Hypoxia-activated prodrugs and (lack of) clinical progress: The need for hypoxia-based biomarker patient selection in phase III clinical trials
Tài liệu tham khảo
Harris, 2002, Hypoxia–a key regulatory factor in tumour growth, Nat Rev Cancer, 2, 38, 10.1038/nrc704
Lomax, 2013, Biological consequences of radiation-induced DNA damage: relevance to radiotherapy, Clin Oncol (R Coll Radiol), 25, 578, 10.1016/j.clon.2013.06.007
Brown, 1999, The hypoxic cell: a target for selective cancer therapy–eighteenth Bruce F. Cain Memorial Award lecture, Cancer Res, 59, 5863
Brown, 2004, Exploiting tumour hypoxia in cancer treatment, Nat Rev Cancer, 4, 437, 10.1038/nrc1367
Cosse, 2008, Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression, Anti-Cancer Agents Med Chem, 8, 790, 10.2174/187152008785914798
Lu, 2010, Hypoxia and hypoxia-inducible factors: master regulators of metastasis, Clin Cancer Res, 16, 5928, 10.1158/1078-0432.CCR-10-1360
Heap, 2014, Spores of Clostridium engineered for clinical efficacy and safety cause regression and cure of tumors in vivo, Oncotarget, 5, 1761, 10.18632/oncotarget.1761
Vaupel, 2008, Hypoxia and aggressive tumor phenotype: implications for therapy and prognosis, Oncologist, 13, 21, 10.1634/theoncologist.13-S3-21
Daruwalla, 2006, Hyperbaric oxygen therapy for malignancy: a review, World J Surg, 30, 2112, 10.1007/s00268-006-0190-6
Song, 2001, Improvement of tumor oxygenation by mild hyperthermia, Radiat Res, 155, 515, 10.1667/0033-7587(2001)155[0515:IOTOBM]2.0.CO;2
Janssens, 2016, Quality-of-life after radiotherapy for advanced laryngeal cancer: results of a phase III trial of the Dutch Head and Neck Society, Radiother Oncol, 119, 213, 10.1016/j.radonc.2016.02.023
Janssens, 2012, Accelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: results of a phase III randomized trial, J Clin Oncol: Off J Am Soc Clin Oncol, 30, 1777, 10.1200/JCO.2011.35.9315
Kelleher, 2010, No sustained improvement in tumor oxygenation after localized mild hyperthermia, Adv Exp Med Biol, 662, 393, 10.1007/978-1-4419-1241-1_57
Mallory, 2015, Therapeutic hyperthermia: The old, the new, and the upcoming, Crit Rev Oncol/Hematol
Wenwu, 2013, Hyperbaric oxygen and cancer: more complex than we expected, Targeted Oncol, 8, 79, 10.1007/s11523-013-0259-8
Overgaard, 2007, Hypoxic radiosensitization: adored and ignored, J Clin Oncol, 25, 4066, 10.1200/JCO.2007.12.7878
van Gisbergen, 2015, How do changes in the mtDNA and mitochondrial dysfunction influence cancer and cancer therapy? Challenges, opportunities and models, Mutat Res Rev Mutat Res, 764, 16, 10.1016/j.mrrev.2015.01.001
Zannella, 2013, Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response, Clin Cancer Res, 19, 6741, 10.1158/1078-0432.CCR-13-1787
Rockwell, 2009, Hypoxia and radiation therapy: past history, ongoing research, and future promise, Curr Mol Med, 9, 442, 10.2174/156652409788167087
Wilson, 2011, Targeting hypoxia in cancer therapy, Nat Rev Cancer, 11, 393, 10.1038/nrc3064
Foehrenbacher, 2013, Design of optimized hypoxia-activated prodrugs using pharmacokinetic/pharmacodynamic modeling, Front Oncol, 3, 314, 10.3389/fonc.2013.00314
Mistry, 2017, Clinical advances of hypoxia-activated prodrugs in combination with radiation, Therapy Int J Radiat Oncol Biol Phys, 98, 1183, 10.1016/j.ijrobp.2017.03.024
Doherty, 1994, Muscle cramping in phase I clinical trials of tirapazamine (SR 4233) with and without radiation, Int J Radiat Oncol Biol Phys, 29, 379, 10.1016/0360-3016(94)90293-3
Reddy, 2009, Tirapazamine: a novel agent targeting hypoxic tumor cells, Expert Opin Invest Drugs, 18, 77, 10.1517/13543780802567250
Wardman, 1995, Radicals from one-electron reduction of nitro compounds, aromatic N-oxides and quinones: the kinetic basis for hypoxia-selective, bioreductive drugs, Biochem Soc Symp, 61, 171, 10.1042/bss0610171
Peterson, 2004, Integrating pharmacology and in vivo cancer models in preclinical and clinical drug development, Eur J Cancer, 40, 837, 10.1016/j.ejca.2004.01.003
Hicks, 2010, Pharmacokinetic/pharmacodynamic modeling identifies SN30000 and SN29751 as tirapazamine analogues with improved tissue penetration and hypoxic cell killing in tumors, Clin Cancer Res, 16, 4946, 10.1158/1078-0432.CCR-10-1439
Hicks, 2007, Oxygen dependence and extravascular transport of hypoxia-activated prodrugs: comparison of the dinitrobenzamide mustard PR-104A and tirapazamine, Int J Radiat Oncol Biol Phys, 69, 560, 10.1016/j.ijrobp.2007.05.049
Wilson, 2007, Bystander effects of bioreductive drugs: potential for exploiting pathological tumor hypoxia with dinitrobenzamide mustards, Radiat Res, 167, 625, 10.1667/RR0807.1
Patterson, 2007, Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA cross-linking agent PR-104, Clin Cancer Res, 13, 3922, 10.1158/1078-0432.CCR-07-0478
Abbattista, 2015, Pre-clinical activity of PR-104 as monotherapy and in combination with sorafenib in hepatocellular carcinoma, Cancer Biol Ther, 16, 610, 10.1080/15384047.2015.1017171
Benito, 2011, Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104, PLoS One, 6, 10.1371/journal.pone.0023108
Hunter, 2014, Dual targeting of hypoxia and homologous recombination repair dysfunction in triple-negative breast cancer, Mol Cancer Ther, 13, 2501, 10.1158/1535-7163.MCT-14-0476
Jameson, 2010, 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, 791, 10.1007/s00280-009-1188-1
McKeage, 2011, A phase I trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid tumour patients, BMC cancer, 11, 432, 10.1186/1471-2407-11-432
Abou-Alfa, 2011, PR-104 plus sorafenib in patients with advanced hepatocellular carcinoma, Cancer Chemother Pharmacol, 68, 539, 10.1007/s00280-011-1671-3
McKeage, 2012, PR-104 a bioreductive pre-prodrug combined with gemcitabine or docetaxel in a phase Ib study of patients with advanced solid tumours, BMC cancer, 12, 496, 10.1186/1471-2407-12-496
Guise, 2010, The bioreductive prodrug PR-104A is activated under aerobic conditions by human aldo-keto reductase 1C3, Cancer Res, 70, 1573, 10.1158/0008-5472.CAN-09-3237
Birtwistle, 2009, 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, 67, 10.1016/j.mrfmmm.2008.12.010
Desmond, 2003, The aldo-keto reductase AKR1C3 is a novel suppressor of cell differentiation that provides a plausible target for the non-cyclooxygenase-dependent antineoplastic actions of nonsteroidal anti-inflammatory drugs, Cancer Res, 63, 505
Millis, 1995, Colvin OM Gamcsik MP. Comparison of the protonation of isophosphoramide mustard and phosphoramide mustard, J Med Chem, 38, 2166, 10.1021/jm00012a017
Guise, 2014, Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia, Chin J Cancer, 33, 80, 10.5732/cjc.012.10285
Phillips, 2016, Targeting the hypoxic fraction of tumours using hypoxia-activated prodrugs, Cancer Chemother Pharmacol, 77, 441, 10.1007/s00280-015-2920-7
Yaromina, 2017, A novel concept for tumour targeting with radiation: Inverse dose-painting or targeting the “Low Drug Uptake Volume”, Radiother Oncol, 124, 513, 10.1016/j.radonc.2017.04.020
Sun, 2012, Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer, Clin Cancer Res, 18, 758, 10.1158/1078-0432.CCR-11-1980
Peeters, 2015, TH-302 in combination with radiotherapy enhances the therapeutic outcome and is associated with pretreatment [18F]HX4 hypoxia PET imaging, Clin Cancer Res, 21, 2984, 10.1158/1078-0432.CCR-15-0018
Stokes, 2016, Hypoxia imaging with pet correlates with antitumor activity of the hypoxia-activated prodrug evofosfamide (TH-302) in rodent Glioma Models, Tomogr: A J Imaging Res., 2, 229, 10.18383/j.tom.2016.00259
Bailey, 2014, Evaluation of the “steal” phenomenon on the efficacy of hypoxia activated prodrug TH-302 in pancreatic cancer, PLoS One, 9, 10.1371/journal.pone.0113586
Takakusagi, 2014, Pyruvate induces transient tumor hypoxia by enhancing mitochondrial oxygen consumption and potentiates the anti-tumor effect of a hypoxia-activated prodrug TH-302, PLoS One, 9, 10.1371/journal.pone.0107995
Wojtkowiak, 2015, Pyruvate sensitizes pancreatic tumors to hypoxia-activated prodrug TH-302, Cancer Metab, 3, 2, 10.1186/s40170-014-0026-z
Meng, 2015, Enhancement of hypoxia-activated prodrug TH-302 anti-tumor activity by Chk1 inhibition, BMC Cancer, 15, 422, 10.1186/s12885-015-1387-6
Sun, 2015, Combination treatment with hypoxia-activated prodrug evofosfamide (TH-302) and mTOR inhibitors results in enhanced antitumor efficacy in preclinical renal cell carcinoma models, Am J Cancer Res, 5, 2139
Borad, 2015, Randomized Phase II Trial of Gemcitabine Plus TH-302 Versus Gemcitabine in Patients With Advanced Pancreatic Cancer, J Clin Oncol: Off J Am Soc Clin Oncol, 33, 1475, 10.1200/JCO.2014.55.7504
Chawla, 2014, Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma, J Clin Oncol: Off J Am Soc Clin Oncol, 32, 3299, 10.1200/JCO.2013.54.3660
Ganjoo, 2011, 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, Oncology, 80, 50, 10.1159/000327739
Weiss, 2011, Phase 1 study of the safety, tolerability, and pharmacokinetics of TH-302, a hypoxia-activated prodrug, in patients with advanced solid malignancies, Clin Cancer Res, 17, 2997, 10.1158/1078-0432.CCR-10-3425
Tap, 2017, Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial, Lancet Oncol, 18, 1089, 10.1016/S1470-2045(17)30381-9
https://gicasym.org/maestro-trial-evofosfamidegemcitabine-pdac-fails-improve-os.
Mortensen, 2010, Identifying hypoxia in human tumors: a correlation study between 18F-FMISO PET and the Eppendorf oxygen-sensitive electrode, Acta Oncol, 49, 934, 10.3109/0284186X.2010.516274
Rasey, 1996, Quantifying regional hypoxia in human tumors with positron emission tomography of [18F]fluoromisonidazole: a pretherapy study of 37 patients, Int J Radiat Oncol Biol Phys, 36, 417, 10.1016/S0360-3016(96)00325-2
Dhani, 2015, Analysis of the intra- and intertumoral heterogeneity of hypoxia in pancreatic cancer patients receiving the nitroimidazole tracer pimonidazole, Br J Cancer, 113, 864, 10.1038/bjc.2015.284
Hay, 2014, Hypoxia-directed drug strategies to target the tumor microenvironment, Adv Exp Med Biol, 772, 111, 10.1007/978-1-4614-5915-6_6
Wilson, 2008, Targeting tumor hypoxia with prodrugs: Challenges and opportunities, AACR Educ Book, 293, 10.1158/AACR.EDB-08-8390
Duan, 2008, Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs, J Med Chem, 51, 2412, 10.1021/jm701028q
Zeman, 1986, SR-4233: a new bioreductive agent with high selective toxicity for hypoxic mammalian cells, Int J Radiat Oncol Biol Phys, 12, 1239, 10.1016/0360-3016(86)90267-1
Hicks, 2006, Use of three-dimensional tissue cultures to model extravascular transport and predict in vivo activity of hypoxia-targeted anticancer drugs, J Natl Cancer Inst, 98, 1118, 10.1093/jnci/djj306
Hicks, 2003, Multicellular resistance to tirapazamine is due to restricted extravascular transport: a pharmacokinetic/pharmacodynamic study in HT29 multicellular layer cultures, Cancer Res, 63, 5970
Hancock, 1996, Clinical evaluation of the hypoxic cytotoxin tirapazamine (SR-4233): phase I experience with repeated dose administration during fractionated irradiation, Int J Radiat Oncol Biol Phys, 36, 184, 10.1016/S0360-3016(97)85393-X
Foehrenbacher, 2013, The role of bystander effects in the antitumor activity of the hypoxia-activated prodrug PR-104, Front Oncol, 3, 263, 10.3389/fonc.2013.00263
Gu, 2010, Metabolism and excretion of the novel bioreductive prodrug PR-104 in mice, rats, dogs, and humans, Drug Metab Dispos, 38, 498, 10.1124/dmd.109.030973
Arnott, 2012, The influence of lipophilicity in drug discovery and design, Expert Opin Drug Discov, 7, 863, 10.1517/17460441.2012.714363
Leeson, 2007, The influence of drug-like concepts on decision-making in medicinal chemistry, Nat Rev Drug Discov, 6, 881, 10.1038/nrd2445
van de Waterbeemd, 2001, Lipophilicity in PK design: methyl, ethyl, futile, J Comput Aided Mol Des, 15, 273, 10.1023/A:1008192010023
Lienhart, 2013, The human flavoproteome, Arch Biochem Biophys, 535, 150, 10.1016/j.abb.2013.02.015
Guise, 2012, Diflavin oxidoreductases activate the bioreductive prodrug PR-104A under hypoxia, Mol Pharmacol, 81, 31, 10.1124/mol.111.073759
Guise, 2007, Identification of human reductases that activate the dinitrobenzamide mustard prodrug PR- 104A: a role for NADPH:cytochrome P450 oxidoreductase under hypoxia, Biochem Pharmacol, 74, 810, 10.1016/j.bcp.2007.06.014
Hunter, 2014, The flavoprotein FOXRED2 reductively activates nitro-chloromethylbenzindolines and other hypoxia-targeting prodrugs, Biochem Pharmacol, 89, 224, 10.1016/j.bcp.2014.03.001
Wang, 2012, The 2-nitroimidazole EF5 is a biomarker for oxidoreductases that activate the bioreductive prodrug CEN-209 under hypoxia, Clin Cancer Res, 18, 1684, 10.1158/1078-0432.CCR-11-2296
Baumann, 2001, Reversal of mitomycin C resistance by overexpression of bioreductive enzymes in Chinese hamster ovary cells, Cancer Res, 61, 7770
Hunter, 2012, Homologous recombination repair-dependent cytotoxicity of the benzotriazine di-N-oxide CEN-209: comparison with other hypoxia-activated prodrugs, Biochem Pharmacol, 83, 574, 10.1016/j.bcp.2011.12.005
Meng, 2012, Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302, Mol Cancer Ther, 11, 740, 10.1158/1535-7163.MCT-11-0634
Su, 2013, FSL-61 is a 6-nitroquinolone fluorogenic probe for one-electron reductases in hypoxic cells, Biochem J, 452, 79, 10.1042/BJ20121695
Bailey, 2001, Involvement of NADPH: cytochrome P450 reductase in the activation of indoloquinone EO9 to free radical and DNA damaging species, Biochem Pharmacol, 62, 461, 10.1016/S0006-2952(01)00674-8
Belcourt, 1996, 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 USA, 93, 456, 10.1073/pnas.93.1.456
Saunders, 2000, The relative importance of NADPH: cytochrome c (P450) reductase for determining the sensitivity of human tumour cells to the indolequinone EO9 and related analogues lacking functionality at the C-2 and C-3 positions, Biochem Pharmacol, 59, 993, 10.1016/S0006-2952(99)00405-0
Saunders, 2000, NADPH:cytochrome c (P450) reductase activates tirapazamine (SR4233) to restore hypoxic and oxic cytotoxicity in an aerobic resistant derivative of the A549 lung cancer cell line, Br J Cancer, 82, 651, 10.1054/bjoc.1999.0977
Patterson, 1995, Stratford IJ. Importance of P450 reductase activity in determining sensitivity of breast tumour cells to the bioreductive drug, tirapazamine (SR 4233), Br J Cancer, 72, 1144, 10.1038/bjc.1995.478
Patterson, 1997, Strafford IJ. Overexpression of human NADPH:cytochrome c (P450) reductase confers enhanced sensitivity to both tirapazamine (SR 4233) and RSU 1069, Br J Cancer, 76, 1338, 10.1038/bjc.1997.558
Tercel, 2009, Hypoxia-activated prodrugs: substituent effects on the properties of nitro seco-1,2,9,9a-tetrahydrocyclopropa[c]benz[e]indol-4-one (nitroCBI) prodrugs of DNA minor groove alkylating agents, J Med Chem, 52, 7258, 10.1021/jm901202b
Walsh, 2014, The clinical importance of assessing tumor hypoxia: relationship of tumor hypoxia to prognosis and therapeutic opportunities, Antioxid Redox Signal, 21, 1516, 10.1089/ars.2013.5378
Hoogsteen, 2007, Tumor microenvironment in head and neck squamous cell carcinomas: predictive value and clinical relevance of hypoxic markers. A review, Head Neck, 29, 591, 10.1002/hed.20543
Milosevic, 2004, The human tumor microenvironment: invasive (needle) measurement of oxygen and interstitial fluid pressure, Semin Radiat Oncol, 14, 249, 10.1016/j.semradonc.2004.04.006
Peerlings, 2017, Hypoxia and hypoxia response-associated molecular markers in esophageal cancer: a systematic review, Methods, 130, 51, 10.1016/j.ymeth.2017.07.002
Dewhirst, 2016, Is a major advance in tumor hypoxia imaging, Cancer Res, 76, 769, 10.1158/0008-5472.CAN-15-2818
O'Connor, 2016, Oxygen-Enhanced MRI Accurately Identifies, Quantifies, and Maps Tumor Hypoxia in Preclinical Cancer Models, Cancer Res, 76, 787, 10.1158/0008-5472.CAN-15-2062
Yaromina, 2011, Exploratory study of the prognostic value of microenvironmental parameters during fractionated irradiation in human squamous cell carcinoma xenografts, Int J Radiat Oncol Biol Phys, 80, 1205, 10.1016/j.ijrobp.2011.02.015
Xu, 2017, (18)F-Fluoromisonidazole in tumor hypoxia imaging, Oncotarget, 8, 94969, 10.18632/oncotarget.21662
Stieb, 2018, Longitudinal PET imaging of tumor hypoxia during the course of radiotherapy, Eur J Nucl Med Mol Imaging, 45, 2201, 10.1007/s00259-018-4116-y
Peeters, 2015, pre-clinical and clinical applications of hypoxia PET imaging using 2-nitroimidazoles, Q J Nucl Med Mol Imaging, 59, 39
Eschmann, 2005, Prognostic impact of hypoxia imaging with 18F-misonidazole PET in non-small cell lung cancer and head and neck cancer before radiotherapy, J Nucl Med, 46, 253
Mortensen, 2012, FAZA PET/CT hypoxia imaging in patients with squamous cell carcinoma of the head and neck treated with radiotherapy: results from the DAHANCA 24 trial, Radiother Oncol, 105, 14, 10.1016/j.radonc.2012.09.015
Wack, 2015, Comparison of [18F]-FMISO, [18F]-FAZA and [18F]-HX4 for PET imaging of hypoxia–a simulation study, Acta Oncol, 54, 1370, 10.3109/0284186X.2015.1067721
Peeters, 2015, A comparative study of the hypoxia PET tracers [18F]HX4, [18F]FAZA, and [18F]FMISO in a preclinical tumor model, Int J Radiat Oncol Biol Phys, 91, 351, 10.1016/j.ijrobp.2014.09.045
Le, 2008, Clinical biomarkers for hypoxia targeting, Cancer Metastasis Rev, 27, 351, 10.1007/s10555-008-9144-9
van Kuijk, 2016, Prognostic significance of carbonic anhydrase IX expression in cancer patients: a meta-analysis, Frontiers in oncology, 6, 69, 10.3389/fonc.2016.00069
Harris, 2015, Expression signatures as biomarkers of tumour hypoxia, Clin Oncol (R Coll Radiol), 27, 547, 10.1016/j.clon.2015.07.004
Toustrup, 2012, Gene expression classifier predicts for hypoxic modification of radiotherapy with nimorazole in squamous cell carcinomas of the head and neck, Radiother Oncol, 102, 122, 10.1016/j.radonc.2011.09.010
Petrik, 2006, Plasma osteopontin is an independent prognostic marker for head and neck cancers, J Clin Oncol: Off J Am Soc Clin Oncol, 24, 5291, 10.1200/JCO.2006.06.8627
Overgaard, 2005, Plasma osteopontin, hypoxia, and response to the hypoxia sensitiser nimorazole in radiotherapy of head and neck cancer: results from the DAHANCA 5 randomised double-blind placebo-controlled trial, Lancet Oncol, 6, 757, 10.1016/S1470-2045(05)70292-8
Toustrup, 2016, Validation of a 15-gene hypoxia classifier in head and neck cancer for prospective use in clinical trials, Acta Oncologica (Stockholm, Sweden), 55, 1091, 10.3109/0284186X.2016.1167959
Alsner, 2018, SP-0550: Clinical Trials, ESTRO 37 Abstr Book, 300
Mack, 2008, Lower osteopontin plasma levels are associated with superior outcomes in advanced non-small-cell lung cancer patients receiving platinum-based chemotherapy: SWOG Study S0003, J Clin Oncol: Off J Am Soc Clin Oncol, 26, 4771, 10.1200/JCO.2008.17.0662
Rischin, 2006, J Clin Oncol: Off J Am Soc Clin Oncol, 24, 2098, 10.1200/JCO.2005.05.2878
Antoniou, 2016, Biomarker-Guided Adaptive Trial Designs in Phase II and Phase III: A Methodological Review, PLoS One, 11, 10.1371/journal.pone.0149803
Parmar, 2008, Speeding up the evaluation of new agents in cancer, J Natl Cancer Inst, 100, 1204, 10.1093/jnci/djn267
Jones, 2007, An adaptive Simon Two-Stage Design for Phase 2 studies of targeted therapies, Contemp Clin Trials, 28, 654, 10.1016/j.cct.2007.02.008
Pusztai, 2007, Pharmacogenomic predictor discovery in phase II clinical trials for breast cancer, Clin Cancer Res, 13, 6080, 10.1158/1078-0432.CCR-07-0809
Simon, 1989, Optimal two-stage designs for phase II clinical trials, Control Clin Trials, 10, 1, 10.1016/0197-2456(89)90015-9
Wilhelm-Benartzi, 2017, Challenges and methodology in the incorporation of biomarkers in cancer clinical trials, Crit Rev Oncol/Hematol, 110, 49, 10.1016/j.critrevonc.2016.12.008
Freidlin, 2010, Randomized clinical trials with biomarkers: design issues, J Natl Cancer Inst, 102, 152, 10.1093/jnci/djp477
Buyse, 2011, Matheson A de Gramont A. Integrating biomarkers in clinical trials, Expert Rev Mol Diagn, 11, 171, 10.1586/erm.10.120
Toustrup, 2012, Hypoxia gene expression signatures as prognostic and predictive markers in head and neck radiotherapy, Semin Radiat Oncol, 22, 119, 10.1016/j.semradonc.2011.12.006