Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-767nl Total loading time: 0 Render date: 2024-07-11T21:21:34.178Z Has data issue: false hasContentIssue false

27 - Development therapeutics

from Section 3 - Evaluation and treatment

Published online by Cambridge University Press:  05 April 2013

Ching-Hon Pui
Affiliation:
St Jude's Children's Research Hospital
Get access

Summary

Introduction

Most agents currently used in the treatment of leukemia were developed over 30 years ago. The traditional approach to drug discovery and development has been classic single-agent phase I trials that determine a maximally tolerated dose at which efficacy is evaluated in phase II studies prior to incorporation into combination regimens. Responses have been defined by the presence (percentage) or absence of leukemic blasts in the bone marrow based on morphology along with trilineage hematopoietic recovery. With recent advances in molecular and immunologic technology, minimal residual disease (MRD) measurement is increasingly applied in defining response, and novel therapeutic strategies are targeting the leukemia-associated antigens and molecular pathways. The development of new oncology drug classes with novel mechanisms of action brings new challenges. New strategies are needed to efficiently combine immunotherapy and molecular therapy with traditional chemotherapeutics and with each other to maximize leukemic cell kill and minimize toxicity. New definitions of response and relapse are needed to effectively evaluate novel treatments in an era where MRD is increasingly used to assess therapeutic efficacy, and transplant is increasingly performed in patients without full peripheral count recovery. A disciplined application of well-designed clinical trials is needed as new therapeutic options emerge in order to optimize and individualize therapy. Paradigms for the design and conduct of preclinical and early clinical trials are discussed, and novel agents in development are presented in this chapter.

Leukemia as a model for drug development

Leukemia cells are easily accessible for study, allowing assessment of a new agent's activity and providing studies in animal models from which drug dose and schedule dependency can be demonstrated. The concepts of combination chemotherapy, remission induction, and maintenance treatment were all developed in pediatric acute lymphoblastic leukemia (ALL) regimens.

Type
Chapter
Information
Childhood Leukemias , pp. 616 - 631
Publisher: Cambridge University Press
Print publication year: 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

DiMasi, JA, Hansen, RW, Grabowski, HG. The price of innovation: new estimates of drug development costs. J Health Econ 2003;22:151–185.CrossRefGoogle Scholar
Kola, I, Landis, J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 2004;3:711–715.CrossRefGoogle ScholarPubMed
Booth, B, Glassman, R, Ma, P. Oncology's trials. Nat Rev Drug Discov 2003;2:609–610.CrossRefGoogle ScholarPubMed
Ma, P, Zemmel, R. Value of novelty? Nat Rev Drug Discov 2002;1:571–572.CrossRefGoogle ScholarPubMed
Gundersen, L. The complex process of naming drugs. Ann Intern Med 1998;129:677–678.CrossRefGoogle ScholarPubMed
Lin, Y, Shih, WJ. Statistical properties of the traditional algorithm-based designs for phase I cancer clinical trials. Biostatistics 2001;2:203–215.CrossRefGoogle ScholarPubMed
O'Quigley, J, Shen, LZ. Continual reassessment method: a likelihood approach. Biometrics 1996;52:673–684.CrossRefGoogle ScholarPubMed
Thall, PF, Lee, JJ, Tseng, CH, Estey, EH. Accrual strategies for phase I trials with delayed patient outcome. Stat Med 1999;18:1155–1169.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Horstmann, E, McCabe, MS, Grochow, L, et al. Risks and benefits of phase 1 oncology trials, 1991 through 2002. N Engl J Med 2005;352:895–904.CrossRefGoogle ScholarPubMed
Roberts, TG, Jr., Lynch, TJ, Jr., Chabner, BA.The phase III trial in the era of targeted therapy: unraveling the “go or no go” decision. J Clin Oncol 2003;21:3683–3695.CrossRefGoogle Scholar
Korn, EL, Arbuck, SG, Pluda, JM, et al. Clinical trial designs for cytostatic agents: are new approaches needed? J Clin Oncol 2001;19:265–272.CrossRefGoogle ScholarPubMed
Thezenas, S, Duffour, J, Culine, S, Kramar, A. Five-year change in statistical designs of phase II trials published in leading cancer journals 1. Eur J Cancer 2004;40:1244–1249.CrossRefGoogle Scholar
Ratain, MJ.Phase II oncology trials: let's be positive. Clin Cancer Res 2005;11:5661–5662.CrossRefGoogle ScholarPubMed
Rubinstein, LV, Korn, EL, Freidlin, B, et al. Design issues of randomized phase II trials and a proposal for phase II screening trials. J Clin Oncol 2005;23:7199–7206.CrossRefGoogle Scholar
Ratain, MJ, Sargent, DJ. Optimising the design of phase II oncology trials: the importance of randomisation. Eur J Cancer 2009;45:275–280.CrossRefGoogle ScholarPubMed
Estey, EH, Thall PF. New designs for phase 2 clinical trials. Blood 2003;102:442–448.CrossRefGoogle ScholarPubMed
Wieand, HS.Randomized phase II trials: what does randomization gain?J Clin Oncol 2005;23:1794–1795.CrossRefGoogle ScholarPubMed
Liu, PY, LeBlanc, M, Desai, M. False positive rates of randomized phase II designs. Control Clin Trials 1999;20:343–352.CrossRefGoogle ScholarPubMed
Cannistra, SA.The ethics of early stopping rules: who is protecting whom?J Clin Oncol 2004;22:1542–1545.CrossRefGoogle ScholarPubMed
Ratain, MJ, Eisen, T, Stadler, WM, et al. Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol 2006;24:2505–2512.CrossRefGoogle ScholarPubMed
DiMasi, JA, Grabowski, HG. Economics of new oncology drug development. J Clin Oncol 2007;25:209–216.CrossRefGoogle ScholarPubMed
Hoekstra, R, Verweij, J, Eskens, FA. Clinical trial design for target specific anticancer agents. Invest New Drugs 2003;21:243–250.CrossRefGoogle ScholarPubMed
Seymour, L, Ivy, SP, Sargent, D, et al. The design of phase II clinical trials testing cancer therapeutics: consensus recommendations from the clinical trial design task force of the national cancer institute investigational drug steering committee. Clin Cancer Res 2010;16:1764–1769.CrossRefGoogle ScholarPubMed
Dancey, JE, Dobbin, KK, Groshen, S, et al. Guidelines for the development and incorporation of biomarker studies in early clinical trials of novel agents. Clin Cancer Res 2010;16:1745–1755.CrossRefGoogle ScholarPubMed
Gelmon, KA, Eisenhauer, EA, Harris, AL, Ratain, MJ, Workman, P. Anticancer agents targeting signaling molecules and cancer cell environment: challenges for drug development? J Natl Cancer Inst 1999;91:1281–1287.CrossRefGoogle ScholarPubMed
LoRusso, PM, Boerner, SA, Seymour, L. An overview of the optimal planning, design, and conduct of phase I studies of new therapeutics. Clin Cancer Res 2010;16:1710–1718.CrossRefGoogle ScholarPubMed
Senderowicz, AM. Information needed to conduct first-in-human oncology trials in the United States: a view from a former FDA medical reviewer. Clin Cancer Res 2010;16:1719–1725.CrossRefGoogle ScholarPubMed
Berry, DA.Adaptive clinical trials: the promise and the caution. J Clin Oncol 2011;29:606–609.CrossRefGoogle ScholarPubMed
Mahoney, MR, Sargent, DJ, O'Connell, MJ, et al. Dealing with a deluge of data: an assessment of adverse event data on North Central Cancer Treatment Group trials. J Clin Oncol 2005;23:9275–9281.CrossRefGoogle ScholarPubMed
Califf, RM.Clinical trials bureaucracy: unintended consequences of well-intentioned policy. Clin Trials 2006;3:496–502.CrossRefGoogle ScholarPubMed
Roche, K, Paul, N, Smuck, B, et al. Factors affecting workload of cancer clinical trials: results of a multicenter study of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2002;20:545–556.CrossRefGoogle ScholarPubMed
Horton, TM, Sposto, R, Brown, P, et al. Toxicity assessment of molecularly targeted drugs incorporated into multiagent chemotherapy regimens for pediatric acute lymphocytic leukemia (ALL): review from an international consensus conference. Pediatr Blood Cancer 2010;54:872–878.Google ScholarPubMed
Smith, MA, Seibel, NL, Altekruse, SF, et al. Outcomes for children and adolescents with cancer: challenges for the twenty-first century. J Clin Oncol 2010;28:2625–2634.CrossRefGoogle ScholarPubMed
Domenech, C, Thomas, X, Chabaud, S, et al. l-Asparaginase loaded red blood cells in refractory or relapsing acute lymphoblastic leukaemia in children and adults: results of the GRASPALL 2005–01 randomized trial. Br J Haematol 2011;153:58–65.CrossRefGoogle ScholarPubMed
Pieters, R, Appel, I, Kuehnel, HJ, et al. Pharmacokinetics, pharmacodynamics, efficacy, and safety of a new recombinant asparaginase preparation in children with previously untreated acute lymphoblastic leukemia: a randomized phase 2 clinical trial. Blood 2008;112:4832–4838.CrossRefGoogle ScholarPubMed
Millar, JL, Millar, BC, Powles, RL, et al. Liposomal vincristine for the treatment of human acute lymphoblastic leukaemia in severe combined immunodeficient (SCID) mice. Br J Haematol 1998;102:718–721.CrossRefGoogle ScholarPubMed
Thomas, DA, Kantarjian, HM, Stock, W, et al. Phase 1 multicenter study of vincristine sulfate liposomes injection and dexamethasone in adults with relapsed or refractory acute lymphoblastic leukemia. Cancer 2009;115:5490–5498.CrossRefGoogle ScholarPubMed
Jabbour, E, O'Brien, S, Kantarjian, H, et al. Neurologic complications associated with intrathecal liposomal cytarabine given prophylactically in combination with high-dose methotrexate and cytarabine to patients with acute lymphocytic leukemia. Blood 2007;109:3214–3218.CrossRefGoogle Scholar
Wagner, E.Programmed drug delivery: nanosystems for tumor targeting. Expert Opin Biol Ther 2007;7:587–593.CrossRefGoogle ScholarPubMed
Dicko, A, Mayer, LD, Tardi, PG. Use of nanoscale delivery systems to maintain synergistic drug ratios in vivo. Expert Opin Drug Deliv 2010;7:1329–1341.CrossRefGoogle ScholarPubMed
Parker, WB, Shaddix, SC, Chang, CH, et al. 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. Cancer Res 1991;51:2386–2394.Google ScholarPubMed
Vahdat, L, Wong, ET, Wile, MJ, et al. Therapeutic and neurotoxic effects of 2-chlorodeoxyadenosine in adults with acute myeloid leukemia. Blood 1994;84:3429–3434.Google ScholarPubMed
Warrell, RP, Jr., Berman E. Phase I and II study of fludarabine phosphate in leukemia: therapeutic efficacy with delayed central nervous system toxicity. J Clin Oncol 1986;4:74–79.CrossRefGoogle ScholarPubMed
Jeha, S, Gaynon, PS, Razzouk, BI, et al. Phase II study of clofarabine in pediatric patients with refractory or relapsed acute lymphoblastic leukemia. J Clin Oncol 2006;24:1917–1923.CrossRefGoogle ScholarPubMed
Ghanem, H, Jabbour, E, Faderl, S, et al. Clofarabine in leukemia. Expert Rev Hematol 2010;3:15–22.CrossRefGoogle ScholarPubMed
Giblett, ER, Ammann, AJ, Wara, DW, Sandman, R, Diamond, LK. Nucleoside-phosphorylase deficiency in a child with severely defective T-cell immunity and normal B-cell immunity. Lancet 1975;1:1010–1013.CrossRefGoogle Scholar
Cohen, A, Lee, JW, Gelfand, EW. Selective toxicity of deoxyguanosine and arabinosyl guanine for T-leukemic cells. Blood 1983;61:660–666.Google ScholarPubMed
Berg, SL, Blaney, SM, Devidas, M, et al. Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: a report from the Children's Oncology Group. J Clin Oncol 2005;23:3376–3382.CrossRefGoogle Scholar
Al-Kali, A, Gandhi, V, Ayoubi, M, Keating, M, Ravandi, F. Forodesine: review of preclinical and clinical data. Future Oncol 2010;6:1211–1217.CrossRefGoogle ScholarPubMed
Kamath, VP, Xue, J, Juarez-Brambila, JJ. Synthesis of analogs of forodesine HCl, a human purine nucleoside phosphorylase inhibitor-Part II. Bioorg Med Chem Lett 2009;19:2627–2629.CrossRefGoogle ScholarPubMed
Kantarjian, H, Garcia-Manero, G, O'Brien, S, et al. Phase I clinical and pharmacokinetic study of oral sapacitabine in patients with acute leukemia and myelodysplastic syndrome. J Clin Oncol 2010;28:285–291.CrossRefGoogle ScholarPubMed
Quintas-Cardama, A, Cortes, J. Evaluation of the l-stereoisomeric nucleoside analog troxacitabine for the treatment of acute myeloid leukemia. Expert Opin Invest Drugs 2007;16:547–557.CrossRefGoogle ScholarPubMed
Lee, CP, de Jonge, MJ, O'Donnell, AE, et al. A phase I study of a new nucleoside analogue, OSI-7836, using two administration schedules in patients with advanced solid malignancies. Clin Cancer Res 2006;12:2841–2848.CrossRefGoogle ScholarPubMed
Strumberg, D, Harstrick, A, Doll, K, Hoffmann, B, Seeber, S. Bendamustine hydrochloride activity against doxorubicin-resistant human breast carcinoma cell lines. Anticancer Drugs 1996;7:415–421.CrossRefGoogle ScholarPubMed
Leoni, LM, Bailey, B, Reifert, J, et al. Bendamustine (Treanda) displays a distinct pattern of cytotoxicity and unique mechanistic features compared with other alkylating agents. Clin Cancer Res 2008;14:309–317.CrossRefGoogle ScholarPubMed
Chow, KU, Boehrer, S, Geduldig, K, et al. In vitro induction of apoptosis of neoplastic cells in low-grade non-Hodgkin's lymphomas using combinations of established cytotoxic drugs with bendamustine. Haematologica 2001;86:485–493.Google ScholarPubMed
Rummel, MJ, Gregory, SA. Bendamustine's emerging role in the management of lymphoid malignancies. Semin Hematol 2011;48(Suppl 1): S24–S36.CrossRefGoogle ScholarPubMed
Seiter, K, Liu, D, Loughran, T, et al. Phase I study of temozolomide in relapsed/refractory acute leukemia. J Clin Oncol 2002;20:3249–3253.CrossRefGoogle ScholarPubMed
Horton, TM, Thompson, PA, Berg, SL, et al. Phase I pharmacokinetic and pharmacodynamic study of temozolomide in pediatric patients with refractory or recurrent leukemia: a Children's Oncology Group Study. J Clin Oncol 2007;25:4922–4928.CrossRefGoogle ScholarPubMed
Gerson, SL. MGMT: its role in cancer aetiology and cancer therapeutics. Nat Rev Cancer 2004;4:296–307.CrossRefGoogle ScholarPubMed
Thomas, DA, O'Brien, S, Faderl, S, et al. Chemoimmunotherapy with a modified hyper-CVAD and rituximab regimen improves outcome in de novo Philadelphia chromosome-negative precursor B-lineage acute lymphoblastic leukemia. J Clin Oncol 2010;28:3880–3889.CrossRefGoogle ScholarPubMed
van Meerten, T, Hagenbeek, A. CD20-targeted therapy: the next generation of antibodies. Semin Hematol 2010;47:199–210.CrossRefGoogle ScholarPubMed
Cheson, BD. Ofatumumab, a novel anti-CD20 monoclonal antibody for the treatment of B-cell malignancies. J Clin Oncol 2010;28:3525–3530.CrossRefGoogle ScholarPubMed
Leonard, JP, Goldenberg, DM. Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies. Oncogene 2007;26:3704–3713.CrossRefGoogle Scholar
Wayne, AS, Kreitman, RJ, Findley, HW, et al. Anti-CD22 immunotoxin RFB4(dsFv)-PE38 (BL22) for CD22-positive hematologic malignancies of childhood: preclinical studies and phase I clinical trial. Clin Cancer Res 2010;16:1894–1903.CrossRefGoogle ScholarPubMed
Wong, BY, Dang, NH. Inotuzumab ozogamicin as novel therapy in lymphomas. Expert Opin Biol Ther 2010;10:1251–1258.CrossRefGoogle ScholarPubMed
Topp, MS, Kufer, P, Gokbuget, N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 2011;29:2493–2498.CrossRefGoogle ScholarPubMed
Ravandi, F.Gemtuzumab ozogamicin: one size does not fit all: the case for personalized therapy. J Clin Oncol 2011;29:349–351.CrossRefGoogle Scholar
Schultz, KR, Bowman, WP, Aledo, A, et al. Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a Children's Oncology Group study. J Clin Oncol 2009;27:5175–5181.CrossRefGoogle ScholarPubMed
Lee, HJ, Thompson, JE, Wang, ES, Wetzler, M. Philadelphia chromosome-positive acute lymphoblastic leukemia: current treatment and future perspectives. Cancer 2011;117:1583–1594.CrossRefGoogle ScholarPubMed
Mullighan, CG, Miller, CB, Radtke, I, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 2008;453:110–114.CrossRefGoogle ScholarPubMed
Zhao, C, Chen, A, Jamieson, CH, et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 2009;458:776–779.CrossRefGoogle ScholarPubMed
Goemans, BF, Zwaan, CM, Harlow, A, et al. In vitro profiling of the sensitivity of pediatric leukemia cells to tipifarnib: identification of T-cell ALL and FAB M5 AML as the most sensitive subsets. Blood 2005;106:3532–3537.CrossRefGoogle ScholarPubMed
Cortes, J, Quintas-Cardama, A, Garcia-Manero, G, et al. Phase 1 study of tipifarnib in combination with imatinib for patients with chronic myelogenous leukemia in chronic phase after imatinib failure. Cancer 2007;110:2000–2006.CrossRefGoogle ScholarPubMed
Niemeyer, CM, Kratz, CP. Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia: molecular classification and treatment options. Br J Haematol 2008;140:610–624.CrossRefGoogle ScholarPubMed
Bercovich, D, Ganmore, I, Scott, LM, et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down's syndrome. Lancet 2008;372:1484–1492.CrossRefGoogle ScholarPubMed
Mullighan, CG, Zhang, J, Harvey, RC, et al. JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci USA 2009;106:9414–9418.CrossRefGoogle ScholarPubMed
Harvey, RC, Mullighan, CG, Chen, IM, et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood 2010;115:5312–5321.CrossRefGoogle Scholar
Verstovsek, S, Kantarjian, H, Mesa, RA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med 2010;363:1117–1127.CrossRefGoogle ScholarPubMed
Auclair, D, Miller, D, Yatsula, V, et al. Antitumor activity of sorafenib in FLT3-driven leukemic cells. Leukemia 2007;21:439–445.CrossRefGoogle ScholarPubMed
Borthakur, G, Kantarjian, H, Ravandi, F, et al. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica 2011;96:62–68.CrossRefGoogle ScholarPubMed
Fenaux, P, Mufti, GJ, Hellstrom-Lindberg, E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol 2010;28:562–569.CrossRefGoogle ScholarPubMed
Issa, JP. DNA methylation as a therapeutic target in cancer. Clin Cancer Res 2007;13:1634–1637.CrossRefGoogle Scholar
Ley, TJ, Ding, L, Walter, MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med 2010;363:2424–2433.CrossRefGoogle ScholarPubMed
Whittaker, SJ, Demierre, MF, Kim, EJ, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 2010;28:4485–4491.CrossRefGoogle ScholarPubMed
Garcia-Manero, G, Yang, H, Bueso-Ramos, C, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood 2008;111:1060–1066.CrossRefGoogle ScholarPubMed
Klimek, VM, Fircanis, S, Maslak, P, et al. Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes. Clin Cancer Res 2008;14:826–832.CrossRefGoogle ScholarPubMed
Ellis, L, Pan, Y, Smyth, GK, et al. Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res 2008;14:4500–4510.CrossRefGoogle ScholarPubMed
Dickinson, M, Ritchie, D, DeAngelo, DJ, et al. Preliminary evidence of disease response to the pan deacetylase inhibitor panobinostat (LBH589) in refractory Hodgkin lymphoma. Br J Haematol 2009;147:97–101.CrossRefGoogle Scholar
Yu, C, Rahmani, M, Almenara, J, et al. Histone deacetylase inhibitors promote STI571-mediated apoptosis in STI571-sensitive and -resistant Bcr/Abl+ human myeloid leukemia cells. Cancer Res 2003;63:2118–2126.Google ScholarPubMed
Fiskus, W, Pranpat, M, Balasis, M, et al. Cotreatment with vorinostat (suberoylanilide hydroxamic acid) enhances activity of dasatinib (BMS-354825) against imatinib mesylate-sensitive or imatinib mesylate-resistant chronic myelogenous leukemia cells. Clin Cancer Res 2006;12:5869–5878.CrossRefGoogle ScholarPubMed
Zhang, B, Strauss, AC, Chu, S, et al. Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate. Cancer Cell 2010;17:427–442.CrossRefGoogle ScholarPubMed
Kuendgen, A, Strupp, C, Aivado, M, et al. Treatment of myelodysplastic syndromes with valproic acid alone or in combination with all-trans retinoic acid. Blood 2004;104:1266–1269.CrossRefGoogle ScholarPubMed
Saunders, P, Cisterne, A, Weiss, J, Bradstock, KF, Bendall, LJ. The mammalian target of rapamycin inhibitor RAD001 (everolimus) synergizes with chemotherapeutic agents, ionizing radiation and proteasome inhibitors in pre-B acute lymphocytic leukemia. Haematologica 2011;96:69–77.CrossRefGoogle ScholarPubMed
Moellering, RE, Cornejo, M, Davis, TN, et al. Direct inhibition of the NOTCH transcription factor complex. Nature 2009;462:182–188.CrossRefGoogle ScholarPubMed
Wu, Y, Cain-Hom, C, Choy, L, et al. Therapeutic antibody targeting of individual Notch receptors. Nature 2010;464:1052–1057.CrossRefGoogle ScholarPubMed
Campana, D, Coustan-Smith, E, Manabe, A, et al. Prolonged survival of B-lineage acute lymphoblastic leukemia cells is accompanied by overexpression of bcl-2 protein. Blood 1993;81:1025–1031.Google ScholarPubMed
Campos, L, Rouault, JP, Sabido, O, et al. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood 1993;81:3091–3096.Google ScholarPubMed
Tauchi, T, Sumi, M, Nakajima, A, et al. BCL-2 antisense oligonucleotide genasense is active against imatinib-resistant BCR-ABL-positive cells. Clin Cancer Res 2003;9:4267–4273.Google ScholarPubMed
Marcucci, G, Byrd, JC, Dai, G, et al. Phase 1 and pharmacodynamic studies of G3139, a Bcl-2 antisense oligonucleotide, in combination with chemotherapy in refractory or relapsed acute leukemia. Blood 2003;101:425–432.CrossRefGoogle ScholarPubMed
Zhang, L, Ming, L, Yu, J. BH3 mimetics to improve cancer therapy; mechanisms and examples. Drug Resist Updat 2007;10:207–217.CrossRefGoogle ScholarPubMed
Parry, D, Guzi, T, Shanahan, F, et al. Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol Cancer Ther 2010;9:2344–2353.CrossRefGoogle Scholar
Goasguen, JE, Dossot, JM, Fardel, O, et al. Expression of the multidrug resistance-associated P-glycoprotein (P-170) in 59 cases of de novo acute lymphoblastic leukemia: prognostic implications. Blood 1993;81:2394–2398.Google ScholarPubMed
Kolitz, JE, George, SL, Marcucci, G, et al. P-glycoprotein inhibition using valspodar (PSC-833) does not improve outcomes for patients younger than age 60 years with newly diagnosed acute myeloid leukemia: Cancer and Leukemia Group B study 19808. Blood 2010;116:1413–1421.CrossRefGoogle ScholarPubMed
Zeng, Z, Shi, YX, Samudio, IJ, et al. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood 2009;113:6215–6224.CrossRefGoogle ScholarPubMed
Nervi, B, Ramirez, P, Rettig, MP, et al. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood 2009;113:6206–6214.CrossRefGoogle ScholarPubMed
Matsunaga, T, Takemoto, N, Sato, T, et al. Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nat Med 2003;9:1158–1165.CrossRefGoogle ScholarPubMed
Jin, L, Hope, KJ, Zhai, Q, Smadja-Joffe, F, Dick, JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 2006;12:1167–1174.CrossRefGoogle ScholarPubMed
Schimmer, AD, Pedersen, IM, Kitada, S, et al. Functional blocks in caspase activation pathways are common in leukemia and predict patient response to induction chemotherapy. Cancer Res 2003;63:1242–1248.Google ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×