Evolution of Anticancer Drug Discovery and the Role Of

Embed Size (px)

Citation preview

  • 7/28/2019 Evolution of Anticancer Drug Discovery and the Role Of

    1/2

    EDITORIALEvolution of Anticancer Drug Discovery and the Role of

    Cell-Based Screening

    Frank M. Balis

    The approach to the discovery of new anticancer drugs has

    recently evolved from a reliance on empiric cell-based screening

    for antiproliferative effects to a more mechanistically based ap-

    proach that targets the specific molecular lesions thought to be

    responsible for the development and maintenance of the malig-

    nant phenotype in various forms of cancer. The ultimate goal of

    the development of molecularly targeted drugs is to improve the

    efficacy and selectivity of cancer treatment by exploiting the

    differences between cancer cells and normal cells. The success

    of recently developed molecularly targeted agents, such as treti-

    noin (all-trans-retinoic acid) for acute promyelocytic leukemia

    (1,2) and imatinib (STI-571) for chronic myelogenous leukemia

    (CML) (3,4) and gastrointestinal stromal tumors (5), providesearly clinical validation for the molecularly targeted approach to

    drug discovery.

    Most of the commonly used cytotoxic anticancer drugs were

    discovered through random high-throughput screening of syn-

    thetic compounds and natural products in cell-based cytotoxicity

    assays. Despite the number and chemical diversity of these

    agents, the mechanisms of action are limited (Table 1), and most

    compounds are DNA-damaging agents with a low therapeutic

    index. With this screening approach, mechanism of action is not

    a primary determinant in selecting agents for further develop-

    ment, and, as a result, none of the current drugs directly targets

    the molecular lesions responsible for malignant transformation.

    The initial National Cancer Institute (NCI) high-throughputscreen used the highly chemosensitive P388 leukemia cell line,

    but this screen failed to identify drugs that were active against

    the common adult solid tumors. In the mid-1980s, the NCI

    implemented a new in vitro disease-oriented screen consisting

    of 60 human tumor cell lines representing nine common forms

    of cancer (6). It remains to be determined whether selective

    activity in vitro against cell lines representing a particular his-

    tologic form of cancer will be predictive for antitumor activity

    in vivo (7).

    As the molecular bases of specific forms of cancer are elu-

    cidated, a variety of new potential therapeutic targets are being

    identified. In the most common forms of cancers, the accumu-

    lation of mutations in multiple genes is required for tumorigen-esis (8,9). These mutations, such as the ras-activating mutations

    and mutations that inactivate p53, allow cancer cells to circum-

    vent intrinsic and extrinsic controls that tightly regulate the cell

    cycle and cell division and apoptosis in normal cells. After iden-

    tification of the genetic alterations in cancer cells, the critical

    transforming mutations must be discerned from genetic alter-

    ations that are the result of the inherent genetic instability in

    cancer cells but that do not play a direct role in tumorigenesis

    (9). This target validation is performed in preclinical cancer

    models. Identification of valid molecular targets has led to ra-

    tional target-based drug discovery at the protein level, as illus-

    trated by the development of imatinib, which was discovered by

    screening compound libraries for inhibitors of the protein kinase

    activity in vitro (10). Many of the proteins involved in cell cycle

    regulation, signal transduction, and the regulation of apoptosis

    are enzymes or receptors and are, therefore, potentially ame-

    nable to inhibition by small molecules (1114). Other examples

    of target-based treatments undergoing clinical evaluation in-

    clude farnesyltranferase inhibitors, which block post-transla-

    tional prenylation of ras, cyclin-dependent kinase inhibitors,

    protein kinase C inhibitors, and epidermal growth factor receptor

    kinase inhibitors (15).

    However, target-based screening for discovery of new mo-

    lecularly targeted cancer treatments has shortcomings. Unlike

    the Bcr-Abl fusion protein in CML and ras oncogenes that rep-resent mutations leading to a gain in function, most mutations in

    cancer cells result in a loss or inactivation of a protein (e.g., p53

    mutations), and screening a group of drugs by use of the protein

    product of mutated tumor suppressor genes is unlikely to dis-

    cover a small molecule that restores protein function. In addi-

    tion, compounds that are active in a target-based screening assay

    may not be specific for the protein used in the screen, and, as a

    result, the pharmacologic effect of the compound at a cellular or

    organism level may be more closely related to its effect on other

    unrelated and potentially higher affinity targets. Finally, most

    molecular targets for new cancer treatments interact with other

    proteins within pathways or networks in the cell, and the phar-

    macologic effect resulting from the inhibition of a specific targetmay be influenced by the expression or relative levels of these

    interacting proteins. Therefore, target-based screening assays

    may not be predictive of drug effect within the context of the

    whole cell (16).

    As demonstrated by Dunstan et al. (17) in this issue of the

    Journal, cell-based screening assays will continue to play an

    important role in drug discovery as we move into the molecular-

    targeting era. Their three-stage cell-based procedure using yeast

    cells that can be genetically manipulated is adaptable to high-

    throughput screening to identify agents that have a selective

    effect against cells with specific mutations (gene deletions). Un-

    like target-based assays, this cell-based assay is not a mechanis-

    tic screen, but determining the mechanism of action of selec-tively toxic agents from this screen may identify new molecular

    targets (e.g., downstream effectors in a pathway that is dere-

    pressed by the deletion of a tumor suppressor gene) for subse-

    quent target-based screening. From a pharmacologic perspec-

    tive, this cell-based screen detects collateral sensitivity. The

    Affiliation of author: Pediatric Oncology Branch, Center for Cancer Research,

    National Cancer Institute, Bethesda, MD.

    Correspondence to: Frank M. Balis, M.D., National Institutes of Health, Bldg.

    10, Rm. 13C103, 10 Center Dr., Bethesda, MD 208921920 (e-mail: balisf@

    nih.gov).

    78 EDITORIAL Journal of the National Cancer Institute, Vol. 94, No. 2, January 16, 2002

  • 7/28/2019 Evolution of Anticancer Drug Discovery and the Role Of

    2/2

    genetic mutation enhances the cells sensitivity to drugs that

    affect related pathways within the cell. The assay is also a direct

    application of the concept of synthetic lethality, which has been

    described previously in yeast (9,18). Two genes are synthetically

    lethal if mutations in either one is survivable, but mutations in

    both genes are lethal. For this cell-based assay, disruption of the

    function of the genes product by a drug is only lethal in cells

    that have a mutation in a second related gene.

    Cell-based assays are also used to confirm the activity of

    agents discovered in target-based screening assays and to assess

    the drugs pharmacologic effects at the cellular level. Unex-pected effects in cellular systems may suggest other targets for

    the agent or interactions of the primary molecular target with

    other vital proteins within the cell. The NCIs panel of 60 human

    tumor cell lines has also been adapted to provide information

    about the mechanism of action or molecular target of new agents

    that are tested based on the drugs profile of activity in the screen

    (19). As new targets are identified, their expression in each of

    the 60 cell lines in the panel can be characterized and correlated

    with the activity profile of the 70 000 compounds screened pre-

    viously without having to retest each agent. The identification of

    the topoisomerases as targets for drugs that were selectively

    active in yeast cells deficient in DNA double-strand break-repair

    proteins was apparently based on their activity profile in the

    human tumor cell line panel.

    Target-based and cell-based screening for new anticancer

    drugs in the molecular targeting era are complementary methods

    of identifying more selective anticancer drugs. They represent a

    dramatic shift in the drug discovery process that is likely to have

    an impact not only on the pharmacologic properties of new

    anticancer agents reaching the clinic but also on our approach to

    clinical drug development and the treatment of cancer.

    REFERENCES

    (1) Sun GL, Ouyang RR, Chen SJ, Gu YZ, Huang LA, Lu JX, et al. Treatment

    of acute promyelocytic leukemia with all-trans retinoic acid. A five-year

    experience. Chin Med J (Engl) 1993;106:7438.

    (2) Tallman MS, Andersen JW, Schiffer CA, Appelbaum FR, Feusner JH,

    Ogden A, et al. All-trans-retinoic acid in acute promyelocytic leukemia.

    N Engl J Med 1997;337:10218.

    (3) Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, et al.

    Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast

    crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with

    the Philadelphia chromosome. N Engl J Med 2001;344:103842.

    (4) Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, et al.

    Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase

    in chronic myeloid leukemia. N Engl J Med 2001;344:10317.

    (5) van Oosterom AT, Judson I, Verweij J, Stroobants S, Donato di Paola E,

    Dimitrijevic S, et al. Safety and efficacy of imatinib (STI571) in metastatic

    gastrointestinal stromal tumours: a phase I study. Lancet 2001;358:14213.

    (6) Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, et al.

    Feasibility of a high-flux anticancer drug screen using a diverse panel of

    cultured human tumor cell lines. J Natl Cancer Inst 1991;83:75766.

    (7) Weinstein JN, Myers TG, OConnor PM, Friend SH, Fornace AJ Jr, Kohn

    KW, et al. An information-intensive approach to the molecular pharmacol-

    ogy of cancer. Science 1997;275:3439.

    (8) Renan MJ. How many mutations are required for tumorigenesis? Implica-

    tions from human cancer data. Mol Carcinog 1993;7:13946.

    (9) Kaelin WG Jr. Choosing anticancer drug targets in the postgenomic era.J Clin Invest 1999;104:15036.

    (10) Drucker BJ, Lydon NB. Lessons learned from the development of an abl

    tyrosine kinase inhibitor for chronic myelogenous leukemia. J Clin Invest

    2000;105:37.

    (11) Drews J. Drug discovery: a historical perspective. Science 2000;287:

    19604.

    (12) Shapiro GI, Harper JW. Anticancer drug targets: cell cycle and checkpoint

    control. J Clin Invest 1999;104:164553.

    (13) Gibbs JB. Anticancer drug targets: growth factors and growth factor sig-

    naling. J Clin Invest 2000;105:913.

    (14) Sellers WR, Fisher DE. Apoptosis and cancer drug targeting. J Clin Invest

    1999;104:165561.

    (15) Gibbs JB. Mechanism-based target identification and drug discovery in

    cancer research. Science 2000;287:196973.

    (16) Sausville EA, Johnson JI. Molecules for the millennium: how will theylook? New drug discovery year 2000. Br J Cancer 2000;83:14014.

    (17) Dunstan HM, Ludlow C, Goehle S, Cronk M, Szankasi P, Evans DR, et al.

    Cell-based assays for identification of novel double-strand break-inducing

    agents. J Natl Cancer Inst 2002;94:8894.

    (18) Hartwell LH, Szankasi P, Roberts CJ, Murray AW, Friend SH. Integrating

    genetic approaches into the discovery of anticancer drugs. Science 1997;

    278:10648.

    (19) Weinstein JN, Buolamwini JK. Molecular targets in cancer drug discovery:

    cell-based profiling. Curr Pharm Des 2000;6:47383.

    Table 1. Mechanism of action of the commonly used natural product

    anticancer drugs*

    Drug class Mechanism of action

    Anthracyclines Topoisomerase II inhibitorsEpipodophyllotoxins Topoisomerase II inhibitorsDactinomycin Topoisomerase II inhibitorsCamptothecins Topoisomerase I inhibitorsTaxanes Tubulin-binding agentsVinca alkaloids Tubulin-binding agents

    *The source and chemical structures of these agents are diverse, but themolecular targets of these agents are limited to the topoisomerases and tubulin.

    Journal of the National Cancer Institute, Vol. 94, No. 2, January 16, 2002 EDITORIAL 79