We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Targeting hypoxia in the leukemia microenvironment

    Juliana Benito

    Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA

    ,
    Zhihong Zeng

    Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA

    ,
    Marina Konopleva

    * Author for correspondence

    Division of Cancer Medicine – Unit 448, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA.

    &
    William R Wilson

    Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland, New Zealand

    Published Online:https://doi.org/10.2217/ijh.13.32

    SUMMARY The bone marrow (BM) microenvironment regulates survival and maintenance of normal hematopoietic stem cells. Within the endosteal niche, hypoxia has an essential role in maintenance of the primitive quiescent hematopoietic stem cell. We and others have demonstrated that in the context of hematologic malignancies the BM is highly hypoxic, and that progression of the disease is associated with expansion of hypoxic niches and stabilization of the oncogenic HIF-1α. This review will provide an overview of the normal and leukemic BM microenvironment with a special emphasis on pathological hypoxia including the development of hypoxia-activated prodrugs and their applicability in hematological malignancies.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Curry JL, Trentin JJ, Wolf N. Hemopoietic spleen colony studies. II. Erythropoiesis. J. Exp. Med.125(4),703–720 (1967).
    • Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells4(1–2),7–25 (1978).
    • Spradling A, Drummond-Barbosa D, Kai T. Stem cells find their niche. Nature414(6859),98–104 (2001).
    • Schofield R. The stem cell system. Biomed. Pharmacother.37(8),375–380 (1983).
    • Lo Celso C, Fleming HE, Wu JW et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature457(7225),92–96 (2009).
    • Calvi LM, Adams GB, Weibrecht KW et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature425(6960),841–846 (2003).
    • Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature481(7382),457–462 (2012).
    • Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell121(7),1109–1121 (2005).
    • Zhang J, Niu C, Ye L et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature425(6960),836–841 (2003).
    • 10  Fujisaki J, Wu J, Carlson AL et al.In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature474(7350),216–219 (2011).
    • 11  Greenbaum A, Hsu YM, Day RB et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature495(7440),227–230 (2013).
    • 12  Notta F, Doulatov S, Laurenti E, Poeppl A, Jurisica I, Dick JE. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science333(6039),218–221 (2011).
    • 13  Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell132(4),598–611 (2008).
    • 14  Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature495(7440),231–235 (2013).▪ Importance of endothelial and osteoblastic niches is shown. Hematopoietic stem cells depend on CXCL12 produced by perivascular and endothelial cells, while lymphoid progenitors rely on that provided by osteoblasts.
    • 15  Ivanovic Z. Hypoxia or in situ normoxia: the stem cell paradigm. J. Cell Physiol.219(2),271–275 (2009).
    • 16  Shapiro BA. Temperature correction of blood gas values. Respir. Care Clin. N. Am.1(1),69–76 (1995).
    • 17  Malatesha G, Singh NK, Bharija A, Rehani B, Goel A. Comparison of arterial and venous pH, bicarbonate, PCO2 and PO2 in initial emergency department assessment. Emerg. Med. J.24(8),569–571 (2007).
    • 18  Chu YC, Chen CZ, Lee CH, Chen CW, Chang HY, Hsiue TR. Prediction of arterial blood gas values from venous blood gas values in patients with acute respiratory failure receiving mechanical ventilation. J. Formos. Med. Assoc.102(8),539–543 (2003).
    • 19  Walkey AJ, Farber HW, O’Donnell C, Cabral H, Eagan JS, Philippides GJ. The accuracy of the central venous blood gas for acid-base monitoring. J. Intensive Care Med.25(2),104–110 (2010).
    • 20  Wolfle D, Jungermann K. Long-term effects of physiological oxygen concentrations on glycolysis and gluconeogenesis in hepatocyte cultures. Eur. J. Biochem.151(2),299–303 (1985).
    • 21  Jungermann K, Kietzmann T. Role of oxygen in the zonation of carbohydrate metabolism and gene expression in liver. Kidney Int.51(2),402–412 (1997).
    • 22  Roy S, Khanna S, Wallace WA et al. Characterization of perceived hyperoxia in isolated primary cardiac fibroblasts and in the reoxygenated heart. J. Biol. Chem.278(47),47129–47135 (2003).
    • 23  Welch WJ, Baumgartl H, Lubbers D, Wilcox CS. Nephron pO2 and renal oxygen usage in the hypertensive rat kidney. Kidney Int.59(1),230–237 (2001).
    • 24  Mik EG, van Leeuwen TG, Raat NJ, Ince C. Quantitative determination of localized tissue oxygen concentration in vivo by two-photon excitation phosphorescence lifetime measurements. J. Appl. Physiol.97(5),1962–1969 (2004).
    • 25  Harrison JS, Rameshwar P, Chang V, Bandari P. Oxygen saturation in the bone marrow of healthy volunteers. Blood99(1),394–394 (2002).
    • 26  Skouby AR. Haematologic adaptation in patients with chronic bronchitis and pulmonary insufficiency. Acta Med. Scand.199(3),185–190 (1976).
    • 27  Parmar K, Mauch P, Vergilio JA, Sackstein R, Down JD. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc. Natl Acad. Sci. USA104(13),5431–5436 (2007).▪▪ Utilizing Hoechst 33342 (Ho) perfusion dye, Parmar et al. suggested the existence of an oxygen gradient from the vasculature to the endosteal niche, with the majority of hematopoietic stem cells having the lowest Ho intensity, that is, being hypoxic in nature.
    • 28  Semenza GL. Oxygen-dependent regulation of mitochondrial respiration by hypoxia-inducible factor 1. Biochem. J.405(1),1–9 (2007).
    • 29  Winkler IG, Barbier V, Wadley R, Zannettino AC, Williams S, Levesque JP. Positioning of bone marrow hematopoietic and stromal cells relative to blood flow in vivo: serially reconstituting hematopoietic stem cells reside in distinct nonperfused niches. Blood116(3),375–385 (2010).
    • 30  Miharada K, Karlsson G, Rehn M et al. Cripto regulates hematopoietic stem cells as a hypoxic-niche-related factor through cell surface receptor GRP78. Cell Stem Cell9(4),330–344 (2011).
    • 31  Takubo K, Goda N, Yamada W et al. Regulation of the HIF-1α level is essential for hematopoietic stem cells. Cell Stem Cell7(3),391–402 (2010).
    • 32  Simsek T, Kocabas F, Zheng J et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell7(3),380–390 (2010).
    • 33  Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene29(5),625–634 (2010).
    • 34  Brown JM, William WR. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer4(6),437–447 (2004).
    • 35  Zhong H, De Marzo AM, Laughner E et al. Overexpression of hypoxia-inducible factor 1α in common human cancers and their metastases. Cancer Res.59(22),5830–5835 (1999).
    • 36  Chen Y, Hu L. Design of anticancer prodrugs for reductive activation. Med. Res. Rev.29(1),29–64 (2009).
    • 37  Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin. Cancer Biol.19(1),12–16 (2009).
    • 38  Chen JQ, Russo J. Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim. Biophys. Acta1826(2),370–384 (2012).
    • 39  Lukashev D, Ohta A, Sitkovsky M. Hypoxia-dependent anti-inflammatory pathways in protection of cancerous tissues. Cancer Metastasis Rev.26(2),273–279 (2007).
    • 40  Das B, Tsuchida R, Malkin D, Koren G, Baruchel S, Yeger H. Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells26(7),1818–1830 (2008).
    • 41  Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin. Cancer Res.16(24),5928–5935 (2010).
    • 42  Zhu Y, Zhao T, Itasaka S et al. Involvement of decreased hypoxia-inducible factor 1 activity and resultant G(1)-S cell cycle transition in radioresistance of perinecrotic tumor cells. Oncogene32(16),2058–2068 (2012).
    • 43  Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist. Updat.14(3),191–201 (2011).
    • 44  Schults MA, Timmermans L, Godschalk RW et al. Diminished carcinogen detoxification is a novel mechanism for hypoxia-inducible factor 1-mediated genetic instability. J. Biol. Chem.285(19),14558–14564 (2010).
    • 45  Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br. J. Cancer9(4),539–549 (1955).
    • 46  Jensen PO, Mortensen BT, Hodgkiss RJ et al. Increased cellular hypoxia and reduced proliferation of both normal and leukaemic cells during progression of acute myeloid leukaemia in rats. Cell Prolif.33(6),381–395 (2000).▪▪ First report associating hypoxia and leukemia using a rat leukemia model, via in vivo pulse-labeling with a theophylline-linked 2-nitroimidazole hypoxia probe.
    • 47  Fiegl M, Samudio I, Clise-Dwyer K, Burks JK, Mnjoyan Z, Andreeff M. CXCR4 expression and biologic activity in acute myeloid leukemia are dependent on oxygen partial pressure. Blood113(7),1504–1512 (2009).
    • 48  Benito J, Shi Y, Szymanska B et al. Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS ONE6(8),e23108 (2011).▪▪ Expansion of hypoxia was found in the bone marrow microenvironment of several xenograft leukemia models. Antileukemia effect of hypoxia activated prodrugs is shown.
    • 49  Wellmann S, Guschmann M, Griethe W et al. Activation of the HIF pathway in childhood ALL, prognostic implications of VEGF. Leukemia18(5),926–933 (2004).
    • 50  Frolova O, Samudio I, Benito J et al. Regulation of HIF-1α signaling and chemoresistance in acute lymphocytic leukemia under hypoxic conditions of the bone marrow microenvironment. Cancer Biol. Ther.13(10),1–13 (2012).
    • 51  Matsunaga T, Imataki O, Torii E et al. Elevated HIF-1α expression of acute myelogenous leukemia stem cells in the endosteal hypoxic zone may be a cause of minimal residual disease in bone marrow after chemotherapy. Leuk. Res.36(6),e122–e124 (2012).
    • 52  Martin SK, Diamond P, Gronthos S, Peet DJ, Zannettino AC. The emerging role of hypoxia, HIF-1 and HIF-2 in multiple myeloma. Leukemia25(10),1533–1542 (2011).
    • 53  Asosingh K, De Raeve H, De Ridder M et al. Role of the hypoxic bone marrow microenvironment in 5T2MM murine myeloma tumor progression. Haematologica90(6),810–817 (2005).
    • 54  Hu J, Handisides DR, Van Valckenborgh E et al. Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug. Blood116(9),1524–1527 (2010).
    • 55  Azab AK, Hu J, Quang P et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood119(24),5782–5794 (2012).
    • 56  Storti P, Bolzoni M, Donofrio G et al. Hypoxia-inducible factor (HIF)-1α suppression in myeloma cells blocks tumoral growth in vivo inhibiting angiogenesis and bone destruction. Leukemia doi:10.1038/leu.2013.24 (2013) (Epub ahead of print).▪ Importance of hypoxia in multiple myeloma. Silencing HIF-1α via shRNA results in tumor suppression due to downregulation of proangiogenic as well as pro-osteoclastogenic cytokines.
    • 57  Xia Y, Choi HK, Lee K. Recent advances in hypoxia-inducible factor (HIF)-1 inhibitors. Eur. J. Med. Chem.49,24–40 (2012).
    • 58  Swietach P, Vaughan-Jones RD, Harris AL. Regulation of tumour pH and the role of carbonic anhydrase 9. Cancer Metastasis Rev.26,299–310 (2007).
    • 59  McDonald PC, Winum JY, Supuran CT, Dedhar S. Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget3(1),84–97 (2012).
    • 60  Rouschop KM, Dubois LJ, Keulers TG et al. PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS. Proc. Natl Acad. Sci. USA110(12),4622–4627 (2013).
    • 61  Atkins C, Liu Q, Minthorn E et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res.73(6),1993–2002 (2013).
    • 62  Chen Y, Hu L. Design of anticancer prodrugs for reductive activation. Med. Res. Rev.29(1),29–64 (2009).
    • 63  Denny WA. Hypoxia-activated prodrugs in cancer therapy: progress to the clinic. Future Oncol.6(3),419–428 (2010).
    • 64  Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer11(6),393–410 (2011).▪▪ Provides an extensive review of the alternative aproaches to target hypoxia including bioreductive agents and inhibitors of molecular targets. Discusses mechanisms of actions, challenges and up-to-date results of the clinical trials carried out to exploit hypoxia as a therapeutic target.
    • 65  Belcourt MF, Hodnick WF, Rockwell S, Sartorelli AC. Differential toxicity of mitomycin C and porfiromycin to aerobic and hypoxic Chinese hamster ovary cells overexpressing human NADPH:cytochrome c (P-450) reductase. Proc. Natl Acad. Sci. USA93(1),456–460 (1996).
    • 66  Guise CP, Wang AT, Theil A, Bridewell DJ, Wilson WR, Patterson AV. Identification of human reductases that activate the dinitrobenzamide mustard prodrug PR-104A: A role for NADPH:cytochrome P450 oxidoreductase under hypoxia. Biochem. Pharmacol.74(6),810–820 (2007).
    • 67  Wang J, Foehrenbacher A, Su J et al. The 2-nitroimidazole EF5 is a biomarker for oxidoreductases that activate bioreductive prodrug CEN-209 under hypoxia. Clin. Cancer Res.18(6),1684–1695 (2012).
    • 68  Mason RP, Holtzman JL. The role of catalytic superoxide formation in the O2 inhibition of nitroreductase. Biochem. Biophys. Res. Commun.67(4),1267–1274 (1975).
    • 69  Plumb JA, Gerritsen M, Workman P. DT-diaphorase protects cells from the hypoxic cytotoxicity of indoloquinone EO9. Br. J. Cancer70(6),1136–1143 (1994).
    • 70  Patterson LH. Bioreductively activated antitumor N-oxides: the case of AQ4N, a unique approach to hypoxia-activated cancer chemotherapy. Drug Metab. Rev.34(3),581–592 (2002).
    • 71  Anderson RF, Denny WA, Ware DC, Wilson WR. Pulse radiolysis studies on the hypoxia-selective toxicity of a colbalt- mustard complex. Br. J. Cancer Suppl.27,S48–S51 (1996).
    • 72  Patel K, Choy SF, Hicks KO, Melink TJ, Holford NHG, Wilson WR. A combined pharmacokinetic model for the hypoxia-targeted prodrug PR-104A in humans, dogs, rats and mice predicts species differences in clearance and toxicity. Cancer Chemother. Pharmacol.67,1145–1155 (2011).
    • 73  Singleton RS, Guise CP, Ferry DM et al. DNA crosslinks in human tumor cells exposed to the prodrug PR-104A: relationships to hypoxia, bioreductive metabolism and cytotoxicity. Cancer Res.69(9),3884–3891 (2009).
    • 74  Guise CP, Abbattista M, Singleton RS et al. The bioreductive prodrug PR-104A is activated under aerobic conditions by human aldo-keto reductase 1C3. Cancer Res.70(4),1573–1584 (2010).
    • 75  Jameson MB, Rischin D, Pegram M et al. A Phase I trial of PR-104, a nitrogen mustard prodrug activated by both hypoxia and aldo-keto reductase 1C3, in patients with solid tumors. Cancer Chemother. Pharmacol.65(4),791–801 (2010).
    • 76  D’Andrea AD. Targeting DNA repair pathways in AML. Best Pract. Res. Clin. Haematol.23(4),469–473 (2010).
    • 77  Gu Y, Patterson AV, Atwell GJ et al. Roles of DNA repair and reductase activity in the cytotoxicity of the hypoxia-activated dinitrobenzamide mustard PR-104A. Mol. Cancer Ther.8(6),1714–1723 (2009).
    • 78  Birtwistle J, Hayden RE, Khanim FL et al. The aldo-keto reductase AKR1C3 contributes to 7,12-dimethylbenz(a)anthracene-3,4-dihydrodiol mediated oxidative DNA damage in myeloid cells: implications for leukemogenesis. Mutat. Res.662(1–2),67–74 (2009).
    • 79  Manesh DM, Szymanska B, Carol H et al. Molecular determinants of T-cell acute lymphoblastic leukaemia sensitivity to the pre-prodrug PR-104. Presented at: 24th Lorne Cancer Conference 2012. Lorne, Victoria, Australia, 9–11 February 2012.
    • 80  Konopleva M, Borthakur G, Thall PF et al. Phase I/II study of PR104, a bioreductive prodrug, in patients with relapsed/refractory acute myeloid leukemia (AML) using patient-specific adaptive dose selection. Blood118(21),661–661 (2011).▪▪ Provides an update of the Phase I/II trial of PR-104, a hypoxia-activated prodrug, in the setting of acute leukemias.
    • 81  Borch RF, Liu J, Joswig C, Baggs RB, Dexter DL, Mangold GL. Antitumor activity and toxicity of novel nitroheterocyclic phosphoramidates. J. Med. Chem.44(1),74–77 (2001).
    • 82  Duan JX, Jiao H, Kaizerman J et al. Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs. J. Med. Chem.51(8),2412–2420 (2008).
    • 83  Meng F, Evans JW, Bhupathi D et al. Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302. Mol. Cancer Ther.11(3),740–751 (2012).
    • 84  Jung D, Lin L, Jiao H, Cai X, Duan JX, Matteucci M. Pharmacokinetics of TH-302: a hypoxically activated prodrug of bromo-isophosphoramide mustard in mice, rats, dogs and monkeys. Cancer Chemother. Pharmacol.69(3),643–654 (2012).
    • 85  Liu Q, Sun JD, Wang J et al. TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules. Cancer Chemother. Pharmacol.69(6),1487–1498 (2012).
    • 86  Benito J, Lu H, Shi Y et al. Hypoxia activated pro-drug TH-302 induces hypoxia-dependent anti-leukemia activity in vitro and in vivo. Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research. Cancer Res.72(8 Suppl.), Abstract 4641 (2012).
    • 87  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. Haematologica96(1),69–77 (2011).
    • 88  Borad MJ, Chiorean EG, Molina JR et al. Clinical benefits of TH-302, a tumor selective hypoxia-activated prodrug, and gemcitabline in first-line pancreatic cancer (PanC). J. Clin. Oncol.29(Suppl. 4), (2011) (Abstract).
    • 89  Ganjoo KN, Cranmer LD, Butrynski JE et al. A Phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology80(1–2),50–56 (2011).
    • 90  Konopleva M, Handisides D, Lorente GA et al. A Phase 1 study of TH-302, a hypoxia-activated cytotoxic prodrug, in subjects with advanced leukemias. J. Clin. Oncol.30(15), (2012) (Abstract 6585).
    • 91  Seow HA, Penketh PG, Shyam K, Rockwell S, Sartorelli AC. 1,2-Bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine: an anticancer agent targeting hypoxic cells. Proc. Natl Acad. Sci. USA102(26),9282–9287 (2005).
    • 92  Reddy SB, Williamson SK. Tirapazamine: a novel agent targeting hypoxic tumor cells. Expert Opin. Investig. Drugs18(1),77–87 (2009).
    • 93  Rischin D, Peters L, Fisher R et al. Tirapazamine, cisplatin, and radiation versus fluorouracil, cisplatin, and radiation in patients with locally advanced head and neck cancer: a randomized Phase II trial of the Trans-Tasman Radiation Oncology Group (TROG 98.02). J. Clin. Oncol.23(1),79–87 (2005).
    • 94  Patterson AV, Jagdish J, Syddall SP et al. Cellular metabolism, murine pharmacokinetics and preclinical antitumor activity of SN29966, a novel hypoxia-activated irreversible pan-HER inhibitor. Mol. Cancer Ther.8(12 Suppl. 1),B79 (2009) (Abstract).
    • 95  Guise CP, Abbattista MR, Tipparaju SR et al. Diflavin oxidoreductases activate the bioreductive prodrug PR-104A under hypoxia. Mol. Pharmacol.81(1),31–40 (2012).
    • 101  PR104 in Treating Patients With Refractory/Relapsed Acute Leukemia. http://clinicaltrials.gov/show/NCT01037556
    • 102  Study of Hypoxia-Activated Prodrug TH-302 to Treat Advanced Leukemias. http://clinicaltrials.gov/show/NCT01149915