Characterization of the p53 tumor suppressor pathway in cell lines of the National Cancer Institute anticancer drug screen and correlations with the growth-inhibitory potency of 123 anticancer agents.

In the present study, we report the characterization of the p53 tumor suppressor pathway in the 60 cell lines of the National Cancer Institute (NCI) anticancer drug screen, as well as correlations between the integrity of this pathway and the growth-inhibitory potency of 123 anticancer agents in this screen. Assessment of p53 status in these lines was achieved through complete p53 cDNA sequencing, measurement of basal p53 protein levels and functional assessment of (a) transcriptional activity of p53 cDNA from each line in a yeast assay, (b) gamma-ray-induced G1 phase cell cycle arrest, and (c) gamma-ray-induced expression of CIP1/WAF1, GADD45, and MDM2 mRNA. Our investigations revealed that p53 gene mutations were common in the NCI cell screen lines: 39 of 58 cell lines analyzed contained a mutant p53 sequence. cDNA derived from almost all of the mutant p53 cell lines failed to transcriptionally activate a reporter gene in yeast, and the majority of mutant p53 lines studied expressed elevated basal levels of the mutant p53 protein. In contrast to most of the wild-type p53-containing lines, cells containing mutant p53 sequence were also deficient in gamma-ray induction of CIP1/WAF1, GADD45, and MDM2 mRNA and the ability to arrest in G1 following gamma-irradiation. Taken together, these assessments provided indications of the integrity of the p53 pathway in the 60 cell lines of the NCI cell screen. These individual p53 assessments were subsequently used to probe a database of growth-inhibitory potency for 123 "standard agents," which included the majority of clinically approved anticancer drugs. These 123 agents have been tested against these lines on multiple occasions, and a proposed mechanism of drug action had previously been assigned to each agent. Our analysis revealed that cells with mutant p53 sequence tended to exhibit less growth inhibition in this screen than the wild-type p53 cell lines when treated with the majority of clinically used anticancer agents: including DNA cross-linking agents, antimetabolites, and topoisomerase I and II inhibitors. Similar correlations were uncovered when we probed this database using most of the other indices of p53 status we assessed in the lines. Interestingly, a class of agents that differed in this respect was the antimitotic agents. Growth-inhibitory activity of these agents tended, in this assay, to be independent of p53 status. Our characterization of the p53 pathway in the NCI cell screen lines should prove useful to researchers investigating fundamental aspects of p53 biology and pharmacology. This information also allows for the large-scale analysis of the more than 60,000 compounds tested against these lines for novel agents that might exploit defective p53 function as a means of preferential toxicity.

[1]  M R Grever,et al.  The National Cancer Institute: cancer drug discovery and development program. , 1992, Seminars in oncology.

[2]  J N Weinstein,et al.  Use of the Kohonen self-organizing map to study the mechanisms of action of chemotherapeutic agents. , 1994, Journal of the National Cancer Institute.

[3]  M. Grever,et al.  Generation of a drug resistance profile by quantitation of mdr-1/P-glycoprotein in the cell lines of the National Cancer Institute Anticancer Drug Screen. , 1995, The Journal of clinical investigation.

[4]  G. Demers,et al.  Loss of normal p53 function confers sensitization to Taxol by increasing G2/M arrest and apoptosis , 1996, Nature Medicine.

[5]  A. Monks,et al.  Site of action of two novel pyrimidine biosynthesis inhibitors accurately predicted by the compare program. , 1995, Biochemical pharmacology.

[6]  G. Gaidano,et al.  Hemi‐ or homozygosity: a requirement for some but not other p53 mutant proteins to accumulate and exert a pathogenetic effect , 1993, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[7]  A. Levine,et al.  The p53 tumour suppressor gene , 1991, Nature.

[8]  W G Alvord,et al.  Enhanced sensitivity to 1-beta-D-arabinofuranosylcytosine and topoisomerase II inhibitors in tumor cell lines harboring activated ras oncogenes. , 1996, Cancer research.

[9]  P. Jeffrey,et al.  Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. , 1994, Science.

[10]  K. Kinzler,et al.  Molecular genetic profiles of colitis-associated neoplasms. , 1994, Gastroenterology.

[11]  M. Grever,et al.  Rhodamine efflux patterns predict P-glycoprotein substrates in the National Cancer Institute drug screen. , 1994, Molecular pharmacology.

[12]  A. Levine,et al.  p53 and its 14 kDa C-terminal domain recognize primary DNA damage in the form of insertion/deletion mismatches , 1995, Cell.

[13]  P. O'Connor,et al.  Cells lacking CIP1/WAF1 genes exhibit preferential sensitivity to cisplatin and nitrogen mustard , 1997, Oncogene.

[14]  C. Prives,et al.  Activation of p53 sequence-specific DNA bindingby short single strands of DNA requires the p53 C-terminus , 1995, Cell.

[15]  A. Fornace,et al.  Genotoxic stress confers preferential and coordinate messenger RNA stability on the five gadd genes. , 1994, Cancer research.

[16]  R. Brown,et al.  Cisplatin, camptothecin, and taxol sensitivities of cells with p53-associated multidrug resistance. , 1996, Molecular pharmacology.

[17]  K. Kinzler,et al.  Identification of p53 as a sequence-specific DNA-binding protein , 1991, Science.

[18]  J N Weinstein,et al.  Neural computing in cancer drug development: predicting mechanism of action. , 1992, Science.

[19]  K D Paull,et al.  Identification of novel antimitotic agents acting at the tubulin level by computer-assisted evaluation of differential cytotoxicity data. , 1992, Cancer research.

[20]  D. Scudiero,et al.  Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. , 1991, Journal of the National Cancer Institute.

[21]  K. Kohn,et al.  p53 gene mutations are associated with decreased sensitivity of human lymphoma cells to DNA damaging agents. , 1994, Cancer research.

[22]  L V Rubinstein,et al.  Multidrug-resistant phenotype of disease-oriented panels of human tumor cell lines used for anticancer drug screening. , 1992, Cancer research.

[23]  S. Friend,et al.  Screening patients for heterozygous p53 mutations using a functional assay in yeast , 1993, Nature genetics.

[24]  A. Fornace,et al.  Induction of fos RNA by DNA-damaging agents. , 1989, Cancer research.

[25]  G. S. Johnson,et al.  An Information-Intensive Approach to the Molecular Pharmacology of Cancer , 1997, Science.

[26]  J N Weinstein,et al.  A protein expression database for the molecular pharmacology of cancer , 1997, Electrophoresis.

[27]  C. Harris,et al.  Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. , 1994, Cancer research.

[28]  K. Kohn,et al.  Disruption of p53 function sensitizes breast cancer MCF-7 cells to cisplatin and pentoxifylline. , 1995, Cancer research.

[29]  Scott W. Lowe,et al.  p53 is required for radiation-induced apoptosis in mouse thymocytes , 1993, Nature.

[30]  L V Rubinstein,et al.  Comparison of in vitro anticancer-drug-screening data generated with a tetrazolium assay versus a protein assay against a diverse panel of human tumor cell lines. , 1990, Journal of the National Cancer Institute.

[31]  D. Brachman,et al.  p53 mutation does not correlate with radiosensitivity in 24 head and neck cancer cell lines. , 1993, Cancer research.

[32]  M. Boyd,et al.  Some practical considerations and applications of the national cancer institute in vitro anticancer drug discovery screen , 1995 .

[33]  D A Scudiero,et al.  An abnormality in the p53 pathway following gamma-irradiation in many wild-type p53 human melanoma lines. , 1996, Cancer research.

[34]  D. Housman,et al.  p53-dependent apoptosis modulates the cytotoxicity of anticancer agents , 1993, Cell.

[35]  R. Slebos,et al.  Loss of a p53-associated G1 checkpoint does not decrease cell survival following DNA damage. , 1993, Cancer research.

[36]  K. Kohn,et al.  Role of the p53 tumor suppressor gene in cell cycle arrest and radiosensitivity of Burkitt's lymphoma cell lines. , 1993, Cancer research.

[37]  J. Minna,et al.  Development of antibodies against p53 in lung cancer patients appears to be dependent on the type of p53 mutation. , 1992, Cancer research.

[38]  B. Vogelstein,et al.  p53 mutations in human cancers. , 1991, Science.

[39]  Edward A. Sausville,et al.  Identification of compounds with preferential inhibitory activity against low-Nm23-expressing human breast carcinoma and melanoma cell lines , 1997, Nature Medicine.

[40]  G. Demers,et al.  Inactivation of p53 enhances sensitivity to multiple chemotherapeutic agents. , 1996, Cancer research.

[41]  R. Shoemaker,et al.  Overlapping phenotypes of multidrug resistance among panels of human cancer‐cell lines , 1996, International journal of cancer.