Cell Cycle Modulators for the Treatment of Lung Malignancies

Cell Cycle Modulators for the Treatment of Lung Malignancies

c omprehensive review Cell Cycle Modulators for the Treatment of Lung Malignancies Adrian M. Senderowicz Abstract It has become clear in the past deca...

693KB Sizes 0 Downloads 22 Views

c omprehensive review Cell Cycle Modulators for the Treatment of Lung Malignancies Adrian M. Senderowicz Abstract It has become clear in the past decade that most human malignancies, including lung neoplasms, have aberrations in cell cycle control. The tumor suppressor gene retinoblastoma is an important player in the G1/S transition and its function is abnormal in most human neoplasms. Retinoblastoma function is lost as a result of phosphorylation by the cyclin-dependent kinases (CDKs). Thus, modulation of CDKs may have an important use for the therapy and prevention of human neoplasms. Direct CDK modulators are small molecules that target specifically the adenosine triphosphate binding site of CDKs. In contrast, indirect CDK modulators affect CDK function by modulation of upstream pathways required for CDK activation. The first example of a direct smallmolecule CDK modulator tested in the clinic, flavopiridol, is a pan-CDK inhibitor that not only promotes cell cycle arrest but also halts transcriptional elongation, promotes apoptosis, induces differentiation, and has antiangiogenic properties. The second example of direct small-molecule CDK modulators tested in clinical trials is UCN-01 (7-hydroxystaurosporine). UCN-01 has interesting preclinical features: it inhibits Ca2+-dependent protein kinase C, promotes apoptosis, arrests cell cycle progression at G1/S, and abrogates checkpoints upon DNA damage. In summary, novel small-molecule CDK modulators are being tested in the clinic with interesting results. Although these small molecules are directed toward a very prevalent cause of carcinogenesis, their role in the clinical armamentarium is still uncertain. Clinical Lung Cancer, Vol. 5, No. 3, 158-168, 2003

Key words: Angiogenesis, Apoptosis, Cyclin-dependent kinases, Differentiation, Flavopiridol, 7-Hydroxystaurosporine, Retinoblastoma

Introduction

INK (p16INK4a, p15INK4b, p18INK4c, p19INK4d) and CIP/KIP families (p21CIP1, p27KIP1, and p57KIP2).1,2 Retinoblastoma proteins are pocket proteins that sequester E2F transcription factors, preventing them from activating critical genes in cell proliferation.3,4 After Rb phosphorylation by CDK4 and/or CDK6 complexes during G1-phase and by CDK2 at G1/S interphase, E2F proteins are released and promote the transcription of genes essential for the transition to S-phase of the cell cycle.5,6 CDK4,6/D-type cyclins therefore execute their critical functions during mid to late G1-phase, as cells cross a G1 restriction point and become independent of mitogens for completion of the division cycle. These features suggest that the fundamental role of these complexes is to integrate extracellular signals with the cell cycle machinery.1,2 Other important points of regulation have been described in G2 and mitosis. In these phases, the specific expression of certain regulators is essential to control the correct sequence of events that leads to cell division. Basically, cyclins B1 and B2 and their partner cdc2 (ie, CDK1), together with other kinases and phosphatases (eg, wee1, cdc25), regulate the final phases of the cell cycle (Figure 1).

Brief Overview of Cell Cycle Regulation Regulation of the cell cycle and cell proliferation has been extensively studied in the past few years, and a consensus paradigm of the regulation of the cell cycle has been developed.1,2 According to this paradigm, the “master switch” of the cell cycle is the retinoblastoma (Rb) family of proteins. Proliferation occurs by the phosphorylation of these proteins by the cyclin-dependent kinases (CDKs; Figure 1).1 At least 9 CDKs are activated by D-type cyclins (D1, D2, and D3) and cyclin E and inhibited by 2 families of CDK inhibitors, the

Molecular Therapeutics Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD Submitted: May 29, 2003; Revised: Sep 23, 2003; Accepted: Oct 1, 2003 Address for correspondence: Adrian M. Senderowicz, MD, Molecular Therapeutics Unit, Oral and Pharyngeal Cancer Branch, National Institute of Craniofacial and Dental Research, National Institutes of Health, 30 Convent Dr, Bldg 30, Rm 212, Bethesda, MD 20892-4330 Fax: 301-402-0823; e-mail: [email protected]

158

Clinical Lung Cancer November 2003

In the past decade, several proteins that participate in the tight control of cell division have been found to be mutated, deleted, amplified, or overexpressed in human tumors. In the first part of this review, the principal points of deregulation found in human tumors will be summarized, with particular emphasis on lung cancer.

Figure 1

Regulation of G1-, S-, G2-, and M-Phases of Cell Cycle Growth Factors (ras, myc, fos, jun) Cyclin B CDK 1

Cell Cycle Alterations in Neoplasia In the past few years, it has become clear that cyclins, CDK complexes, and other cell cycle regulators are mechanistically involved in the development of human tumors.7-11 This is consistent with a large body of literature showing the importance of inactivation of the Rb pathway in tumor development.9,12,13 The inactivation of Rb can be produced by direct mutation of the Rb protein, but this is a relatively rare event, occurring only in Rbs, osteosarcomas, and a minority of breast and some other tumors.1,9,14 More frequent alterations of this pathway occur by functional inactivation of Rb by hyperphosphorylation. This is normally the result of increased CDK activities caused by overexpression of cyclins. For example, several laboratories have reported that some tumors show loss of Rb or, alternatively, overexpression of cyclin D1.15-17 Similarly, in other tumors, loss of p16INK4a and Rb are mutually exclusive.18-20 This observation led to the hypothesis that inactivation of the cyclin D/CDK/p16/Rb protein pathway can promote tumor development and that either (1) loss of the suppressor activity of Rb or p16INK4a or (2) overexpression of cyclin D1 can override this checkpoint.1,9 Several reports have implicated D-type cyclins in the neoplastic development, although limited information is available on the participation of its partner, CDK4, in these events. The involvement of CDK4 in the neoplastic process was suggested by the fact that CDK4 amplification and/or overexpression was detected in human glioblastomas, but overexpression and/or amplification of D-type cyclins was not detected in these tumors.21,22 In addition, CDK4 mutations were identified in patients with familial melanoma,23 and recently, amplification and overexpression of CDK4 were also detected in sporadic breast carcinomas,24 ovarian carcinomas,25 and sarcomas.26 In light of all these findings, proteins that govern cell cycle control are reasonable targets for cancer therapy.13,27

Specific Cell Cycle Aberrations in Lung Cancer The Rb tumor-suppressor gene is located on chromosome 13q14. Cytogenetic abnormalities of chromosome 13 and loss of heterozygosity at the Rb locus have been reported in a variety of human cancers, including non–small-cell lung cancer (NSCLC), and the frequency of Rb abnormalities detected by immunohistochemistry in NSCLC has reached 30%.28,29 Furthermore, functional Rb protein is absent in 90% of cases of small-cell lung cancer (SCLC) and in as many as 30% of NSCLC primary lesions and cell lines.28 Thus, the G1/S checkpoint transition is lost in the majority of patients with lung cancer. As mentioned earlier, the molecular events that lead to deregulated Rb function include sustained activity of CDKs as a result of loss of CDK inhibitors such as p16INK4a,

p16INK4a family

M Rb

p21CIP1 family P

CDK 4/6

P

G2 Rb~ P E2F

Cyclin A

Cyclin D

CDK 1

G1 Cyclin E CDK 2

S Cyclin A CDK 2

p21CIP1 family

Abbreviations: CDK = cyclin-dependent kinase; Rb = retinoblastoma

and persistent upregulation of several cyclins such as cyclin D1, cyclin A, and cyclin E. Therefore, sustained hyperphosphorylation and inactivation of Rb also contribute to the transformation of normal bronchial epithelium to autonomously growing cancer cells. Additional mechanisms of Rb inactivation include Rb point mutations and chromosomal deletions.30,31 Although Rb plays an important role in lung tumorigenesis, it appears that Rb status does not represent a significant prognostic factor in adenocarcinoma of the lung. Indeed, among 90 patients, 56.7% were shown to have reduced expression of Rb as measured by immunohistochemistry; however, there were no statistical differences among expression of Rb and clinical survival.32 Similar results were observed in a larger study in 207 surgically resected primary NSCLCs.33 Members of the INK4 protein family such as p16, p15, p18, and p19 inhibit CDK4- and CDK6-mediated phosphorylation of Rb;34,35 inactivation of p16INK4a in tumors expressing wild-type Rb is required for malignant cells to enter the Sphase or escape senescence.36 The occurrence of p16INK4a lesions is second only to p53 abnormalities in human cancer and is a frequent event in premalignant lesions of the upper digestive tract.37 The CDKN2A gene is inactivated by a 2-hit mechanism that can involve CpG island methylation, 9p21 loss of heterozygosity, mutation, or homozygous deletion.14,38 The CDKN2A locus on chromosome 9p21 encodes 2 proteins translated by alternative splicing of messenger RNA (mRNA): p16INK4a, which inhibits phosphorylation of Rb through cyclin D1/CDK4 and p14ARF, a protein that binds MDM2, thereby stabilizing wild-type p53.39 Analysis of p16INK4a, p14ARF, and p53 from 38 primary NSCLC specimens showed that p16INK4a was inactivated in 58% of tumors by homozy-

Clinical Lung Cancer November 2003

159

Cyclin-Dependent Kinase Inhibitors in Lung Cancer Figure 2 Modes of Action for Cell Cycle Modulators

A P

CDK7

T160 PO4-

CDK

CKI

ATP Y15

Cyclin

T14

wee1/myt1

P

PO4-

cdc25

B Perifosine

Manipulation of Cell Cycle Machinery for Therapeutic Purposes

UCN-01 T160

CKI CKI CKI CDK

Flavopiridol Alsterpaullone

ATP

Cyclin

Y15 T14

Loss in CDK function may occur as a result of loss in mass of catalytic subunit and/or cofactors, increased endogenous inhibitors, increased wee1/myt1, or loss in CDK7 or cdc25 activity (A). However, the most successful way to modulate CDK is by competing with ATP binding in CDK and upregulating endogenous CDK inhibitors (B). Abbreviations: ATP = adenosine triphosphate; CDK = cyclin-dependent kinase, CKI = cyclin-dependent kinase inhibitor

gous deletions, promoter hypermethylation, and point mutation in exon 2.40 Fourteen tumors had simultaneous p16INK4a and p14ARF inactivation, most frequently because of homozygous deletions extending into the INK4a/ARF locus. A confirmatory study by Spanakis et al demonstrated decreased p16INK4a in 46% of NSCLCs examined.41 Gorgoulis et al investigated the alterations of G1-phase proteins using immunohistochemical and molecular methods in a series of 68 NSCLCs and performed correlation with clinicopathologic findings and prognosis.42 Abnormal expression in p16INK4a and Rb was observed in 33 (49%) and 27 (40%) cases, respectively. The main mechanisms responsible for loss in p16INK4a were deletions and transcriptional silencing by methylation. In addition, similar deregulation may occur in other tumor-suppressor genes that reside at the 9p21-22 region, CDKN2/MTS1/p16INK4a, p14ARF, and MTS2/p15INK4b. The lack of correlation with clinical tumor-stage and prevalence aberrations in several G1 proteins suggests that multiple hits of this network may represent an early event in lung carcinogenesis. In another interesting study, Shapiro et al compared the expression of G1 checkpoint proteins in NSCLC, which is predominantly Rb-positive, and in SCLC, which is Rb-negative.43 Most NSCLC and SCLC resection specimens and cell lines overexpress cyclin D1 (indicating that cyclin D1 overexpression and Rb inactivation can coexist in SCLC). However, 9 of 9 Rbpositive NSCLC cell lines have absent or low levels of p16, whereas an Rb-negative NSCLC line and 5 of 5 SCLC cell lines have high levels of p16. In primary resection specimens, p16

160

was undetectable in 18 of 27 NSCLC samples and abundant in 4 of 5 SCLC samples. Thus, a reciprocal relationship between Rb inactivation and p16 expression is common in several malignancies (including NSCLC), and a clear distinction is apparent in the expression of p16 in SCLC and NSCLC. In summary, most lung carcinomas have aberrations in the G1/S transition. Although decreased expression in Rb occurs in a significant number of cases, the majority of lung tumors have loss in Rb function caused by aberrations in proteins that regulate the activity of CDKs, leading to inactivation and phosphorylation of Rb. This information suggests that smallmolecule CDK modulators may be useful for the prevention or treatment of lung cancer.

Clinical Lung Cancer November 2003

Several strategies could be considered to modulate CDK activity (Figure 2). These strategies are divided into direct effects on the catalytic CDK subunit and indirect modulation of regulatory pathways that govern CDK activity.44,45 As depicted in Figure 2A, the small molecular CDK inhibitors are compounds that directly target the catalytic CDK subunit. Most of these compounds modulate CDK activity by interacting specifically with the adenosine triphosphate (ATP) binding site of CDKs.44-48 Examples of this class include flavopiridol and alsterpaullone. The second class of CDK inhibitors are compounds that inhibit CDK activity by targeting the regulatory upstream pathways that modulate CDK activity by altering the expression and synthesis of the CDK/cyclin subunits or the CDK inhibitory proteins; by modulating the phosphorylation of CDKs; by targeting CDK-activating kinase, cdc25, and wee1/myt1; or by manipulating the proteolytic machinery that regulates the catabolism of CDK/cyclin complexes or their regulators (Figure 2B).44,45,49 Examples of this class of compounds include perifosine and UCN-01.

Modulators of Cyclin-Dependent Kinase Activity As mentioned previously, CDK can be modulated by direct effects on the catalytic subunit and/or by disruption of upstream regulatory pathways. Several examples and mechanisms are described in Table 145,47,50-66 and elsewhere.13,27,44,49,67-70

Flavopiridol Mechanism of Antiproliferative Effects. Flavopiridol (L868275; HMR 1275) is a semisynthetic flavonoid derived from rohitukine, an indigenous plant of India. Initial studies with flavopiridol demonstrated modest in vitro inhibitory activity with respect to epidermal growth factor receptor (EGFR) and protein kinase A (PKA; inhibitory concentrations [IC50s] of 21 and 122 μmol/L, respectively).71 However, when this compound was tested in the NCI 60 cell line anticancer drug screen panel, it demonstrated very potent growth inhibition (IC50, 66 nmol/L), a concentration approximately 1000 times lower than the concentration required to inhibit PKA and EGFR.71 Initial studies with this flavonoid revealed clear evidence of G1/S or G2/M arrest caused by loss in CDK1 and

Adrian M. Senderowicz CDK2.72-74 Studies using purified CDKs showed that the inhibition observed is reversible and competitively blocked by ATP, with a Ki of 41 nmol/L.72-76 Furthermore, the crystal structure of the complex of deschloroflavopiridol and CDK2 showed that flavopiridol binds to the ATP binding pocket, with the benzopyrene occupying the same region as the purine ring of ATP,77 confirming the results of earlier biochemical studies with flavopiridol.73 Flavopiridol inhibits all CDKs thus far examined (IC50, approximately 100 nmol/L), but the inhibition potential of flavopiridol on CDK7 (CDK-activating kinase) was found to be much lower than that against other CDKs (IC50, approximately 300 nmol/L).73,75,76 In addition to directly inhibiting CDKs, flavopiridol promotes a decrease in the level of cyclin D1, an oncogene that is overexpressed in many human neoplasms. Of note, neoplasms that overexpress cyclin D1 have a poor prognosis.78-80 Depletion of cyclin D1 appears to lead to the loss of CDK activity.81 Cyclin D1 decrease is caused by depletion of cyclin D1 mRNA and was associated with a specific decrease in cyclin D1 promoter as measured by a luciferase reporter assay. The transcriptional repression of cyclin D1 observed after treatment with flavopiridol is consistent with the effects of flavopiridol on yeast cells (as described earlier) and underscores the conserved effect of flavopiridol on eukaryotic cyclin transcription.50 In summary, flavopiridol can induce cell cycle arrest by at least these 3 mechanisms: (1) direct inhibition of CDK activities by binding to the ATP binding site; (2) prevention of the phosphorylation of CDKs at threonine-160/161 by inhibition of CDK7/cyclin H;74,75 and (3) decrease in the amount of cyclin D1, an important cofactor for CDK4 and CDK6 activation (G1/S arrest only). Another effect of flavopiridol on transcription is attenuation of the induction of vascular endothelial growth factor (VEGF) mRNA in monocytes after hypoxia (as described later). This effect is a result of alterations in the stability of VEGF mRNA.82 We recently demonstrated that flavopiridol potently inhibits P-TEFb (also known as CDK9/cyclin T), with a Ki of 3 nmol/L, leading to inhibition of transcription by RNA polymerase II by blocking the transition into productive elongation. Interestingly, in contrast with all CDKs tested so far, flavopiridol was not competitive with ATP in this reaction. P-TEFb is a required cellular cofactor for the human immunodeficiency virus (HIV)–1 transactivator Tat. Consistent with its ability to inhibit P-TEFb, flavopiridol blocked Tat transactivation of the viral promoter in vitro. Furthermore, flavopiridol blocked HIV1 replication in single-round and viral spread assays with an IC50 of < 10 nmol/L.83 These actions of the drug led to the testing of flavopiridol in clinical trials in patients with HIV-related malignancies.84 Apoptosis. An important biochemical effect involved in the antiproliferative effect of flavopiridol is the induction of apoptotic cell death. Hematopoietic cell lines are often quite sensitive to flavopiridol-induced apoptotic cell death,85-88 but the mechanism(s) by which flavopiridol induces apoptosis have not yet been elucidated. Flavopiridol does not modulate topoi-

Table 1

Direct Cyclin-Dependent Kinase Modulators

Kinase Modulated

CDK1/CDK2/CDK5

Examples Purvalanol and Compound 5250,63 Roscovitine55,56 Olomoucine55,57,59 Butyrolactone I58 CVT-31360

Nonspecific CDK

Flavopiridol45,65 UCN-0145,51-54 Staurosporine53,65 Oxindole I61

Unknown

Paullones47,55,64 Toyocamycin62 Myricetin66

Abbreviation: CDK = cyclin-dependent kinase

somerase I/II activity.87 In certain hematopoietic cell lines, neither Bcl-2/BAX nor p53 appeared to be affected;87,89 whereas, in other systems, Bcl-2 may be inhibited.88 Preliminary evidence demonstrated that flavopiridol-induced apoptosis in leukemia cells is associated with early activation of the mitogen-activated protein kinase family of proteins (MEK, p38, and JNK).90 This activation may lead to activation of caspases.90 As seen in this and other models, caspase inhibitors prevent flavopiridol-induced apoptosis.86,90 It is unclear whether the putative flavopiridol-induced inhibition of CDK activity is required for induction of apoptosis. Clear evidence of cell cycle arrest along with apoptosis was observed in a panel of squamous head and neck cancer cell lines, including a cell line (HN30) that is refractory to several DNA-damaging agents, such as γ-irradiation and bleomycin.91 Again, the apoptotic effect was independent of p53 status and was associated with the depletion of cyclin D1. These findings have been corroborated in other preclinical models.89,92-94 Efforts to understand flavopiridol-induced apoptosis are under intense investigation. Angiogenesis. Flavopiridol targets not only tumor cells but also angiogenesis pathways. Brusselbach et al incubated primary human umbilical vein endothelial cells with flavopiridol and observed apoptotic cell death even in cells that were not cycling, leading to the notion that flavopiridol may have antiangiogenic properties as a result of endothelial cytotoxicity.95 In other model systems, Kerr et al tested flavopiridol in an in vivo Matrigel™ model of angiogenesis and found that flavopiridol decreased blood vessel formation, which is a surrogate marker for antiangiogenic effect of this compound.96 Furthermore, as mentioned earlier, Melillo et al demonstrated that, at low nanomolar concentrations, flavopiridol prevented the induction of VEGF by hypoxic conditions in human monocytes.82 This effect was caused by a decreased stability of VEGF mRNA, which paralleled the decrease in VEGF protein. Thus, the antitumor activity of flavopiridol observed may be caused in part by antiangiogenic effects. Whether various antiangiogenic actions of flavopiridol result from its interaction with a CDK target or other targets requires further study.

Clinical Lung Cancer November 2003

161

Cyclin-Dependent Kinase Inhibitors in Lung Cancer Tumor Cell Differentiation. The antitumor effect observed with flavopiridol can also be explained by activation of differentiation pathways. It has become clear that cells become differentiated when exit of the cell cycle (G0) and loss of CDK2 activity occurs. Based on this information, Lee and colleagues tested flavopiridol and roscovitine, both known CDK2 inhibitors, to determine whether they induce a differentiated phenotype.97 For this purpose, NCI-H358 lung carcinoma cell lines were exposed to CDK2 antisense construct, flavopiridol, or roscovitine. Clear evidence of mucinous differentiation along with loss in CDK2 activity was observed in this lung carcinoma model. Thus, it is plausible that the antitumor effect of flavopiridol in lung carcinoma models may be a result of induction of differentiation, among other causes. Synergistic Effect with Chemotherapy. Several investigators have attempted to determine whether flavopiridol has synergistic effects with standard chemotherapeutic agents. For example, synergistic effects in A549 lung carcinoma cells were demonstrated when treatment with flavopiridol followed treatment with paclitaxel, cytarabine, topotecan, doxorubicin, or etoposide.98,99 In contrast, a synergistic effect was observed with 5-fluorouracil (5FU) only when cells were treated with flavopiridol for 24 hours before addition of 5-FU. Furthermore, synergistic effects with cisplatin were not schedule dependent.99 However, Chien et al failed to demonstrate a synergistic effect between flavopiridol and cisplatin and/or γ-irradiation in bladder carcinoma models.92 One important issue to mention is that most of these studies were performed in in vitro models. Thus, confirmatory in vivo studies in animal models are needed. Animal Studies. Experiments using colorectal (Colo205) and prostate (LnCaP/DU-145) carcinoma xenograft models in which flavopiridol was administered frequently over a protracted period demonstrated that flavopiridol is cytostatic.71,100 These demonstrations led to human clinical trials of flavopiridol administered as a 72-hour continuous infusion every 2 weeks101. Subsequent studies in human leukemia/lymphoma xenografts demonstrated that flavopiridol administered intravenously as a bolus rendered animals tumor free, whereas flavopiridol administered as an infusion only delayed tumor growth.85 Moreover, in a head and neck cancer xenograft (HN12), flavopiridol administered as an intraperitoneal bolus daily at 5 mg/kg for 5 days demonstrated substantial growth delay.91 Again, apoptotic cell death and cyclin D1 depletion were observed in tissues from xenografts treated with flavopiridol.85 Based on these results, a phase I trial of 1hour daily infusional flavopiridol every 3 weeks was conducted at the National Cancer Institute (NCI).102 Phase I Trial of 72-Hour Continuous Infusion of Flavopiridol. Two phase I clinical trials of flavopiridol administered as a 72-hour continuous infusion every 2 weeks have been completed.101,103 In the NCI phase I trial of infusional flavopiridol (N = 76), the dose-limiting toxicity (DLT) was diarrhea, with a maximum tolerated dose (MTD) of 50 mg/m2/day for 3 days. In the presence of antidiar-

162

Clinical Lung Cancer November 2003

rheal prophylaxis (a combination of cholestyramine and loperamide), patients tolerated higher doses and a second MTD of 78 mg/m2/day for 3 days was defined. The DLTs observed at the higher dose level were a substantial proinflammatory syndrome (fever, fatigue, local tumor pain, and modulation of acute phase reactants) and reversible hypotension.101 Minor responses were observed in patients with non-Hodgkin's lymphoma (NHL), colon cancer, and kidney cancer for > 6 months. Moreover, 1 patient with refractory renal cancer experienced a partial response for > 8 months. Of 14 patients who received flavopiridol for > 6 months, 5 patients received flavopiridol for > 1 year and 1 patient received flavopiridol for > 2 years. Plasma concentrations of 300-500 nmol/L flavopiridol, which inhibit CDK activity in vitro, were safely achieved during this trial. In a complementary phase I trial exploring the same schedule (72-hour continuous infusion every 2 weeks), Thomas et al found that the DLT was diarrhea, corroborating the NCI experience.103 Moreover, plasma concentrations of 300-500 nmol/L flavopiridol were also observed. Interestingly, there was one patient in this trial with refractory metastatic gastric cancer that progressed after a treatment regimen containing 5-FU. When treated with flavopiridol, this patient experienced a sustained complete response without any evidence of disease for > 2 years after treatment was completed. Phase I Trial of 1-Hour Flavopiridol Infusion for 5 Days. The first phase I trial of a daily 1-hour infusion of flavopiridol for 5 consecutive days every 3 weeks was recently completed.101,102 This schedule was based on antitumor results observed in leukemia/lymphoma and head and neck cancer xenografts treated with flavopiridol.85,91 A total of 55 patients were treated in this trial. The recommended phase II dose is 37.5 mg/m2 per day for 5 consecutive days. Dose-limiting toxicities observed at 52.5 mg/m2/day are nausea/vomiting, neutropenia, fatigue, and diarrhea.101,102 Other (non–dose-limiting) side effects are local tumor pain and anorexia. To reach higher flavopiridol concentrations, the protocol was amended to administer flavopiridol for 3 days and then for 1 day only every 3 weeks. With these protocol modifications, we were able to achieve concentrations necessary to induce apoptosis in xenograft models (approximately 4 μmol/L).85,91,102,104 Of note, the half-life observed in this trial was much shorter (approximately 3 hours) than that observed in the infusional trial (approximately 10 hours). Thus, the high micromolar concentrations achieved in the 1-hour infusional trial could be maintained for short periods of time.102,104 Several phase 2 trials in patients with refractory head and neck cancer, chronic lymphocytic leukemia (CLL), or mantle-cell lymphoma (MCL) are currently being tested using this schedule (described in later sections). A phase I trial testing the combination of paclitaxel and 24-hour infusional flavopiridol demonstrated good tolerability with a pulmonary DLT.105 Phase II Trials of 72-Hour Continuous Infusion of Flavopiridol. Current research includes phase II trials of flavopiridol given as a 72-hour continuous infusion in CLL, NSCLC, NHL, and colon, prostate, gastric, head and neck, and kidney cancers;

Adrian M. Senderowicz and phase I trials of flavopiridol administered on novel schedules and in combination with standard chemotherapeutic agents.84,106-109 In a recently published phase II trial of flavopiridol in metastatic renal cancer, 2 objective responses were observed (response rate, 6%; 95% CI, 1%-20%). Most patients developed grade 1/2 diarrhea and asthenia.108 In this trial, patients who demonstrated glucuronide flavopiridol metabolites in plasma as measured by high-performance liquid chromatography had less pronounced diarrhea than patients who demonstrated no glucuronide flavopiridol metabolites.110 Thus, it may be possible that patients with higher metabolic rates may tolerate higher doses of flavopiridol. Phase II trials of shorter infusions of flavopiridol (ie, 1 hour) are being conducted in MCL, CLL, and head and neck squamous cell carcinoma. Of interest, several patients with refractory CLL or MCL have demonstrated clear evidence of responses (partial responses) in these trials (J.R. Suarez, MD, oral communication, May 2002). Preclinical Studies of Flavopiridol in Lung Carcinoma Models. A panel of NSCLC cell lines expressing wild-type Rb protein and lacking p16INK4A were exposed to flavopiridol for 72 hours.89 Flavopiridol demonstrated cytotoxicity in all 7 cell lines, with evidence of apoptosis. As mentioned before, the induction of apoptosis was p53-independent, a clinical situation that occurs in ≥ 50% of solid tumors. Although cells in all phases are sensitive to flavopiridol, cells in S-phase are preferentially sensitive to this agent. In addition, cells are sensitized to flavopiridol following recruitment to S-phase, whether this is accomplished by synchronization or by treatment with noncytotoxic concentrations of chemotherapy agents that impose an S-phase delay.89 Combinations of gemcitabine or cisplatin, followed by flavopiridol at concentrations that correlate with CDK inhibition, produce sequence-dependent cytotoxic synergy. Thus, administration of flavopiridol after chemotherapy agents that cause S-phase accumulation may be an efficacious antitumor strategy. Another interesting feature observed with flavopiridol is the induction of differentiation in lung carcinoma models. Lee and colleagues tested whether flavopiridol and roscovitine, both known CDK2 inhibitors or antisense CDK2 constructs, block cell cycle progression in NCI-H358 lung carcinoma cell lines.97 Clear evidence of mucinous differentiation, along with loss in CDK2 activity, was observed in this lung carcinoma model. Clinical Studies of Flavopiridol in NSCLC. Based on the interesting results obtained in preclinical lung carcinoma models, a phase II trial in patients with metastatic lung carcinoma was performed.111 A total of 20 patients were treated with a 72-hour continuous infusion of flavopiridol every 14 days at a dosage of 50 mg/m2/day. The most common toxicities included grade ≤ 2 diarrhea, asthenia, and venous thromboses. Median overall survival for the 20 patients who received treatment was 7.5 months. Of note, the median survival of 7.5 months achieved with flavopiridol is similar to the range of 7.4-8.2 months reported recently in a randomized trial of 4 chemotherapy regimens containing platinum analogues in combination with tax-

anes or gemcitabine.112 Moreover, a similar overall survival was obtained with gefitinib, a recently approved EGFR inhibitor for the treatment of advanced lung cancer.113 Based on these encouraging results, a phase III trial comparing standard combination chemotherapy versus combination chemotherapy plus flavopiridol is currently under investigation.

UCN-01 Mechanism of Antiproliferative Activity. Staurosporine is a potent nonspecific protein and tyrosine kinase inhibitor with a very low therapeutic index in animals.114 Thus, efforts to find staurosporine analogues have identified compounds specific for protein kinases. One staurosporine analogue, UCN-01 (7hydroxystaurosporine), has potent activity against several protein kinase C isoenzymes, particularly the Ca2+-dependent protein kinase C, with an IC50 of approximately 30 nmol/L.52,115,116 In addition to its effects on protein kinase C, UCN-01 has antiproliferative activity in several human tumor cell lines.51,53,54,117,118 In contrast, another highly selective potent protein kinase C inhibitor, GF-109203X, has minimal antiproliferative activity, despite a similar capacity to inhibit protein kinase C in vitro.117 These results suggest that the antiproliferative activity of UCN-01 cannot be explained solely by inhibition of protein kinase C. Although UCN-01 moderately inhibited the activity of immunoprecipitated CDK1 (cdc2) and CDK2 (IC50, 300-600 nmol/L), exposure of UCN-01 to intact cells leads to “inappropriate activation” of the same kinases.117 This phenomenon correlates with the G2 abrogation checkpoint observed with this agent. Experimental evidence suggests that DNA damage leads to cell cycle arrest to allow DNA repair. In cells in which the G1-phase checkpoint is not active because of p53 inactivation, irradiated cells accumulate in G2-phase as a result of activation of the G2 checkpoint (inhibition of cdc2). In contrast, Wang et al exposed CA46 cell lines to radiation followed by UCN-01, promoting the inappropriate activation of cdc2/cyclin B and early mitosis with the onset of apoptotic cell death.119 These effects could be partially explained by the inactivation of wee1, the kinase that negatively regulates the G2/M-phase transition.120 Moreover, UCN-01 can have a direct effect on chk1, a protein kinase that regulates the G2 checkpoint.121-123 Thus, although UCN-01 at high concentrations can directly inhibit CDKs in vitro, UCN-01 can modulate cellular upstream regulators at much lower concentrations, leading to inappropriate CDC2 activation. Studies from other groups suggest that not only is UCN-01 able to abrogate the G2 checkpoint induced by DNA-damaging agents, but also, in some circumstances, it is able to abrogate the DNA damage–induced S-phase checkpoint.124,125 Cell Arrests in G1 Phase. Another interesting property of UCN-01 is its ability to arrest cells in G1-phase of the cell cycle.51,54,97,118,126-130 When human epidermoid carcinoma A431 cells (mutated p53) or HN12 head and neck carcinoma cell lines are incubated with UCN-01, these cells were arrested in G1-phase with Rb hypophosphorylation and p21waf1/p27kip1

Clinical Lung Cancer November 2003

163

Cyclin-Dependent Kinase Inhibitors in Lung Cancer accumulation.118,131 Chen et al suggested that Rb function, but not p53 function, is essential for UCN-01–mediated G1 arrest.129 However, Shimizu et al demonstrated that lung carcinoma cell lines with absent, mutant, or wild-type Rb exposed to UCN-01 displayed G1 arrest and antiproliferative effects irrespective of Rb function.128 Thus, the exact role of Rb or p53 in the G1 arrest induced by UCN-01 is still unknown. Further studies of the putative target(s) for UCN-01 in the G1-phase arrest of cells are warranted. Cytotoxicity in Tumor Cells with Mutated p53. Another interesting pharmacologic feature of UCN-01 is the observed increased cytotoxicity in cells that harbor mutated p53.119 In CA46 and HT-29 tumor cell lines carrying mutated p53 genes, potent cytotoxicity results following exposure to UCN-01. To further extend these observations, the MCF-7 cell line with no endogenous p53 because of the ectopic expression of E6, a human papilloma virus (HPV) type-16 protein, showed enhanced cytotoxicity when treated with a DNA-damaging agent, such as cisplatin, and UCN-01, compared with the isogenic wild-type MCF-7 cell line. Thus, a common feature observed in > 50% of human neoplasias that is associated with poor outcome and refractoriness to standard chemotherapies may render tumor cells more sensitive to UCN-01.132,133 Inhibition of PI3 Kinase/Akt Survival Pathway. A very exciting recent development is that of the effects of UCN-01 on the PI3 kinase/Akt survival pathway.134,135 UCN-01 displays a very potent inhibition in vitro of the phosphoinositide-dependent kinase 1 serine/threonine kinase, leading to dephosphorylation and inactivation of Akt.135 Although this is an exciting novel feature of UCN-01, it is of utmost importance to demonstrate whether the antitumor effects of UCN-01 are mediated by this action. Moreover, demonstration that these effects also occur in in vivo settings is crucial. Synergy with Chemotherapy. As previously mentioned, synergistic effects of UCN-01 have been observed with many chemotherapeutic agents, including mitomycin-C, 5-FU, carmustine, and camptothecin, among others.124,136-143 Therefore, it is possible that combining UCN-01 with these or other agents could improve the agents’ therapeutic indexes. Clinical trials exploring these possibilities are currently being developed. Animal Studies. UCN-01 administered by an intravenous or intraperitoneal route displayed antitumor activity in xenograft model systems with breast carcinoma (MCF-7 cells), renal carcinoma (A498 cells), and leukemia (MOLT-4 and HL-60 cells; A.S., unpublished data, 2003). The antitumor effect was greater when UCN-01 was given over a longer period of time. This requirement for a longer period of treatment was also observed in in vitro models, with the greatest antitumor activity observed when UCN-01 was present for 72 hours.51 Thus, a clinical trial using a 72-hour continuous infusion every 2 weeks was conducted.

164

Clinical Lung Cancer November 2003

Clinical Trials of UCN-01. The first phase I trial of UCN-01 was recently completed.101,144 UCN-01 was initially administered as a 72-hour continuous infusion every 2 weeks based on data from in vitro and xenograft preclinical models. However, it became apparent in the first few patients that the drug had an unexpectedly long half-life (approximately 30 days). This half-life was 100 times longer than the half-life observed in preclinical models, most likely because of the avid binding of UCN-01 to α1-acid glycoprotein.145,146 Thus, the protocol was modified to administer UCN-01 every 4 weeks (1 halflife), and on subsequent courses, the duration of infusion was decreased by half (total of 36 hours). Thus, it was possible to reach similar peak plasma concentrations in subsequent courses with no evidence of drug accumulation. There was no evidence of myelotoxicity or gastrointestinal toxicity, which were prominent side effects observed in animal models, despite very high plasma concentrations achieved (35-50 μmol/L).101,144-146 Dose-limiting toxicities were nausea/vomiting (amenable to standard antiemetic treatment), symptomatic hyperglycemia associated with an insulin-resistant state (increase in insulin and c-peptide levels while receiving UCN-01), and pulmonary toxicity characterized by substantial hypoxemia without obvious radiologic changes. The recommended phase II dosage of UCN-01 on a 72-hour continuous infusion schedule was 42.5 mg/m 2/day. 144 One patient with refractory metastatic melanoma showed a partial response that lasted 8 months. Another patient with refractory anaplastic large-cell lymphoma that had failed multiple chemotherapeutic regimens including high-dose chemotherapy had no evidence of disease 4 years after the initiation of UCN-01 therapy. Moreover, several patients with leiomyosarcoma, NHL, or lung cancer demonstrated stable disease for ≥ 6 months.144,147 In order to estimate free UCN-01 concentrations in body fluids, several efforts were considered. Plasma ultracentrifugation and salivary determination of UCN-01 revealed similar results. At the recommended phase II dosage (37.5 mg/m2/day over 72 hours), concentrations of free salivary UCN-01 that may cause G2 checkpoint abrogation (approximately 100 nmol/L) can be achieved. As mentioned earlier, UCN-01 is a potent protein kinase C inhibitor. In order to determine the putative signaling effects of UCN-01 in tissues, bone marrow aspirates and tumor cells were obtained from patients before and during the first cycle of UCN-01 administration. Western blot studies were performed in those samples against phosphorylated adducin, a cytoskeletal membrane protein, a specific substrate phosphorylated by protein kinase C.148 Clear loss in phosphoadducin content in the posttreatment samples was observed in all tumor and bone marrow samples tested, and the conclusion was drawn that UCN-01 could modulate protein kinase C activity in tissues from patients in this trial.144,147 Several groups are conducting shorter-duration (3 hours) infusional trials of UCN-01. Interestingly, the toxicity profiles of shorter infusions are similar to the toxicities observed in the 72hour infusion trials.149,150 However, with shorter infusions, more pronounced hypotension was observed. Determination of free UCN-01 in these trials is of utmost importance because

Adrian M. Senderowicz higher free concentrations for shorter periods may be more or less beneficial than the free concentrations observed in the 72hour infusion trial. Based on the unique pharmacologic features and anecdotal clinical evidence of synergistic effects in 1 patient with refractory disease,151 several combination trials with standard chemotherapeutic agents recently commenced. A phase I/II trial of gemcitabine followed by 72-hour infusional UCN-01 in CLL was started at the NCI. Other studies of UCN-01 in combination with cisplatin and 5-FU, among other agents, also commenced recently. Studies of UCN-01 in Preclinical Lung Carcinoma Models. The use of UCN-01 in lung cancer models was investigated by several groups, particularly in combination studies with DNAdamaging agents.127,128,130,152-159 Shimizu et al studied the cell cycle effects of UCN-01 in a large panel of NSCLC cell lines.128 Clear loss in Rb phosphorylation was associated with G1/S arrest in the majority of cells exposed to UCN-01. In another study, Usuda et al investigated the expression of cell cycle proteins in sensitive and resistant SB3 NSCLC clones.130 They observed that SB3-resistant clones have significantly lower levels of baseline and induced p21waf1/cip1, a known endogenous CDK inhibitor. They concluded that the lack of p21waf1/cip1 may result from loss of IRF-1, a protein that may be associated with modulation of p21waf1/cip1.130 Another group using A549 cells demonstrated that A549-resistant clones were not cross-resistant to doxorubicin, paclitaxel, staurosporine, and UCN-02, but they displayed significant resistance to cisplatin and mitomycin-C.160 The expression of cyclin A, cyclin B1, Rb, and CDK2 proteins were downregulated in the resistant clones. Moreover, the antiapoptotic protein bcl-2 was apparently upregulated in A549/UCN cells; however, bcl-xL, another antiapoptotic protein, was downregulated and there were no changes in bak and bax.160 Thus, multiple cell cycle and apoptotic proteins may be associated with UCN01 resistance in lung cancer models. Mack et al studied several NSCLC cell lines with different genotypes (ie, Rb and p53 mutant, null, or wild type) using the combination of cisplatin and UCN-01.158 Clear synergistic effects (apoptosis and G2 checkpoint abrogation) were observed when NSCLC cells were treated with cisplatin followed by UCN-01, particularly in cells with mutant p53, a situation that occurs in the majority of cases of NSCLC.158 Similar synergism was observed in another study using UCN-01 and 5-fluorodeoxyuridine in A431 human epidermoid cancer cells.159 Finally, Xiao et al combined UCN-01 with γ-irradiation in several wild-type and mutant p53 lung carcinoma models.157 They observed, as expected, that cells with loss in p53 resulting from transfection in the HPV E6 protein were more resistant to radiation. However, the same radiation-resistant cells were more sensitive to the combination of UCN-01 and radiation.157 Taken together, trials using a combination of DNA-damaging agents followed by UCN-01 (particularly in p53 mutant lung cancer) are very reasonable to consider. Clinical trials in several tumor types including lung cancer are being planned using cisplatin or γ-irradiation in combination with UCN-01.

Conclusion Based on the frequent aberration in cell cycle regulatory pathways in human cancer by CDK hyperactivation, novel ATPcompetitive CDK inhibitors are being developed. The first 2 tested in clinical trials, flavopiridol and UCN-01, showed promising results with evidence of antitumor activity and plasma concentrations sufficient to inhibit CDK-related functions. The best administration schedule, combination with standard chemotherapeutic agents, tumor types to be targeted, and demonstration of CDK modulation from tumor samples from patients in these trials are important issues that need to be answered in order to advance these agents to the clinic.

References 1. Sherr CJ. Cancer cell cycles. Science 1996; 274:1672-1677. 2. Morgan DO. Cyclin-dependent kinases: engines, clocks, and microprocessors. Annu Rev Cell Dev Biol 1997; 13:261-291. 3. Wang C, Fu M, Mani S, et al. Histone acetylation and the cell-cycle in cancer. Front Biosci 2001; 6:D610-629. 4. Yu JT, Foster RG, Dean DC. Transcriptional repression by RB-E2F and regulation of anchorage-independent survival. Mol Cell Biol 2001; 21:3325-3335. 5. Lees JA, Saito M, Vidal M, et al. The retinoblastoma protein binds to a family of E2F transcription factors. Mol Cell Biol 1993; 13:7813-7825. 6. DeGregori J, Leone G, Ohtani K, et al. E2F-1 accumulation bypasses a G1 arrest resulting from the inhibition of G1 cyclin-dependent kinase activity. Genes Dev 1995; 9:2873-2887. 7. Robles AI, Rodriguez-Puebla ML, Glick AB, et al. Reduced skin tumor development in cyclin D1-deficient mice highlights the oncogenic ras pathway in vivo. Genes Dev 1998; 12:2469-2474. 8. Rodriguez-Puebla ML, LaCava M, Conti CJ. Cyclin D1 overexpression in mouse epidermis increases cyclin-dependent kinase activity and cell proliferation in vivo but does not affect skin tumor development. Cell Growth Differ 1999; 10:467-472. 9. Weinberg RA. The molecular basis of carcinogenesis: understanding the cell cycle clock. Cytokines Mol Ther 1996; 2:105-110. 10. Motokura T, Arnold A. Cyclin D and oncogenesis. Curr Opin Genet Dev 1993; 3:5-10. 11. Jacks T, Weinberg RA. The expanding role of cell cycle regulators. Science 1998; 280:1035-1036. 12. Hatakeyama M, Weinberg RA. The role of RB in cell cycle control. Prog Cell Cycle Res 1995; 1:9-19. 13. Senderowicz AM. Cyclin-dependent kinases as new targets for the prevention and treatment of cancer. Hematol Oncol Clin North Am 2002; 16:1229-1253. 14. Hunter T, Pines J. Cyclins and cancer. II: cyclin D and CDK inhibitors come of age. Cell 1994; 79:573-582. 15. Bartek J, Staskova Z, Draetta G, et al. Molecular pathology of the cell cycle in human cancer cells. Stem Cells 1993; 11(suppl 1):51-58. 16. Bartkova J, Lukas J, Guldberg P, et al. The p16-cyclin D/Cdk4-pRb pathway as a functional unit frequently altered in melanoma pathogenesis. Cancer Res 1996; 56:5475-5483. 17. Bartkova J, Lukas J, Muller H, et al. Abnormal patterns of D-type cyclin expression and G1 regulation in human head and neck cancer. Cancer Res 1995; 55:949-956. 18. Aagaard L, Lukas J, Bartkova J, et al. Aberrations of p16Ink4 and retinoblastoma tumour-suppressor genes occur in distinct sub-sets of human cancer cell lines. Int J Cancer 1995; 61:115-120. 19. Lukas J, Bartkova J, Rohde M, et al. Cyclin D1 is dispensable for G1 control in retinoblastoma gene-deficient cells independently of cdk4 activity. Mol Cell Biol 1995; 15:2600-2611. 20. Lukas J, Aagaard L, Strauss M, et al. Oncogenic aberrations of p16INK4/CDKN2 and cyclin D1 cooperate to deregulate G1 control. Cancer Res 1995; 55:4818-4823. 21. Ichimura K, Schmidt EE, Goike HM, et al. Human glioblastomas with no alterations of the CDKN2A (p16INK4A, MTS1) and CDK4 genes have frequent mutations of the retinoblastoma gene. Oncogene 1996; 13:1065-1072. 22. Sonoda Y, Yoshimoto T, Sekiya T. Homozygous deletion of the MTS1/p16 and MTS2/p15 genes and amplification of the CDK4 gene in glioma. Oncogene 1995; 11:2145-2149. 23. Wolfel T, Hauer M, Schneider J, et al. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 1995; 269:1281-1284. 24. An HX, Beckmann MW, Reifenberger G, et al. Gene amplification and overexpression of CDK4 in sporadic breast carcinomas is associated with high tumor cell proliferation. Am J Pathol 1999; 154:113-118. 25. Masciullo V, Scambia G, Marone M, et al. Altered expression of cyclin D1 and CDK4 genes in ovarian carcinomas. Int J Cancer 1997; 74:390-395. 26. Kanoe H, Nakayama T, Murakami H, et al. Amplification of the CDK4 gene in sarcomas: tumor specificity and relationship with the RB gene mutation. Anticancer Res 1998; 18:2317-2321. 27. Meijer L. Cyclin-dependent kinases inhibitors as potential anticancer, antineurodegenerative, antiviral and antiparasitic agents. Drug Resist Update 2000; 3:83-88.

Clinical Lung Cancer November 2003

165

Cyclin-Dependent Kinase Inhibitors in Lung Cancer 28. Wistuba, II, Gazdar AF, Minna JD. Molecular genetics of small cell lung carcinoma. Semin Oncol 2001; 28:3-13. 29. Geradts J, Hu SX, Lincoln CE, et al. Aberrant RB gene expression in routinely processed, archival tumor tissues determined by three different anti-RB antibodies. Int J Cancer 1994; 58:161-167. 30. Cordon-Cardo C. Mutations of cell cycle regulators. Biological and clinical implications for human neoplasia. Am J Pathol 1995; 147:545-560. 31. Goodrich DW, Lee WH. Molecular characterization of the retinoblastoma susceptibility gene. Biochim Biophys Acta 1993; 1155:43-61. 32. Sugio K, Tsukamoto S, Ushijima C, et al. Clinical significance of the Rb expression in adenocarcinoma of the lung. Anticancer Res 2001; 21:1931-1935. 33. Lee YC, Chang YL, Luh SP, et al. Significance of P53 and Rb protein expression in surgically treated non-small cell lung cancers. Ann Thorac Surg 1999; 68:343-347; discussion 348. 34. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev 1995; 9:1149-1163. 35. Hirai H, Roussel MF, Kato JY, et al. Novel INK4 proteins, p19 and p18, are specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6. Mol Cell Biol 1995; 15:2672-2681. 36. Sherr CJ, DePinho RA. Cellular senescence: mitotic clock or culture shock? Cell 2000; 102:407-410. 37. Wong DJ, Paulson TG, Prevo LJ, et al. p16(INK4a) lesions are common, early abnormalities that undergo clonal expansion in Barrett's metaplastic epithelium. Cancer Res 2001; 61:8284-8289. 38. Pines J. Cyclin-dependent kinase inhibitors: the age of crystals. Biochim Biophys Acta 1997; 1332:M39-42. 39. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 1999; 13:1501-1512. 40. Sanchez-Cespedes M, Reed AL, Buta M, et al. Inactivation of the INK4A/ARF locus frequently coexists with TP53 mutations in non-small cell lung cancer. Oncogene 1999; 18:5843-5849. 41. Spanakis NE, Gorgoulis V, Mariatos G, et al. Aberrant p16 expression is correlated with hemizygous deletions at the 9p21-22 chromosome region in non-small cell lung carcinomas. Anticancer Res 1999; 19:1893-1899. 42. Gorgoulis VG, Zacharatos P, Kotsinas A, et al. Alterations of the p16-pRb pathway and the chromosome locus 9p21-22 in non-small-cell lung carcinomas: relationship with p53 and MDM2 protein expression. Am J Pathol 1998; 153:1749-1765. 43. Shapiro GI, Edwards CD, Kobzik L, et al. Reciprocal Rb inactivation and p16INK4 expression in primary lung cancers and cell lines. Cancer Res 1995; 55:505-509. 44. Senderowicz AM. Small molecule modulators of cyclin-dependent kinases for cancer therapy. Oncogene 2000; 19:6600-6606. 45. Senderowicz AM, Sausville EA. Preclinical and clinical development of cyclin-dependent kinase modulators. J Natl Cancer Inst 2000; 92:376-387. 46. Meijer L, Kim SH. Chemical inhibitors of cyclin-dependent kinases. Methods Enzymol 1997; 283:113-128. 47. Zaharevitz DW, Gussio R, Leost M, et al. Discovery and initial characterization of the paullones, a novel class of small-molecule inhibitors of cyclin-dependent kinases. Cancer Res 1999; 59:2566-2569. 48. De Azevedo WF, Leclerc S, Meijer L, et al. Inhibition of cyclin-dependent kinases by purine analogues: crystal structure of human cdk2 complexed with roscovitine. Eur J Biochem 1997; 243:518-526. 49. Senderowicz AM. The cell cycle as a target for cancer therapy: basic and clinical findings with the small molecule inhibitors flavopiridol and UCN-01. Oncologist 2002; 7(suppl 3):12-19. 50. Gray NS, Wodicka L, Thunnissen AM, et al. Exploiting chemical libraries, structure, and genomics in the search for kinase inhibitors. Science 1998; 281:533-538. 51. Seynaeve CM, Stetler-Stevenson M, Sebers S, et al. Cell cycle arrest and growth inhibition by the protein kinase antagonist UCN-01 in human breast carcinoma cells. Cancer Res 1993; 53:2081-2086. 52. Seynaeve CM, Kazanietz MG, Blumberg PM, et al. Differential inhibition of protein kinase C isozymes by UCN-01, a staurosporine analogue. Mol Pharmacol 1994; 45:1207-1214. 53. Akinaga S, Gomi K, Morimoto M, et al. Antitumor activity of UCN-01, a selective inhibitor of protein kinase C, in murine and human tumor models. Cancer Res 1991; 51:4888-4892. 54. Akinaga S, Nomura K, Gomi K, et al. Effect of UCN-01, a selective inhibitor of protein kinase C, on the cell-cycle distribution of human epidermoid carcinoma, A431 cells. Cancer Chemother Pharmacol 1994; 33:273-280. 55. Gray N, Detivaud L, Doerig C, et al. ATP-site directed inhibitors of cyclin-dependent kinases. Curr Med Chem 1999; 6:859-875. 56. Meijer L, Borgne A, Mulner O, et al. Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem 1997; 243:527-536. 57. Buquet-Fagot C, Lallemand F, Montagne MN, et al. Effects of olomoucine, a selective inhibitor of cyclin-dependent kinases, on cell cycle progression in human cancer cell lines. Anticancer Drugs 1997; 8:623-631. 58. Kitagawa M, Okabe T, Ogino H, et al. Butyrolactone I, a selective inhibitor of cdk2 and cdc2 kinase. Oncogene 1993; 8:2425-2432. 59. Schutte B, Nieland L, van Engeland M, et al. The effect of the cyclin-dependent kinase inhibitor olomoucine on cell cycle kinetics. Exp Cell Res 1997; 236:4-15. 60. Brooks EE, Gray NS, Joly A, et al. CVT-313, a specific and potent inhibitor of CDK2 that prevents neointimal proliferation. J Biol Chem 1997; 272:2920729211.

166

Clinical Lung Cancer November 2003

61. Kent LL, Hull-Campbell NE, Lau T, et al. Characterization of novel inhibitors of cyclin-dependent kinases. Biochem Biophys Res Commun 1999; 260:768-774. 62. Park S, Cheon J, Lee Y, et al. A specific inhibitor of cyclin-dependent protein kinases, CDC2 and CDK2. Mol Cell 1996; 6:679-683. 63. Rosania GR, Merlie J Jr, Gray N, et al. A cyclin-dependent kinase inhibitor inducing cancer cell differentiation: biochemical identification using Xenopus egg extracts. Proc Natl Acad Sci U S A 1999; 96:4797-4802. 64. Schultz C, Link A, Leost M, et al. Paullones, a series of cyclin-dependent kinase inhibitors: synthesis, evaluation of CDK1/cyclin B inhibition, and in vitro antitumor activity. J Med Chem 1999; 42:2909-2919. 65. Senderowicz AM. Flavopiridol: the first cyclin-dependent kinase inhibitor in human clinical trials. Invest New Drugs 1999; 17:313-320. 66. Walker DH. Small-molecule inhibitors of cyclin-dependent kinases: molecular tools and potential therapeutics. Curr Top Microbiol Immunol 1998; 227:149-165. 67. Senderowicz AM. Cyclin-dependent kinase modulators: a novel class of cell cycle regulators for cancer therapy. Cancer Chemother Biol Response Modif 2001; 19:165-188. 68. Mettey Y, Gompel M, Thomas V, et al. Aloisines, a new family of CDK/GSK-3 inhibitors. SAR study, crystal structure in complex with CDK2, enzyme selectivity, and cellular effects. J Med Chem 2003; 46:222-236. 69. Schoepfer J, Fretz H, Chaudhuri B, et al. Structure-based design and synthesis of 2benzylidene-benzofuran-3-ones as flavopiridol mimics. J Med Chem 2002; 45:17411747. 70. Ortega MA, Montoya ME, Zarranz B, et al. Pyrazolo[3,4-b]quinoxalines. A new class of cyclin-dependent kinases inhibitors. Bioorg Med Chem 2002; 10:21772184. 71. Sedlacek H, Czech J, Naik R, et al. Flavopiridol (L86-8275, NSC-649890), a new kinase inhibitor for tumor therapy. Int J Oncol 1996; 9:1143-1168. 72. Kaur G, Stetler-Stevenson M, Sebers S, et al. Growth inhibition with reversible cell cycle arrest of carcinoma cells by flavone L86-8275. J Natl Cancer Inst 1992; 84:1736-1740. 73. Losiewicz MD, Carlson BA, Kaur G, et al. Potent inhibition of CDC2 kinase activity by the flavonoid L86-8275. Biochem Biophys Res Commun 1994; 201:589-595. 74. Worland PJ, Kaur G, Stetler-Stevenson M, et al. Alteration of the phosphorylation state of p34cdc2 kinase by the flavone L86-8275 in breast carcinoma cells. Correlation with decreased H1 kinase activity. Biochem Pharmacol 1993; 46:1831-1840. 75. Carlson B, Pearlstein R, Naik R, et al. Inhibition of CDK2, CDK2 and CDK7 by flavopiridol and structural analogs. Proc Am Assoc Cancer Res 1996; 424. 76. Carlson BA, Dubay MM, Sausville EA, et al. Flavopiridol induces G1 arrest with inhibition of cyclin-dependent kinase (CDK) 2 and CDK4 in human breast carcinoma cells. Cancer Res 1996; 56:2973-2978. 77. De Azevedo WF Jr, Mueller-Dieckmann HJ, Schulze-Gahmen U, et al. Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase. Proc Natl Acad Sci U S A 1996; 93:2735-2740. 78. Gansauge S, Gansauge F, Ramadani M, et al. Overexpression of cyclin D1 in human pancreatic carcinoma is associated with poor prognosis. Cancer Res 1997; 57:16341637. 79. Fredersdorf S, Burns J, Milne AM, et al. High level expression of p27(kip1) and cyclin D1 in some human breast cancer cells: inverse correlation between the expression of p27(kip1) and degree of malignancy in human breast and colorectal cancers. Proc Natl Acad Sci U S A 1997; 94:6380-6385. 80. Michalides R, van Veelen N, Hart A, et al. Overexpression of cyclin D1 correlates with recurrence in a group of forty-seven operable squamous cell carcinomas of the head and neck. Cancer Res 1995; 55:975-978. 81. Carlson B, Lahusen T, Singh S, et al. Down-regulation of cyclin D1 by transcriptional repression in MCF-7 human breast carcinoma cells induced by flavopiridol. Cancer Res 1999; 59:4634-4641. 82. Melillo G, Sausville EA, Cloud K, et al. Flavopiridol, a protein kinase inhibitor, down-regulates hypoxic induction of vascular endothelial growth factor expression in human monocytes. Cancer Res 1999; 59:5433-5437. 83. Chao SH, Fujinaga K, Marion JE, et al. Flavopiridol inhibits P-TEFb and blocks HIV-1 replication. J Biol Chem 2000; 275:28345-28348. 84. Wright J, Blatner GL, Cheson BD. Clinical trials referral resource: clinical trials of flavopiridol. Oncology (Huntingt) 1998; 12:1018, 1023-1014. 85. Arguello F, Alexander M, Sterry JA, et al. Flavopiridol induces apoptosis of normal lymphoid cells, causes immunosuppression, and has potent antitumor activity in vivo against human leukemia and lymphoma xenografts. Blood 1998; 91:24822490. 86. Byrd JC, Shinn C, Waselenko JK, et al. Flavopiridol induces apoptosis in chronic lymphocytic leukemia cells via activation of caspase-3 without evidence of bcl-2 modulation or dependence on functional p53. Blood 1998; 92:3804-3816. 87. Parker BW, Kaur G, Nieves-Neira W, et al. Early induction of apoptosis in hematopoietic cell lines after exposure to flavopiridol. Blood 1998; 91:458-465. 88. Konig A, Schwartz GK, Mohammad RM, et al. The novel cyclin-dependent kinase inhibitor flavopiridol downregulates Bcl-2 and induces growth arrest and apoptosis in chronic B-cell leukemia lines. Blood 1997; 90:4307-4312. 89. Shapiro GI, Koestner DA, Matranga CB, et al. Flavopiridol induces cell cycle arrest and p53-independent apoptosis in non-small cell lung cancer cell lines. Clin Cancer Res 1999; 5:2925-2938. 90. Lahusen J, Loaiza-Perez A, Sausville E, et al. Flavopiridol-induced apoptosis is associated with p38 and MEK activation and is prevented by caspase and MAPK inhibitors. Proc Am Soc Clin Oncol 2000; 19:313 (Abstract #1991). 91. Patel V, Senderowicz AM, Pinto D Jr, et al. Flavopiridol, a novel cyclin-dependent

Adrian M. Senderowicz 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104.

105. 106. 107. 108. 109. 110. 111. 112. 113.

114. 115. 116. 117. 118.

119. 120.

kinase inhibitor, suppresses the growth of head and neck squamous cell carcinomas by inducing apoptosis. J Clin Invest 1998; 102:1674-1681. Chien M, Astumian M, Liebowitz D, et al. In vitro evaluation of flavopiridol, a novel cell cycle inhibitor, in bladder cancer. Cancer Chemother Pharmacol 1999; 44:81-87. Schrump DS, Matthews W, Chen GA, et al. Flavopiridol mediates cell cycle arrest and apoptosis in esophageal cancer cells. Clin Cancer Res 1998; 4:2885-2890. Bible KC, Kaufmann SH. Flavopiridol: a cytotoxic flavone that induces cell death in noncycling A549 human lung carcinoma cells. Cancer Res 1996; 56:4856-4861. Brusselbach S, Nettelbeck DM, Sedlacek HH, et al. Cell cycle-independent induction of apoptosis by the anti-tumor drug Flavopiridol in endothelial cells. Int J Cancer 1998; 77:146-152. Kerr JS, Wexler RS, Mousa SA, et al. Novel small molecule alpha v integrin antagonists: comparative anti-cancer efficacy with known angiogenesis inhibitors. Anticancer Res 1999; 19:959-968. Lee HR, Chang TH, Tebalt MJ III, et al. Induction of differentiation accompanies inhibition of Cdk2 in a non-small cell lung cancer cell line. Int J Oncol 1999; 15:161-166. Schwartz GK, Farsi K, Maslak P, et al. Potentiation of apoptosis by flavopiridol in mitomycin-C-treated gastric and breast cancer cells. Clin Cancer Res 1997; 3:1467-1472. Bible KC, Kaufmann SH. Cytotoxic synergy between flavopiridol (NSC 649890, L86-8275) and various antineoplastic agents: the importance of sequence of administration. Cancer Res 1997; 57:3375-3380. Drees M, Dengler WA, Roth T, et al. Flavopiridol (L86-8275): selective antitumor activity in vitro and activity in vivo for prostate carcinoma cells. Clin Cancer Res 1997; 3:273-279. Senderowicz AM, Headlee D, Stinson SF, et al. Phase I trial of continuous infusion flavopiridol, a novel cyclin-dependent kinase inhibitor, in patients with refractory neoplasms. J Clin Oncol 1998; 16:2986-2999. Tan AR, Headlee D, Messmann R, et al. Phase I clinical and pharmacokinetic study of flavopiridol administered as a daily 1-hour infusion in patients with advanced neoplasms. J Clin Oncol 2002; 20:4074-4082. Thomas JP, Tutsch KD, Cleary JF, et al. Phase I clinical and pharmacokinetic trial of the cyclin-dependent kinase inhibitor flavopiridol. Cancer Chemother Pharmacol 2002; 50:465-472. Senderowicz AM, O’Reilly E, Ilson D, et al. A Phase I trial of 1 hour infusion of flavopiridol (FLA), a novel cyclin-dependent kinase inhibitor flavopiridol in combination with paclitaxel in patients with advanced solid tumors. Proc Am Soc Clin Oncol 2000; 19:204a (Abstract #796). Schwartz GK, O'Reilly E, Ilson D, et al. Phase I study of the cyclin-dependent kinase inhibitor flavopiridol in combination with paclitaxel in patients with advanced solid tumors. J Clin Oncol 2002; 20:2157-2170. Werner J, Kelsen D, Karpeh M, et al. The cyclin-dependent kinase inhibitor flavopiridol is an active and unexpectedly toxic agent in advanced gastric cancer. Proc Am Soc Clin Oncol 1998; 17:234a (Abstract #896). Shapiro G, Patterson A, Lynch C, et al. A Phase II Trial of flavopiridol in patients with stage IV non-small cell lung cancer. Proc Am Soc Clin Oncol 1999; 18:522a (Abstract #2013). Stadler WM, Vogelzang NJ, Amato R, et al. Flavopiridol, a novel cyclin-dependent kinase inhibitor, in metastatic renal cancer: a University of Chicago Phase II Consortium study. J Clin Oncol 2000; 18:371-375. Bennett P, Mani S, O'Reilly S, et al. Phase II Trial of flavopiridol in metastatic colorectal cancer: preliminary results. Proc Am Soc Clin Oncol 1999; 18:277a (Abstract #1065). Innocenti F, Stadler W, Iyer L, et al. Flavopiridol-induced diarrhea is related to the systemic metabolism of flavopiridol to its glucuronide. Proc Am Soc Clin Oncol 2000; 19:179a (Abstract #694). Shapiro GI, Supko JG, Patterson A, et al. A phase II trial of the cyclin-dependent kinase inhibitor flavopiridol in patients with previously untreated stage IV non-small cell lung cancer. Clin Cancer Res 2001; 7:1590-1599. Schiller JH, Harrington D, Belani CP, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002; 346:92-98. Kris M, Natale N, Herbst R, et al. A phase II trial of ZD1839 ('Iressa') in advanced non-small cell lung cancer (NSCLC) patients who had failed platinum- and docetaxel-based regimens (IDEAL 2). Proc Am Soc Clin Oncol 2002; 21:292a (Abstract #1166). Tamaoki T. Use and specificity of staurosporine, UCN-01, and calphostin C as protein kinase inhibitors. Methods Enzymol 1991; 201:340-347. Takahashi I, Kobayashi E, Asano K, et al. UCN-01, a selective inhibitor of protein kinase C from Streptomyces. J Antibiot (Tokyo) 1987; 40:1782-1784. Takahashi I, Saitoh Y, Yoshida M, et al. UCN-01 and UCN-02, new selective inhibitors of protein kinase C. II. Purification, physico-chemical properties, structural determination and biological activities. J Antibiot (Tokyo) 1989; 42:571-576. Wang Q, Worland PJ, Clark JL, et al. Apoptosis in 7-hydroxystaurosporine-treated T lymphoblasts correlates with activation of cyclin-dependent kinases 1 and 2. Cell Growth Differ 1995; 6:927-936. Akiyama T, Yoshida T, Tsujita T, et al. G1 phase accumulation induced by UCN-01 is associated with dephosphorylation of Rb and CDK2 proteins as well as induction of CDK inhibitor p21/Cip1/WAF1/Sdi1 in p53-mutated human epidermoid carcinoma A431 cells. Cancer Res 1997; 57:1495-1501. Wang Q, Fan S, Eastman A, et al. UCN-01: a potent abrogator of G2 checkpoint function in cancer cells with disrupted p53. J Natl Cancer Inst 1996; 88:956-965. Yu L, Orlandi L, Wang P, et al. UCN-01 abrogates G2 arrest through a Cdc2-de-

121. 122. 123. 124.

125. 126.

127.

128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. 146. 147.

148.

pendent pathway that is associated with inactivation of the Wee1Hu kinase and activation of the Cdc25C phosphatase. J Biol Chem 1998; 273:33455-33464. Sarkaria JN, Busby EC, Tibbetts RS, et al. Inhibition of ATM and ATR kinase activities by the radiosensitizing agent, caffeine. Cancer Res 1999; 59:4375-4382. Graves PR, Yu L, Schwarz JK, et al. The Chk1 protein kinase and the Cdc25C regulatory pathways are targets of the anticancer agent UCN-01. J Biol Chem 2000; 275:5600-5605. Busby EC, Leistritz DF, Abraham RT, et al. The radiosensitizing agent 7-hydroxystaurosporine (UCN-01) inhibits the DNA damage checkpoint kinase hChk1. Cancer Res 2000; 60:2108-2112. Shao RG, Cao CX, Shimizu T, et al. Abrogation of an S-phase checkpoint and potentiation of camptothecin cytotoxicity by 7-hydroxystaurosporine (UCN-01) in human cancer cell lines, possibly influenced by p53 function. Cancer Res 1997; 57:4029-4035. Bunch RT, Eastman A. 7-Hydroxystaurosporine (UCN-01) causes redistribution of proliferating cell nuclear antigen and abrogates cisplatin-induced S-phase arrest in Chinese hamster ovary cells. Cell Growth Differ 1997; 8:779-788. Akiyama T, Shimizu M, Okabe M, et al. Differential effects of UCN-01, staurosporine and CGP 41 251 on cell cycle progression and CDC2/cyclin B1 regulation in A431 cells synchronized at M phase by nocodazole. Anticancer Drugs 1999; 10:67-78. Kawakami K, Futami H, Takahara J, et al. UCN-01, 7-hydroxyl-staurosporine, inhibits kinase activity of cyclin-dependent kinases and reduces the phosphorylation of the retinoblastoma susceptibility gene product in A549 human lung cancer cell line. Biochem Biophys Res Commun 1996; 219:778-783. Shimizu E, Zhao MR, Nakanishi H, et al. Differing effects of staurosporine and UCN-01 on RB protein phosphorylation and expression of lung cancer cell lines. Oncology 1996; 53:494-504. Chen X, Lowe M, Keyomarsi K. UCN-01-mediated G1 arrest in normal but not tumor breast cells is pRb-dependent and p53-independent. Oncogene 1999; 18:5691-5702. Usuda J, Saijo N, Fukuoka K, et al. Molecular determinants of UCN-01-induced growth inhibition in human lung cancer cells. Int J Cancer 2000; 85:275-280. Patel V, Lahusen T, Leethanakul C, et al. Antitumor activity of UCN-01 in carcinomas of the head and neck is associated with altered expression of cyclin D3 and p27(KIP1). Clin Cancer Res 2002; 8:3549-3560. Marchetti A, Buttitta F, Merlo G, et al. p53 alterations in non-small cell lung cancers correlate with metastatic involvement of hilar and mediastinal lymph nodes. Cancer Res 1993; 53:2846-2851. Lowe SW, Bodis S, Bardeesy N, et al. Apoptosis and the prognostic significance of p53 mutation. Cold Spring Harb Symp Quant Biol 1994; 59:419-426. Testa JR, Bellacosa A. AKT plays a central role in tumorigenesis. Proc Natl Acad Sci U S A 2001; 98:10983-10985. Sato S, Fujita N, Tsuruo T. Interference with PDK1-Akt survival signaling pathway by UCN-01 (7-hydroxystaurosporine). Oncogene 2002; 21:1727-1738. Akinaga S, Nomura K, Gomi K, et al. Enhancement of antitumor activity of mitomycin C in vitro and in vivo by UCN-01, a selective inhibitor of protein kinase C. Cancer Chemother Pharmacol 1993; 32:183-189. Bunch RT, Eastman A. Enhancement of cisplatin-induced cytotoxicity by 7-hydroxystaurosporine (UCN-01), a new G2-checkpoint inhibitor. Clin Cancer Res 1996; 2:791-797. Hsueh CT, Kelsen D, Schwartz GK. UCN-01 suppresses thymidylate synthase gene expression and enhances 5-fluorouracil-induced apoptosis in a sequence-dependent manner. Clin Cancer Res 1998; 4:2201-2206. Husain A, Yan XJ, Rosales N, et al. UCN-01 in ovary cancer cells: effective as a single agent and in combination with cis-diamminedichloroplatinum(II)independent of p53 status. Clin Cancer Res 1997; 3:2089-2097. Pollack IF, Kawecki S, Lazo JS. Blocking of glioma proliferation in vitro and in vivo and potentiating the effects of BCNU and cisplatin: UCN-01, a selective protein kinase C inhibitor. J Neurosurg 1996; 84:1024-1032. Tsuchida E, Urano M. The effect of UCN-01 (7-hydroxystaurosporine), a potent inhibitor of protein kinase C, on fractionated radiotherapy or daily chemotherapy of a murine fibrosarcoma. Int J Radiat Oncol Biol Phys 1997; 39:1153-1161. Sugiyama K, Shimizu M, Akiyama T, et al. UCN-01 selectively enhances mitomycin C cytotoxicity in p53 defective cells which is mediated through S and/or G(2) checkpoint abrogation. Int J Cancer 2000; 85:703-709. Jones CB, Clements MK, Wasi S, et al. Enhancement of camptothecin-induced cytotoxicity with UCN-01 in breast cancer cells: abrogation of S/G(2) arrest. Cancer Chemother Pharmacol 2000; 45:252-258. Sausville EA, Arbuck SG, Messmann R, et al. Phase I trial of 72-hour continuous infusion UCN-01 in patients with refractory neoplasms. J Clin Oncol 2001; 19:2319-2333. Sausville EA, Lush RD, Headlee D, et al. Clinical pharmacology of UCN-01: initial observations and comparison to preclinical models. Cancer Chemother Pharmacol 1998; 42(suppl):S54-59. Fuse E, Tanii H, Kurata N, et al. Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human alpha1-acid glycoprotein. Cancer Res 1998; 58:3248-3253. Senderowicz AM, Headlee D, Lush R, et al. Phase I trial of infusional UCN-01, a novel protein kinase inhibitor, in patients with refractory neoplasms. In: Proceedings of the 10th National Cancer Institute–European Organization for Research on Treatment of Cancer Symposium; June 1998; Amsterdam, The Netherlands. Page 78. Fowler L, Dong L, Bowes RC III, et al. Transformation-sensitive changes in expression, localization, and phosphorylation of adducins in renal proximal tubule epithe-

Clinical Lung Cancer November 2003

167

Cyclin-Dependent Kinase Inhibitors in Lung Cancer lial cells. Cell Growth Differ 1998; 9:177-184. 149. Tamura T, Sasaki Y, Minami H, et al. Phase I study of UCN-01 by 3-hour infusion. Proc Am Soc Clin Oncol 1999; 18:159a (Abstract #611). 150. Dees E, O'Reilly S, Figg W, et al. A phase I and pharmacologic study of UCN-01, a protein kinase C inhibitor. Proc Am Soc Clin Oncol 2000; 19:205a (Abstract #797). 151. Wilson WH, Sorbara L, Figg WD, et al. Modulation of clinical drug resistance in a B cell lymphoma patient by the protein kinase inhibitor 7-hydroxystaurosporine: presentation of a novel therapeutic paradigm. Clin Cancer Res 2000; 6:415-421. 152. Courage C, Budworth J, Gescher A. Comparison of ability of protein kinase C inhibitors to arrest cell growth and to alter cellular protein kinase C localisation. Br J Cancer 1995; 71:697-704. 153. Courage C, Snowden R, Gescher A. Differential effects of staurosporine analogues on cell cycle, growth and viability in A549 cells. Br J Cancer 1996; 74:1199-1205. 154. Courage C, Bradder SM, Jones T, et al. Characterisation of novel human lung carcinoma cell lines selected for resistance to anti-neoplastic analogues of staurosporine. Int J Cancer 1997; 73:763-768. 155. Akiyama T, Sugiyama K, Shimizu M, et al. G1-checkpoint function including a cy-

168

Clinical Lung Cancer November 2003

156. 157. 158. 159. 160.

clin-dependent kinase 2 regulatory pathway as potential determinant of 7-hydroxystaurosporine (UCN-01)-induced apoptosis and G1-phase accumulation. Jpn J Cancer Res 1999; 90:1364-1372. Mack PC, Gandara DR, Bowen C, et al. RB status as a determinant of response to UCN-01 in non-small cell lung carcinoma. Clin Cancer Res 1999; 5:2596-2604. Xiao HH, Makeyev Y, Butler J, et al. 7-Hydroxystaurosporine (UCN-01) preferentially sensitizes cells with a disrupted TP53 to gamma radiation in lung cancer cell lines. Radiat Res 2002; 158:84-93. Mack PC, Gandara DR, Lau AH, et al. Cell cycle-dependent potentiation of cisplatin by UCN-01 in non-small-cell lung carcinoma. Cancer Chemother Pharmacol 2003; 51:337-348. Grem JL, Danenberg KD, Kao V, et al. Biochemical and molecular effects of UCN01 in combination with 5-fluorodeoxyuridine in A431 human epidermoid cancer cells. Anticancer Drugs 2002; 13:259-270. Sugiyama K, Akiyama T, Shimizu M, et al. Decrease in susceptibility toward induction of apoptosis and alteration in G1 checkpoint function as determinants of resistance of human lung cancer cells against the antisignaling drug UCN-01 (7-hydroxystaurosporine). Cancer Res 1999; 59:4406-4412.