Thiotepa85.183.0
Melphalan, Fludarabine and Thiotepa81.872.7
Total Body Irradiation, Thiotepa and Cyclophosphamide80.868.3

[back to top]

Number of Participants With Acute and Chronic GVHD Following T-cell Depleted, CD34+ Progenitor Cell Enriched Transplants Fractionated by the CliniMACS System.

Standard BMT-CTN and IBMTR systems clinical criteria as defined by Rowlings, et al will be used to establish and grade acute GvHD. Chronic GvHD will be diagnosed and graded according to the criteria of Sullivan (CIBMTR). (NCT01119066)
Timeframe: 3 years

,,,
Interventionparticipants (Number)
aGVHD II-IVcGVHD, LimitedcGVHD, Extensive
Busulfan, Melphalan and Fludarabine6041
Clofarabine, Melphalan and Thiotepa1011
Melphalan, Fludarabine and Thiotepa201
Total Body Irradiation, Thiotepa and Cyclophosphamide3428

[back to top] [back to top]

The Incidence of Durable Hematopoietic Engraftment for T-cell Depleted Transplants Fractionated by the CliniMACS System Administered After Each of the Four Disease Targeted Cytoreduction Regimens.

(NCT01119066)
Timeframe: 3 years

,,,
InterventionParticipants (Count of Participants)
EngraftedPrimary Graft FailureLate graft failureNot Evaluable Engraftment
Busulfan, Melphalan and Fludarabine205050
Clofarabine, Melphalan and Thiotepa45020
Melphalan, Fludarabine and Thiotepa10010
Total Body Irradiation, Thiotepa and Cyclophosphamide118011

[back to top]

Number of Patients With Dose-Limiting Toxicity (DLT)

Treatment related toxicities that preclude proceeding to HSCT by day 56 of the treatment course. (NCT01158885)
Timeframe: Beginning with the first dose of investigational product until day 56 of treatment course, an average of 1 year

InterventionParticipants (Count of Participants)
Single Arm0

[back to top]

Number of Participants in Phase I With First Cycle Dose Limiting Toxicities (DLT) Observed

Dose limiting toxicity (DLT) consists of participants who developed DLT during maximum tolerated dose (MTD) estimation period where DLTs observed during dose escalation were used to develop MTD. The MTD is the highest dose level in which <2 participants of 6 develop first cycle. DLT. Toxicity graded according to the NCI Common Toxicity Criteria Version 3.0. The timeframe to assess dose-limiting toxicities (DLT's) will be the first cycle of treatment, i.e. the first 4 weeks on study. The Plerixafor dose to be used in Phase II of the protocol is the highest dose at which no more than 1 of 6 patients experience a DLT in the Phase I part of the protocol or a lower dose selected at the end of dose escalation. (NCT01160354)
Timeframe: First cycle of treatment, i.e. first 4 weeks on study

InterventionParticipants (Count of Participants)
Plerixafor 240 mcg/kg + Clofarabine0
Plerixafor 320 mcg/kg + Clofarabine0
Plerixafor 400 mcg/kg + Clofarabine0

[back to top]

Part 2: Number of Participants With Relapsed Disease

Number of high risk patients with relapsed disease after receiving the maximum dose of clofarabine. Relapse is defined as the presence of >5% aberrant blasts by morphology on a marrow aspirate or the presence of circulating blasts in the peripheral blood. (NCT01252667)
Timeframe: 6 months post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)3

[back to top]

Pharmacokinetics (PK) of Clofarabine

Pharmacokinetic measurement of the volume of plasma from which clofarabine is completely removed per unit time. Clearance normalized to actual body weight. Pharmacokinetic analyses were performed on only a subset of patients and no aggregate calculations were made using the data. (NCT01252667)
Timeframe: Blood was drawn on day -6 before the first clofarabine dosing, at the end of the infusion, and at 3, 4, 5, 6 and 24 hours after the start of infusion. Only pre-dose samples were drawn on days -5, -4, and -3.

InterventionL/h/kg (Number)
ID 1ID 2
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)0.530.37

[back to top]

Pharmacokinetics (PK) of Clofarabine

Pharmacokinetic measurement of the volume of plasma from which clofarabine is completely removed per unit time. Clearance normalized to actual body weight. Pharmacokinetic analyses were performed on only a subset of patients and no aggregate calculations were made using the data. (NCT01252667)
Timeframe: Blood was drawn on day -6 before the first clofarabine dosing, at the end of the infusion, and at 3, 4, 5, 6 and 24 hours after the start of infusion. Only pre-dose samples were drawn on days -5, -4, and -3.

InterventionL/h/kg (Number)
ID 4ID 5ID 6
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0.290.400.50

[back to top]

Number of Participants With Minimal Residual/Recurring Disease (MRD) Post-transplant

Number of patients with MRD post-transplant, detected in the bone marrow as cytogenetic abnormalities or <5% monoclonal blasts by flow cytometry. (NCT01252667)
Timeframe: 1 Year post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)2
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)6

[back to top]

Number of Participants Surviving Overall

Number of patients surviving overall post-transplant. (NCT01252667)
Timeframe: 1 Year post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)2
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)1
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)22

[back to top]

Part 1: Number of Participants With Dose Limiting Toxicities (DLT)

"The primary objective of part 1 is to determined the highest dose of clofarabine that can be tolerated safely in conjunction with nonmyeloablative transplant. If three patients successfully transplant without DLT, dose escalation occurs. If one of those three patients experiences DLT an additional three patients will be treated using the same dose. If a second DLT is observed in the first 3-6 patients, or if three patients are successfully transplanted without DLT at the highest dose the study will proceed to Part 2 using the maximum tolerated dose.~A Clofarabine related dose-limiting toxicity is defined as a grade 4 toxicity involving the lungs, heart, liver (not resolving in 48 hours), kidneys (not resolving in 48 hours), gastrointestinal tract, and/or central nervous system." (NCT01252667)
Timeframe: 14 days post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)0

[back to top]

Number of Participants Surviving Progression-free.

Number of patients surviving without progressive disease post-transplant. Progression is defined as the presence of >5% aberrant blasts by morphology on a marrow aspirate or the presence of circulating blasts in the peripheral blood. (NCT01252667)
Timeframe: 1 Year post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)2
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)1
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)21

[back to top]

Number of Participants Who Graft Rejected.

Number of patients who graft rejected post-transplant. Graft rejection is defined as <5% donor peripheral blood T cells (CD3+). (NCT01252667)
Timeframe: 1 Year post-transplant.

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)0

[back to top]

Number of Non-Relapse Mortalities (NRM)

Number of patients who expired without disease progression/relapse. (NCT01252667)
Timeframe: 100 days post-transplant

InterventionParticipants (Count of Participants)
Part 1 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 1 - Dose 2 (40 mg/m^2 Clofarabine)1
Part 1 - Dose 3 (50 mg/m^2 Clofarabine)0
Part 2 - Dose 1 (30 mg/m^2 Clofarabine)0
Part 2 - Dose 2 (40 mg/m^2 Clofarabine)0
Part 2 - Dose 3 (50 mg/m^2 Clofarabine)1

[back to top]

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT01289457)
Timeframe: up to 2 years

InterventionMonths (Median)
Group 1 CIA14.5
Group 2 FLAI15.1

[back to top]

Response Rates of Clofarabine, Idarubicin, and Cytarabine (CIA) Versus Fludarabine, Idarubicin, and Cytarabine (FLAI)

NCI & Myelodysplastic syndromes (MDS) International Working Group (IWG) Definitions: Complete Response (CR): Neutrophil count ≥1.0 ×10^9/L, Platelet count ≥100 ×10^9/L, Bone marrow aspirate 10% after initial response. Response assessed Day 28 of every 2-3 cycles during treatment. (NCT01289457)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Group 1 CIA107
Group 2 FLAI76

[back to top]

Event-Free Survival (EFS) at 2 Years

Comparison of the event-free survival (EFS) between treatment CIA and FLAI, where an event is defined to be resistance to treatment, relapse (after response) or death, whichever occurred first. (NCT01289457)
Timeframe: Up to 2 years or until relapse/death

InterventionMonths (Median)
Group 1 CIA7.1
Group 2 FLAI8.4

[back to top]

Maximum Tolerated Dose (MTD) of Clofarabine, Idarubicin, and Cytarabine

MTD is highest dose level in which <2 patients of 6 develop first cycle dose limiting toxicities (DLT). Toxicity defined as any treatment-related grade 3 or greater non-hematological toxicities. (NCT01289457)
Timeframe: 28 days

Interventionmg/m^2 (Number)
Clofarabine + Idarubicin + Cytarabine15

[back to top]

Overall Survival (OS)

Overall survival defined as the time interval from study entry date to the date of death due to any cause, measured in days/months. Bayesian time-to-event model used to monitor overall survival. (NCT01444742)
Timeframe: 5 years

InterventionMonths (Median)
Clofarabine + Cytarabine10.3

[back to top]

Number of Participants With Complete Response (CR)

Complete Response Criteria (CR must last for at least 4 weeks): Marrow: /= 11 g/dL (untransfused, patient not on EPO); Neutrophils >/= 1x109/L (not on myeloid growth factor); Platelets >/= 100 * 109/L (not on thrombopoietic agent); No blasts. Bone marrow aspirate and/or biopsy at the end of course 1 (day 28 +/- 7 days). The method of Thall, Simon, and Estey used to monitor response. (NCT01444742)
Timeframe: 4 weeks after first cycle

InterventionParticipants (Count of Participants)
Clofarabine + Cytarabine19

[back to top]

Minimal Residual Disease (MRD)

Detectable disease in bone marrow or blood: A binary measure, positive (detectable), negative (non-detectable) (NCT01451515)
Timeframe: Day 8

,,
Interventionparticipants (Number)
NegativePositive
Stratum 180
Stratum 241
Stratum 304

[back to top]

Probability of Event-free Survival (EFS)

"For EFS, relapse and second malignancies are considered as failures in addition to death in complete remission. The time to EFS will be set to 0 for patients who fail to achieve complete remission. Kaplan-Meier estimates of the OS and EFS curves are computed, along with estimates of standard errors by Peto's method.~Please note the unit of measurement of probabilities are percentages." (NCT01451515)
Timeframe: Two years post therapy.

Interventionpercentage of event-free patients (Number)
Stratum 191.7
Stratum 271.4
Stratum 3100
All Enrollments86.96

[back to top]

Minimal Disseminated Disease (MDD)

Detectable disease in bone marrow or blood: A binary measure, positive (detectable), negative (non-detectable) (NCT01451515)
Timeframe: At Diagnosis

,,
Interventionparticipants (Number)
NegativePositive
Stratum 141
Stratum 223
Stratum 304

[back to top]

Probability of Overall Survival (OS)

"For OS, only deaths are considered failures for OS. Kaplan-Meier estimates of the OS curves are computed along with estimates of standard errors by Peto's method.~Please note the unit of measurement of probabilities are percentages." (NCT01451515)
Timeframe: Two years post therapy.

Interventionpercentage of patients alive (Number)
Stratum 191.7
Stratum 271.4
Stratum 3100
All Enrollments86.96

[back to top]

The Percentage of Patients Alive at 1 Year

Overall survival was calculated following transplant using a CloBu4 conditioning regimen for patients with non-remission AML (NCT01457885)
Timeframe: 1 year

Interventionpercentage of patients (Number)
CloBu4 Regimen32

[back to top]

Incidence of Relapse

(NCT01457885)
Timeframe: 2 years

Interventionpercentage of patients (Number)
CloBu4 Regimen55

[back to top]

Cumulative Incidence of Non Relapse Mortality (NRM)

Percentage of patients passed without relapse/recurrence at 1 year. (NCT01457885)
Timeframe: 1 year

Interventionpercentage of patients (Number)
CloBu4 Regimen21

[back to top]

Cumulative Incidence of Relapse (CIR)

Cumulative incidence of relapse (CIR) at 1 year, it will be calculated from the date of achievement of the first CR, using the cumulative incidence method, considering death in CR as a competing risk. Patients still alive, without a date of relapse, will be censored at the time of the last follow-up. In this case, the CIR curve will be truncated at 1 year. (NCT01462253)
Timeframe: At one year from therapy completion.

InterventionPercentage of patients (Number)
Clofarabine + Cyclophosphamide31.25

[back to top]

Number of Participants With Toxicity of Grade 2 or Greater

Referring to CTCAE (Common Toxicity Criteria Events), version 4.0 (NCT01462253)
Timeframe: At 13 months from study entry

InterventionParticipants (Count of Participants)
Clofarabine + Cyclophosphamide10

[back to top]

The Primary End-point is the Number of Patients in CR After Induction Therapy.

Disappearance of any clinical and laboratoristic sign of ALL. The patient must be transfusion-free with neutrophils >1.0 x109/L and platelets >100 x109/L. BM examination must show absence or reduction of blast cell content (< 5%, none of which obviously leukemic), with cellularity in the normal or slightly hypocellular range and with evidence of trilineage hemopoiesis. BM is examined on day 28 from start of chemotherapy cycle 1, or later as clinically indicated in ill/cytopenic patients, and after cycle 2 in patients with PR proceeding to this treatment. (NCT01462253)
Timeframe: At day +28 from start of chemotherapy cycle 1 and after cycle 2 in pts with PR

InterventionParticipants (Count of Participants)
Clofarabine + Cyclophosphamide9

[back to top]

Overall Survival (OS)

Overall Survival (OS) at 1 year; defined as the time interval between inclusion and death for any cause; patients still alive will be censored at the time of the last follow-up. In this case, the OS curve will be truncated at 1 year. (NCT01462253)
Timeframe: At one year from therapy completion.

InterventionPercentage of patients (Number)
Clofarabine + Cyclophosphamide28.6

[back to top]

Number of Participants With Minimal Residual Disease (MRD) Response in Remission.

(NCT01462253)
Timeframe: At week 10, 16 and 22 from start of treatment and the, every three months till study completion

InterventionParticipants (Count of Participants)
Clofarabine + Cyclophosphamide1

[back to top]

Disease-free Survival (DFS)

Disease-free survival (DFS) at 1 year, defined as the time interval between the evaluation of CR and relapse of the disease or death in first CR; patients still alive, in first CR, will be censored at the time of the last follow-up. In this case, the DFS curve will be truncated at 1 year (NCT01462253)
Timeframe: At one year from completion of chemotherapy

InterventionPercentage of patients (Number)
Clofarabine + Cyclophosphamide31.25

[back to top]

Number of Participants in the Study Who Are With no Grade 3 or 4 Acute Graft-versus-host Disease at Any Time During the First 100 Days Post Transplant.

Number of participants in the study who are with no Grade 3 or 4 acute graft-versus-host disease at any time during the first 100 days post transplant. (NCT01471444)
Timeframe: 100 days post transplant

InterventionParticipants (Count of Participants)
Arm A (Flu+Bu)125
Arm B (Flu+Clo+Bu)115

[back to top]

Overall Survival (OS) Post Transplant at 1, 3 and 5 Years

Number of participants in the study who are alive and disease free at 1, 3 and 5 years post transplant. (NCT01471444)
Timeframe: Post transplant after 1, 3 and 5 years

,
InterventionParticipants (Count of Participants)
1 Year Post Transplant3 Year Post Transplant5 Year Post Transplant
Arm A (Flu+Bu)836964
Arm B (Flu+Clo+Bu)826963

[back to top]

Progression-Free Survival (PFS)

Number of events with progression free survival. (Progression is defined as more than 5% blast in the peripheral blood or bone marrow biopsy.) or expired from treatment related mortality post transplant. (NCT01471444)
Timeframe: From day of transplant to disease of progression or death of any cause, whichever came first, assessed up to 5 years

InterventionNumber of events (Number)
Arm A (Flu+Bu)69
Arm B (Flu+Clo+Bu)61

[back to top]

Number of Participants With Non Relapse Mortality at 100 Day Post Transplant

Number of participants expired from complications other than relapsed disease at 100 day Post Transplant. (NCT01471444)
Timeframe: 100 day Post Transplant

InterventionParticipants (Count of Participants)
Arm A (Flu+Bu)3
Arm B (Flu+Clo+Bu)6

[back to top] [back to top]

Engraftment Rate of Patients With Non-malignant Diseases (Stratum A)

Engraftment will be defined as the development of an Absolute Neutrophil Count (ANC) >500 for 3 consecutive days plus donor CD14/15 cells >70%. The engraftment rate in the population used for historical control is 40%. If 3 patients in Stratum A experience graft rejection, this stratum will close early for failing to achieve superior engraftment compared to standard-of-care. (NCT01596699)
Timeframe: Participants will have engraftment blood studies starting approximately Day 30 post hematopoietic stem cell transplant and then monthly until stable. Average study participation is approximately 5 years.

Interventionpercentage of participants (Number)
Stratum A: Patients With Non-Malignancies66.67

[back to top]

Mixed-donor Chimerism Rate of Patients With High-risk Myeloid Malignancies (Stratum B)

Full-donor chimerism will be defined by as ≥99% donor cells by Short Tandem Repeat (STR) analysis in all cell lines (CD3, CD14/15, and CD19) in peripheral blood. The historic control for Stratum B was determined using the 20 patients who were transplanted from 2005 - 2010 with Busulfan (BU)-based regimens and who retrospectively would have been eligible for the current trial. Of these 20 patients, at 100 days post-HCT, only 8 (40%) patients had full-donor chimerism. If 5 patients in Stratum B experienced mixed-donor chimerism at Day 100, we will close this stratum early for failing to achieve superior donor cell engraftment compared to standard-of-care. (NCT01596699)
Timeframe: Participants will have peripheral blood chimerism assessed at Day 100 post hematopoietic stem cell transplant and then monthly until stable.

Interventionpercentage of participants (Number)
Stratum B: Patients With Myeloid Malignancies66.67

[back to top]

Incidence and Severity of Chronic Graft Versus Host Disease (GVHD)

The severity of chronic GVHD will be described. Chronic GVHD was evaluated using NIH Consensus Global Severity Scoring. The number of participants with chronic GVHD is given, organized by severity. (NCT01621477)
Timeframe: 100 days post transplant

Interventionparticipants (Number)
No Chronic GVHDMildModerateSevere
Treatment15020

[back to top]

Incidence and Severity of Acute Graft Versus Host Disease (GVHD)

The number of participants with acute GVHD is given, organized by grade. Participants are graded on a scale from 1 to 4, with 1 being mild and 4 being severe. (NCT01621477)
Timeframe: 100 days post transplant

Interventionparticipants (Number)
No Acute GVHDGrade IGrade IIGrade IIIGrade IV
Treatment93131

[back to top] [back to top]

One-year Survival (OS)

Evaluate the number of participants alive at 1 year. The number of participants surviving to one-year post-transplantation is given. (NCT01621477)
Timeframe: One year post transplant

Interventionparticipants (Number)
Treatment7

[back to top]

Incidence of Malignant Relapse

Estimate the incidence of malignant relapse at one year post-transplant. The number of participants with malignant relapse or progressive disease is given. Relapse was evaluated using standard WHO criteria for each disease. (NCT01621477)
Timeframe: One year post transplantation.

Interventionparticipants (Number)
Treatment10

[back to top]

Event-Free Survival (EFS)

Estimate the EFS at one-year post-transplantation. The event is defined as relapse or death due to any cause. The number of participants who were alive without relapse at one year post-transplant is reported. (NCT01621477)
Timeframe: one year post transplant

Interventionparticipants (Number)
Treatment4

[back to top]

Disease-Free Survival (DFS)

Estimate the DFS at one-year post-transplantation. The event is defined as relapse or death due to relapse. The number of participants who did not relapse up to one year post transplant is reported. (NCT01621477)
Timeframe: one year post transplant

Interventionparticipants (Number)
Treatment7

[back to top]

Donor (Allogeneic) Hematopoietic Engraftment

Number of patients who achieve hematopoietic engraftment - assessment of nucleated peripheral blood cells for donor (allogeneic) chimerism following this reduced-intensity HCT. (NCT01626092)
Timeframe: Day 100 Following Hematopoietic Cell Transplant (HCT)

Interventionparticipants (Number)
Intent-To-Treat Patients1

[back to top] [back to top] [back to top]

Time-to-engraftment

The number of days until participants by dose level reach engraftment. (NCT01629511)
Timeframe: 30 days post transplant

,,
Interventionparticipants (Number)
Day 10Day 11Day 12Day 13Day 15Day 17<10 days - Graft Failure
Gemcitabine Dose Level 11210000
Gemcitabine Dose Level 32401001
Gemcitabine Dose Level 40010110

[back to top]

Overall Survival

Will be estimated by the method of Kaplan and Meier. Time-to-event distributions as function of patient baseline covariates will be evaluated using Bayesian time-to-event regression modeling. (NCT01629511)
Timeframe: Up to 1 year post transplant

InterventionParticipants (Count of Participants)
Gemcitabine Dose Level 13
Gemcitabine Dose Level 20
Gemcitabine Dose Level 34
Gemcitabine Dose Level 42

[back to top]

Incidence of Hepatic Veno-occlusive Disease

The number of participants that experienced hepatic veno-occlusive disease (VOD). VOD is a condition in which some of the small veins in the liver are obstructed. (NCT01643668)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Treatment Arm0

[back to top] [back to top]

Assessment of Donor Stem Cell Engraftment: ANC Count

Patients are considered to have achieved donor cell engraftment if they have an absolute neutrophil count (ANC) of at least 500 cells/uL of blood for 3 consecutive measurements and at least 75% of hematopoietic elements are donor-derived as determined by chimerism assays from peripheral blood prior to day +40 after Busulfan/Clofarabine (BuClo) reduced intensity allogeneic stem cell transplantation (NCT01643668)
Timeframe: 1, 2, 3, and 4 weeks after transplantation

InterventionParticipants (Count of Participants)
Neutrophil Engraftment AchievedNeutrophil Engraftment Not Achieved
Treatment Arm330

[back to top]

Cumulative Incidence and Severity of Acute GVHD Within 100 Days Post Transplant

The percentage of participants who experienced grades 2-4 and grades 3-4 acute graft-versus-host disease (GVHD) by 100 days post transplantation. GVHD is a condition that can occur following an allogenic stem cell transplantation when the donated bone marrow or peripheral stem cells view the recipients body as foreign and the donated cell/marrow attack the body. Acute GVHD is generally observed within the first 100 days post transplant. Acute GVHD is associated with increased treatment related morbidity and mortality. Grade I GVHD is characterized as mild disease, grade II GVHD as moderate, grade III as severe, and grade IV life-threatening. The grade of the GVHD is determined by grading GHVD associated adverse events. Associated adverse events were graded using Common Terminology Criteria for Adverse Events (CTCAE) version 4 which uses the same mild, moderate, severe, life threatening grading system as the overall GHVD assessment. (NCT01643668)
Timeframe: 100 days

Interventionpercentage of Participants (Number)
Grades II-IV acute GVHDGrades III-IV acute GVHD
Treatment Arm2112

[back to top]

Cumulative Incidence of Non-relapse Mortality

The percentage of participants that experienced non-relapse mortality (NRM) at day 100 and 1 year after BuClo RIC SCT. Non-relapse mortality is any mortality that is not associated with or proceeded by disease progression of prior cancers. (NCT01643668)
Timeframe: 100 days, 1 year

Interventionpercentage of participants who died (Number)
100 Days1 Year
Treatment Arm5.924

[back to top]

Donor Stem Cell Engraftment: Platelet Count

Platelet recovery was defined as having a platelet count of at least 20,000 platelets/uL of blood on 2 consecutive measurements without transfusional support prior to day +100 after BuClo RIC HSCT. (NCT01643668)
Timeframe: 1, 2, 3, 4, 8, and 14 weeks post transplant

InterventionParticipants (Count of Participants)
Platelet Engraftmnet AchievedPlatelet Engraftmnet Not Achieved
Treatment Arm330

[back to top]

Progression-Free and Overall Survival

The 1-year and 2-year progression-free and overall survival measured from the time of stem cell transplantation. Progression is the recurrence or increase in the number of cancer cells found in the body. (NCT01643668)
Timeframe: 1 year, 2 years

Interventionpercentage of participants (Number)
Progression Free Survival at 1 yearProgression Free Survival at 2 yearsOverall Survival at 1 yearOverall Survival at 2 years
Treatment Arm50505656

[back to top]

Grade 3 or 4 Toxicities

The number of participants that experienced the specified grade 3 and 4 non-hematological toxicities during treatment and follow-up as assessed by Common Terminology Criteria for Adverse Events version 4(CTAE v 4.0). Grade 3 toxicity is considered to be severe and grade 4 is considered to be life threatening. (NCT01643668)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Bacterial InfectionViral InfectionFungal InfectionMucositisIncreased bilirubinIncreased alanine aminotransferaseIncreased aspartate aminotransferaseEngraftment SyndromeFebrile NeutropeniaSepsisAcute Renal FailureDiffuse alveolar hemorrhageIdiopathic pneumonia syndrome
Treatment Arm10121111021100

[back to top]

Cumulative Incidence of Chronic GVHD at One Year

The percentage of participants who experienced chronic Graft Versus Host Disease (GVHD) by one year. GVHD is a condition that can occur following an allogenic stem cell transplantation when the donated bone marrow or peripheral stem cells view the recipients body as foreign and the donated cell/marrow attack the body. Chronic GVHD normally occurs after the first 100 days post transplantation. Chronic GVHD can adversely influence long term survival. (NCT01643668)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Treatment Arm44

[back to top]

Measure Patient Response to High-dose Cytarabine Followed by Clofarabine in Adult Patients With Relapsed or Refractory AML

Response to the therapy is measured by a defined improvement in Neutrophil and platlet counts, along with improved cellularity of bone marrow biopsy (>20% with maturation of all cell lines), <5% blasts, auer rods must not be detectable and extramedullary leukemia or soft tissue involvment must not be present. (NCT01656031)
Timeframe: 5 weeks

Interventionparticipants (Number)
High-Dose Cytarabine and Clofarabine17

[back to top]

Median CD20 Expression Levels

To estimate median levels of CD20 expression at baseline, during treatment with dexamethasone-containing chemotherapy and following rituximab treatment in Block I of remission induction therapy for relapsed precursor B-cell ALL. (NCT01700946)
Timeframe: Baseline and at the end of Block I (approximately 5 weeks after the on-study date)

,,
Interventionpercentage of CD20 Antigen (Median)
At BaselineAt Block 1
All Patients Enrolled on the Study22.015.58
HIGH RISK23.1319.58
STANDARD RISK16.4011.83

[back to top]

3-year Event-free Survival Rates in Patients With Relapsed ALL

Estimate the 3-year event-free survival rate of participants with first relapse or primary refractory precursor B-cell ALL treated with risk-directed therapy. (NCT01700946)
Timeframe: 3 years of follow-up since the on-study date

Interventionpercentage of participants (Number)
STANDARD RISK83.3
HIGH RISK55.7
All Patients Enrolled on the Study67.83

[back to top]

Mean of CD20 Expression Levels

To estimate mean levels of CD20 expression at baseline, during treatment with dexamethasone-containing chemotherapy and following rituximab treatment in Block I of remission induction therapy for relapsed precursor B-cell ALL. (NCT01700946)
Timeframe: Baseline and at the end of Block I (approximately 5 weeks after the on-study date)

,,
Interventionpercentage of CD20 Antigen (Mean)
At BaselineAt Block I
All Patients Enrolled on the Study36.2319.43
HIGH RISK39.8220.10
STANDARD RISK31.1018.54

[back to top]

3-year Overall Survival Rate of Patients With Relapsed ALL

Estimate the 3-year survival rate of participants with first relapse or primary refractory precursor B-cell ALL treated with risk-directed therapy. (NCT01700946)
Timeframe: 3 years of follow-up since the on-study date

Interventionpercentage of participants (Number)
STANDARD RISK94.4
HIGH RISK55.5
All Patients Enrolled on the Study72.63

[back to top]

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT01794702)
Timeframe: Up to 2 years after participants off study date

InterventionMonths (Median)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin7.7

[back to top]

Number of Participants With a Response

Primary endpoint is overall response defined as the best response either complete response, complete remission without platelet recovery, or complete remission without incomplete blood count recovery within 56 days. (NCT01794702)
Timeframe: 56 days

InterventionParticipants (Count of Participants)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin20

[back to top]

Maximum Tolerated Dose (MTD) of Clofarabine

Maximum tolerated dose (MTD) defined as the highest dose schedule in which 6 patients were treated with at most 1 experiencing a dose-limiting toxicity (DLT). Clofarabine 15 mg/m2 IV over approximately 1 hour daily (number of days selected based on Phase I portion). (NCT01794702)
Timeframe: After second, 33 day cycle

Interventionmg/m^2 x 4 days (6-9) (Number)
Period 115

[back to top]

To Determine the Disease-free Survival (DFS).

Time from date of treatment start until the date of first objective documentation of return of disease. (NCT01794702)
Timeframe: Up to 2 years after participants off study date

Interventionmonths (Median)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin17.9

[back to top]

Overall Survival at 2 Years

Overall survival (OS) is defined as time from start of protocol treatment to death from any cause. It was estimated using the Kaplan-Meier method; the 95% confidence interval was calculated using Greenwood's formula. (NCT01885689)
Timeframe: From start of protocol treatment to death due to any cause, or last follow-up, whichever comes first, assessed up to 2 years.

Interventionpercent probability (Number)
Treatment (Clofarabine, Melphalan, Transplant)67

[back to top]

Progression-free Survival at 2 Years

Progression-free survival (PFS) is defined as time from start of protocol treatment to disease relapse/progression, death or last contact, whichever occurs first. Progression-free survival was estimated using the Kaplan-Meier method; the 95% confidence interval was calculated using Greenwood's formula. (NCT01885689)
Timeframe: From start of protocol treatment to death due to any cause, disease relapse/progression, or last follow-up, whichever comes first, assessed up to 2 years.

Interventionpercent probability (Number)
Treatment (Clofarabine, Melphalan, Transplant)54

[back to top]

Complete Clinical Response

Number of patients with newly diagnosed Acute Myeloid Leukemia who achieved Complete Response to therapy as determined by bone marrow biopsy evaluation. A CR designation required that the patient achieved a morphologic leukemia-free state and an absolute neutrophil count greater than or equal to 1.0 x 10^9/l, a platelet count greater than or equal to 100 x 10^9/l, and no evidence of extramedullary disease. (NCT01960387)
Timeframe: Between 14 and 28 days from start of study treatment

Interventionparticipants (Number)
Clofarabine (40mg/m^2/Day) + Cytarabine (1g/m^2/Day)2

[back to top]

Proportion of Patients With Complete Remission

"Patients are required to have all of the following to be considered as having a completion remission (CR).~Peripheral Blood Counts~Neutrophil count > 1.0 x 10^9 /L~Platelet count ≥ 100 x 10^9 /L~Reduced hemoglobin concentration or hematocrit has no bearing on remission status~Leukemic blasts must not be present in the peripheral blood~Bone Marrow Aspirate and Biopsy~Cellularity of bone marrow biopsy must be > 20% with maturation of all cell lines~< 5% blasts by morphologic review~Auer rods must not be detectable~Extramedullary leukemia, such as CNS or soft tissue involvement, must not be present" (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionproportion of participants (Number)
A (Step 1:Daunorubicin/Cytarabine; Step 2:Cytarabine; Step 3: Observation/Decitabine/Transplant)0.446
B (Step 1: Clofarabine; Step 2: Clofarabine; Step 3: Observation/Decitabine/Transplant)0.453

[back to top]

Overall Survival by Donor Status

Overall survival is defined as time between achieving leukemia-free state after induction therapy to death from any cause or date last known alive. The association between overall survival and donor status was evaluated regardless of assigned treatment arms. Patients with transplant donor information (either had donor or did not have a donor) reported after achieving CR/Cri/leukemia-free state post induction therapy were included in this analysis. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
With Transplant Donor27.2
Without Transplant Donor12.9

[back to top]

Overall Survival

Overall survival is defined as the time from randomization to death or date last known alive. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
A (Step 1:Daunorubicin/Cytarabine; Step 2:Cytarabine; Step 3: Observation/Decitabine/Transplant)12.9
B (Step 1: Clofarabine; Step 2: Clofarabine; Step 3: Observation/Decitabine/Transplant)11.6

[back to top]

Disease-free Survival for Maintenance

DFS for maintenance comparison is defined as the time from maintenance randomization until relapse or death of any cause. The censored follow-up time for patients without relapse and death information is the date of last contact. Only patients who remained in CR or CRi after completion of consolidation therapy that were randomized to either observation or decitabine in the maintenance Step were included in this analysis. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
Maintenance : Observation8.2
Maintenance : Decitabine16.3

[back to top]

Minimal Residual Disease

A bone marrow evaluation to determine study response and remission status will be performed on study Day 30 or upon adequate blood count recovery (ANC > 0.50 and platelet > 50,000), whichever occurs first. If the marrow is hypocellular and without evidence of normal tri-lineage hematopoiesis the marrow should be repeated at Day 42. (NCT02349178)
Timeframe: Day 30 or adequate blood recovery

InterventionParticipants (Count of Participants)
MRD PositiveMRD Negative
Bridging Arm33

[back to top]

Response Rate (OR) of LCH Cohort

The OR was defined as the proportion of participants achieving certain response on treatment among LCH patients, criteria were defined per protocol. Better response were defined as achieving complete disease resolution (NAD) or disease regression (AD better); intermediate response defined as stable or unchanged disease status; worse response defined as disease progression. (NCT02425904)
Timeframe: Disease was evaluated at baseline and the end of cycle 2. Treatment continued after cycle 2 until disease progression or unacceptable toxicity. LCH treatment duration has a median of 5.8 months with range 2.1-7 months.

Interventionproportion of participants (Number)
BetterIntermediateWorse
Recurrent or Refractory Langerhans Cell Histiocytosis (LCH) + Clofarabine0.850.150

[back to top] [back to top]

1-year Progression Free Survival (PFS)

1-year PFS defined as the proportion of patients that survival progression free at 1 year. PFS based on the duration of time from study entry to documented disease progression (PD) or death. Per protocol criteria: where time to event for PFS is the time from study enrollment until the time of first occurrence of new lesions, progressive disease, or death from any cause, or until last contact if no event occurs. (NCT02425904)
Timeframe: At 1 year

Interventionproportion of patients (Number)
Recurrent or Refractory Langerhans Cell Histiocytosis (LCH) + Clofarabine0.89
LCH-related Disorders + Clofarabine0.5

[back to top]

1-year Overall Survival (OS)

1-year OS defined as the proportion of patients that survival at 1 year. OS calculated as the time from enrollment until death or censored at date last known alive. (NCT02425904)
Timeframe: At 1 year

Interventionproportion of patients (Number)
Recurrent or Refractory Langerhans Cell Histiocytosis (LCH) + Clofarabine1
LCH-related Disorders + Clofarabine0.75

[back to top] [back to top]

Time to Engraftment

Number of days from transplant when participants achieved engraftment measure by ANC of 0.5 for three consecutive days. (NCT03096782)
Timeframe: Up to 12 months after transplant

InterventionParticipants (Count of Participants)
Twenty Two DaysTwenty Nine DaysThirty DaysNine DaysThirty Two Days
Chemotherapy Plus Cord Blood Transplant21111

[back to top]

Disease-free Survival

Number of participants that were in remission post transplant. (NCT03096782)
Timeframe: Up to12 months

InterventionParticipants (Count of Participants)
Complete Remission at 30 daysComplete Remission at 12 months
Chemotherapy Plus Cord Blood Transplant64

[back to top]

Overall Survival

Number of participants alive 1 year post transplant. (NCT03096782)
Timeframe: Up to 12 months after transplant

InterventionParticipants (Count of Participants)
Chemotherapy Plus Cord Blood Transplant4

[back to top]

Complete Remission (CR) Rate at Day 30 Post HSCT

The CR rate at 30 days (Day +30) post stem cell transplant infusion (NCT04002115)
Timeframe: 30 days

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^2100

[back to top]

Neutrophil Engraftment

Rates of engraftment, defined as the first day of Absolute Neutrophil Count (ANC) greater than 500 for the first of three consecutive days (NCT04002115)
Timeframe: 1 year

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^20

[back to top] [back to top]

Rate of Acute Graft-versus-host Disease (GVHD)

The rate of any grade (1-4) of acute GvHD as measured from day of transplantation to Day +100 using the Glucksberg criteria. (NCT04002115)
Timeframe: 100 days

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^20

[back to top]

Rate of Chronic GVHD

The rate of any grade (1-4) of Chronic GvHD as measured from Day +100 to Year 1 post-transplantation using the Glucksberg criteria. (NCT04002115)
Timeframe: 1 year

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^20

[back to top]

Severity of Acute Graft-versus-host Disease (GVHD)

The highest grade (1-4) of acute GvHD experienced by participants as measured from day of transplantation to Day +100 using the Glucksberg criteria (NCT04002115)
Timeframe: 100 days

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^20

[back to top]

Severity of Chronic GVHD

The highest overall grade (1-4) of chronic GvHD experienced by participants as measured from Day +100 to Year 1 post-transplantation using the Glucksberg criteria (NCT04002115)
Timeframe: 1 year

Interventionpercentage of Participants (Number)
Clofarabine 30 mg/m^20

[back to top]