Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 2/2019

17.05.2019 | Original Article

Exposure time versus cytotoxicity for anticancer agents

verfasst von: David M. Evans, Jianwen Fang, Thomas Silvers, Rene Delosh, Julie Laudeman, Chad Ogle, Russell Reinhart, Michael Selby, Lori Bowles, John Connelly, Erik Harris, Julia Krushkal, Larry Rubinstein, James H. Doroshow, Beverly A. Teicher

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 2/2019

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Time is a critical factor in drug action. The duration of inhibition of the target or residence time of the drug molecule on the target often guides drug scheduling.

Methods

The effects of time on the concentration-dependent cytotoxicity of approved and investigational agents [300 compounds] were examined in the NCI60 cell line panel in 2D at 2, 3, 7 and in 3D 11 days.

Results

There was a moderate positive linear relationship between data from the 2-day NCI60 screen and the 3-, 7- and 11-day and a strong positive linear relationship between 3-, 7- and 11-day luminescence screen IC50s by Pearson correlation analysis. Cell growth inhibition by agents selective for a specific cell cycle phase plateaued when susceptible cells were growth inhibited or killed. As time increased the depth of cell growth inhibition increased without change in the IC50. DNA interactive agents had decreasing IC50s with increasing exposure time. Epigenetic agents required longer exposure times; several were only cytotoxic after 11 days’ exposure. For HDAC inhibitors, time had little or no effect on concentration response. There were potency differences amongst the three BET bromodomain inhibitors tested, and an exposure duration effect. The PARP inhibitors, rucaparib, niraparib, and veliparib reached IC50s < 10 μM in some cell lines after 11 days.

Conclusions

The results suggest that variations in compound exposure time may reflect either mechanism of action or compound chemical half-life. The activity of slow-acting compounds may optimally be assessed in spheroid models that can be monitored over prolonged incubation times.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Eastman A (2017) Improving anticancer drug development begins with cell culture: misinformation perpetrated by the misuse of cytotoxicity assays. Oncotarget 8:8854–8866CrossRefPubMed Eastman A (2017) Improving anticancer drug development begins with cell culture: misinformation perpetrated by the misuse of cytotoxicity assays. Oncotarget 8:8854–8866CrossRefPubMed
2.
Zurück zum Zitat Perego P, Hempel G, Linder S, Bradshaw TD, Larsen AK, Peters GJ, Phillips RM, on behalf of the EORTC PAMM Group (2018) Cellular pharmacology studies of anticancer agents: recommendations from the EORTC-PAMM group. Cancer Chemother Pharmacol 81:427–41 Perego P, Hempel G, Linder S, Bradshaw TD, Larsen AK, Peters GJ, Phillips RM, on behalf of the EORTC PAMM Group (2018) Cellular pharmacology studies of anticancer agents: recommendations from the EORTC-PAMM group. Cancer Chemother Pharmacol 81:427–41
3.
Zurück zum Zitat Liston DR, Davis M (2017) Clinically relevant concentrations of anticancer drugs: a guide for nonclinical studies. Clin Cancer Res 23:3489–3498CrossRefPubMedPubMedCentral Liston DR, Davis M (2017) Clinically relevant concentrations of anticancer drugs: a guide for nonclinical studies. Clin Cancer Res 23:3489–3498CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Henderson ES, Adamson RH, Denham C, Oliverio VT (1965) The metabolic fate of tritiated methotrexate I. absorption, excretion and distribution in mice, rats, dogs and monkeys. Cancer Res 25:1008–1017 Henderson ES, Adamson RH, Denham C, Oliverio VT (1965) The metabolic fate of tritiated methotrexate I. absorption, excretion and distribution in mice, rats, dogs and monkeys. Cancer Res 25:1008–1017
5.
Zurück zum Zitat Ludwig R, Alberts DS (1984) Chemical and biological stability of anticancer drugs used in a human tumor clonogenic assay. Cancer Chemother Pharmacol 12:142–145PubMed Ludwig R, Alberts DS (1984) Chemical and biological stability of anticancer drugs used in a human tumor clonogenic assay. Cancer Chemother Pharmacol 12:142–145PubMed
6.
Zurück zum Zitat Beijnen JH, Van der Nat JM, Labadie RP, Underberg WJ (1986) Decomposition of mitomycin and anthracycline cytostatics in cell culture media. Anticancer Res 6:39–43PubMed Beijnen JH, Van der Nat JM, Labadie RP, Underberg WJ (1986) Decomposition of mitomycin and anthracycline cytostatics in cell culture media. Anticancer Res 6:39–43PubMed
7.
Zurück zum Zitat Niell HD, Webster KC, Smith EE (1985) Anticancer drug activity in platin in human bladder tumor cell lines. Cancer 56:1039–1044CrossRefPubMed Niell HD, Webster KC, Smith EE (1985) Anticancer drug activity in platin in human bladder tumor cell lines. Cancer 56:1039–1044CrossRefPubMed
8.
Zurück zum Zitat Schuldes H, Bade S, Knobloch J, Jonas D (1997) Loss of in vitro cytotoxicity of cisplatin after storage as stock solution in cell culture medium at various temperatures. Cancer 79:1723–1728CrossRefPubMed Schuldes H, Bade S, Knobloch J, Jonas D (1997) Loss of in vitro cytotoxicity of cisplatin after storage as stock solution in cell culture medium at various temperatures. Cancer 79:1723–1728CrossRefPubMed
9.
Zurück zum Zitat Karahoca M, Momparler RL (2013) Pharmacokinetic and pharmacodynamic analysis of 5-aza-2’-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin Epigen 5:3–19CrossRef Karahoca M, Momparler RL (2013) Pharmacokinetic and pharmacodynamic analysis of 5-aza-2’-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin Epigen 5:3–19CrossRef
10.
Zurück zum Zitat Du L, Musson DG, Wang AQ (2006) Stability studies of vorinostat and its two metabolites in human plasma, serum and urine. J Pharmaceut Biomed Anal 42:556–564CrossRef Du L, Musson DG, Wang AQ (2006) Stability studies of vorinostat and its two metabolites in human plasma, serum and urine. J Pharmaceut Biomed Anal 42:556–564CrossRef
11.
Zurück zum Zitat Plumb JA, Finn PW, Williams RJ, Bandara MJ, La Thangue NB, Brown R (2003) Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol Cancer Therap 2:721–728 Plumb JA, Finn PW, Williams RJ, Bandara MJ, La Thangue NB, Brown R (2003) Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol Cancer Therap 2:721–728
12.
Zurück zum Zitat Wang H, Yu N, Chen D, Lee KCL, Lye PL, Chang JWW, Deng W, Ng MCY, Lu T et al (2011) Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile. J Med Chem 54:4694–4720CrossRefPubMed Wang H, Yu N, Chen D, Lee KCL, Lye PL, Chang JWW, Deng W, Ng MCY, Lu T et al (2011) Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile. J Med Chem 54:4694–4720CrossRefPubMed
13.
Zurück zum Zitat Konsoula R, Jung M (2008) In vitro plasma stability, permeability and solubility of mercaptoacetamide histone deacetylase inhibitors. Int J Pharm 361:19–25CrossRefPubMedPubMedCentral Konsoula R, Jung M (2008) In vitro plasma stability, permeability and solubility of mercaptoacetamide histone deacetylase inhibitors. Int J Pharm 361:19–25CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Holbeck SL, Camalier R, Crowell JA, Govinharajulu JP, Hollingshead M, Anderson LW, Polley E, Rubenstein L, Srivastava A, Wilsker D, Collins JM, Doroshow JH (2017) The national cancer institute ALMANAC: a comprehensive screening resource for the detection of anticancer drug pairs with enhanced therapeutic activity. Cancer Res 77:3564–3576CrossRefPubMedPubMedCentral Holbeck SL, Camalier R, Crowell JA, Govinharajulu JP, Hollingshead M, Anderson LW, Polley E, Rubenstein L, Srivastava A, Wilsker D, Collins JM, Doroshow JH (2017) The national cancer institute ALMANAC: a comprehensive screening resource for the detection of anticancer drug pairs with enhanced therapeutic activity. Cancer Res 77:3564–3576CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Lorenzi PL, Reinhold WC, Varma S, Hutchinson AA, Pommier Y, Chanock SJ, Weinstein J (2009) DNA fingerprinting of the NCI-60 cell line panel. Mol Cancer Ther 8:713–724CrossRefPubMedPubMedCentral Lorenzi PL, Reinhold WC, Varma S, Hutchinson AA, Pommier Y, Chanock SJ, Weinstein J (2009) DNA fingerprinting of the NCI-60 cell line panel. Mol Cancer Ther 8:713–724CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Selby M, Delosh R, Laudeman J, Ogle C, Reinhart R, Silvers T, Lawrence S, Kinders R, Parchment R, Teicher BA, Evans DM (2017) 3D models of the NCI60 cell lines for screening oncology compounds. SLAS Discovery 22:473–483PubMed Selby M, Delosh R, Laudeman J, Ogle C, Reinhart R, Silvers T, Lawrence S, Kinders R, Parchment R, Teicher BA, Evans DM (2017) 3D models of the NCI60 cell lines for screening oncology compounds. SLAS Discovery 22:473–483PubMed
18.
Zurück zum Zitat Shoemaker RH (2006) The NCI60 human tumor cell line anticancer drug screen. Nat Rev Cancer 6:813–823CrossRefPubMed Shoemaker RH (2006) The NCI60 human tumor cell line anticancer drug screen. Nat Rev Cancer 6:813–823CrossRefPubMed
19.
Zurück zum Zitat Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, Hose C, Langley J, Cronise P, Vaigro-Wolff A, Gray-Goodrich M, Campbell P, Mayo J, Boyd M (1991) Feasibility of a High-flux anticancer drug screen using a diverse panel of cultured tumor cell lines. J Natl Cancer Inst 83:757–766CrossRefPubMed Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, Hose C, Langley J, Cronise P, Vaigro-Wolff A, Gray-Goodrich M, Campbell P, Mayo J, Boyd M (1991) Feasibility of a High-flux anticancer drug screen using a diverse panel of cultured tumor cell lines. J Natl Cancer Inst 83:757–766CrossRefPubMed
20.
Zurück zum Zitat Weinstein JN, Myers TG, O’Connor PM, Friend SH, Fornace AJ, Kohn KW, Fojo T, Bates SE, Rubinstein LV, van Osdol WW, Monks AP et al (1997) An informative-intensive approach to the molecular pharmacology of cancer. Science 275:343–349CrossRefPubMed Weinstein JN, Myers TG, O’Connor PM, Friend SH, Fornace AJ, Kohn KW, Fojo T, Bates SE, Rubinstein LV, van Osdol WW, Monks AP et al (1997) An informative-intensive approach to the molecular pharmacology of cancer. Science 275:343–349CrossRefPubMed
21.
Zurück zum Zitat Bertino JR (2009) Cancer research: from folate antagonism to molecular targets. Best Prac Res Clin Hematol 22:577–582CrossRef Bertino JR (2009) Cancer research: from folate antagonism to molecular targets. Best Prac Res Clin Hematol 22:577–582CrossRef
22.
Zurück zum Zitat Li H, Li W, Liu S, Zong S, Wang W, Ren J, Li Q, Hou F, Shi Q (2016) DNMT1, DNMT3A and DNMT3B polymorphisms associated with gastric cancer risk: a systemic review and meta-analysis. EBioMed 13:125–131CrossRef Li H, Li W, Liu S, Zong S, Wang W, Ren J, Li Q, Hou F, Shi Q (2016) DNMT1, DNMT3A and DNMT3B polymorphisms associated with gastric cancer risk: a systemic review and meta-analysis. EBioMed 13:125–131CrossRef
23.
Zurück zum Zitat Uysal F, Akkoyunlu G, Ozturk S (2015) Dynamic expression of DNA methyltransferase (DNMTs) in oocytes and early embryos. Biochimie 116:103–113CrossRefPubMed Uysal F, Akkoyunlu G, Ozturk S (2015) Dynamic expression of DNA methyltransferase (DNMTs) in oocytes and early embryos. Biochimie 116:103–113CrossRefPubMed
24.
Zurück zum Zitat Fahy J, Jeltsch A, Arimondo PB (2012) DNA methyltransferase inhibitors in cancer: a chemical and therapeutic patent overview and selected clinical studies. Exp Opin Ther Patents 22:1427–1442CrossRef Fahy J, Jeltsch A, Arimondo PB (2012) DNA methyltransferase inhibitors in cancer: a chemical and therapeutic patent overview and selected clinical studies. Exp Opin Ther Patents 22:1427–1442CrossRef
25.
Zurück zum Zitat Hollenbach PW, Nguyen AN, Brady H, Williams M, Ning Y, Richard N, Krushel L, Aukerman SL, Heise C, MacBeth KJ (2010) A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS ONE 5:e9001CrossRefPubMedPubMedCentral Hollenbach PW, Nguyen AN, Brady H, Williams M, Ning Y, Richard N, Krushel L, Aukerman SL, Heise C, MacBeth KJ (2010) A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS ONE 5:e9001CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Yoo J, Choi S, Medina-Franco JL (2013) Molecular modeling studies of the novel inhibitors of DNA methyltransferases SGI-1027 and CBC12: implications for the mechanism of inhibition of DNMTs. PLoS ONE 8:e62152CrossRefPubMedPubMedCentral Yoo J, Choi S, Medina-Franco JL (2013) Molecular modeling studies of the novel inhibitors of DNA methyltransferases SGI-1027 and CBC12: implications for the mechanism of inhibition of DNMTs. PLoS ONE 8:e62152CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Peters GJ, Smid K, Vecchi L, Kathmann I, Sarksjan D, Honeywell RJ, Losekoot N, Ohne O, Orbach A, Blaugrund E, Jeong LS, Lee YB, Ahn CH, Kim DJ (2013) Metabolism, mechanism of action and sensitivity profile of fluorocyclopentenylcytosine (RX-3117; TV-1360). Invest New Drugs 31:1444–1457CrossRefPubMed Peters GJ, Smid K, Vecchi L, Kathmann I, Sarksjan D, Honeywell RJ, Losekoot N, Ohne O, Orbach A, Blaugrund E, Jeong LS, Lee YB, Ahn CH, Kim DJ (2013) Metabolism, mechanism of action and sensitivity profile of fluorocyclopentenylcytosine (RX-3117; TV-1360). Invest New Drugs 31:1444–1457CrossRefPubMed
28.
Zurück zum Zitat Eckschlager T, Plch J, Stiborova M, Hrabeta J (2017) Histone deacetylase inhibitors as anticancer drugs. Int J Molec Sci 18:1414–1439CrossRef Eckschlager T, Plch J, Stiborova M, Hrabeta J (2017) Histone deacetylase inhibitors as anticancer drugs. Int J Molec Sci 18:1414–1439CrossRef
29.
Zurück zum Zitat Shen L, Orillion A, Pili R (2016) Histone deacetylase inhibitors as immunomodulators in cancer therapeutics. Epigenomics 8:415–428CrossRefPubMed Shen L, Orillion A, Pili R (2016) Histone deacetylase inhibitors as immunomodulators in cancer therapeutics. Epigenomics 8:415–428CrossRefPubMed
31.
Zurück zum Zitat Italiano A (2016) Role of the EZH2 histone methyltransferase as a therapeutic target in cancer. Pharm Therap 165:26–31CrossRef Italiano A (2016) Role of the EZH2 histone methyltransferase as a therapeutic target in cancer. Pharm Therap 165:26–31CrossRef
32.
Zurück zum Zitat Lu C, Figueroa JA, Liu Z, Konala V, Aulakhy A, Verma R, Cobos E, Chiriva-Internati M, Gao W (2015) Nuclear export as a novel therapeutic target: the CRM1 connection. Curr Drug Targets 15:575–592CrossRef Lu C, Figueroa JA, Liu Z, Konala V, Aulakhy A, Verma R, Cobos E, Chiriva-Internati M, Gao W (2015) Nuclear export as a novel therapeutic target: the CRM1 connection. Curr Drug Targets 15:575–592CrossRef
33.
Zurück zum Zitat El-Tanani M, Dakir E, Raynor B, Morgan R (2016) Mechanisms of nuclear export in cancer and resistance to chemotherapy. Cancers 8:35–46CrossRefPubMedCentral El-Tanani M, Dakir E, Raynor B, Morgan R (2016) Mechanisms of nuclear export in cancer and resistance to chemotherapy. Cancers 8:35–46CrossRefPubMedCentral
34.
Zurück zum Zitat Garg M, Kanojia D, Mayakonda A, Said JW, Doan NB, Chien W, Ganesan TS, Chuang LSH et al (2017) Molecular mechanism and therapeutic implications of selinexor (KPT-330) in liposarcoma. Oncotarget 8:7521–7532PubMed Garg M, Kanojia D, Mayakonda A, Said JW, Doan NB, Chien W, Ganesan TS, Chuang LSH et al (2017) Molecular mechanism and therapeutic implications of selinexor (KPT-330) in liposarcoma. Oncotarget 8:7521–7532PubMed
35.
Zurück zum Zitat Magina KN, Pregartner G, Zebisch A, Wolfler A, Neumeister P, Greinix HT, Berghold A, Sill H (2017) Cytarabine dose in the consolidation of AML: a systematic review and meta-analysis. Blood 130:946–948CrossRefPubMed Magina KN, Pregartner G, Zebisch A, Wolfler A, Neumeister P, Greinix HT, Berghold A, Sill H (2017) Cytarabine dose in the consolidation of AML: a systematic review and meta-analysis. Blood 130:946–948CrossRefPubMed
36.
Zurück zum Zitat Zhenchuk A, Lotfi K, Juliusson G, Albertioni F (2009) Mechanisms of anti-cancer action and pharmacology of clofarabine. Biochem Pharmacol 78:1351–1359CrossRefPubMed Zhenchuk A, Lotfi K, Juliusson G, Albertioni F (2009) Mechanisms of anti-cancer action and pharmacology of clofarabine. Biochem Pharmacol 78:1351–1359CrossRefPubMed
37.
Zurück zum Zitat Jung M, Gelato KA, Fernandez-Montalvan A, Siegel S, Haendler B (2015) Targeting BET bromodomains for cancer treatment. Epigenomics 7:487–501CrossRefPubMed Jung M, Gelato KA, Fernandez-Montalvan A, Siegel S, Haendler B (2015) Targeting BET bromodomains for cancer treatment. Epigenomics 7:487–501CrossRefPubMed
38.
Zurück zum Zitat Andrieu G, Belkina AC, Denis GV (2016) Clinical trials for BET inhibitors run ahead of the science. Drug Discov Today 19:45–50CrossRef Andrieu G, Belkina AC, Denis GV (2016) Clinical trials for BET inhibitors run ahead of the science. Drug Discov Today 19:45–50CrossRef
39.
40.
Zurück zum Zitat Xu Y, Vakoc CR (2017) Targeting cancer cells with BET bromodomain inhibitors. Cold Spring Harbor Perspect Med 7:a026674CrossRef Xu Y, Vakoc CR (2017) Targeting cancer cells with BET bromodomain inhibitors. Cold Spring Harbor Perspect Med 7:a026674CrossRef
41.
Zurück zum Zitat Brown JS, O’Carrigan B, Jackson SP, Yap TA (2017) Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov 7:20–37CrossRefPubMed Brown JS, O’Carrigan B, Jackson SP, Yap TA (2017) Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discov 7:20–37CrossRefPubMed
43.
Zurück zum Zitat Probst BL, Liu L, Ramesh V, Li L, Sun H, Minna JD, Wang L (2010) Smac mimetics increase cancer cell response to chemotherapeutics in a TNF-α-dependent manner. Cell Death Differ 17:1645–1654CrossRefPubMed Probst BL, Liu L, Ramesh V, Li L, Sun H, Minna JD, Wang L (2010) Smac mimetics increase cancer cell response to chemotherapeutics in a TNF-α-dependent manner. Cell Death Differ 17:1645–1654CrossRefPubMed
44.
45.
Zurück zum Zitat Rimkus TK, Carpenter RI, Qasem S, Chan M, Lo HW (2016) Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors. Cancers 8:22–45CrossRefPubMedCentral Rimkus TK, Carpenter RI, Qasem S, Chan M, Lo HW (2016) Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors. Cancers 8:22–45CrossRefPubMedCentral
46.
Zurück zum Zitat Derakhshan A, Chen Z, Van Waes C (2016) Therapeutic small molecules target inhibitor of apoptosis proteins in cancers with deregulation of extrinsic and intrinsic cell death pathways. Clin Cancer Res 23:1379–1387CrossRefPubMedPubMedCentral Derakhshan A, Chen Z, Van Waes C (2016) Therapeutic small molecules target inhibitor of apoptosis proteins in cancers with deregulation of extrinsic and intrinsic cell death pathways. Clin Cancer Res 23:1379–1387CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Peery RC, Liu JY, Zhang JT (2017) Targeting survivin for therapeutic discovery: past, present, and future promises. Drug Discov Today 22:1466–1477CrossRefPubMed Peery RC, Liu JY, Zhang JT (2017) Targeting survivin for therapeutic discovery: past, present, and future promises. Drug Discov Today 22:1466–1477CrossRefPubMed
48.
Zurück zum Zitat Altieri DC (2010) Survivin and IAP proteins in cell-death mechanisms. Biochem J 430:199–205CrossRefPubMed Altieri DC (2010) Survivin and IAP proteins in cell-death mechanisms. Biochem J 430:199–205CrossRefPubMed
49.
Zurück zum Zitat Zhang H (2016) Three generations of epidermal growth factor receptor tyrosine kinase inhibitors developed to revolutionize the therapy of lung cancer. Drug Design Dev Therapy 10:3867–3872CrossRef Zhang H (2016) Three generations of epidermal growth factor receptor tyrosine kinase inhibitors developed to revolutionize the therapy of lung cancer. Drug Design Dev Therapy 10:3867–3872CrossRef
50.
Zurück zum Zitat Santarpia M, Ligori A, Karachaliou N, Gonzalez-Cao M, Daffina MG, D’Aveni A, Marabello G, Altavilla G, Rosell R (2017) Osimertinib in the treatment of non-small cell lung cancer: design, development and place in therapy. Lung Cancer Targets Therapy 8:109–125CrossRefPubMedPubMedCentral Santarpia M, Ligori A, Karachaliou N, Gonzalez-Cao M, Daffina MG, D’Aveni A, Marabello G, Altavilla G, Rosell R (2017) Osimertinib in the treatment of non-small cell lung cancer: design, development and place in therapy. Lung Cancer Targets Therapy 8:109–125CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Gaumann AKA, Kiefer F, Alfer J, Lang SA, Geissler EK, Breier G (2016) Receptor tyrosine kinase inhibitors: are they real tumor killers? Int J Cancer 138(540–54):52 Gaumann AKA, Kiefer F, Alfer J, Lang SA, Geissler EK, Breier G (2016) Receptor tyrosine kinase inhibitors: are they real tumor killers? Int J Cancer 138(540–54):52
52.
Zurück zum Zitat Wengner AM, Siemeister G, Koppitz M, Schulze V, Kosemund D, Klar U, Stoeckigt D, Neuhaus R, Lienau P et al (2016) Novel MPS1 kinase inhibitors with potent antitumor activity. Molec Cancer Therap 15:583–592CrossRef Wengner AM, Siemeister G, Koppitz M, Schulze V, Kosemund D, Klar U, Stoeckigt D, Neuhaus R, Lienau P et al (2016) Novel MPS1 kinase inhibitors with potent antitumor activity. Molec Cancer Therap 15:583–592CrossRef
53.
Zurück zum Zitat Ma WW (2011) Development of focal adhesion kinase inhibitors in cancer therapy. Anti-cancer Agents Med Chem 11:638–642CrossRef Ma WW (2011) Development of focal adhesion kinase inhibitors in cancer therapy. Anti-cancer Agents Med Chem 11:638–642CrossRef
54.
Zurück zum Zitat Ray GR, Hahn GM, Bagshaw MA, Kurkjian S (1973) Cell survival and repair of plateau phase cultures after chemotherapy: relevance to tumor therapy and to the in vitro screening of new agents. Cancer Chemother Rep 57:473–475PubMed Ray GR, Hahn GM, Bagshaw MA, Kurkjian S (1973) Cell survival and repair of plateau phase cultures after chemotherapy: relevance to tumor therapy and to the in vitro screening of new agents. Cancer Chemother Rep 57:473–475PubMed
55.
Zurück zum Zitat Keyes K, Cox K, Treadway P, Mann L, Shih C, Faul MM, Teicher BA (2002) An in vitro tumor model: analysis of angiogenic factor expression after chemotherapy. Cancer Res 62:5597–5602PubMed Keyes K, Cox K, Treadway P, Mann L, Shih C, Faul MM, Teicher BA (2002) An in vitro tumor model: analysis of angiogenic factor expression after chemotherapy. Cancer Res 62:5597–5602PubMed
56.
Zurück zum Zitat Bale SS, Moore L, Yarmush M, Jindal R (2016) Emerging in vitro liver technologies for drug metabolism and inter-organ interactions. Tissue Engineer B 22:383–394CrossRef Bale SS, Moore L, Yarmush M, Jindal R (2016) Emerging in vitro liver technologies for drug metabolism and inter-organ interactions. Tissue Engineer B 22:383–394CrossRef
57.
Zurück zum Zitat Pelkonen O, Turpeinen M, Hakkola J, Abass K, Pasanen M, Raunio H, Vahakangas K (2013) Preservation, induction or incorporation of metabolism into the in vitro cellular system—views to current opportunities and limitations. Toxicol In Vitro 27:1578–1583CrossRefPubMed Pelkonen O, Turpeinen M, Hakkola J, Abass K, Pasanen M, Raunio H, Vahakangas K (2013) Preservation, induction or incorporation of metabolism into the in vitro cellular system—views to current opportunities and limitations. Toxicol In Vitro 27:1578–1583CrossRefPubMed
59.
Zurück zum Zitat Halsall JA, Turner BM (2016) Histone deacetylase inhibitors for cancer therapy. An evolutionarily ancient resistance response may explain their limited success. Bioessays 38:1102–1110 Halsall JA, Turner BM (2016) Histone deacetylase inhibitors for cancer therapy. An evolutionarily ancient resistance response may explain their limited success. Bioessays 38:1102–1110
60.
Zurück zum Zitat Zhou MM, Dhalliun C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM (1999) Structure and ligand of a histone acetyltransferase bromodomain. Nature 399:491–496CrossRefPubMed Zhou MM, Dhalliun C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM (1999) Structure and ligand of a histone acetyltransferase bromodomain. Nature 399:491–496CrossRefPubMed
61.
Zurück zum Zitat McGlynn O, Lloyd B (2002) Recombinational repair and restart of damaged replication forks. Nat Rev 3:859–870CrossRef McGlynn O, Lloyd B (2002) Recombinational repair and restart of damaged replication forks. Nat Rev 3:859–870CrossRef
62.
Zurück zum Zitat Fleuren EDG, Zhang L, Wu J, Daly RJ (2016) The kinome ‘at large’ in cancer. Nat Rev Cancer 16:83–98CrossRefPubMed Fleuren EDG, Zhang L, Wu J, Daly RJ (2016) The kinome ‘at large’ in cancer. Nat Rev Cancer 16:83–98CrossRefPubMed
63.
Zurück zum Zitat Rotow J, Bivona TG (2017) Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer 17:637–658CrossRefPubMed Rotow J, Bivona TG (2017) Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer 17:637–658CrossRefPubMed
64.
Metadaten
Titel
Exposure time versus cytotoxicity for anticancer agents
verfasst von
David M. Evans
Jianwen Fang
Thomas Silvers
Rene Delosh
Julie Laudeman
Chad Ogle
Russell Reinhart
Michael Selby
Lori Bowles
John Connelly
Erik Harris
Julia Krushkal
Larry Rubinstein
James H. Doroshow
Beverly A. Teicher
Publikationsdatum
17.05.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 2/2019
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-019-03863-w

Weitere Artikel der Ausgabe 2/2019

Cancer Chemotherapy and Pharmacology 2/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.