You are on page 1of 32

Critical Reviews in Oncology/Hematology 38 (2001) 139– 170

www.elsevier.com/locate/critrevonc

Review
Mechanisms of action of flavopiridol
H.H. Sedlacek *
A6entis Pharma Deutschland GmbH, Central Biotechnology, P.O. Box 1140, 35001 Marburg, Germany

Received 28 February 2000; received in revised form 30 September 2000; accepted 23 October 2000

Contents
1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140

2. Inhibition of kinases in relation to intracellular concentration of flavopiridol . . . . . . . . . . 141

3. Function and inhibition of cyclin dependent kinases . . . . . . . . . . . . . . . . . . . . . . . . 142


3.1. *Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.2. Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144

4. Characteristics of the antiproliferative activity of flavopiridol . . . . . . . . . . . . . . . . . . . 145


4.1. Induction of apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
4.2. Interaction with cytostatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
4.3. Activity in resting cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
4.4. Activity in relation to pRb expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
4.5. Activity in relation to p53 expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
4.6. Activity in relation to Bcl-2 expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152

5. Inhibition of other kinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154


5.1. Interaction with receptors for the EGF family . . . . . . . . . . . . . . . . . . . . . . . . . 154
5.2. Inhibition of receptor associated protein kinases . . . . . . . . . . . . . . . . . . . . . . . . 155
5.3. Inhibition of protein kinase C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156

6. Modulation of multidrug resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158

7. Antiangiogenic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159

8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

Biography. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170

* Tel.: +49-6421-392768; fax: + 49-6421-394663.


E-mail address: hans-harald.sedlacek@aventis.com (H.H. Sedlacek).

1040-8428/01/$ - see front matter © 2001 Elsevier Science Ireland Ltd. All rights reserved.
PII: S 1 0 4 0 - 8 4 2 8 ( 0 0 ) 0 0 1 2 4 - 4
140 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

Abstract

Flavopiridol inhibits phosphokinases. Its activity is strongest on cyclin dependent kinases (cdk-1, -2, -4, -6, -7) and less on
receptor tyrosine kinases (EGFR), receptor associates tyrosine kinases (pp60 Src) and on signal transducing kinases (PKC and
Erk-1). Although the inhibiting activity of flavopiridol is strongest for cdk, the cytotoxic activity of flavopiridol is not limited to
cycling cells. Resting cells are also killed. This fact suggests that inhibition of cdks involved in the control of cell cycle is not the
only mechanism of action. Inhibition of cdk’s with additional functions (i.e. involved in the control of transcription or function
of proteins that do not control cell cycle) may contribute to the antitumoral effect. Moreover, direct and indirect inhibition of
receptor activation (EGFR) and/or a direct inhibition of kinases (pp60 Src, PKC, Erk-1) involved in the signal transduction
pathway could play a role in the antiproliferative activity of flavopiridol. From pharmacokinetic data in patients it can be
concluded that the inhibitory activity (IC50) of flavopiridol on these kinases is in the range of concentrations that might be
achieved intracellularly after systemic application of non-toxic doses of flavopiridol. However, no in situ data from flavopiridol
treated cells have been published yet that prove that by inhibition of EGFR, pp60 Src, PKC and/or Erk-1 (in addition to
inhibition of cdk’s) flavopiridol is able to induce apoptosis. Thus many questions regarding the detailed mechanism of antitumoral
action of flavopiridol are still open. For the design of protocols for future clinical studies this review covers the essential
information available on the mechanism of antitumoral activity of flavopiridol. The characteristics of this antitumoral activity
include: High rate of apoptosis, especially in leukemic cells; synergy with the antitumoral activity of many cytostatics;
independence of its efficacy on pRb, p53 and Bcl-2 expression; lack of interference with the most frequent multidrug resistance
proteins (P-glycoprotein and MRP-190); and a strong antiangiogenic activity. Based on these pharmacological data it can be
concluded that flavopiridol could be therapeutically active in tumor patients: independent on the genetic status of their tumors or
leukemias (i.e. mutations of the pRb and/or p53, amplification of bcl-2 ); in spite of drug resistance of their tumors induced by first
line treatment (and caused by enhanced expression of multidrug resistance proteins); in combination with conventional
chemotherapeutics preferentially given prior to flavopiridol; and due to a complex mechanism involving cytotoxicity on cycling
and on resting tumor cells, apoptosis and antiangiogenic activity. In consequence, flavopiridol is a highly attractive, new
antitumoral compound and deserves further elucidation of its clinical potency. © 2001 Elsevier Science Ireland Ltd. All rights
reserved.

Keywords: cdk; Phospokinases; Inhibitors; Flavopiridol; Mechanism of action; Apoptosis; Cell cycle; Multidrug resistance; Antiangiogenic activity

1. Introduction cdk7. The broadness of its activity on the different


cdk’s and the strengths of its activity on cdk-4 has been
The new compound flavopiridol (L868 275, regarded as unique [2].
NSC649 890, HMR1275) was selected for further stud- The essential role of cdk’s in driving cells through the
ies [1,2] because of: cell cycle [10] implies that flavopiridol should act on
1. Its high potency to inhibit the proliferation of a cycling cells via inhibition of cdk’s. Several groups
broad range of human tumor cell lines after pro- could confirm this assumption, others, however, found
longed exposure time that flavopiridol is also cytotoxic for resting cells.
2. Its potency to inhibit tyrosine kinases and serine To explain this discrepancy many experiments were
kinases [1,3] performed in order to elucidate the mechanism of ac-
3. The discrepancies between its high degree of cytoxi- tion of flavopiridol. Tests were thus conducted to estab-
city and
lish whether the cytotoxic activity of flavopiridol is
4. Its potency to inhibit known kinases as well as the
dependent on pRb expression or on the expression of
lack of correlation between its cytoxicity and the
other proteins being involved in cell survival, i.e. p53 or
sensitivity of the respective test cells to growth fac-
Bcl-2 or on the cell line being investigated, its func-
tors and
5. Its potency to inhibit in vivo a broad type range of tional state or any other experimental condition.
human tumors, leukemias and lymphomas [1,2,4 –8]. The overall results of these experiments indicate that
In view of its strong antitumoral activity the question the mechanism of action of flavopiridol seems to be
of the mechanism of action of flavopiridol was first more complex than predicted and to depend on various
taken up in 1991 [2,9]. It was speculated [1] that inhibi- experimental conditions including the type of the target
tion of both types of kinases, i.e. tyrosine as well as cells chosen.
serine phosphokinases may synergistically contribute to Clinical studies with flavopiridol have already been
the strong antiproliferative potency of flavopiridol. started and clinical tumor responses could be observed
In addition it was found that flavopiridol inhibits the in most of the phase I [11 –13] and phase II studies [14]
cell cycle and is a strong inhibitor of cyclin dependent on different types of progressive tumors refractory to
kinases (cdk’s), including cdk1, cdk2, cdk4, cdk6 and conventional treatment.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 141

However, another clinical phase II study failed to bind to intracellular and extracellular lipids and
show any effect of flavopiridol on metastatic renal lipoproteins.
carcinoma [15]. Pharmacokinetic studies in mice and rats [2] showed
The reason for this discrepancy is unknown yet, but that after i.v. injection plasma levels of flavopiridol
it could be that the dose and treatment schedule applied decline quickly (T1/2a of 16 min (mice) and 10.5 min
may not have been appropriate in view of the complex (rats)). However, the elimination of flavopiridol from
mechanisms of action of flavopiridol. Thus the aim of the blood plasma was prolonged (T1/2b of 201 min
this review is to report in a critical manner all the (mice) and 112 min (rats)) and the volume of distribu-
various suggested mechanisms so that the reader may tion at steady state (Vdss) was relatively high (8.3 l/kg
be able to design further clinical studies by taking the (rats)) [2]. The short T1/2a and the high Vdss value in
complexity of the mechanisms of action of flavopiridol rodents suggest a high distribution of the drug into
into account. extravascular compartments [17].
Clinical phase I studies in tumor patients, however,
showed non-homogenous pharmacokinetic results.
Most ( : 70%) patients exhibited a course of blood
2. Inhibition of kinases in relation to intracellular
plasma elimination of flavopiridol similar to the one
concentration of flavopiridol
found in mice and rats with a relatively long half-life
T1/2b of 11.6 (range 1.3 –29.1 h) and a high Vdss of
Mammalian protein kinases include receptor kinases,
131.16 l/m2. However, some (approximately about one
receptor associated protein kinases, signal transducing
third) of patients showed aberrant pharmacokinetic
kinases and cyclin dependent kinases [16].
profiles that included postinfusion single or saw tooth
Flavopiridol has a proven inhibiting activity on a transient elevations of flavopiridol blood plasma con-
variety of such cellular protein kinases (see Table 1) [2]. centrations [12].
The potency of this inhibiting activity, however, differs The median maximal steady state plasma flavopiridol
among the different types of kinases. Moreover, the concentration (Cpss) in patients was 0.27 (range 0.17 –
inhibition of which kinase at which concentration is 2.94) mM (after the administration of the maximal
relevant for the antitumoral activity in vitro and in tolerated dose of 50 mg/m2/dx3) and 0.55 (range 0.36 –
vivo, particularly in patients, is unknown. One way to 4.21) mM (after infusion of the toxic dose low (TDL:
answer this question is to estimate the concentration of 122.5 mg/m2/dx3) administered together with antidi-
flavopiridol in the extravascular compartments after arrheal prophylaxis (Loperamide) and causing hypoten-
systemic application. As flavopiridol is not only soluble sion as the dose limiting toxicity) [12,13,18].
in water but also slightly to sparingly soluble in etha- In another phase I study the mean Cpss after the
nol, n-octanol or methanol [2], it can be expected that administration of 56 mg/m2/dx3 was 0.469 0.16 mM
flavopiridol is able to penetrate cell membranes and to [19].

Table 1
Activity of Flavopiridol on cellular kinases

Family Kinase IC50 (mM) Ref.

50.3 B40 ]40

EGF receptor family EGF receptor (TPK) 21–25 Sedlacek et al. [1]
Losiewicz et al. [81]
Receptor associated src family kinases pp60 Src 34 Czech et al. [3]
In T-Ly Lck 50 Czech et al. [3]
In Mo/PMN Hck 500 Czech et al. [3]
In T/B-Ly Fyn 500 Czech et al. [3]
Signal transducing kinases PKA 122–145 Sedlacek et al. [1]
PKC 6 Losiewicz et al. [81]
Czech et al. [5]
Erk-1 16 Webster et al. [320]
Cyclin dependent kinases (direct inhibition) cdk1/cyclin B 0.03–0.4 Losiewicz et al. [81]
cdk2/cyclin A 0.1 Carlson et al. [86]
cdk2/cyclin E 0.1 Carlson et al. [87]
cdk4/cyclin D 0.02–0.04 Carlson et al. [86]
cdk6/cyclin D 0.06 Singh et al. [88]
cdk7/cyclin H 0.11–0.3 Carlson et al. [36]
142 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

The high distribution volume of flavopiridol at steady Using these parameters it can be concluded that
state (Vdss) in rats and patients indicates that the distri- flavopiridol might be active in vivo via inhibition of
bution of flavopiridol leads to a higher concentration of Cdk’s, PKC, EGFR, pp60 Src and Erk-1 but probably
flavopiridol in the extravascular compartments com- not via inhibition of Lck, Hck, Fyn and PKA (see Table
pared to blood plasma. As the concentration in blood 1).
plasma of flavopiridol 12 h after infusion into patients
was very similar to the concentration found in their
pleura or pericardial fluids (dose 16 mg/m2/x3: 59 nmol/l 3. Function and inhibition of cyclin dependent kinases
in plasma; 57 nmol/l in pleura; dose 122.5 mg/m2/x3: 528
nmol/l in plasma; 409 nmol/l in pleura) [12] it can be 3.1. Function
concluded that the concentration of flavopiridol in the
interstitial (i.e. extravascular and extracellular) compart-
Cyclin dependent kinases consist of a catalytic subunit
ments is similar to that found in the plasma. In conse-
(cdk) and a regulatory subunit (cyclin). More than eight
quence, it can be assumed that the high Vdss is mainly
human cdk’s have been described so far. The most
caused by an easy cell membrane passage possibly
important ones involved in the cell division cycle are:
leading to an enrichment of flavopiridol in the intracel-
lular compartment. However, no data on intracellular cdk1 (=cdc2), cdk2, cdk4, cdk5, cdk6, cdk7 and cdk8.
concentrations of flavopiridol are available yet. Indeed, All of these cdk’s are regulated by the transient associ-
such data respresent a cornerstone in the discussion ation with one member of the cyclin family including
concerning potentially relevant mechanisms of action of cyclin A, cyclin B1 –B3, cyclin D1 –D3, cyclin E, cyclin
flavopiridol and should be performed as early as possible. H and cyclin C. Other cdk1-related kinases have been
As the intracellular concentration of flavopiridol is sequenced and await identification of their regulatory
unknown the area under the blood plasma concentration partners and cell cycle regulatory functions [10,20].
curve (AUC) is taken as a surrogate parameter. The Progression through each step of the cell cycle is
AUC should reflect the amount of flavopiridol ex- thought to be regulated by cdk-complexes. The impor-
changed between the extravascular compartments and tant ones are:
blood over time. The AUC for flavopiridol varied
considerably between the individual patients and dosage
groups and was in the range between 16.8 mM h (after cdk4/cyclin D (1–3), Mid G1
infusion of 50 mg/m2/dx3), 38.3 mM h (after infusion of cdk6/cyclin D (1–3)
78 mg/m2/dx3; this dose is the MTD of flavopiridol plus cdk2/cyclin E Late G1
antidiarrheal prophylaxis) and 144 mM h (after infusion phase
of the TDL of 122.5 mg/m2/dx3, i.e. the highest dose that cdk2/cyclin E, cdk2/cyclin A G1/S
was administered) [12]. transition
Taking these pharmacokinetic data into consideration cdk2/cyclin A S-phase
the following concentrations can be chosen as thresholds cdk1/cyclin B, cdk1/cyclin A G2/M
to decide whether the in vitro data of flavopiridol may transition
have some relevance for antitumoral activity in vivo.
Concentrations that are achievable intracellularly in
vivo:
Cyclin-dependent kinases are for their part regulated
With 50.3 mM Mean Cpss in patients af-
through ‘check-point control’ at defined restriction
certainty ter administration of the
points that ensure that each step of the cell cycle is
MTD (50–56 mg/m2/dx3)
completed before the cell can proceed through the next
With some 55.0 mM Mean Cpss in patients af-
step [21,22].
probability ter administration of the
TDL under antidiarrheal The mechanisms regulating cyclin-dependent kinases
therapy (122.5 mg/m2/ [10,20,23 –29] include:
dx3) 1. Post-translational modifications by activating en-
With low 540 mM Derived from the AUC zymes (cdc25C phosphatase) and inhibitory enzymes
probability in patients after adminis- (wee1 kinase);
tration of the MTD un- 2. Formation of cyclin complexes;
der antidiarrheal therapy 3. Association with inhibitory subunits (e.g. p16INK4A,
(78 mg/m2/dx3) p15INK4B, p21Cip1, p27Kip1, p18 (for cdk4, -2, and
Probably not \40 mM -1)), some of them (p16, p15, p18) inhibiting by
displacing the cyclin subunit.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 143

Fig. 1. Inhibition of cdk’s and cell cycle progression.

4. Association with regulatory subunits of unknown, during cell cycle seems to have an essential role in the
yet essential functions (p9CDShs, p15cdkBP). pathogenesis of diverse tumor diseases. Cyclins D1, D2,
5. Ubiquitin-dependent degradation of the cyclin A and E have been reported to be overexpressed in
subunit; many different types of cancer [45 –50] and to be corre-
6. Intracellular translocations and association with lated directly and quantitatively with tumor aggressive-
subcellular structures (centrosomes, microtubules). ness and invasiveness [47,51]. Cdk1, cdk4 or cdk7 have
The regulatory sites of cyclin-dependent kinases in- been found to be overexpressed or mutated in several
clude the ATP-binding pocket in the catalytic subunit tumors [46,49,52,53]. Likewise p16 or p15 were found
of the cdk/cyclin complexes that contain two amino to be deleted, mutated or aberrantly methylated in
acids that inhibit kinase activity when phosphorylated many tumor types, both in vitro and in vivo [49,54,55].
[30 – 32]. The protein substrate binding site is located in Main cellular substrates for cyclin dependent kinases
the cleft between the two lobes of the catalytic subunit. are proteins of the retinoblastoma protein family, i.e.
Its phosphorylation by cdk7/cyclin H appears to be pRb, p107 and p130 (see Fig. 1). In the hypoposphory-
essential for kinase activity [33 – 36]. lated state all three so-called pocket proteins are capa-
In addition cdk7/cyclin H complexes are also inti- ble of inhibiting growth in certain cell types by forming
mately associated with the transcriptional machinery: complexes with and inhibiting a number of partner
specifically, they are found within the basal transcrip- molecules (see Fig. 1). The functional inactivation of
tion factor TFIIH in mammalian cells. Cdk7/cyclin H is pocket proteins takes place by hyperphosphorylation
constitutively active and capable of phosphorylating the [56 –61]. Loss-of-function mutations of pocket proteins,
carboxy-terminal domain of RNA polymerase II [37]. i.e. of pRb are very common in small cell lung cancer
Cyclin D molecules have redundant cell functions, [62] and other tumor types [63 –66], indicating their
however, their tissue expression and regulation are very function as tumor suppressors [58,67 –74].
different: cyclin D1 is more prevalent in epithelial tis- pRb and p130 are detected in resting cells as low
sues whereas cyclins D2 and D3 are more prevalent in phosphorylated forms. When cells progress through the
hematopoietic tissues [38]. G0/G1 transition and through mid G1, p130 is hyper-
The expression of cyclin D1 in normal cells varies phosphorylated and levels of hyperphosphorylated
with the phase of the cell cycle with a peak in early G1 p130 are downregulated as cells move to S phase
followed by a decline during S- and G2-M phase. The [58,67 –74]. In constrast very little-to-no p107 is de-
expression of cyclin D is regulated by transcriptional tected in quiescent cells. However, p107 accumulates
and translational mechanisms [39]. In contrast, the significantly as cells progress through the cell cycle
expression of most of the other cyclins (i.e. cyclin B1, [67,75,76].
A, E) is regulated by degradation processes [40 –44]. Cdk’s phosphorylate multiple sites (primarily serine
The excessive, aberrant or inappropriate expression threonine sites) in pRb and most likely in p107 and
of cdk’s and/or cyclins and their regulatory proteins p130 [76]. Phosphorylation of pRb, p107 and p130 is
144 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

associated with the loss of their growth suppressive This result is in marked contrast to other growth
activities. Such activities are mediated through the asso- inhibitory agents such as nitrogen mustard, methotrex-
ciation of pocket proteins with a number of cellular ate, camptothecin or etoposide that are known to in-
proteins including the members of the E2F family duce a preserved or increased tyrosine phosphorylation
(E2F-1, -2, -3, -4, -5) of transcription factors and other of cdk1 [84].
transcription factors involved in cell proliferation (i.e. Control experiments including Western blot on cdk1
c-Jun, ATF-2, Sp1, AhR, TF II D, c-Myc, N-Myc), immunoprecipitates showed that the relative mass of
transcription factors controlling expression of RNA cdk1 was not altered during the time of flavopiridol
polymerases (TFIIIB, UBF)), tissue specific transcrip- exposure (in contrast to cdk1 protein reduction after
tion factors involved in the differentiation of myocytes, exposure to protein synthesis inhibitors emetine and
adipocytes and epithelial cells, cdk-subunits (cyclin cycloheximide) during which phosphorylation and cdk1
D1 – 3, cyclin A, E) and inhibitors of the tumor sup- kinase activity were decreased. Furthermore, high doses
pressor p53 (MDM-2) [76]. of flavopiridol did not inhibit [32S]-methionine incorpo-
For phosphorylation D-type cyclin/cdk complexes ration into immunoprecipitated cdk1 or into proteins in
interact with all three pocket proteins [45,77,78], while whole cell lysates [83].
cyclin E and cyclin A/cdk2 complexes form stable In addition, no decrease of cyclin B was detected by
complexes with p107 and p130, but not with pRb. an anti-cyclin B antibody in cdk1 immunoprecipitates
For inactivation of the growth suppressor activity of after flavopiridol treatment of aphidocolin synchro-
pRb, phosphorylation of its Ser-795 by cdk4/cyclin D1 nized cells [83]. Thus it may be concluded that flavopiri-
complexes is essential [79]. dol inhibits phosphorylation and kinase activity of
Cdk also phosphorylate E2F. Phosphorylation by cdk1/cyclin B but does not alter the protein level of
cdk4/cyclin (D1 –3) in the G1 phase prevents interac- either protein by a specific or a global effect on protein
tion of E2F with and inhibition by pocket proteins
synthesis.
whereas hyperphosphorylation of E2F by cdk2/cyclin A
Using breast carcinoma cells (the same as used for
complexes in the late S-phase leads to its inactivation
the cdk1 experiments) synchronized through block in
[21,22].
the M-phase by nocodazole [85 –87] and using specific
antisera to cdk2 it was revealed that cdk2 was likewise
3.2. Inhibition
directly inhibited by flavopiridol. Phosphoamino acid
analysis showed both threonine and tyrosine phospho-
Activity of flavopiridol is strongest on cdk’s (see
rylation of cdk2 to be abrogated in the presence of
Table 1). Flavopiridol directly inhibits purified acti-
flavopiridol [82,86,87]. Thus, it could be concluded that
vated cdk1/cyclin B complexes from sea star and to the
flavopiridol inhibits the normal phosphorylation and
same extent human cdk1.
activity of cdk2 and by this mechanism prevents S-
This could be proven by experiments in which the
kinase activity of cdk1 antibody immunoprecipitates phase entry of cycling cells (see Fig. 1).
(obtained (at 9–12 h) after release of breast carcinoma Moreover, using immune complex kinase assays (sim-
cells from aphidicolin block of DNA polymerase [8] ilar to cdk1 and cdk2) for cdk4 and cdk7, flavopiridol
into drug-free medium when the cells are in late G2 or was shown to inhibit both enzymes very efficiently (see
passing into M-phase) was potently inhibited by Table 1) [86,87].
flavopiridol. Neither autophosphorylation of cdk1 ki- In tumor cells known to lack cdk4, flavopiridol in-
nase nor any ATPase activity was detected [9,80 –82]. hibited the activity of cdk6 [88] in exponentially grow-
Inhibition of cdk1 by flavopiridol could be reduced by ing cells. Such cdk4 deficient cells synchronized at
an excessive concentration of ATP [81]. G0/G1 by serum starvation were unable to progress to
Analysis of cdk1 kinase activity in cells synchronized S phase after serum stimulation when flavopiridol was
at the G1/S transition by aphidicolin and treated with present. This G1 arrest also coincided with dephospho-
flavopiridol revealed inhibition of cdk1 kinase activity rylation of the cdk6 (and cdk4) specific phosphoryla-
relative to control activity at every time in the S-, G2- tion site of pRb, detected by a specific antibody [88].
or M-phase and concomitantly retardation in cell cycle Altogether, it can be concluded that flavopiridol can
progression through S-phase and accumulation in G2 inhibit cell cycle progression by a variety of mecha-
[83]. nisms related to cdk inactivation (see Fig. 1) including:
Cdk1 immunoprecipitates isolated from such syn- 1. Inhibition of cdk4/cyclin D and cdk6/cyclin D lead-
chronized and flavopiridol-treated cells either by a C- ing to
terminal specific or by a PSTAIRE motif specific (a) Inhibition of phosphorylation of pRb, p107
antiserum showed a decrease in [32P]-orthophosphate and p130, which induces binding to their part-
labeling, which indicates that the activity of those ki- ner molecules and results in inhibition of their
nases phosphorylating cdk1 (i.e. cdk7/cyclin H) is also transcriptional activity (see Fig. 1)
inhibited by flavopiridol [83]. (b) Inhibition of activation of E2F and
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 145

(c) Stop in G1 progression and cdk2, respectively, and the buried surface for
2. Inhibition of cdk2/cyclin E and cdk2/cyclin A lead- L868 276 amounts to 75% of the buried surface in cdk2
ing to [92].
(a) Inhibition of further phosphorylation and inac- Flavopiridol has a chlorophenyl instead of the phenyl
tivation of pRb, p107 and p130 in the L868 276 molecule. This modification increases
(b) Inhibition of inactivation of E2F (by cdk2/cy- the kinase inhibition by a factor of about 10, probably
clin A) and due to the new possible contacts that the chlorine
(c) Retardation in cell cycle progression through makes with residues Leu10, Phe82 and Leu93, increasing
the S-phase and accumulation in G1 and G2 the total number of contacts between flavopiridol and
3. Inhibition of cdk1/cyclin A and cdk1/cyclin B lead- enzymes to 61 [92].
ing to When cytosolic soluble tumor cell extracts were
(a) Inhibition of activation of topoisomerase acti- tested for proteins binding to immobilized flavopiridol
vators, of lamin proteoglycans histone 1 it was revealed [94] that cdk’s (i.e. cdk1, -2, -4, -7) bind
protein and chromatin condensation and only weakly (possibly for steric reasons, because
(b) Retardation in cell cycle progression through flavopiridol via its hydroxy groups was bound to Sep-
S-phase and accumulation in G2 phase harose 6B and one of the hydroxy groups of flavopiri-
4. Inhibition of cdk7/cyclin H complexes leading to dol mediates contacts with the enzyme [92]) when ATP
(a) Reduced activation of other cdk’s (i.e. cdk4/cy- was absent but not in its presence. In contrast several
clin D, cdk1/cyclin B) other proteins including cytosolic aldehyde dehydroge-
(b) Reduced activation of RNA polymerase nase class-1 (ALDH-1) could be identified to bind
In addition to cdk7 some other cdk’s, i.e. cdk8 and strongly to flavopiridol. ALDH-1 did not metabolize
cdk9 have been proposed to participate directly in the flavopiridol and flavopiridol did not inhibit the enzy-
control of transcription and activation of RNA-poly- matic activity of ALDH-1. Nevertheless, the intracellu-
merase [89,90]. Inhibition of such cdk’s by flavopiridol lar level of ALDH-1 correlated with the resistance of
may directly influence expression of cyclin D1 and D3 tumor lung cells to flavopiridol [94].
[91]. Thus, there might exist still unknown intracellular
To understand how flavopiridol binds to and inhibits binding partners for flavopiridol. Noteworthy in this
cyclin dependent kinases the X-ray structure of cdk2 in context is the recent finding that flavopiridol is able to
complex with the deschlorophenyl derivative L868 276 decrease the stability of mRNA encoding VEGF in
of flavopiridol was determined and compared with the mononuclear blood cells [95].
X-ray structure of the cdk2-ATP-complex [92].
The deschlorophenyl flavopiridol derivative L868 276
is about ten times less active on cdk’s compared to 4. Characteristics of the antiproliferative activity of
flavopiridol. flavopiridol
Binding of L868 276 to cdk2 is characterized by
predominantly hydrophobic and van der Waals interac- 4.1. Induction of apoptosis
tions with the same hydrophobic enzyme residues that
form the pocket for the adenine base in the ATP As has already been outlined (Section 3.1) expression
complex structure [93]. However, there are more con- and function of cyclin dependent kinases and their
tacts between the enzyme and the benzopyran ring of substrates are regulated and controlled in a very com-
deschloro flavopiridol (34 contacts) than were seen plex way [10,20,76,96 –98]. In cancer cells this control is
between the enzyme and the adenine ring of ATP (26 obviously dysregulated either by loss of proapoptotic
contacts). Many of the contacts between L868 276 and proteins or by gain of functions of proteins inhibiting
cdk2 are made by only three residues, Ile10, Leu83 and apoptosis [99,100].
Leu134, which form a total of 24 contacts, correspond- Flavopiridol seems to interfer with this dysregulated
ing to 43% of the observed contacts. Two of the cdk2 system in various ways not only by directly inhibiting
residues in contact with phenyl rings, His84 and Lys89, cdk’s involved in cell cycle but also by inhibiting the
are unique to the cdk2-L868 276 complex [92]. expression of cyclin D1, cyclin D3 and possibly as a
The shape complementarity of the contact surfaces of consequence of this activity by reducing expression of
both partners is described as solvent-accessible surfaces cdk4 [91].
that become buried upon ligand binding. In the cdk2- In consequence, by inhibition of cdk’s flavopiridol
ATP complex the buried areas of ATP (352 A, 2) and may reduce or block function of antiapoptotic, cell
cdk2 (435 A, 2) show a close fit. ATP is almost com- cycle promoting proteins, enabling their antagonists to
pletely inaccessible to solvent and its buried surface inhibit cell proliferation, to induce differentiation [101]
amounts to 80% of the buried surface in cdk2. Corre- and/or to induce apoptosis. All these phenomena may
sponding values are 301 A, 2 and 399 A, 2 for L868 276 contribute to the considerable cytotoxicity, shown for
146 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

flavopiridol [1,2]. Thus as shown in Fig. 1, by inhibition and/or annexin V binding) after 6–48 h in vitro expo-
of phosphorylation of the pocket proteins pRb, p107 sure to 0.1 –0.4 mM flavopiridol varied considerably (see
and p130, flavopiridol is able to give rise to hypophos- Table 2). Most sensitive to flavopiridol’s activity to
phorylated active pocket proteins able to bind to and to induce apoptosis are human leukemia cells independent
inhibit cyclins (cyclin D, A, E), transcription factors of their origin (cultured cells, fresh patient’s isolates)
involved in promoting cell cycle (E2F, c-Jun, Myc) and and type (B-cell type, T-cell type) followed by head and
nuclear proteins (i.e. MDM-2) inhibiting oncogene sup- neck tumors, breast carcinomas and non-small cell lung
pressor products (p53). Inhibition of all these proteins carcinomas. Flavopiridol even induced apoptosis in
has been shown to promote apoptosis [102 – 106]. In those tumor cells that were insensitive to apoptosis due
addition, inhibition of cdk2/cyclin A by flavopiridol to DNA damaging agents (g-irradiation and bleomycin)
inhibits phosphorylation and inactivation of E2F in the [108,7]. Induction of apoptosis by flavopiridol was ac-
late S-phase of the cell cycle that may lead to apoptosis companied by activation of caspase 3 and increase of
[107]. cleavage products of its substrate poly (ADP-ribose)
As expected flavopiridol is able to induce apoptosis polymerase (PARP) (see Table 3).
of tumor cells and normal cells in vitro (see Table 2) as In contrast to tumor cells, normal cells of different
well as of tumor cells in vivo [108,7]. The percentage of types showed a considerable variation in their sensitiv-
cells showing signs of apoptosis (chromatin condensa- ity to flavopiridol (see Table 2). Whereas human pe-
tion, internucleosomal cleavage, DNA fragmentation ripheral lymphocytes died after exposure to flavopiridol

Table 2
Induction of apoptosis in cell lines by Flavopiridol

Numbers Flavopiridol Number of cell lines with apoptosis (% of Ref.


(mM) apoptotic cells)

B10 10–30 \30–60 \60–90 \90

Tumor cells
T cell leukemia 3 0.1 3 Bible and Kaufmann [133];
Parker et al. [186]
B cell leukemia 1 0.3 1 Guedez et al. [137]
(mantel)
1 0.01–0.1 1 Parker et al. [186], Kitada et al.
[187]
3 0.4 3 König et al. [320]
(EBV+)
12 0.1–0.2 7 5 Byrd et al. [109], Kitada et al.
patients [187]
Head+neck 5 0.4 5 Patel et al. [108,7]
Breast 2 0.3 1 1 Schwartz et al. [119]
1 (low Erb–2) 0.3 1 Li et al. [216]
2 (high Erb–2) 0.3 2
NSC lung 7 0.1–0.5 7 Kaur et al. [9]; Shapiro et al.
[134,321–323]
Prostate 1 0.2 1 Parker et al. [186]
Gastric 2 0.3 1 1 Schwartz et al. [119]; Werner et
al. [324]
Oesophageal 4 0.1 1 3 Schrump et al. [135]
Bladder 2 1.0 1 1 Chien et al. [136]
Normal cells
Peripheral 9 0.1–0.2 9 Byrd et al. [109]
lymphocytes patients
Endothelial 1 0.1 1 Brüsselbach et al. [110]
(HUVEC)
Cortical neurons 2 1.0 2 Giovanni et al. [111]; Stefanis et
(rat) al. [127]
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 147

Table 3
Activation of caspase by Flavopiridol

Number Flavopiridol PARPa cleavage Ref.


products

Exposure time Concentration Caspase-3


(h) (mM)

Tumor cells
B-CLL 3 pat. 4–2 0.18 ++ (]4 h) ++ (]24 h) Byrd et al. [109]
NSC-lung 1 4 0.1–1.0 n.d. ++ Kaur et al. [80]
7 \4 0.1–0.5 ++++ Shapiro et al. [323]
Breast 1 24 0.3 ++ ++ Motwani et al. [116,117]
3 24 0.3 ++ ++ Li et al. [216]
Gastric 1 24 0.3 ++ ++ Motwani et al. [116,117]
Normal cells
Endothelial 1 12 0.5 n.d. ] 50% of total PARP Brüsselbach et al. [110]
(HUVEC)

a
PARP, poly(ADP-ribose) polymerase.

to a similar high degree as human leukemic cells [109], 4.2. Interaction with cytostatics
endothelial cells were less sensitive than most of the
tumor cells [110]. Apoptosis of human endothelial cells The percentage of apoptotic cells as well as the
was accompanied by increase of caspase 3 and increase degree of cytotoxicity could significantly be increased
of cleavage products of its substrate PARP [110]. Sur- by treatment of tumor cells with cytostatic drugs (taxol,
prisingly, the induction of apoptosis by flavopiridol in mitomycin, doxorubicin, etoposide, cisplatin, cytara-
endothelial cells was independent of expression of cy- bine or 5FU) 24 h prior to flavopiridol treatment
clin dependent kinases (cdk1, cdk2), which suggests a [113 –121] (see Table 4).
mechanism of action beyond simple inhibition of cdk’s Simultaneous treatment showed a significant syner-
[110]. gistic effect only with antimetabolites (5FU, cytarabine)
In addition, embryonic cortical neurons are quite [113,118 –121] and flavopiridol treatment 24 h prior to
resistant to treatment with flavopiridol alone [111]. treatment with cytostatics still showed synergistic ef-
No indications exist that the flavopiridol mediated fects with antimetabolites but abrogated considerably
antiproliferative activity is more selective for trans- the cytotoxic effect of taxol.
formed cells per se than for normal cells. Thus lung All the cytotoxic drugs used in these combination
cancer cells, diploid lung fibroblasts and normal human experiments are known to induce apoptosis of tumor
bronchial epithelial cells respond similarly to flavopiri- cells in all phases of the cell cycle [122]. However, most
dol [112]. sensitive to apoptosis induced by cytotoxic drugs seem
However, cycling transformed cells including tumor to be cells in the G2/M-phase [123]. The mechanism of
cells, especially those in the S-phase, undergo apoptosis the cell cycle independent apoptotic effect of cytostatics
much earlier and to a higher degree than the corre- seems to include damage of mitochondria, as measured
sponding non-transformed cells. Recruitment of tumor by the mitochondrial inner transmembrane potential
cells to S-phase can be done by treatment with cytostat- [122].
ics (see Section 4.2). The mechanism of S-phase sensi- Flavopiridol’s mechanism of action in synergy with
tivity and selectivity of transformed cells may be related cytostatic drugs seems to be different dependent on the
to cyclin A-cdk2 inhibition by flavopiridol which, as drug. Damage of mitochondria could be one of the
has already been pointed out in Section 3.1, results in preconditions for promoting apoptosis by the subse-
inappropriately persistant E2F activity. In transformed quent treatment with flavopiridol. For instance in pacli-
cells, in which the E2F activity is already high, the taxel treated cells, flavopiridol is able to enhance
additional E2F activity may lead to cell death [112]. activation of caspases, thus intensifying paclitaxel in-
In conclusion flavopiridol is able to induce apoptosis duced apoptosis [124]. Another precondition could be
in tumor cells as well as in normal cells. Proliferating the effect of cytostatic drugs on the expression of cdk’s.
transformed cells seem to be more sensitive to flavopiri- Paclitaxel-induced apoptosis is preceded by a transient
dol than proliferating normal cells. Inhibition of cdk’s mitotic block with elevation of cyclin B1 expression,
by flavopiridol seems to be the prominent, but not the cdk1 kinase activity and MPM-2 reactivity, followed by
only mechanism of action. a decrease in these markers, hypophosphorylated Rb
148 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

and induction of G1 markers but without cytokinesis Synergy with flavopiridol has also been observed
[124]. By inhibition of cdk1 kinase flavopiridol may with irinotecan. Exposure of human cancer cells to the
accelerate mitotic exit after paclitaxel therapy and active metabolite of irinotecan induced p21 expression
hereby enhance apoptosis. but no apoptosis, whereas subsequent treatment with
In contrast, when administered together or before flavopiridol reduced p21 expression, inhibition of cdk1
paclitaxel, flavopiridol could override the paclitaxel ef- and cdk2 and apoptosis [126].
fect of cdk1 kinase activation but may inhibit entry into The effect of flavopiridol on enhancing mitomycin C
M-phase, a prerequisite for paclitaxel-induced apopto- induced apoptosis seems to have quite a different mech-
anism. This synergic effect can at least partially be
sis [124].
reversed by pretreating the cells with the PKC activator
An other way of action of flavopiridol in synergy
PMA under conditions proven to be associated with
with cytostatic drugs could be its direct interaction with
activation of PKC. Thus, in combination with mito-
DNA, which may cause death of cells pretreated with mycin C, flavopiridol mediates its synergistic effect at
cytostatics [125]. least partly by inhibition of PKC [115 –117,120].
The synergy in cytotoxicity of paclitaxel and PKC has been proposed to be involved in phospho-
flavopiridol seems to offer an attractive clinical treat- rylation and activation of the P-glycoprotein, one of the
ment opportunity, i.e. a patient with prostate car- drug transporter proteins, which can be overexpressed
cinoma, who failed prior paclitaxel treatment, in drug resistant cells (see Section 6).
significantly responded to the combination of paclitaxel However, pretreatment of tumor cells with flavopiri-
and flavopiridol [11]. dol does not result in a synergism with most cytostatics

Table 4
Synergistic effect of Flavopiridol with cytostatics on tumor cells

Tumor cells Cytostatic (day 0) Flavopiridol treatmenta Ref.

+24 h 0 −24 h

NSC lung
Cytoarabine +++ 0 (+) Bible and Kaufmann [113]
A549 5-FU ++ + +++
Doxorubicin + (+) (+) 0
BCNU ++ 0 (+)
Etoposide ++ (+) (+)
Paclitaxel + − −
A549 Paclitaxel (−) n.d. 0 Edelman et al. [114]
Calu-1 Paclitaxel ++ n.d. ++
Gastric
MKN-74 Mitomycin +++ n.d. + Schwartz et al. [120]
Paclitaxel +++ − − Motwani et al. [117]
Paclitaxel +++ n.d. + Motwani and Schwartz [115]
Cisplatin +++ n.d. n.d. Motwani and Schwartz [116,117]; Werner et al. [324]
Gemcitabine +++ n.d. n.d. Jung et al. [325]
Pancreatic
Capan-2 Gemcitabine ++ n.d. 0 Jung et al. [236]
Colon
Hct 116 Irinotecan (active ++ n.d. n.d. Motwani and Schwartz [125]
metabol.)
Breast
MDA-MB-468 Mitomycin n.d. n.d. ++ Motwani and Schwartz [115–117]; Schwartz et al. [120]
MCF-7 Paclitaxel ++ (−) Motwani et al. [117]
MDA-MB-468 Paclitaxel ++ (−)
O6arian
Ov429, Ov432, SKOv-3, Cisplatin ++ Makhija et al. [160]
SKOv-3R
Osteosarcoma
Sa OS-2 Doxorubicin ++ Li et al. [138]
Sa Os-2/9 Doxorubicin 0

a
In relation to treatment with cytostatics.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 149

(see Table 4). Thus it seems unlikely that inhibition of 4.3. Acti6ity in resting cells
PKC by flavopiridol could reduce expression and acti-
vation of P-glycoprotein. The strong inhibiting activity on nearly all cdk/cyclin
An additional aspect of the synergism between cyto- complexes involved in cell cycle progression led to the
static drugs and flavopiridol is the prevention of en- assumption that flavopiridol is mainly or even exclu-
doreduplication and/or apoptosis. sively active on cycling cells and that the cytotoxicity of
Breast and colon carcinoma cells with intact G1-S flavopiridol might be caused by inhibition of cdk/cyclin
check points (p53 + / + , pRb + / + ) arrest in G1 with 4n complexes in cycling cells. Direct as well as indirect (via
DNA content in response to spindle inhibitors like cdk7/cyclin H) inhibition of cdk/cyclin complexes in
paclitaxel, whereas cells with defective G1-S check- cycling cells should lead to reduced phosphorylation of
points (p5 − / − , pRb − / − ) endoreduplicate, become E2F as well as of the pocket proteins leading to in-
creased binding to and inhibition of E2F, other tran-
polyploid and display dysregulated and persistent acti-
scription factors and nuclear proteins involved in cell
vation of cdk1/cyclin B1 and cdk2/cyclin E kinase
proliferation. It is assumed that such a blockage of
activity. Surprisingly, flavopiridol prevents the dysregu-
proteins involved in the cell cycle process could force
lated and persistent activation of cdk kinases, en-
the cycling cells, especially tumor cells, into pro-
doreduplication and development of polyploids when grammed cell death. If so, resting cells should be rela-
administered after paclitaxel. Thus flavopiridol could tively resistant to flavopiridol.
have the potential to prevent the occurrence of neo- Several experiments (see Table 5) indeed demon-
plasia [127]. strated that after exposure times of 24–28 h cycling
Neuronal cells undergo apoptosis when treated with tumor cells are much more sensitive for flavopiridol
B-amyloid protein (ABP) or camptothecin [111,128]. than the same density-arrested cells [9,86,87,110].
Surprisingly, however, subsequent treatment with However, in contrast to the expectations according to
flavopiridol did not enhance apoptosis (as shown for the proposed mechanism of action of flavopiridol, it
tumor cells) but prevented loss of cytochrome C and could also be shown both by using the same as well as
mitochondrial transmembrane potential as well as cas- different tumor cell lines that resting tumor cells ex-
pase activation and processing [128] and provided a hibited the same extent of sensitivity during a 24 h
long-term rescue from death [111,128]. In comparison, exposure to flavopiridol as proliferating cells
general caspase inhibitors rescued neurons from this [109,110,133,134] (see Table 5).
rapid apoptotic death but had no effect on mitochon- Exposure of cells for a longer period of time (i.e. for
drial damage and delayed death. As cdk’s are known to 3 days or longer) increased the cytotoxicity by flavopiri-
participate in death of neurons induced by DNA dam- dol significantly [1,5,86,87,110] and considerably re-
aging agents [129 –132] the protective effect of flavopiri- duced cell-type or laboratory-related differences in
dol for neuronal cells can be attributed to its potency to cytotoxicity of flavopiridol between resting and prolif-
inhibit cdk’s, i.e. cdk4 and cdk6, to inhibit hyperphos- erating tumor cells [9,110,133].
phorylation of pRb and p107 and to inhibit the tran- Surprisingly, normal endothelial cells (HuVEC) were
scriptional activity of E2F/Dp [111]. sensitive to flavopiridol, irrespective whether they were
In conclusion, flavopiridol shows a synergistic effect arrested by density or methionine deprivation or
with cytostatics on tumor cells, prevents development whether they were proliferating or their expression of
cyclin A,-B, cdk1, cdk2, cdc25C, pRb or p27 was high
of polyploid DNA content and protects neuronal cells
or low [110]. In contrast, embryonic cortical neurons
from apoptosis induced by cytotoxic compounds.
were resistant to flavopiridol [111].
An explanation of this discrepancy could be that
These observations raise the question of whether the
dependent on whether the target cell is cycling or not,
mechanism that leads to cytotoxicity after flavopiridol
the effect of flavopiridol might be different. Tumor cells exposure is related to the cell cycle of the target cells
are programmed to proliferate and undergo apoptosis and to inhibition of cdk’s involved in cell cycle regula-
after exposure to flavopiridol and inhibition of the tion. The possibility that flavopiridol-induced cell death
cdk’s expressed during cycling. In contrast, neuronal involves other cellular targets must be considered [133].
cells are more programmed to be resting cells. DNA In this respect it is interesting to note that flavopiridol
damage in such resting cells stimulates expression of binds to genomic DNA to similar extent as ethidium
cdk’s, which induce apoptosis if they are not inhibited, bromide and that the equilibrium dissociation constant
i.e. by flavopiridol. of the flavopiridol –DNA-complex is in the same range
However, we also have to consider that the mecha- observed for binding of the intercalators doxorubicin to
nisms, underlying apoptosis and cytotoxicity induced DNA [124]. However, results of these studies also
by flavopiridol in synergy with cytostatics, could be showed that flavopiridol does not inhibit topoisomerase
much more complex. I and II.
150 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

Table 5
Cell cycle dependence of the cytotoxicity of Flavopiridol

Flavopiridol Method for synchronizing IC50 (mM) Cell cycle Ref.


exposure time cells dependence
Resting cells Proliferating cells

Tumor cells
Breast 2 days Density arrest \0.3 – + Kaur et al. [80]
(MDA468)
6 days No 0.1 Carlson et al. [86]
24 h Aphidicolin (S-phase) 0.2
Nocodazole (M-phase) 0.2
Lung (A549) 24 h Density arrest :10.0 0.1 + Brüsselbach et al.
[110]
3 days Density arrest 0.6
24 days Starvation 0.3 0.3 − Shapiro et al.
[134,323]
24 h Density arrest 0.25 0.25 − Bible and
Kaufmann [133]
B-CLL (7 24 h No (Facs-analysis: DNA 0.18 0.18 − Byrd et al. [109]
patients) contents)
Normal cells
Endothelial cells 24 h Density arrest 0.1 0.09 − Brüsselbach et al.
(HuVEC) Methionine deprivation 0.08 [110]
Cortical neurons No \1.0 − Giovanni et al.
[111]

4.4. Acti6ity in relation to pRb expression osteosarcoma cell lines lacking Rb protein were more
sensitive to the proapoptotic effect of taxol derivative in
If cytotoxicity of flavopiridol on tumor cells is mainly combination with flavopiridol than osteosarcoma cells
caused by inhibition of cdk’s, hypophosphorylation of transduced to express the Rb protein [138]. These ob-
pocket proteins and inhibition of E2F and other tran- servations are consistent with other data that have
scriptional activators involved in cell cycle, cells with demonstrated that Rb expression enhances resistance to
loss of function of pRb should be less sensitive to apoptosis mediated by diverse stimuli including IFNg,
flavopiridol. radiation and cytostatics [139 –142].
However, no significant difference in the cytotoxic Reduction of cyclin D1 expression after flavopiridol
activity of flavopiridol could be found between cell lines treatment can at least partly be explained by the re-
expressing pRb and cell lines defective in Rb expression duced level of hyperphosphorylated pRb. pRb is
(see Table 6) as was shown in esophageal cancer cells
mainly hyperphosphorylated by cdk4/cyclin D com-
[135], breast carcinoma cells [86,87] and in bladder
plexes. Hyperphosphorylated pRb is proposed to in-
carcinoma cells [136]. Thus, the antiproliferative action
crease expression of cyclin D1 [143] and to produce a
of flavopiridol does not seem to be dependent on the
feedback inhibition of cdk4/cyclin D activities by en-
presence of functional pRb. On the other hand, how-
ever, flavopiridol treatment induced a significant hy- hanced expression of the cdk4 inhibitor p16 [10,143].
pophosphorylation of pRb [86 – 88] and a significant On the other hand hypophosphorylated pRb binds to
reduction in the expression of cyclin D1 [88,135,137], and inactivates cyclin D and as a final consequence
pRb and p107 [135]. Thus the phenotype of flavopiri- leads to a reduced level of both proteins. In cells with
dol-treated cells resembles that of cells in the G0/early no pRb expression p16 is overexpressed and inhibits
G1 phase of the cell cycle. Expression of p107 and complex formation between cdk4 and cyclin D
cyclin D1 were reduced by flavopiridol irrespective [144,145]. In addition flavopiridol has been shown spe-
whether the cancer cells were negative for pRb but cifically to repress the activity of the cyclin D1 pro-
positive for p16 or vice versa [135]. moter [100]. Thus, in cell lines that lack cdk4
Esophageal cancer cell lines that lack detectable Rb flavopiridol inhibited G1 progression to S phase by
protein expression showed a more pronounced rate of inhibiting cdk6 activity as well as cyclin D1 expression
apoptosis after flavopiridol treatment [135]. Similarly, [100].
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 151

Overexpression of cyclin D is known to be associated However, cytotoxicity is correlated with suppression


with several types of cancer [46 – 48,146 – 148]. More- of cyclin D1. Overexpression of cyclin D1 is known to
over, transgenic mice overexpressing cyclin D1 spon- be associated with tumor diseases. In tumor cells that
tanously develop tumor diseases [149]. Reduction of are deficient in the expression cyclin D1, cdk4 and Rb,
cyclin D1 level by antisense cyclin D1 [150] or wild-type flavopiridol may be active via inhibiton of cdk2 [86]
p16 gene [151] delivered via viral vectors mediate and inhibition of the phosphorylation and inactivation
growth inhibition, cell cycle arrest and apoptosis in of E2F.
carcinoma cells. Thus it may be concluded that reduc-
tion of cyclin D1 expression may be one essential 4.5. Acti6ity in relation to p53 expression
mechanism by which flavopiridol induces its
cytotoxicity. Expression and function of p53 are in several ways
In tumor cells that lack cdk4/cyclin D1 complexes dependent on the activity of cdk’s. The question was,
and are negative for pRb and in which therefore the
whether flavopiridol is modulating the expression of
regulatory interplay between cdk4, cyclin D, p16 and
p53 and whether the cytoxicity of flavopiridol is depen-
pRb is lost flavopiridol seems to be active mainly by
dent on the expression of p53.
inhibiting cdk2/cyclin E and cdk2/cyclin A complexes
without altering cdk2 levels or cyclin E and cyclin A p53 plays a central role in the cellular response to
levels as dramatically as shown for cyclin D1 [86,87]. DNA damage, providing by induction of a cell cycle
High expression of cyclin A has been shown to corre- delay or apoptosis a protective effect against tumorige-
late with tumor agressiveness [50]. Inhibition of cdk2/ nesis. Targets of p53 include genes associated with
cyclin A reduces phosphorylation mediated inactivation growth control and cell cycle check points (e.g. p21
of E2F in the late S-phase of the cell cycle that may (WAF-1), GADD45, MDM2, EGFR, PCNA, cyclin
lead to tumor cell apoptosis [107]. D1, cyclin G), DNA repair (e.g. GADD45, PCNA,
In summary cytotoxicity of flavopiridol on tumor p21) and apoptosis (e.g. Bax, Bcl-XL, FAS, FASL)
cells and normal cells is independent of the genetic [152].
status of the target cells, i.e. whether they are express- Mutations of p53 are estimated to contribute to
ing pRb or p16 or not. around 50% of all cancers [153,154]. p53 is activated by

Table 6
Cytotoxicity of Flavopiridol in correlation with the genetic status for Rb and p16

Rb/p16 status No. Cytotoxicity Reduction of expression to No change Ref.


(IC50 mM) (100%)

B30% \30%, B80%

Tumor cells
Rb+/+ B-CLL 7 pat. 0.2 – – p27 Byrd et al. [109]
B-CLL (mantel) 3 0.3 Cyclin D1 – p27 Guedez et al.
[137]
Breast 1 0.3 Cyclin D1 Cyclin A, E cdk2, -4 Carlson et al.
[86]
1 0.3 Cyclin D1 Cyclin D3 Cyclin E, D2 Carlson et al.
[91]
Osteosarcoma 1 0.3 – – cdk2, p21 Li et al. [138]
NSC lung 4 0.2 – cdk2 cdk1 Kaur et al. [80]
Head+neck 1 0.1 Cyclin D1 Cyclin D3, E, Patel et al. [108]
cdk1, -2
Bladder (p16−/−) 2 B1.0 – – – Chien et al. [136]
Rb−/− Breast (cdk4−/−, 1 0.2 – Cyclin A, E cdk2 Carlson et al.
cyclin D1−/−) [86]
Oesophageal 2 0.1 Cyclin D1, p107 – – Schrump et al.
[135]
Bladder 2 B1.0 – – – Chien et al. [136]
p16m/m Head+neck 4 0.1 Cyclin D1 – – Patel et al. [108]
p16−/− Oesophageal 2 0.1 Cyclin D1, pRb, – – Schrump et al.
p107 [135]
Bladder 2 B1.0 – – – Chien et al. [136]
Normal cells
Rb++ Breast 1 0.2 Cyclin D1, D3 – n.t. Singh et al. [88]
152 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

Fig. 2. Inhibition of cdk’s by flavopiridol may effect pocket proteins, p53 and bcl-2.

DNA damage (i.e. double strand breaks, mismatches Whereas the cytotoxic activity of flavopiridol in most
single stranded DNA) [155,156]. Its half-life time and cases was independent of p53 expression, the synergistic
transactivation activity is increased by phosphorylation cytotoxic effect of taxol and flavopiridol (given 24 h
through DNA-PK, MAP kinases, SAP kinases and after taxol) was shown to be strongest in tumor cells
cdk’s [157]. Phosphorylation of the sites on the C-termi- with deletion in p53 expression [114]. This indicates a
nal end of p53 by cdk’s has been shown to activate the mechanism of synergistic action that is counteracted by
sequence specific binding of p53 in a manner specific p53.
for the promoters of stress-responsive genes [158,159].
p53 induces the expression of and is inactivated by 4.6. Acti6ity in relation to Bcl-2 expression
MDM-2. Inhibitors of MDM-2 include hypophospho-
rylated pRb [76]. Thus, flavopiridol, by inhibiting the In view of the fact that targets of p53 include genes
phosphorylation of pRb, may promote inhibition of of the bcl-2 family [152,162] the interesting question
MDM-2 and release of active p53. Active p53 in turn was whether cytotoxicity of flavopiridol is correlated
can enhance expression of MDM-2 and of pRb [152]. with the expression of these proteins.
On the other hand, by inhibition of cdk2 flavopiridol The Bcl-2 family of proteins consists of both in-
may inhibit phosphorylation and activation of p53 (see hibitors and promoters of apoptosis [163,164] via its
Fig. 2). function as an ion channel and as an adapter or dock-
However, in spite of the many interactions between ing protein, e.g. for calcineurin and p53 binding protein
cdk’s and p53 nearly all studies on different cell lines [165].
and clinical isolates showed that the antiproliferative Inhibitors of apoptosis (including Bcl-2, Bcl-X, Bcl-
activity of flavopiridol is clearly independent on the w, Bfl-1, Brag-1, Mcl-1 and A1) contain four domains:
genetic p53 status of the cell (see Table 7). Cells with 1. The hydrophobic C-terminal transmembrane do-
wild-type p53, inactivated p53 (by HPV-16-E6), mu- main that allows their insertion into intracellular
tated p53 or without p53 expression responded equally membrane, e.g. mitochondrial membranes;
to flavopiridol (see Table 7). In p53 null ovarian car- 2. The pore formation domain formed by BH1 and
cinoma, transfection of p53 even enhanced the cyto- BH2
toxic activity of flavopiridol [160] and, on the other 3. The ligand domain BH3 and
hand, deletion in p53 increased the resistance rate of 4. The docking domain BH4, by which Bcl-2 and other
embryonic fibroblasts to flavopiridol [161]. inhibitors of programmed cell death (PCD) of the
In most cases flavopiridol had no effect on the level Bcl-2 family can bind Raf-1 and other regulatory
of p53 expression. However, a significant reduction of proteins and target these proteins to mitochondria.
p53 expression could be seen in B cell lymphoma, which Promoters of apoptosis (including Bax, BAD, Bak,
might be the result of a higher degree of ubiquination Bcl-Xs, Bid, Bik and Hrk) lack at least one of these
and degradation of the less phosphorylated p53, caused domains, in most cases (except Bcl-Xs) the docking
by inhibition of cdk’s through flavopiridol. domain BH4.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 153

All pro-survival bcl-2 like genes are potentially onco- there to phosphorylate (and hereby to inactivate) BAD
genic, and some mutations probably increase their ex- and possibly other protein substrates promoting apop-
pression indirectly. In hematopoietic cells oncoproteins tosis [183].
such as Myb, Ras and AML1-ETO induce Bcl-2 ex- For pro-survival members, phosphorylation may
pression [166,167] and that of Bcl-2 and A1 is often both augment and suppress activity [183]. Bcl-2 may be
elevated in myeloid leukemia and in esophageal, col- activated by Ser70 phosphorylation but inactivated or
orectal and stomach cancer, and reciprocal to p53 and otherwise altered by phosphorylation of several loop
c-myc expression [168 – 171]. sites, perhaps by Jun kinase (JNK) [184,185]. Sustained
Pro-apoptotic Bcl-2 family members may act as tu- activation of the JNK or p38 kinase pathways, perhaps
mor suppressors. Loss-of-function mutation of Bax can after caspase activation, has been implicated in
be found in human gastrointestinal cancer and some apoptosis.
leukemias [172 –175]. Activation of Bax expression by Expression of Bcl-2 family members is induced tran-
p53 can provoke apoptosis [162,176,177]. scriptionally by certain cytokines and Bax is induced in
All members of the Bcl-2 family form homo- or
some cells as part of the p53 mediated damage response
selective pairs of heterodimers. Dimerization is per-
[162].
formed with the BH1, BH2 or BH3 domain and intra-
Flavopiridol seems to downregulate Bcl-2 expression
cellularly inhibited by phosphorylation of the different
in several B-cell leukemia cell lines [120,186,187] but
proteins of the Bcl-2 family and/or by the action of
not in primary B-CLL cells from patients [109,186]. In
proteases [163,178].
cell lines of ovarian carcinoma [120] and prostate car-
Formation of heterodimers of apoptosis inhibitors
with promoters of apoptosis neutralize the antiapop- cinoma [186] the expression of Bcl-2 was likewise sig-
totic effect of inhibitors. Thus, it depends on the ratio nificantly reduced (see Table 8).
of inhibitors and promoters in the cell, whether a cell As flavopiridol did not reduce expression of Bax, it
will survive and proliferate or whether apoptosis is may be assumed that reduction of Bcl-2 by flavopiridol
induced either via direct activation of proteases and/or in some cell lines could allow Bax and other proapop-
through the opening of the mitochondrial permeability totic family member to induce apoptosis possibly via
transition pores and the mitochondrial release of activation of caspase-3 [80,109,110].
protease activators [179 – 183]. However, it is also evident from the results summa-
Phosphorylation of pro-apoptotic members of the rized in Table 8 that reduction of Bcl-2 level is not an
Bcl-2 family seems to be performed by Raf-1. Raf-1 can essential requirement for the antiproliferative activity of
bind with its C-terminal catalytic domain to the BH-4 flavopiridol as there seems to exist no correlation be-
domain in Bcl-2. Via this binding, Raf-1 is targeted to tween the cytotoxic action of flavopiridol and its po-
mitochondrial membranes, possibly allowing Raf-1 tency to reduce expression of Bcl-2.

Table 7
Cytotoxicity of Flavopiridol in correlation to the genetic status p53

p53 status No. IC50 (mM) Expression of p53 (%) Ref.

Tumor cells
p53+/+ B-CLL 5 pat. 0.2 B5 Byrd et al. [109]
1 0.1 B5 Parker et al. [186]
Breast (MCF-7) 1 B0.3 100 Carlson et al. [86]
Ovarian (p53 transduc.) 1 0.1 100 Makhija et al. [160]
NSCL lung 1 0.1 100 Kaur et al. [80]
Oeseophageal 4 0.3 100 Shapiro et al. [322,323]
p53m/m Breast 1 0.2 100 Schrump et al. [135]
B0.3 0 Carlson et al. [86]
NSCL lung 3 0.2–0.5 0 Shapiro et al. [322]
6 0.1–0.5 0 Shapiro et al. [323]
p53−/− Myeloid leukemia 2 0.1 0 Parker et al. [186]
T-ALL 1 0.1 0 Parker et al. [186]
prostate 1 0.2 0 Parker et al. [186]
ovarian 1 0.3 0 Makhija et al. [160]
Normal cells
p53+/+ Murine splenocytes 1 0.3 100 Byrd et al. [109]
p53−/− Murine splenocytes (null type) 1 0.3 0
154 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

Table 8
Modulation of the expression of Bcl-2 family proteins by flavopiridol

No. Cytotoxicity IC50 Expression reduced to (%) Ref.


(mM)
Bcl-2 Bax

Tumor cells
Ovarian 1 0.1 0 n.d. Schwarz et al. [120]
Prostate 1 0.1 30 100 Parker et al. [186]
B-CLL 2 0.4 B50 100 Kitada et al. [187]; König et al. [326]
(unchanged)
B-CLL 1 0.4 :80 n.d. Schwartz et al. [120]
Breast (low Erb-2) 1 0.3 :80 ]100 Li et al. [216]
Breast (high Erb-2) 2 0.3
B-CLL 7 pat. 0.2 Â Byrd et al. [109]
Ã
1 0.2 Parker et al. [186]
Ã
Gastric 4 0.8 100 100 Schwartz et al. [120]
Ì
Oesophageal 4 0.1 (unchanged) (unchanged) Schrump et al. [135]
Ã
NSCL 4 0.2 Kaur et al. [80]
Ã
Mesotheliom 1 0.3 Å Waheed et al. [183]
Normal cells
Blood mononuclear 9 volunt. 0.2 100 n.d. Byrd et al. [109]
cells (unchanged)

On the other hand, neither transfection of tumor cells effect. Although the inhibiting activity on EGFR ki-
to express Bcl-2 does provide resistance of those cells to nase, pp60 Src, PKC and Erk-1 is at least 50 times less
flavopiridol [161,188] nor does inhibition of expression than on cdk’s, considering the intracellular concentra-
of Bcl-2 by antisense RNA increase cytotoxicity of tion estimated to be achieved after systemic application
flavopiridol [188]. of flavopiridol, inhibition of such kinases may still
In summary, there exists no evidence that the expres- contribute to the proapoptotic and cytotoxic activity of
sion of Bcl-2 is causatively involved in the mechanism flavopiridol.
of flavopiridol induced cytotoxicity. Moreover, expres-
sion of Bcl-2 does not provide resistance of cells to 5.1. Interaction with receptors for the EGF family
flavopiridol.
The fact that flavopiridol is not cytotoxic for cultured The EGFR serves to regulate the proliferation of
embryonic cortical neurons but prevents camptothecin multiple tissues during fetal development, non-repro-
induced loss of cytochrome C and mitochondrial ductive tissues in adult life, and multiple tissues during
transmembrane potential as well as caspase activation pregnancy [190]. Overexpression of EGFR results in
and processing [128,189] may support the assumption EGF-dependent transformation [191,192]. Many tu-
that cytotoxicity caused by flavopiridol is not per se mors of epithelial origin including breast, colorectal,
induced by interference with Bcl-2, Bax and function of non-small cell lung, head and neck, ovarian, bladder
mitochondria. and prostate carcinomas show an aberrant, enhanced
and/or constitutive expression of EGFR [14,192] and
respond to EGFR binding growth factors (e.g. EGF,
5. Inhibition of other kinases TGFa, amphiregulin, heparin binding EGF, epiregulin
and betacellulin) [193]. Expression of high levels of
The present data indicate that inhibition of cell cycle EGFR in tumors strongly correlates with a poor
cdk’s is most probably the main mechanism for the prognosis.
proapoptotic and cytotoxic effect of flavopiridol on The co-expression of EGFR binding growth factors
cycling cells. However, its cytotoxic activity on resting and EGFR frequently occurs in human carcinomas,
cells is difficult to explain on the basis of that suggesting that autocrine activation of the receptor
mechanism. might play a role in cancer cell growth [194 –196].
Possibly, inhibition of other kinases in addition to The c-ErbB-2 protein is a transmembrane protein
cdk’s may be essential for this kind of antitumoral with substantial homology to EGFR. The normal func-
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 155

tion of the c-ErbB-2 protein is not completely known on the one site and direct and indirect inhibition of the
[197]. The normal functions of the structurally related kinase activity of the activated EGFR on the other site
c-ErbB-3 or c-ErbB-4 receptors are even less certain may force cells (especially tumor cells) into pro-
[197 –200]. grammed cell death. In agreement with this assumption
In tumors including stomach, prostate, breast, lung it could be shown that flavopiridol can interrupt an
(adeno carcinoma), pancreas and endometrium consti- EGF-activated survival pathway [215].
tutive activation by point mutations [201] or amplifica- An additional mechanism of action of flavopiridol
tion [193,202] and/or overexpression of the c-ErbB-2 or could be downregulation of c-ErbB-2 expression in
the c-ErbB-3 proteins (prostate carcinoma) has been tumor cells. Breast carcinoma cells showed a 55–90%
found [14] and positively correlated with invasion and decrease in c-ErbB-2 expression after exposure to
metastasis [203,204]. flavopiridol compared to untreated control [216].
In their inactive state the receptors for the EGF Downregulation was much more pronounced in (wild-
family are monomeric. Receptor activation usually in- type) parental cells compared to those stably trans-
volves dimerization induced by the binding of growth duced to overexpress c-ErbB-2 [216]. However, after
factor polypeptides or an increase in the association flavopiridol treatment neither the dose dependent
between already dimerized subunits. growth inhibition nor the degree of apoptosis was
Ligand binding brings into close contact tyrosine influenced by the status of c-ErbB-2 expression. Thus
kinase cytoplasmic domains resulting in the autophos- ErbB-2 expression in breast cancer cells induces neither
phorylation and/or cross-phosphorylation [205] of sev- resistance nor sensitivity to flavopiridol. However,
eral receptor C-terminal tyrosine residues. Receptor flavopiridol may reduce malignancy of breast car-
tyrosine phosphorylation generates sites for binding of cinoma cells by reducing c-ErbB-2 expression.
SH2 (Src homology) domain adapter and signal trans-
ducing proteins that function as substrates for the 5.2. Inhibition of receptor associated protein kinases
phosphorylation by the receptor kinases and hereby
gain kinase activity [190,205 – 210]. Cell membrane receptors lacking intrinsic kinase ac-
The cytoplasmic domain of the EGFR owns six tivity need association with and activation of mem-
phosphorylation sites. The T654 is located in the N-ter- brane-associated kinases for signaling. Such receptor
minal, juxtamembrane region. It is specifically phos- associated protein kinases can be different depending
phorylated by PKC [211] whereas the T669 is on the type of receptors [14]. However, there seems to
phosphorylated by Erk1/2 [212]. Phosphorylation of exist no complete restriction, i.e. several growth factor
T654 inhibits signal transduction of the EGF receptor. receptors exhibiting intrinsic tyrosine kinase activity
Thus the PKC plays the main cellular opponent of have been additionally shown to form heterodimeric
ligand induced EGF-R-activation [213]. complexes with multiple signaling and bridging
The cytoplasmic domain of the c-ErbB-2 also owns molecules via (tyrosine) phosphorylated Src homology
one juxtamembrane site (T686) for phosphorylation by region interactions [197]. For example, the insulin re-
PKC and three additional sites for autophosphoryla- ceptor can be hyperactivated with the help of c-Src
tion [214]. [217]. Moreover, isolated c-Src SH2 domain can bind
Flavopiridol has been shown to inhibit the kinase activated EGFR specifically and directly [218,219] and
activity of ligand-activated EGF receptors isolated is required for EGF-dependent mitogenesis [220]. c-Src
from cell membranes (A431 cells) [1,5]. Any interfer- can phosphorylate the Tyr-845 site of the EGFR in
ence by flavopiridol with binding of the ligand to its addition to its autophosphorylation [219]. In addition,
EGF receptor could clearly be excluded [5]. the phosphorylation of the EGFR or of c-ErB-2 may
The direct inhibition of receptors for the EGF family involve other Src family members, or the JAK kinases
may be an additional mechanism by which flavopiridol [221].
can inhibit proliferation of tumors. This effect could be On the other hand PTKs like c-Src can interact with
tumor selective insofar as many tumors show overex- substrates independent from receptor [222 –224]. Bind-
pression, amplification, constitutive or autocrine activa- ing of proteins of the Src family is mediated by their
tion of such receptors. myristilated N-termini (i.e. to the inner surface of the
In addition, through the inhibition of Erk-1 flavopiri- plasma membrane), by their SH2 domains to sequences
dol may indirectly inhibit the activation of the EGFR containing phosphotyrosine and by their SH3 domains
as well as the signal transduction pathway stimulated to motifs rich in proline residues [225].
by activated EGFR. In contrast, by the inhibition of The Src family of PTKs includes pp60 c-Src, Yes
PKC flavopiridol may simultaneously inhibit phospho- (expressed in a variety of tissues, especially epithelial
rylation of T654 and hereby block the inactivation of cells and brain), Fyn (mainly expressed in T- and
the EGFR (see Fig. 3). It is quite possible that the B-cells), Hck (mainly expressed in monocytes and gran-
conflicting situation arising by blockage of inactivation ulocytes) Fgr (mainly expressed in monocytes) and Lck
156 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

(mainly expressed in T-cells) [225]. As components of immune suppressive effect observed in vitro and in vivo
the receptor system their stimulation initiates a cascade on antigen as well as mitogen stimulated T-cells and
of events leading to cellular proliferation [205,226]. B-cells [4].
Overexpression of proteins of the c-Src family is
associated with a considerable number of tumor dis- 5.3. Inhibition of protein kinase C
eases [225] and may synergistically act with modified
growth factor receptor in its oncogenic activity Protein kinase C (PKC) is a family of serine/
[219,225,227]. threonine specific single chain protein kinases, consist-
Inhibition of Src kinase activity is known to induce ing of at least ten isoenzyme species. ATP binding sites
arrest of mitotic progression of cancer cells [227]. are highly conserved between isoenzymes and resemble
Flavopiridol is able to inhibit Src and to a lesser degree those of other protein kinases. The regulatory domains
Lck but exhibits no relevant activity on Hck and Fyn differ between isoenzymes. In the ‘conventional’ isoen-
[3]. This implies that flavopiridol is not only directly zyme family it contains binding sites for Ca2 + , for
active on receptors for the EGF family but also on anionic phospholipids, for diacylglycerol (DAG) and
receptors that lack intrinsic tyrosine kinase activity and also for phorbol esters [228].
transfer their activation signal via the receptor associ- DAG is the physiological activator of PKC and
ated protein kinase Src. Through this mechanism physiological activation of PKC appears to involve its
flavopiridol could be able to inhibit proliferation of translocation from the cytosol to the cell membrane
tumor cells. In addition, although the IC50 of flavopiri- [229,230]. DAG is generated by various signaling path-
dol for Lck is slightly beyond the threshold of 40 mM ways including those activated by binding of a variety
(Table 1) the slight inhibition of Lck and the stronger of cytokines, hormones, neurotransmitters and growth
inhibition of pp60 Src by flavopiridol may explain its factors to their respective receptors.
strong apoptotic activity on normal lymphoid cells, on The signal transduction pathway that operates via
human lymphoma and leukemia cells and its significant PKC appears to be essentially ubiquitous and regulates

Fig. 3. Inhibition of kinases involved in receptor activation and signal transduction.


H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 157

many cellular processes, including cell proliferation DNA that together with proteins forms a protective
and differentiation [231,232]. The extraordinary diver- cap known as telomeres for eukaryotic linear chro-
sity of response to PKC activation can be explained mosomes [270 –272]. In most human cancers telom-
by the multiple gene products that constitute the eres stop shortening because of the de novo
PKC family, their functional heterogeneity and their synthesis of telomeric DNA by activated telomerase
tissue-selective distribution [231,233 – 236]. [273].Inhibition of PKC should lead to a reduced
Significant increases of PKC expression has been activation of telomerase, a reduced synthesis and
observed in human leukemias, melanomas and breast, maintenance of telomeric DNA and an inhibition of
colon, prostate and lung cancers. The metastatic po- tumor cell proliferation.
tential, aggressiveness and/or growth rate of tumor 5. To phosphorylate DNA methyltransferase, which
cells seem to correlate with their PKC activity [237 – alters gene expression by changing DNA methyla-
242]. tion patterns [274].
Surprisingly in human colonic adenoma or car- The involvement of signal PKC in the transduction
cinoma and in early emergence papillomas, PKC ac- pathway can regulate cell death mediated by a variety
tivity is significantly reduced as compared to adjacent of proapoptotic stimuli. The effects of PKC modula-
mucosa tissue and mucosa in control subjects [243 – tors, however, vary significantly depending on the pro-
246]. Nevertheless, downregulation of PKC in col- apoptotic stimuli [275 –277].
orectal tumor cells [247] by antisense oligonucleotides Nevertheless, the many ways by which PKC con-
induced apoptosis or inhibited DNA synthesis indicat- tributes to signal transduction from the cell membrane
ing distinct roles for PKC in growth and survival to the nucleus makes this enzyme an attractive target
[236]. for the search of inhibitors [276]. Flavopiridol inhibits
Many substrates of PKC have an essential function PKC to a lesser degree than cdk, but nevertheless at a
concentration that seems to be achievable in vivo
in cell proliferation including receptors for the EGF
whithin the cells after application of non-toxic doses of
family, for transferrin, insulin and acetylcholin, the
flavopiridol [2]. Via inhibition of PKC flavopiridol may
signal transducing proteins, several transcription fac-
inhibit signaling of many receptors including receptors
tors, DNA methyltransferase, ribosomal protein S6,
for antigens, neuropeptides (bombesin, vasopressin),
lamin B, telomerase and histone (type III) [248 –250].
endothelin, IL-8, adhesive glycoproteins (fibronectin,
PKC can substitute for or synergize with signal trans-
vitronectin, laminin, collagen IV), peptide hormones
ducing proteins to promote transformation. Many of
(GH, prolactin) and many cytokines and interleukins.
the proteins that act at the signal transduction cas-
Such receptors transduce their activation signal via
cade, such as Ras, Src and ErbB2 are known to in-
receptor-associated kinase including pp60 Src and other
crease cellular DAG that activates PKC [251 –256].
Src family members including FAK, Fps, Fes, Fer,
Activated PKC can activate Src as well as Ras and
Jak1/2/3 or Tyk2. Such kinases activate PLCg, which
by that mechanism reinforce its own activation. Acti- cleaves inositol phospholipids to generate inositol
vated PKC is able: triphosphate and diacylglycerol (DAG), which is the
1. To stimulate the Ras-Raf signal transducing path- physiological activator of PKC [228]. By inhibition of
way that includes activation of Erk-1 [236,257 –259]. PKC flavopiridol may block cellular signaling in a
2. To activate the transcription factors AP-1 [260] and central and decisive way and may block activation of
c-jun [261,262] and those involved in activation of transcription factors (AP-1, c-jun) or expression of
the c-fos and c-jun genes [263]. Enhanced activation transcription factors (c-fos, c-jun) or activation of in-
of the fos promoter results into constitutive fos/jun hibitors of transcription factors (IkB) involved in cell
expression and renders the cells growth factor inde- cycle progression and apoptosis (see Fig. 3). Abroga-
pendent [238]. tion of Bcl-2 expression may be one of the many
3. To phosphorylate and inactivate IkB, the inhibitor consequences of inhibition of PKC by flavopiridol
of the transcription factor NFkB [70,71,264,265]. In [275]. The inhibition through flavopiridol of PKC pos-
many tumor types, overexpression or constitutive sibly in concert with the inhibition of cdk’s, EGF-R,
activation of NFkB could be observed [14]. Expres- pp60 Src and/or Erk-1 may be the mechanism by which
sion of IkBa is known to induce apoptosis in tumor programmed cell death could be induced.
cells overexpressing NFkB [266,267]. If inhibition of PKC is important for induction of
4. To phosphorylate both subunits (telomerase associ- apoptosis of tumor cells by flavopiridol, its way of
ated protein 1 (TEP1) and telomerase reverse tran- action should be different to the PKC inhibitor 7-hy-
scriptase (TERT)) and hereby to activate human droxy staurosporine, which prevents lamin B phospho-
telomerase [248,268,269]. Telomerase is responsible rylation and degradation in and apoptosis of human
for the synthesis and maintenance of telomeric leukemia cells induced by camptothecin [230].
158 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

6. Modulation of multidrug resistance drug resistance [300 –303] and transfection of cells with
the gene for PKC-a can enhance drug resistance several
Resistance of tumor cells to chemotherapy limits its fold [304,305].
therapeutic effect. Resistance can be mediated by In contrast, however, compounds known to reverse
known as well as still unknown mechanisms. Resistance MDR like verapamil, also promote phosphorylation of
phenotypes include the overexpression of drug trans- P-glycoproteins [306]. Thus the possibility exists that
porter proteins [278] like the P-glycoprotein (PG-170) phosphorylation of P-glycoprotein can also inhibit
encoded by the multidrug resistance-1 gene (mdr-1 ) MDR.
[279], the multidrug resistance protein (MRP-190) en- Flavopiridol showed no cross-resistance to many cy-
coded by the mrp-1 gene [280 – 282] or the expression of tostatics (see Table 9), i.e. it was cytotoxic on tumor
the breast cancer resistance protein (BCRP) [161]. Re- cells irrespective of whether these cells were made resis-
sistance may also be caused by disruption of apoptosis tant in vitro to doxorubicin [1,161] or cisplatin [160] or
activation due to overexpression of cyclin D1, Bcl-2 or were clinical isolates from patients refractory to treat-
inactivation of p53 [283 – 286]. ment with chlorambucil, cyclophosphamide, vincristine,
Drug transporter proteins belong to the superfamily doxorubicin, cisplatin and antimetabolites [109] or g-ir-
of ATP binding cassette transporters [287 – 289], which radiation and bleomycin [7,108].
appear to cause cellular resistance to chemotherapeutics Flavopiridol’s absence of cross-resistance in respect
such as anthracyclines, vinca alkaloids and taxol via to most of the conventional chemotherapeutic agents
transport of those drugs across cellular membranes may contribute to the clinically responses to flavopiri-
[279]. dol observed in patients with treatment refractory neo-
Drugs with neutral charge can be transported by plasms and prior disease progression [11 –13].
PG-170 as well as MRP-190, whereas anionic com- One of the molecular mechanisms of this cytotoxic
pounds are thought to be transported by MRP-190 effect of flavopiridol on drug resistant tumor cells could
[287,288,290] and cationic compounds by PG-170 [291]. be inhibition of PKC resulting in a reduced expression
and activation of drug transporter proteins.
Overexpression of MRP-190 induced by doxorubicin
However, the fact that pretreatment of tumor cells
revealed resistance of cells to anthracyclines, epipodo-
with flavopiridol 24 h prior to the addition of cytostat-
phyllotoxins and vinca alkaloids but not to antimetabo-
ics does not in the case of most cytostatics result in
lites or platinum-containing drugs and they show only a
their enhanced activity argues against blockage of the
low level of resistance to taxol.
PG-170 protein by flavopiridol (see Section 4.2).
Overexpression of PG-170 can be induced by the
Moreover, colon carcinoma cell lines that have been
same types of drugs inducing overexpression of MRP-
exposed to increasing concentrations of flavopiridol
190 but the spectrum of the resulting resistance to drugs
and possess 8-fold resistance to flavopiridol showed an
is slightly different. What determines whether MRP-190 increased drug uptake, no difference in the expression
or PG-170 will be overexpressed in a particular instance of PG-170 but a 4-fold resistance to taxol [307].
is not yet clear [278]. On the other hand, a human ovarian carcinoma cell
Overexpression of drug transporter proteins and acti- line that spontaneously developed resistance to
vation of proteins involved in signal transduction and flavopiridol (and to cisplatin) was just as sensitive as
control of cell cycle and apoptosis are connected with the parental cells to cytostatics that are exported by
each other in many ways, i.e. PG-170 or by MPR-190 [308].
1. Expression of Ras and p53 proteins appear to stim- Thus it is unlikely that flavopiridol is active on
ulate PG-170 expression [292], and cell lines resis- resistant cells by direct action on the drug transporter
tant to cisplatin have been shown to overexpress the proteins PG-170 or MPR-190. Like other flavonoids
c-fos gene [293]. being cross-resistant to cytostatics flavopiridol is able to
2. Moreover, c-Myc overexpression can reverse the increase the ATPase activity of MRP-190. But at low
MDR phenotype [294] and the promoter of the concentration, flavopiridol is the only of these drugs
mdr-1 gene harbors a Raf-1 responsive element that inhibits the MRP-190 mediated transport of
[295]. Raf-1 is known to enhance cytoprotection by daunorubicin and is cytotoxic for MRP-190 overex-
Bcl-2 [165]. Raf-1 is activated by Ras and PKC is a pressing cells. This cytotoxicity is slightly reduced (fac-
strong activator of Ras (see Fig. 2) [14]. tor 3) compared to non-MRP-190 overexpressing cells
Whether PKC is involved in MDR is controversial. but still 103 × stronger than the cytotoxicity of the
PKC seems to be directly involved in phosphorylation other flavonoids [309,310].
and activation of the P-glycoprotein [296]. Phosphory- Nevertheless, the intracellular concentration of
lation by PKC of PG-170 seems to be correlated with flavopiridol in the resistant cell line was lower com-
its drug transport activity [297]. Clonally selected MDR pared to that in the parental cell, a fact suggestive to a
cell lines exhibit overexpression of PKC [118,298,299]. drug transporter mechanism involved in cellular resis-
Activation of PKC-a in cancer cells can induce multi- tance to flavopiridol [308].
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 159

Table 9
Activity of Flavopiridol on cells resistant to chemotherapy or radiotherapy

Tumor cells No. Drug resistance Degree/type IC50 mM (Flavopiridol) Ref.

Non-resistant Resistant cell line


cell line

Murine 1 Doxorubicin 110× 0.1 0.25 Sedlacek et al.


leukemia [1]
B-CLL 5 pat. Chlorambucil, prednisone, fludarabine, Clinically 0.12 (0.02–0.22) 0.15 (0.03–0.24) Byrd et al.
cyclophosphamide, vincristine, refractory [109]
doxorubicin, cisplatin
6 pat. fludarabine Clinically – 0.17 (0.09–0.21)
refractory
2 Doxorubicin MRP-1 (after 0.04 0.12 Hooijberg et al.
transfection) [309]
AML Doxorubicin MRP-1 0.09 0.13 Hooijberg et al.
[309]
Myeloma 1 Doxorubicin MDR 0.1–0.01 0.1–0.01 Schlegel et al.
[161]
Leukemia 1 Doxorubicin MDR 0.1–0.01 0.1–0.01 Schlegel et al.
[161]
Leukemia 1 Doxorubicin MPR-1 0.1–0.01 0.1–0.01 Schlegel et al.
[161]
Ovarian ca. 1 Cisplatin 6× – 0.3 Makhija et al.
[160]
Head and neck 4 g-Irradiation+bleomycin No apoptosis 0.01–0.07 B0.3 Patel et al.
ca. (IC95 =0.3) (IC95 =0.3) [108]
NSC lung ca. 1 7-Hydroxy-staurosporine No apoptosis – B0.3 Sugiyama et al.
[327]
Breast ca. 1 Mitomycin BCRP :0.1 :0.5 Schlegel et al.
[161]
Colon ca. 2 MRP transfection MRP 0.045–0.170 0.045–0.170 Smith et al.
[307]

Such mechanisms of resistance to flavopiridol, i.e. known why endothelial cells are in vitro more sensitive
overexpression of the drug transporter protein called to flavopiridol than all the other normal resting and
breast cancer resistance protein [161] or other mecha- proliferating cells tested.
nisms, like overexpression of aldehyde dehydrogenase-1 Flavopiridol is also able to block completely in hu-
or of glycogenphosphorylase [307], which are high man monocytes hypoxia-induced VEGF mRNA and
affinity cytosolic binding proteins for flavopiridol (but protein expression by decreasing the VEGF mRNA
do not enzymatically degrade it) [94] have already been stability [95]. Neither transcriptional activation of the
reported. 6egf promoter nor the hypoxia inducible factor 1 activa-
tion sequence were affected and the constitutive levels
of VEGF mRNA were only slightly reduced by
7. Antiangiogenic activity flavopiridol. Via inhibition of PKC and fos promoter
activation flavopiridol might also reduce c-fos induced
In several test systems flavopiridol showed a signifi- VEGFD expression [312]. Another pathway could be
cant antiangiogenic activity (Table 10). Of special im- the inhibition by flavopiridol of Src, because activation
portance may be the ability of flavopiridol to induce of Src is involved in the upregulation of VEGF expres-
apoptosis in resting as well as in proliferating human sion [313].
endothelial cells [110]. The sensitivity of endothelial The fact that flavopiridol is cytotoxic to endothelial
cells to flavopiridol is higher (IC50:0.1 mM) than has cells as well as inhibits hypoxia-induced VEGF expres-
been reported for human normal fibroblasts (no induc- sion suggests that inhibition of tumor angiogenesis
tion of apoptosis) [311], for normal bone marrow cells could play a considerable role in the antitumoral activ-
[6] and for normal peripheral lymphocytes [109]. ity of flavopiridol [110].
Flavopiridol’s mechanism of action in inducing Indeed, in the in vivo mouse matrigel model of
apoptosis in resting endothelial cells is not completely angiogenesis flavopiridol was more potent than the
understood (as in other cells) and likewise it is not yet known anti angiogenic compound TNP-470 in inhibit-
160 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

ing neovascularization. Flavopiridol also showed at 1, ICAM-2, VCAM-1, E-selectin and CD34 on en-
least equivalent activity to TNP-470 in reducing mean dothelial cells in the tumor vasculature and to induce
tumor volume in nude mice bearing human colon car- resistance to inflammatory signals such as TNFa, IL-1
cinoma xenografts [314]. and IFNg [316 –319]. In addition, antiangiogenic fac-
The antiangiogenic effect of flavopiridol is supported tors like platelet factor 4 (PF4), thrombospondin-1 and
by its potency to decrease MMP activity of tumor cells g-interferon inducible protein-10 can prevent or reverse
(i.e. breast carcinoma expressing low c-ErbB-2 or trans- downregulation of the expression of adhesion molecules
duced to express high c-ErbB-2) and to inhibit their on endothelial cells [317]. It may well be that a reduced
invasion through matrigel [216]. By secretion of degra- expression of adhesion molecules in the tumor vascula-
dative enzymes tumors support invasion and attach- ture may enhance the sensitivity of tumor endothelial
ment of endothelial cells [315]. cells to flavopiridol.
Surprisingly, no side effects of flavopiridol on the The fact that in patients hypotension is the dose
vasculature have been reported from preclinical as well limiting toxicity of i.v. infused flavopiridol in combina-
as clinical studies [2,12,13], although flavopiridol is tion with antidiarrheal prophylaxis [12,13] may indicate
similarly active on both cycling and non-cycling en- that flavopiridol could damage the endothelial cell lin-
dothelial cells. It may be possible that in vivo normal ing of the cardiovasculature at higher dosages.
blood vessels are less accessible for flavopiridol due to Altogether, in addition to direct its direct cytotoxicity
thicker layer of bloodborne macromolecules that may for tumor cells flavopiridol may also inhibit tumor
inhibit transmembrane transport of flavopiridol into growth by inhibition of tumor angiogenesis as well as
endothelial cells. In contrast, several clinical studies detoriation of tumor endothelial cells and tumor vascu-
have observed a high incidence of venous thrombotic lature. As antiangiogenic compounds are known to
events that can be caused by a direct action of flavopiri- enhance in vivo the antitumoral activity of cytotoxic
dol on endothelial cells. drugs, the combination of flavopiridol with cytostatic
Proangiogenic factors in tumors are known to induce drugs might provide therapeutic advantages in tumor
downregulation of adhesion molecules such as ICAM- diseases.

Table 10
Antiangiogenic activity of Flavopiridola

Test systems Results Comments Ref.

In 6itro
Cytotoxicity for
endothelial cells
Resting cells IC50: 0.08–0.1 Not dependent on expression of cdk1, Brüsselbach et al. [110]
cdk2, cyclin A, -B, p27, pRb
Proliferating cells IC50: 0.09
Downregulation of
VEGF expression
Normoxic M ¥ Slight effect Reduction of mRNA half-life Melillo et al. [95]
Hypoxic M ¥ Complete blockage
Reduction of MMP
activity
Breast ca. Â Decreased levels of MMP Unclear, whether decline of MMP is a Li et al. [216]
(low Erb-2) Ã after 0.3 mM flavopiridol direct effect or caused by
Breast ca. Ì downregulation of Erb-2
(high Erb-2) ÃÅ
In 6i6o
Matrigel model Reduction of angiogenesis  Robinson et al. [314]
(mouse) Ì Equivalent to TNP-470
Colon carcinoma Tumor growth inhibition Å Robinson et al. [314]
xeno(mouse)
Clinical studies
Phase I study Hypotension catheter related Senderowicz et al. [12,13];
thrombosis induction of inflammatory Werner et al. [14]
markers

a
Senderowicz et al. [12]; Werner et al. [13].
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 161

8. Conclusion 1. Independent on the genetic status of their tumors or


leukemias (i.e. mutations of the pRb and/or p53,
Flavopiridol is a strong inhibitor of cyclin dependent amplification of bcl-2)
kinases (cdk-1, -2, -4, -6, -7) and less active on receptor 2. In spite of drug resistance of their tumors induced
tyrosine kinases (EGFR), receptor associates tyrosine by first line treatment (and caused by enhanced
kinases (pp60 Src) and on signal transducing kinases expression of multidrug resistance proteins)
(PKC and Erk-1). 3. In combination with conventional chemotherapeu-
Although the inhibiting activity of flavopiridol is tics given prior to flavopiridol and
strongest for cdk, the cytotoxic activity of flavopiridol 4. Due to a complex mechanism involving cytotoxicity
is not limited to cycling cells. Resting cells are killed, on cycling and on resting tumor cells, apoptosis and
too. This indicates that inhibition of cdks involved in antiangiogenic activity.
the control of cell cycle is not the only mechanism of In consequence, flavopiridol is a highly attractive,
action. Inhibition of cdk’s with additional functions new antitumoral compound and deserves further eluci-
(i.e. involved in the transcriptional control of the ex- dation of its clinical potency.
pression of cyclins or transcription and/or function of
proteins that do not control the cell cycle) could con- Reviewers
tribute to the antitumoral activity of flavopiridol. In
Professor Dr H.J. Broxterman, Academisch of Medi-
addition, direct and indirect inhibition of receptor acti-
cal Oncology, PO Box 7057, NL-1007 MB Amsterdam,
vation (EGFR) and/or a direct inhibition of kinases
The Netherlands.
(pp60 Src, PKC, Erk-1) involved in the signal transduc-
Walter M. Staler, M.D., Assistant Professor of
tion pathway could participate in the antiproliferative
Medicine, Section of Hematology/Oncology, University
activity of flavopiridol.
of Chicago Medical center 5841 S. Maryland Avenue,
From pharmacokinetic data in patients it can be
MC 2215, Chicago, IL.
concluded that the inhibitory activity (IC50) of
Professor Cr G. Eisenbrand, Universität Kaiser-
flavopiridol on these kinase is in the range of concen-
trations that might be achieved intracellularly after slautern, Fachbereich Chemie, Gebäude 52/322 Erwin-
systemic application of non-toxic doses of flavopiridol. Schrödinger-Strasse, D-67663 Kaiserslautern, FRG
However, no in situ data from flavopiridol treated cells Germany.
have been published yet that prove that by inhibition of
EGFR, pp60 SRC, PKC and/or Erk-1 (in addition to
inhibition of cdk’s) flavopiridol is able to induce apop- Acknowledgements
tosis. Thus many questions regarding the detailed
mechanism of antitumoral action of flavopiridol are The author is indebted to Manuela Rogala for her
still open. Nevertheless, the assumption seems to be skillful secretarial assistance in preparing the
attractive that the strong antitumoral activity of manuscript.
flavopiridol on cycling and resting malignant as well as
on normal cells could be caused by a complex mecha-
nism. Prominent seems to be the inhibition of cdk’s References
involved in control of the cell cycle and/or activation of
[1] Sedlacek HH, Hoffmann D, Czech J, Kolar C, Seemann G,
transcription. Inhibition of receptors and kinases in-
Güssow D, Bosslet K. The change in research for the therapy of
volved in signal transduction may support the antitu- tumors. Chimia 1991;45:311– 6.
moral effect of flavopiridol. This antitumoral activity [2] Sedlacek HH, Czech J, Naik R, Kaur G, Worland P, Losiewicz
shows M, Parker B, Carlson B, Smith A, Senderowicz A, Sausville E.
1. High rate of apoptosis, especially in leukemic cells Flavopiridol (L868 275; NSC649 890), a new kinase inhibitor
2. Synergy with the antitumoral activity of many for tumor therapy. Int J Oncol 1996;9:1143.
[3] Czech J, Hoffmann D, Myers CE, Horak ID, Sausville EA,
cytostatics
Sedlacek HH. Proceedings of the 4th International Congress on
3. Independence of its efficacy on pRb, p53 and Bcl-2 Hormones and Cancer, Amsterdam, 1991.
expression [4] Arguello F, Alexander M, Sterry JA, Tudo X, Smith EM,
4. Lack of interference with the most frequent multi- Kalavar NT, Grenne JF, Koss W, Morgan CD, Stinson SF,
drug resistance proteins (P-glycoprotein and MRP- Siford TJ, Alvord WG, Klabansky RL, Sausville EA.
190) and Flavopiridol induces apoptosis of normal lymphoid cells, causes
immunosuppression, and has potent antitumor activity in vivo
5. A strong antiangiogenic activity.
against human leukemia and lymphoma xenografts. Blood
Based on these pharmacological data it can be con- 1998;91:2482– 90.
cluded that flavopiridol could be therapeutically active [5] Czech J, Hoffmann D, Naik R, Sedlacek HH. Antitumoral
in tumor patients activity of flavone L86-8275. Int J Oncol 1995;6:31– 6.
162 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

[6] Drees M, Dengler WA, Roth T, Labonte H, Mayo J, Malspeis p16INK4/MTS1- and p14 INK4B/MTS2-related cdk6 in-
L, Grever M, Sausville EA, Fiebig HH. Flavopiridol (L86- hibitor, correlates with wild-type pRb function. Genes Dev
2875): selective antitumor activity in vitro and activity in vivo 1994;8:2939– 52.
for prostate carcinoma cells. Clin Cancer Res 1997;3:273– 9. [25] Hannon GJ, Beach D. p15INK4B is a potential effector of
[7] Patel V, Senderowicz AM, Pinto DJ, Igishi T, Ensley JF, TGFfl-induced cell cycle arrest. Nature 1994;371:257–61.
Sausville E, Gutkind SJ. Flavopiridol a novel cyclin dependent [26] Hunter T, Pines J. Cyclins and cancer. II: cyclin D and cdk
kinase inhibitor prevents the growth of head and neck inhibitors come of age. Cell 1994;79:573– 82.
squamous cell carcinomas by inducing apoptosis. Proc Am [27] Noda A, Ning Y, Venable SF, Pereira-Smith OM, Smith JR.
Assoc Cancer Res 1998;39:291. Cloning of senescent cell-derived inhibitors of DNA synthesis
[8] Pedrali-Noy G, Spadari S, Miller-Faures A, Miller AO, Kruppa using an expression screen. Exp Cell Res 1994;211:90–8.
J, Koch G. Synchronization of HeLa cell cultures by inhibition [28] Peter M, Herskowitz I. Joining the complex: cyclin-dependent
of DNA polymerase-a with aphidicolin. Nucl Acids Res kinase inhibitory proteins and the cell cycle. Cell 1994;7:181–4.
1980;8:377. [29] MacLachlan TK, Sang N, Giordano A. Cyclins, cyclin-depen-
[9] Kaur G, Stetler-Stevenson M, Sebers S, Worland P, Sedlacek dent kinases and cdk inhibitors: implications in cell cycle con-
HH, Myers C, Czech J, Naik R, Sausville E. Growth inhibition trol and cancer. Crit Rev Eukary Gene Expr 1995;5:127–56.
with reversible cell cycle arrest of carcinoma cells by flavone [30] DeBondt HL, Rosenblatt J, Jancarik J, Jones HD, Morgan
L86 8275. J Natl Cancer Inst 1992;84:1736–40. DO, Kim SH. Crystal structure of cyclin-dependent kinase 2.
[10] Müller R. Transcriptional regulation during the mammalian Nature 1993;363:595– 602.
cell cycle. Trends Genet 1995;11:173–8. [31] Endicott JA, Nurse P, Johnson LN. Mutational analysis sup-
[11] Schwartz GK, Werner JL, Maslak P, Saltz L, O’Reilly E, ports a structural model for the cell cycle kinase p34. Protein
Keslen DP, Inzeo D, Sugarman A, Tong W, Spriggs D. Eng 1994;7:243– 53.
Flavopiridol enhances the biological effects of paclitaxel a [32] Lees EM, Harlow E. Sequences within the conserved cyclin box
phase I trial in patients with advanced solid tumors. Proc of human cyclin A are sufficient for binding to and activation
ASCO 1998;17:188. of cdc2 kinase. Mol Cell Biol 1993;13:1194– 201.
[12] Senderowicz AM, Headlee D, Stinson SF, Lush RM, Kalil N, [33] Draetta GF. Cell cycle: will the real cdk-activating kinase
Villalba L, Hill K, Steinberg SM, Figg WD, Tompkins A, please stand up. Curr Biol 1997;7:R50– 2.
Arbuck SG, Sausville EA. Phase I trial of continuous infusion [34] Desai D, Essling HC, Fisher RP, Morgan DO. Effects of
Flavopiridol, a novel cyclin-dependent kinase inhibitor, in pa- phosphorylation by CAK on cyclin binding by cdc2 and cdk2.
tients with refractory neoplasms. J Clin Oncol 1998;16:2986– Mol Cell Biol 1995;15:345– 50.
99. [35] Ducommun B, Brambilla P, Felix MA, Franza BRJ, Karsenti
[13] Senderowicz AM. Flavopiridol: the first cyclin-dependent ki- E, Draetta G. cdc2 phosphorylation is required for its interac-
nase inhibitor in human clinical trials. Invest. New Drugs tion with cyclin. EMBO J 1991;10:3311– 9.
1999;17:313– 20. [36] Zhang J, Sanchez RJ, Wang S, Guarnaccia C, Tossi A, Za-
[14] Werner JL, Kelsen DP, Karpeh M, Inzeo D, Barazzuol J, hariev S, Pongor S. Substrate specificity of cdc2 kinase from
Sugarman A, Schwartz GK. The cyclin-dependent kinase in- human HeLa cells as determined with synthetic peptides and
hibitor Flavopiridol is an active and unexpectedly toxic agent in molecular modelling. Arch Biochem Biophys 1994;351:415–24.
advanced gastric cancer. Proc ASCO 1998;17:234. [37] Rickert P, Seghezzi W, Shanahan F, Cho H, Lees E. Cyclin
[15] Stadler WM, Vogelzang NJ, Amato R, Sosman J, Taber D, C/cdk8 is a novel CTD kinase associated with RNA polymerase
Liebowitz D, Vokes EE. Flavopiridol, a novel cyclin-dependent II. Oncogene 1996;12:2631– 40.
kinase inhibitor, in metastatic renal cancer: a University of [38] Bartkova J, Lukas J, Strauss M, Bartek J. Cell cycle-related
Chicago phase II consortium study. J Clin Oncol 2000;18:371– variation and tissue-restricted expression of human cyclin D1
5. protein. J Pathol 1994;172:237– 45.
[16] Sedlacek HH. Kinase inhibitors in cancer therapy: a look [39] Choi YH, Lee SJ, Nguyen P, Jang JS, Lee J, Wu ML, Takano
ahead. Drugs 2000;59:435–76. E, Maki M, Henkart PA, Trepel JB. Regulation of cyclin D1 by
[17] Forth W, Henschler D, Rummel W, editors. Pharmakologie calpain protease. J Biol Chem 1997;272:28479– 84.
und Toxikologie. Zürich: Bibliographisches Institut, 1980:54– 8. [40] King RW, Deshaies RJ, Peters JM, Kirschner MW. How
[18] Stinson SF, Hill K, Siford TJ, Phillips LR, Daw TW. Determi- proteolysis drives the cell cycle. Science 1996;274:1652–9.
nation of flavopiridol (L86 8275; NSC 649 890) in human [41] Hershko A, Ganoth D, Sudakin V, Dahan A, Cohen LH, Luca
plasma by reversed-phase liquid chromatography with electro- FC, Ruderman JV, Eytan E. Components of a system that
chemical detection. Cancer Chemother Pharmacol ligated cyclin to Ubiquitin and their regulation by the protein
1998;42:261– 5. kinase cdc2. J Biol Chem 1994;269:4940– 6.
[19] Thomas J, Cleary J, Tutsch K, Arzoomanian R, Alberti D, [42] King RW, Jackson PK, Rischner MW. Mitosis in transition.
Simon K, Feier-abend C, Morgan K, Wilding G. Phase I Cell 1994;79:563– 71.
clinical and pharmacokinetic trial of flavopiridol. Proc Am [43] Lorca T, Devault A, Colas P, van Loon A, Fesquet D, Lazaro
Assoc Cancer Res 1997;38:222. JB, Dorée M. Cyclin A-Cys41 does not undergo cell cycle-de-
[20] Müller R, Mumberg D, Lucibello FC. Signals and genes in the pendent degradation in Xenopus extracts. FEBS Lett
control of cell-cycle progression. Biochim Biophys Acta 1992;306:90– 3.
1993;1155:151– 79. [44] Holloway SL, Glotzer M, King RW, Murray AV. Anaphase is
[21] Hartwell LH, Kastan MB. Cell cycle control and cancer. Sci- initiated by proteolysis rather than by the inactivation of matu-
ence 1994;266:1821–8. ration-promoting factor. Cell 1993;79:1393– 402.
[22] Murray A. Cell cycle checkpoints. Curr Opin Cell Biol [45] Kato J, Matsushime H, Hiebert SW, Ewen ME, Sherr CJ.
1994;6:872– 6. Direct binding of cyclin D to the retinoblastoma gene product
[23] Bürger C, Wick M, Müller R. Lineage-specific regulation of cell (pRb) and pRb phosphorylation by the cyclin D-dependent
cycle gene expression in differentiating myeloid cells. J Cell Sci kinase cdk4. Genes Dev 1993;7:331– 42.
1994;107:2047– 54. [46] Bartkova J, Lukas J, Muller H, Lutzhoft D, Strauss M, Bartek
[24] Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, O’Keefe J. Cyclin D 1 protein expression and function in human breast
CL, Matera AG, Xiong Y. Growth suppression by p18, a cancer. Int J Cancer 1994;57:353– 61.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 163

[47] Keyomarsi K, Pardee AB. Redundant cyclin overexpression Minna JD, Gazdar AF, Kaye FJ. RB protein status and clinical
and gene amplification in brest cancer cells. Proc Natl Acad Sci correlation from 171 cell lines representing lung cancer, extra-
USA 1993;90:1112–26. pulmonary small cell carcinoma, and mesothelioma. Oncogene
[48] Motokura T, Bloom T, Kim HG, Juppner H, Ruderman JV, 1994;9:2441– 8.
Kronenberg HM, Arnold A. A novel cyclin encoded by a [67] Beijersbergen RL, Carlee L, Kerkhoven RM, Bernards R.
bcl1-linked candidate oncogene. Nature 1991;350:512–5. Regulation of the retinoblastoma protein-related p107 by G1
[49] Hirama T, Koeffler HP. Role of the cyclin-dependent kinase cyclin complexes. Genes Dev 1995;9:1340– 53.
inhibitors in the development of cancer. Blood 1995;86:841– 54. [68] Mayol X, Garriga J, Grana X. Cell cycle-dependent phosphory-
[50] Huuhtanen RL, Blomqvist CP, Böhling TO, Wiklund TA, lation of the retinoblastoma-related protein p130. Oncogene
Tukiainen EJ, Virolainen M, Tribukait B, Andersson LC. Ex- 1995;11:801– 8.
pression of cyclin A in soft tissue sarcomas correlates with [69] Richon VM, Lyle RE, McGehee RE. Regulation and expres-
tumor aggressiveness. Cancer Res 1999;59:2885–90. sion of retinoblastoma proteins p107 and p130 during 3T3-L1
[51] Keyomarsi K, O’Leary N, Molnar G, Lees E, Fingert HJ, adipocyte differentiation. J Biol Chem 1997;272:10117–24.
Pardee AB. Cyclin E, a potential prognostic marker for breast [70] Garriga J, Limon A, Mayol X, Rane SG, Albrecht JH, Reddy
cancer. Cancer Res 1994;54:380–5. EP, Andres V, Grana X. Differential regulation of the
[52] He J, Allen JR, Collins VP, Allalunis-Turnver MJ, Godbout R, retinoblastoma family of proteins during cell proliferation and
Day RS, James CD. CDK4 amplification is an alternative differentiation. Biochem J 1998;333:645– 54.
mechanism to p16 gene homozygous deletion in glioma cell [71] Ghosh S, Baltimore D. Activation in vitro of NF-kB by phos-
lines. Cancer Res 1994;54:5804–7. phorylation of its inhibitor IkB. Nature 1990;344:678–82.
[53] Wolfel T, Hauer M, Schneider J, Serrano M, Wolfel C, Kleh- [72] Kiess M, Gill RM, Hamel PA. Expression and activity of the
mann-Hieb E, De Plaen E, Hankeln T, Buschenfelde KH, retinoblastoma protein (pRb)-family proteins, p107 and p130,
Beach D. A p16INK4a-insensitive CDK4 mutant targeted by during L6 myoblast differentiation. Cell Growth Differ
cytolytic T lymphocytes in a human melanoma. Science 1995;6:1287– 98.
1995;269:1281– 4. [73] Smith EJ, Leone G, Degregori J, Jakoi L, Nevins JR. The
[54] Costello JF, Merger MS, Huang HJ, Cavenee WK. Silencing of accumulation of an E2F-p130 transcriptional repressor distin-
p16/CDKN2 expression in human gliomas by methylation and guishes a G0 cell state from a G1 cell state. Mol Cell Biol
chromatin condensation. Cancer Res 1996;56:2405–10. 1996;16:6965– 76.
[55] Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, [74] Smith EJ, Leone G, Nevins JR. Distinct mechanisms control
Tavtigian SV, Stockert E, Day RS, Johnson BE, Scolnick MH. the accumulation of the Rb-related p107 and p130 proteins
A cell cycle regulator potentially involved in genesis of many during cell growth. Cell Growth Differ 1998;9:297– 303.
tumor types. Science 1994;264:436–40. [75] Hurford RK, Cobrinik D, Lee MH, Dyson N. pRb and P107/
[56] Ludlow JW, Nelson DA. Control and activity of type-1 serine/ p130 are required for the regulated expression of different sets
threonine protein phosphatase during the cell cycle. Semin of E2F responsive genes. Genes Dev 1997;11:1447– 63.
Cancer Biol 1995;6:195–202. [76] Grana X, Garriga J, Mayol X. Role of the retinoblastoma
[57] Lundberg AS, Weinberg RA. Functional inactivation of the protein family, pRb, p107 and p130 in the negative control of
retinoblastoma protein requires sequential modification by at cell growth. Oncogene 1998;17:3365– 83.
least two distinct cyclin-cdk complexes. Mol Cell Biol [77] Dowdy SF, Hinds PW, Louie K, Reed SI, Arnold A, Weinberg
1998;18:753– 61. RA. Physical interaction of the retinoblastoma protein with
[58] Mayol X, Garriga J, Grana X. G1 cyclin/cdk-independent human D cyclins. Cell 1993;73:499– 511.
phosphorylation and accumulation of p130 during the transi- [78] Ewen ME, Sluss HK, Sherr CJ, Matsushime H, Kato J, Liv-
tion from G1 to G0 lead to its association with E2F-4. Onco- ingston DM. Functional interactions of the retinoblastoma
gene 1996;13:237– 46. protein with mammalian D-type cyclins. Cell 1993;73:487–97.
[59] Suzuki-Takahashi I, Kitagawa M, Saijo M, Higashi H, Ogino [79] Connell-Crowley L, Harper JW, Goodrich DW. Cyclin D1/
H, Matsumoto H, Taya Y, Nishimura S, Okuyama A. The cdk4 regulates retinoblastoma protein-mediated cell cycle arrest
interactions of E2F with pRb and with p107 are regulated via by site-specific phosphorylation. Mol Biol Cell 1997;8:287–301.
the phosphorylation of pRb and 107 by a cyclin-dependent [80] Kaur G, Hursey M, Sausville EA. Cell cycle arrest and apopto-
kinase. Oncogene 1995;10:1691–8. sis in NSCLC cell lines by cyclin-dependent kinase inhibitor.
[60] Zhu L, Harlow E, Dynlacht BD. p107 uses a p21CIP1-related Proc Am Assoc Cancer Res 1998;39:605– 6.
domain to bind cyclin/cdk2 and regulate interactions with E2F. [81] Losiewicz MD, Carlson BA, Kaur G, Sausville EA, Worland
Genes Dev 1995;9:1740–52. PJ. Potent inhibition of cdc2 kinase activity by the flavonoid
[61] Zhu L, Zhu L, Xie E, Chang LS. Differential roles of two L86-8275. Biochem Biophys Res Commun 1994;201:589–95.
tandem E2F sites in repression of the human p107 promoter by [82] Losiewicz MD, Carlson BA, Sausville EA, Naik RG,
retinoblastoma and p107 proteins. Mol Cell Biol 1995;15:3552– Narayanan VL, Worland PJ. Inhibition of cdk2 by Flavopiridol
62. and structural analoga. Proc Am Assoc Cancer Res 1995;36:35.
[62] Hartwell LH, Kastan MB. Cell cycle control and cancer. Sci- [83] Worland PJ, Kaur G, Stetler-Stevenson M, Sebers S, Sartor O,
ence 1994;266:1821–8. Sausville EA. Alteration of the phosphorylation state of p34cdc2
[63] Mulligan GJ, Jacks T. The retinoblastoma gene family: cousins kinase by the flavone L86-8275 in breast carcinoma cells.
with overlapping interests. Trends Genet 1998;14:223–9. Biochem Pharmacol 1993;46:1831– 40.
[64] Weinberg RA. The retinoblastoma protein and cell cycle con- [84] Meyerson M, Enders GH, Wu CL, Su LK, Gorka C, Nelson C,
trol. Cell 1995;81:323–30. Harlow E, Tsai LH. A family of human cdc2-related protein
[65] Ishii H, Igarashi T, Saito T, Nakano T, Mori M, Ohyama H, kinases. EMBO J 1992;11:2909– 17.
Miyamoto T, Saito Y, Oh H. Retinoblastoma protein expressed [85] Zieve GW, Turnbull D, Mullins JM, McIntosh JR. Production
in human non-Hodgkin’s lymphoma cells generates resistance of large numbers of mitotic mammalian cells by use of the
against radiation-induced apoptosis. Am J Hematol reversible microtubuline inhibitor nocodazole. Nocodazole ac-
1997;55:46– 8. cumulated mitotic cells. Exp Cell Res 1980;126:397– 405.
[66] Shimizu E, Coxon A, Otterson GA, Steinberg SM, Kratzke [86] Carlson BA, Dubay MM, Sausville EA, Brizuela L, Worland
RA, Kim YW, Fedorko J, Oie H, Johnson BE, Mulshine JL, PJ. Flavopiridol induces G1 arrest with inhibition of cyclin-de-
164 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

pendent kinase (cdk) 2 and cdk4 in human breast carcinoma ity to 1-b-D-arabinofuranosylcytosine and topoisomerase II in-
cells. Cancer Res 1996;56:2973–8. hibitors in tumor cell lines harboring activated ras oncogenes.
[87] Carlson BA, Pearlstein RA, Naik RG, Sedlacek HH, Sausville Cancer Res 1996;56:5211– 6.
EA, Worland PJ. Inhibition of cdk2, cdk4 and cdk7 by [106] Koo HM, Gray-Goodrich M, Kohlhagen G, McWilliams MJ,
Flavopiridol and structural analogs. Proc Am Assoc Cancer Jeffers M, Vaigro-Wolff A, Alvord WG, Monks A, Paull KD,
Res 1996;37:424. Pommier Y, Vande Woude GF. The ras oncogene-mediated
[88] Singh SS, Sausville EA, Senderowicz AM. Cyclin D1 and Cdk6 sensitization of human cells to topoisomerase II inhibitor-in-
are the targets for Flavopiridol-mediated G1 block in MCF10A duced apoptosis. J Natl Cancer Inst 1999;19:236– 44.
breast epithelial cell line. Proc Am Assoc Cancer Res [107] Krek W, Ewen ME, Shirodka S, Arany Z, Kaelin WJ, Liv-
1999;40:28. ingston DM. Negative regulation of the growth-promoting
[89] Grana X, De Luca A, Sang N, Fu Y, Claudio P, Rosenblatt P, transcription factor E2F-1 by a stably bound cyclin A-depen-
Morgan DO, Giordano A. PITALRE, a nuclear CDC2-related dent protein kinase. Cell 1994;78:161– 71.
protein kinase that phosphorylates the retinoblastoma protein [108] Patel V, Senderowicz AM, Pinto D, Igishi T, Raffeld M,
in vitro. Proc Natl Acad Sci USA 1994;91:3834–8. Quintanilla-Martinez L, Ensley JF, Sausville EA, Gutkind JS.
[90] Gold MO, Rise AP. Targeting of cdk8 to a promoter-proximal Flavopiridol, a novel cyclin-dependent kinase inhibitor, sup-
RNA element demonstrates catalysis-dependent activation of presses the growth of head and neck squamous cell carcinomas
gene expression. Nucl Acids Res 1998;26:3784–8. by inducing apoptosis. J Clin Invest 1998;102:1674– 81.
[91] Carlson B, Lahusen T, Singh S, Loaiza-Perez A, Worland PJ, [109] Byrd JC, Shinn C, Waselenko JK, Fuch EJ, Lehman TA,
Pestell R, Albanese C, Sausville EA, Senderowicz AM. Down- Nguyen PL, Flinn IW, Dieh LF, Sausville E, Grever MR.
regulation of cyclin D1 by transcriptional repression in MCF-7 Flavopiridol induces apoptosis in chronic lymphocytic leukemia
human breast carcinoma cells induced by Flavopiridol. Cancer cells via activation of caspase-3 without evidence of bcl-2
Res 1999;59:4634– 41. modulation or dependence on functional p53. Blood
[92] De Azevedo WF, Mueller-Dieckmann HJ, Schulze-Gahmen U, 1998;92:3804– 16.
Worland PJ, Sausville E, Kim SH. Structural basis for specific- [110] Brüsselbach S, Nettelbeck DM, Sedlacek HH, Müller R. Cell
ity and potency of a flavonoid inhibitor of human cdk2, a cell cycle-independent induction of apoptosis by the anti-tumor
cycle kinase. Proc Natl Acad Sci USA 1996;93:2735–40. drug Flavopiridol in endothelial cells. Int J Cancer
[93] Jeffrey PD, Russo AA, Polyak K, Gibbs E, Hurwitz J, Mas- 1998;77:146– 52.
[111] Giovanni A, Wirtz-Brugger F, Keramaris E, Slack R, Park DS.
sague J, Pavletich NP. Mechanism of cdk activation revealed by
Involvement of cell cycle elements, cyclin-dependent kinases,
the structure of a cyclin A-cdk2 complex. Nature
pRb and E2F DP, in B-amyloid-induced neuronal death. J Biol
1995;376:313– 20.
Chem 1999;274:19011– 6.
[94] Schnier JB, Kaur G, Kaiser A, Stinson SF, Sausville EA,
[112] Matranga CB, Shapiro GI. Selective sensitization of trans-
Gardner J, Nishi K, Bradbury EM, Senderowicz AM. Identifi-
formed cells to flavopiridol-induced apoptosis during S phase.
cation of cytosolic aldehyde dehydrogenase 1 from non-small
Proc Am Assoc Cancer Res 2000;41:32.
cell lung carcinomas as a flavopiridol-binding protein. FEBS
[113] Bible KC, Kaufman SH. Cytotoxic synergy between Flavopiri-
Lett 1999;454:100–4.
dol (NSC 649 890, L86-2875) and various antineoplastic agents:
[95] Melillo G, Sausville EA, Cloud K, Lahusen T, Varesio L,
the importance of sequence of administration. Cancer Res
Senderowicz AM. Flavopiridol, a protein kinase inhibitor,
1997;57:3375– 80.
down-regulates hypoxic induction of vascular endothelial
[114] Edelman MJ, Mullins B, Mack PC, Gumerlock PH, Gandara
growth factor expression in human monocytes. Cancer Res
DR. Synergistic interaction between Flavopiridol (FLAVO)
1999;59:5433– 7.
and Taxol (TAX) in a p53-null non-small cell lung carcinoma
[96] Jacks T, Weinberg RA. The expanding role of cell regulators. (NSCLC) cell line. Proc Am Assoc Cancer Res 1998;39:309–10.
Science 1998;280:1035–6. [115] Motwani M, Schwartz GK. Inappropriate cell cycle progression
[97] Leone G, DeGregori J, Sears R, Jakoi L, Nevins JR. Myc and enhances the induction of apoptosis by Flavopiridol (Flavo) in
ras collaborate in inducing accumulation of active cyclin E/ Taxol treated gastric cancer cells. Proc Am Assoc Cancer Res
Cdk2 and E2F. Nature 1997;387:422–6. 1998;39:190.
[98] Hoang AT, Cohen KJ, Barrett JF, Bergstrom DA, Dang CV. [116] Motwani M, Li XK, Schwartz GK. The cyclin-dependent ki-
Participation of cyclin A in Myc-induced apoptosis. Proc Natl nase (CDK) inhibitor Flavopiridol (Flavo) inhibits polyploidy
Acad Sci USA 1994;91:6875–9. in paclitaxel(P) treated p53 − / − breast cancer cells. Proc Am
[99] Strauss M, Lukas J, Bartek J. Unrestricted cell cyclin and Assoc Cancer Res 1999;40:24.
cancer. Nat Med 1995;1:1245–6. [117] Motwani M, Delohery TM, Schwartz GK. Sequential depen-
[100] Sherr CJ. Cancer cell cycles. Science 1996;274:1672–7. dent enhancement of caspase activation and apoptosis by
[101] Lee HR, Chang TH, Tebalt MJ, Senderowicz AM, Szabo E. Flavopiridol on Paclitaxel-treated human gastric and breast
Induction of differentiation accompanies inhibition of cdk2 in a cancer cells. Clin Cancer Res 1999;5:1876– 83.
non-small cell lung cancer cell lines. Int J Oncol 1999;15:161– 6. [118] Schwartz GK, Arkin H, Holland J, Ohnuma T. Protein kinase
[102] Sauter ER, Nesbit M, Litwin S, Klein-Szanto AJ, Cheffetz S, C activity and multidrug resistance on MOLT-3 human
Herlyn M. Antisense cyclin D1 induces apoptosis and tumor lymphoblastic leukemia cells resistant to trimethotrexate. Can-
shrinkage in human squamous carcinoma. Cancer Res cer Res 1991;51:55– 61.
1999;59:4876– 81. [119] Schwartz GK, Farsi K, Daso D, et al. The protein kinase C
[103] Sandig V, Brand K, Herwig S, Lukas J, Bartek J, Strauss M. (PKC) inhibitors UCN-01 and Flavopiridol (FLAVO) signifi-
Adenovirally transferred p16INK4/CDKN2 and p53 genes co- cantly enhance the cytotoxic effect of chemotherapy by promot-
operate to induce apoptotic tumor cell death. Nat Med ing apoptosis in gastric and breast cancer cells. Proc AmSoc
1997;3:313– 9. Oncol 1996;15:501.
[104] Mesner JP, Budihardjo II, Kaufmann SH. Chemotherapy-in- [120] Schwartz GK, Farsi K, Majhija S, Konig A, Gabrilove JL,
duced apoptosis. Adv Pharmacol 1997;41:461–98. Spriggs D. Sensitivity of tumor cells to the cyclin dependent
[105] Koo HM, Monks A, Mikheev A, Rubinstein LV, Gray- kinase (CDK) inhibitor Flavopiridol (FLAVO) correlates to
Goodrich M, McWilliams MJ, Alvord WG, Oie HK, Gazdar loss of bcl-2 expression. Proc Am Assoc Cancer Res
AF, Paull KD, Zarbl H, Vande Woude GF. Enhanced sensitiv- 1997;38:472– 3.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 165

[121] Schwartz GK, Farsi K, Maslak P, Kelsen DP, Spriggs D. [140] Ishii H, Igarashi T, Saito T, Nakano T, Mori M, Ohyama H,
Potentiation of apoptosis by flavopiridol in mitomycin-C- Miyamoto T, Saito Y, Oh H. Retinoblastoma protein expressed
treated gastric and breast cancer cells. Clin Cancer Res in human non-Hodgkin’s lymphoma cells generates resistance
1997;3:1467– 72. against radiation-induced apoptosis. Am J Hematol
[122] Decaudin D, Marzo I, Brenner C, Kroemer G. Mitochondria in 1997;55:46– 8.
chemotherapy-induced apoptosis: a prospective novel target of [141] Shimizu E, Coxon A, Otterson GA, Steinberg SM, Kratzke
cancer therapy (review). Int J Oncol 1998;12:141–52. RA, Kim YW, Fedorko J, Oie H, Johnson BE, Mulshine JL,
[123] Cotter TG, Glynn JM, Echeverri F, Green DR. The induction Minna JD, Gazdar AF, Kaye FJ. RB protein status and clinical
of apoptosis by chemotherapeutic agents occurs in all phases of correlation from 171 cell lines representing lung cancer, extra-
the cell cycle. Anticancer Res 1992;12:773–80. pulmonary small cell carcinoma, and mesothelioma. Oncogene
[124] Motwani M, Delohery TM, Schwartz GK. Sequential depen- 1994;9:2441– 8.
dent enhancement of caspase activation and apoptosis by [142] Berry DE, Lu, Schmidt B, Fallon PG, O’Connell C, Hu SX, Xu
flavopiridol on paclitaxel-treated human gastric and breast HJ, Blanck G. Retinoblastoma protein inhibits IFN-g induced
cancer cells. Clin Cancer Res 1999;5:1876–83. apoptosis. Oncogene 1996;12:1809– 19.
[125] Bible KC, Bible RH, Kottke TJ, Svingen PA, Xu K, Pang YP, [143] Müller H, Lukas J, Schneider A, Warthoe P, Bartek J, Eilers
Hajdu E, Kaufmann SH. Flavopiridol binds to duplex DNA. M, Strauss M. Cyclin D1 expression is regulated by the
Cancer Res 2000;60:2419–28. retinoblastoma protein. Proc Natl Acad Sci USA
[126] Motwani M, Li X-K, Schwartz GK. Flavopiridol, a cyclin-de- 1994;91:2945– 9.
pendent kinase inhibitor, prevents spindle inhibitor-induced [144] Lukas J, Bartkova J, Rohde M, Strauss M, Bartek J. Cyclin D1
endoreduplication in human cancer cells. Clin Cancer Res is dispensable for G1 control in retinoblastoma gene-deficient
2000;6:924– 32. cells independently of cdk4 activity. Mol Cell Biol
[127] Stefanis L, Park DS, Friedman WJ, Greene LA. Caspase-de- 1995;15:2600– 11.
pendent and -independent death of camptothecin-treated em- [145] Otterson GA, Kratzke RA, Coxon A, Kim YW, Kaye FJ.
bryonic cortical neurons. J Neurosci 1999;19:6235–47. Absence of p16ink4 protein is restricted to the subset of lung
[128] Park DS, Farinelli SE, Greene LA. Inhibitors of cyclin-depen- cancer lines that retains wildtype Rb. Oncogene 1994;9:3375–8.
dent kinases promote survival of post-mitotic neuronally differ- [146] Dreyling MH, Bulinger L, Ott G, Stilgenbauer S, Muller-Her-
melink HK, Benth M, Hiddemann W, Dohner H. Alterations
entiated cells and sympathetic neurons. J Biol Chem
of the cyclin D1/p16-pRB pathway in mantle cell lymphoma.
1996;271:21898– 905.
Cancer Res 1997;57:4608– 14.
[129] Park DS, Morris EJ, Stefanis L, Troy CM, Shelanski ML,
[147] Arber N, Lightdale C, Rotterdam H, Han EKH, Sgambato A,
Geller HM, Greene LA. Multiple pathways of neuronal death
Yap E, Ahsan H, Finegold J, Steves PD, Green PHR, Hib-
induced by DNA-damaging agents, NGF deprivation, and
shoosh H, Neugut AI, Holt PR, Weinstein IB. Increased ex-
oxidative stress. J Neurosci 1998;18:830–40.
pression of the cyclin D1 gene in Barrett’s esophagus. Cancer
[130] Courtney MJ, Coffey ET. The mechanism of Ara-C-induced
Epidemiol Biomark Prev 1996;5:457– 9.
apoptosis of differentiating cerebellar granule neurons. Eur J
[148] Cordon-Cardo C. Mutations of cell cycle regulators. Biological
Neurosci 1999;11:1073–84.
and clinical implications for human neoplasia. Am J Pathol
[131] Park DS, Morris EJ, Greene LA, Geller HM. G1/S cell cycle
1995;147:545– 60.
blockers and inhibitors of cyclin-dependent kinases suppress
[149] Nakagawa H, Wang TC, Zukerberg L, Odze R, Togawa K,
camptothecin-induced apoptosis. J Neurosci 1997;17:1256– 70.
May GHW, Wilson J, Rustgi AK. The targeting of the cyclin
[132] Park DS, Morris EJ, Padmanabhan J, Shelanski ML, Geller H,
D1 oncogene by an Epstein– Barr virus promoter in transgenic
Greene LA. Cyclin-dependent kinases participate in death of mice causes dysplasia in the tongue, esophagus and forestom-
neurons evokes by DNA-damaging agents. J Cell Biol ach. Oncogene 1997;14:1185– 90.
1998;143:457– 67. [150] Zhou P, Jiang W, Zhang YJ, Kahn SM, Schieren I, Santella R,
[133] Bible KC, Kaufmann SH. Flavopiridol: a cytotoxic flavone that Weinstein IB. Antisense to cyclin D1 inhibits growth and
induces cell death in noncyclin A549 human lung carcinoma reverses the transformed phenotype of human esophageal can-
cells. Cancer Res 1996;56:4856–61. cer cells. Oncogene 1995;11:571– 80.
[134] Shapiro GI. Potentiation of Flavopiridol-mediated apoptosis in [151] Schrump DS, Chen A, Consuli U, Jin X, Roth RA. Inhibition
non-small cell lung cancer cell lines following recruitment of of esophageal cancer proliferation by adenoviral-mediated de-
cells to S phase. Proc Am Assoc Cancer Res 1999;40:305. livery of p16INK4. Cancer Gene Ther 1996;3:357– 64.
[135] Schrump DS, Matthews W, Chen GA, Mixon A, Altorki NK. [152] Amundson SA, Myers TG, Fornace AJ. Roles for p53 in
Flavopiridol mediates cell cycle arrest and apoptosis in growth arrest and apoptosis: putting on the brakes after
esophageal cancer cells. Clin Cancer Res 1998;4:2885–90. genotoxic stress. Oncogene 1998;17:3287– 99.
[136] Chien M, Astumian M, Liebowitz D, Rinker-Schaeffer C, [153] Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 muta-
Stadler WM. In vitro evaluation of flavopiridol, a novel cell tions in human cancers. Science 1991;253:49– 53.
cycle inhibitor, in bladder cancer. Cancer Chemother Pharma- [154] Levine AJ, Momand J, Finlay CA. The p53 tumour suppressor
col 1999;44:81– 7. gene. Nature 1991;351:453– 6.
[137] Guedez L, Quintanilla-Martinez L, Lahusen T, Davies T, Singh [155] Lee S, Elenbaas B, Levine A, Griffith J. p53 and its 14 kDa
SS, Barotto N, Vistica D, Raffeld M, Sausville EA, Senderow- C-terminal domain recognize primary DNA damage in the
icz AM. Flavopiridol-induced apoptosis is associated with a form of insertion/deletion mismatches. Cell 1995;81:1013–20.
decrease in cyclin D1 in mantle lymphoma cell lines. Proc Am [156] Reed M, Woelker B, Wang P, Wang Y, Anderson ME, Tegt-
Assoc Cancer Res 1999;40:580. meyer P. The C-terminal domain of p53 recognizes DNA
[138] Li WW, Fan JG, Bertino JR. Flavopiridol enhances doxoru- damaged by ionizing radiation. Proc Natl Acad Sci USA
bicin-induced apoptosis in human sarcoma cells lacking Rb 1995;92:9455– 9.
protein. Proc Am Assoc Cancer Res 1999;40:7. [157] Meek DW. Multisite phosphorylation and the integration of
[139] Berry DE, Lu Y, Schmidt B, Fallon PG, O’Connell C, Hu SX, stress signals at p53. Cell Signal 1998;10:159– 66.
Xu HJ, Blanck G. Retinoblastoma protein inhibits IFN-g in- [158] Hecker D, Page G, Lohrum M, Weiland S, Scheidtmann KH.
duced apoptosis. Oncogene 1996;12:1809–19. Complex regulation of the DNA-binding activity of p53 by
166 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

phoshphorylation: differential effects of individual phosphory- [179] Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y,
lation sites on the interaction with different binding motifs. Greenberg ME. Akt phosphorylation of BAD couples survival
Oncogene 1996;12:953–61. signals to the cell intrinsic death machinery. Cell 1997;91:231–
[159] Wang HG, Rapp UR, Reed JC. Bcl-2 targets the protein kinase 41.
Raf-1 to mitochondria. Cell 1996;87:629–38. [180] Green DR, Reed JC. Mitochondria and apoptosis. Science
[160] Makhija S, Husain A, Schwartz GK, Spriggs DR. Cytotoxicity 1998;281:1309– 12.
of Flavopiridol in ovarian cancer cells, alone and in combina- [181] Fridman JS, Benedict MA, Maybau J. bcl-Xs-induced cell
tion with CDDP. Proc Am Assoc Cancer Res 1997;38:320. death in 3T3 cells does not require or induce caspase activation.
[161] Schlegel S, Klimecki W, List AF. Breast cancer resistance Cancer Res 1999;59:5999– 6004.
protein (BCRP) associated drug export and p53 inactivation [182] delPeso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G.
impact Flavopiridol (FLA) antitumor activity. Proc Am Assoc Interleukin-3-induced phosphorylation of BAD through the
Cancer Res 1999;40:669. protein kinase AKT. Science 1997;278:687– 9.
[162] Han J, Sabbatini E, Perez D, Rao L, Modha D, White E. The [183] Waheed I, Guo S, Mathews W, Chen A, Mixon A, Schrump
E1B 19K protein block apoptosis by interaction with and DS. Expression of T-antigen in pleural mesothelioma delays
inhibiting the p53-inducibble and death-promoting Bax protein. Flavopiridol mediated cell cycle arrest and apoptosis. Proc Am
Genes Dev 1996;10:461–77. Assoc Cancer Res 1999;40:447.
[163] Kroemer G. The proto-oncogene Bcl-2 and its role in regulating [184] Ling YH, Tornos C, Perez-Soler R. Phosphorylation of Bcl-2 is
apoptosis. Nature Med 1997;3:614. a marker of M phase events and not a determinant of apopto-
[164] Chang BS, Minn AJ, Muchmore SW, Fesik SW, Thompson sis. J Biol Chem 1998;273:18984– 91.
CB. Identification of a novel regulatory domain in Bcl-X(L) [185] Maundrell K, Antonsson B, Magnenat E, Camps M, Muda M,
and Bcl-2. EMBO J 1997;16:968–77. Chabert C, Gillieron C, Boschert U, Vial-Knecht E, Martinou
[165] Reed JC. Double identity for proteins of the Bcl-2 family. JC, Arkinstall S. J Biol Chem 1997;272:25238– 42.
Nature 1997;387:773. [186] Parker BW, Kaur G, Nieves-Neira W, Taimi M, Kohlhagen G,
[166] Klampfer L, Zhang J, Zelenetz AO, Uchida H, Nimer SD. The Shimizu T, Losiewicz MD, Pommier Y, Sausville EA,
AML1/ETO fusion protein activates transcription of BCL-2. Senderowicz AM. Early induction of apoptosis in hematopoi-
Proc Natl Acad Sci USA 1996;93:14059–64. etic cell lines after exposure to Flavopiridol. Blood
[167] Taylor D, Badiani P, Weston K. A dominant interfering Myb 1998;91:458– 65.
mutant causes apoptosis in T cells. Genes Dev 1996;10:2732– [187] Kitada S, Tamm I, Andreeff M, Reed JC. Protein kinase C
44. inhibitor 7OH-staurosporine (UCN-01) and CDK-family ki-
[168] Choi SS, Park IC, Yun JW, Sun YC, Hong SI, Shin HS. A nase inhibitor flavopiridol down regulation expression of sur-
novel Bcl-2 related gene, Bfl-1, is overexpressed in stomach vival genes and induce apoptosis in B-CLL chronic lymphocyte
cancer and preferentially expressed in bone marrow. Oncogene leukemia (B-CLL). Blood 1998;92(suppl.):102a.
1995;11:1693– 8. [188] Slater EP, Achenbach TV, Müller R. Flavopiridol: finally a
[169] Delia D, Aiello A, Soligo D, Fontanella E, Melani C, Pezzella chemotherapeutic that is Bcl-2 independent. Proc Am Assoc
F, Pierotti MA, Della Porta G. bcl-2 proto-oncogene expres- Cancer Res 2000;41:739.
sion in normal and neoplastic human myeolid cells. Blood [189] Stefanis L, Troy CM, Qi H, Shelanski ML, Greene LA. Cas-
1992;79:1291– 8. pase-2 (Nedd-2) processing and death of trophic factor-de-
[170] Popescu RA, Lohri A, de Kant E, Thiede C, Reuter J, Her- prived PC12 cells and sympathetic neurons occur independently
rmann R, Rochlitz CF. bcl-2 expression is reciprocal to p53 and of caspase-3 (CPP32)-like activity. J Neurosci 1998;18:9204–15.
c-myc expression in metastatic human colorectal cancer. Eur J [190] Fisher DA, Lakshmanan J. Metabolism and the effects of
Cancer 1998;34:1268–73. epidermal growth factor and related growth factors in mam-
[171] Torzewski M, Sarbia M, Heep H, Dutkowski P, Willers R, mals. Endocr Rev 1990;11:418– 45.
Gabbert HE. Expression of Bcl-XL, an antiapoptotic member [191] Kolibaba KS, Druker BJ. Protein tyrosine kinases and cancer.
of the Bcl-2 family, in esophageal squamous cell carcinoma. Biochim Biophys Acta 1997;1333:217– 48.
Clin Cancer Res 1998;4:577–83. [192] Velu TJ, Beguinot L, Vass WC, Willingham MC, Merlino GT,
[172] Meijerink JP, Mensink EJ, Wang K, Sedlak TW, Sloetjes AW, Pastan I, Lowy DR. Epidermal growth factor dependent trans-
de Witte T, Waksam G, Korsmeyer SJ. Hematopoietic malig- formation by a human EGF receptor proto-oncogene. Science
nancies demonstrate loss-of-function mutations of BAX. Blood 1987;238:1408– 10.
1998;91:2991– 7. [193] Dickson RB, Lippman ME. Growth factors in breast cancer.
[173] Ouyang H, Furukawa T, Abe T, Kato Y, Horii A. The BAX Endocrine Rev 1995;16:559.
gene, the promoter of apoptosis, is mutated in genetically [194] Barrett-Lee P, Travers M, Luqmani Y, Coombes RC. Tran-
unstable cancers of the colorectum, stomach, and endometrium. scripts for transforming growth factors in human breast cancer:
Clin Cancer Res 1998;4:1071–4. clinical correlates. Br J Cancer 1999;61:612– 7.
[174] Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC, [195] Murphy LC, Dotslaw H, Wong MSJ. Epidermal growth factor:
Perucho M. Somatic frameshift mutations in the BAX gene in receptor and ligand expression in human breast cancer. Semin
colon cancers of the microsatellite mutator phenotype. Science Cancer Biol 1990;1:305– 15.
1997;275:967– 9. [196] Murray PA, Barrett-Lee P, Travers M, Luqmani Y, Powles T,
[175] Yamamoto H, Sawai H, Perucho M. Frameshift somatic muta- Coombes RC. The prognostic significance of transforming
tions in gastrointestinal cancer of the microsatellite mutator growth factors in human breast cancer. Br J Cancer
phenotype. Cancer Res 1997;57:4420–6. 1993;67:1408– 12.
[176] Evan G, Littlewood T. A matter of life and cell death. Science [197] Carraway KL, Cantley LC. A new acquaintance for erbB3 and
1998;281:1317– 22. erbB4: a role for receptor heterodimerization in growth sig-
[177] Miyashita T, Reed JC. Tumor suppressor p53 is a direct nalling. Cell 1994;78:5– 8.
transcriptional activator of the human bax gene. Cell [198] Goldman R, Levy RB, Peles E, Yarden Y. Heterodimerization
1995;80:293– 9. of the erbB-1 and erbB-2 receptors in human breast carcinoma
[178] Adams JM, Cory S. The Bcl-2 protein family: arbiters of cell cells: a mechanism for receptor transregulation. Biochemistry
survival. Science 1998;281:1322–6. 1990;29:11024– 8.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 167

[199] Qian X, Decker SJ, Greene MI. p185c-neu and epidermal [220] Wilson LK, Luttrell DK, Parsons JT, Parsons SJ. pp60c-src
growth factor receptor associate into a structure composed of tyrosine kinase, myristylation, and modulatory domaons are
activated kinases. Proc Natl Acad Sci USA 1992;89:1330– 4. required for enhanced mitogenic responsiveness to epdidermal
[200] Soltoff SP, Carraway KL, Prigent SA, Gullick WG, Cantley growth factor seen in cells overexpressing c-src. Mol Cell Biol
LC. ErbB3 is involved in activating phosphatidylinositol 3-ki- 1989;9:1526– 44.
nase by epidermal growth factor. Mol Cell Biol 1994;14:3550– [221] Bolen JB. Nonreceptor tyrosine protein kinases. Oncogene
8. 1993;8:2025– 31.
[201] Bergman CL, Hung MC, Weinberg RA. Multiple independent [222] Chang JH, Wilson LK, Moyers JS, Zhang K, Parsons SJ.
activation of the neu oncogene by point mutations altering the Increased levels of b21ras-GTP and enhanced DNA synthesis
transmembrane domain of p185. Cell 1986;46:649–57. accompany elevated tyrosyl phosphorylation of GAP-associ-
[202] Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal ated proteins, p190 and p62, in c-src overexpressors. Oncogene
growth factor-related peptides and their receptors in human 1993;8:959– 67.
malignancies. Crit Rev Oncol 1995;19:183–232. [223] Maa MC, Wilson LK, Moyers JS, Vines RR, Parsons JT,
[203] Lacroix H, Iglehart JD, Skinner MA, Kraus MH. Overexpres- Parsons SJ. Identification and characterization of a cytoskele-
sion of ErbB-2 or EGF receptor proteins present in early stage ton-associated, epidermal growth factor sensitive pp60c-src sub-
mammary carcinoma is detected simultaneously in matched strate. Oncogene 1992;7:2429– 38.
primary tumors and regional metastases. Oncogene [224] Wu JK, Chikaraishi DM. Differential expression of ros onco-
1989;4:145– 51. gene in primary huan astrocytomas and astrocytoma cell lines.
[204] Yu D, Wang SS, Dulski KM, Tsai CM, Nicolson GL, Hung Cancer Res 1990;50:3032– 5.
MC. c-ErbB-2/neu overexpression enhances metastatic poten- [225] Hardie G, Hanks S. The Protein Kinase Facts Book. Academic
tial of human lung cancer cells by induction of metastasis-asso- Press, 1995.
ciated properties. Cancer Res 1994;54:3260–6. [226] Zhang W, Sloan-Lancaster J, Kitchen J, Trible RP, Samelson
[205] Boulikas T. The phosphorylation connection to cancer (review). LE. LAT: the ZAP-70 tyrosine kinase substrate that links T cell
Int J Oncol 1995;6:271–8. receptor to cellular activation. Cell 1998;92:83– 92.
[206] Cohen S, Ushiro, Stoscheck C, et al. A native 170 000 epider- [227] Moasser MM, Srethapakdi M, Sachar KS, Kraker AJ, Rosen
mal growth factor receptor-kinase complex from shed plasma N. Inhibition of Src kinase by a selective tyrosine kinase
membrane vesicles. J Biol Chem 1982;257:1523–31. inhibitor cases mitotic arrest. Cancer Res 1999;59:6145–52.
[207] Li Y, Nichols MA, Shay JW, Xiong Y. Transcriptional repres- [228] Harris W, Hill CH, Lewis EJ, Nixon JS, Wilkinson SE. Protein
sion of the D-type cyclin-dependent kinase inhibitor p167 by kinase C inhibitors. Drugs Future 1993;18:727– 35.
the retinoblastoma susceptibility gene product pRb. Cancer Res [229] Shimizu Y, Shimizu N. Cell genetic evidence of correlation of
1994;54:6078– 82. intracellular translocation of protein kinase C (PKC) and PKC-
[208] Gale NW, Kaplan S, Lowenstein EJ, Schlessinger J, Bar-Sagi mediated phosphorylation of 80-kDa protein with mitogenic
D. Grb2 mediated the EGF-dependent activation of guanine action of tumor promoters. Somat Cell Mol Genet
nucleotide exchange on Ras. Nature 1993;363:88–90. 1989;15:321– 9.
[209] Medema RH, De Vries-Smits AMM, Van Der Zon GCM. Ras [230] Shimizu T, Cao CX, Shao RG, Pommier Y. Lamin B phospho-
activation by insulin and epidermal growth factor through rylation by protein kinase calpha and proteolysis during apop-
enhanced exchange of guanine nucleotides on p21ras. Mol Cell tosis in human leukemia HL60 cells. J Biol Chem
Biol 1993;13:155– 62. 1998;273:8669– 74.
[210] Ellis C, Moran M, McCormick F, Pawson T. Phosphorylation [231] Nishizuka Y. The molecular heterogeneity of protein kinase C
of GAP and GAP-associated proteins by transforming and and its implications for cellular regulation. Nature
mitogenic tyrosine kinases. Nature 1990;343:377–81. 1988;334:661– 5.
[211] Carpenter G. Receptors for epidermal growth factor and other [232] Nishizuka Y. The protein kinase C family: heterogeneity and its
polypeptide mitogens. Annu Rev Biochem 1987;56:881– 914. implications. Annu Rev Biochem 1989;58:31– 44.
[212] Countaway JL, Northwood IC, Davis RJ. Mechanism of epi- [233] Parker PJ, Kour G, Marais RM, Mitchell F, Pears CJ, Schaap
dermal growth factor receptor at threonine 669. J Biol Chem D, Stabel S, Webster C. Protein kinase C — a family affair.
1989;264:10828– 35. Mol Cell Endocrinol 1989;65:1– 11.
[213] Hunter T, Cooper JA. Annu Rev Biochem 1985;54:897. [234] Stabel S, Parker PJ. Protein kinase C. Pharmac Ther
[214] Jallal B, Ullrich A. In: Hardie G, Hanks S, editors. The Protein 1991;51:71– 95.
Kinase Facts Book. Academic Press, 1995:126. [235] Schaap D, Parker PJ. Expression, purification, and characteri-
[215] Rueller S, Stahl C, Koehler G, Eickhoff B, Breder J, Schlaak zation of protein kinase C-o. J Biol Chem 1990;265:7301–7.
M, van der Bosch J. Sensitization of tumor cells to ribotoxic [236] Marais RM, Parker PJ. Purification and characterisation of
stress-induced apoptotic cell death: A new therapeutic strategy. bovine brain protein kinase C isotypes a, b and g. Eur J
Clin Cancer Res 1999;5:2714–25. Biochem 1989;182:129– 37.
[216] Li Y, Bhuiyan M, Alhasan S, Senderowicz AM, Sarkar FH. [237] Aflalo E, Wolfson M, Ofir R, Weinstein Y. Elevated activities
Induction of apoptosis and inhibition of c-erbB-2 in breast of protein kinase C and tyrosine kinase correlate to leukemic
cancer cells by flavopiridol. Clin Cancer Res 2000;5:223– 9. cell aggressiveness. Int J Cancer 1992;50:136– 41.
[217] Cooper JA, Howell B. The when and how of Src regulation. [238] O’Brian CA. Protein kinase C-a: a novel target for the therapy
Cell 1993;73:1051– 4. of androgen-independent prostate cancer? Oncol Reports
[218] Luttrell DK, Lee A, Lansing TJ, Crosby RM, Jung KD, 1998;5:305– 9.
Willard D, Luther M, Rodriguez M, Berman J, Gilmer TM. [239] Connolly JM, Rose DP. Expression of the invasive phenotype
Involvement of pp60c-src with two major singlaing pathways in by MCF7 human breast cancer cells transfected to overexpress
human breast cancer. Proc Natl Acad Sci USA 1994;91:83– 7. protein kinase C-a or the erbB2 proto-oncogene. Int J Oncol
[219] Maa MC, Leu TH, McCarley DJ, et al. Potentiation of epider- 1997;10:71– 6.
mal growth factor receptor-mediated oncogenesis by c-Src: [240] Liu B, Maher RJ, Hannun YA, Porter AT, Honn KV. 12(S)-
implications for the etiology of multiple human cancers. Proc HETE enhancement of prostate tumor cell invasion: selective
Natl Acad Sci USA 1995;92:6981–5. role of PKC-a. J Natl Cancer Inst 1994;86:1145– 51.
168 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

[241] Ways DK, Kukoly CA, de Vente J, Hooker JL, Bryant WO, [258] Borner C, Guadagno SN, Fabbro D, Weinstein IB. Expression
Posekany KJ, Fletcher DJ, Cook PP, Parker PJ. MCF7 breast of four protein kinase C isoforms in rat fibroblasts: distinct
cancer cells transfected with protein kinase C-a exhibit altered subcellular distribution and regulation by calcium and phorbol
expression of other protein kinase C isoforms and display a esters. J Biol Chem 1992;267:12892– 9.
more aggressive neoplastic phenotype. J Clin Invest [259] Borner C, Guadagno SN, Hsiao WLW, Fabbro D, Barr M,
1995;95:1906– 15. Weinstein IB. Expression of four protein kinase C isoforms in
[242] Baxter G, Oto E, Daniel-Issakani S, Strulovici B. Constitutive rat fibroblasts: differential alterations in ras-, src- and fos-trans-
presence of a catalytic fragment of protein kinase Co in a small formed cells. J Biol Chem 1992;267:12900– 10.
cell lung carcinoma cell line. J Biol Chem 1992;267:1910– 7. [260] Lee W, Mitchell P, Tjian R. Purified transcription factor AP-1
[243] Guillem JG, O’Brian CA, Fitzer CJ, Forde KA, LoGerfo P, interacts with TPA-inducible enhancer elements. Cell
Treat M, Weinstein IB. Altered levels of protein kinase C and 1987;49:741– 52.
Ca2 + -dependent protein kinases in human colon carcinomas. [261] Adler V, Polotskaya A, Wagner F, Kraft AS. Affinity-purified
Cancer Res 1987;47:2036–9. c-jun amino-terminal protein kinase requires serine/threonine
[244] Kopp R, Noelke B, Sauter G, Schildberg FW, Paumgartner G, phosphorylation for activity. J Biol Chem 1992;267:17001–5.
Pfeiffer A. Altered protein kinase C activity in biopsies of [262] Boyle WJ, Smeal T, Defize LHK, Angel P, Woodget JR, Karin
human colonic adenomas and carcinomas. Cancer Res M, Hunter T. Activation of protein kinase C decreases phos-
1991;51:205– 10. phorylation of c-jun at sites that negatively regulate its DNA-
[245] Sakanoue Y, Hatada T, Kusunoki M, Yanagi H, Yamamura T, binding activity. Cell 1991;64:573– 84.
Utsunomiya J. Protein kinase C activity as marker for col- [263] Krongrad A, Bai G. C-fos promoter insensitivity to phorbol
orectal cancer. Int J Cancer 1991;48:803–6. ester and possible role of protein kinase C in androgen-indepen-
[246] Mills KJ, Bocckino SB, Burns DJ, Loomis CR, Smart RC. dent cancer cells. Cancer Res 1994;54:6073– 7.
Alterations in protein kinase C isozymes a and b2 in activated [264] DiDonato JA, Hayakawa, Rothwarf DM, Zandi E, Karin M.
Ha-ras containing papillomas in the absence of an increase in A cytokine-responsive IkappaB kinase that activates the tran-
diacylglycerol. Carcinogenesis 1992;13:1113–20. scription factor NF-kappaB. Nature 1997;388:548– 54.
[247] Marian B, Hochegger K, Partik G, Schörkhuber M. Inhibition [265] Li JJ, Westergaard C, Ghosh P, Colburn NH. Inhibitors of
of PCK inhibits growth an induces apoptosis in colorectal both nuclear factor-kappaB and activator protein-1 activation
tumor cells. Proc Am Assoc Cancer Res 1999;40:165. block the neoplastic transformation response. Cancer Res
[248] Li H, Zhao L, Yang Z, Funder JW, Liu JP. Telomerase is 1997;57:3569– 76.
controlled by protein kinase Ca in human breast cancer cells. J [266] Sumitomo M, Tachibana M, Ozu C, Asakura H, Murai M,
Biol Chem 1998;273:33436–42. Hayakawa M, Nakamura H, Takayanagi A, Shimizu N. Induc-
[249] Ohno S, Suzuki K. Protein kinase C (vertebrates). In: Hardie tion of apoptosis of cytokine-producing bladder cancer cells by
G, Hanks S, editors. The Protein Kinase Facts Book. Academic adenovirus-mediated IkBa overexpression. Human Gene Ther
Press, 1995:80– 8. 1999;10:37– 47.
[250] Hsiao WLW, Housey GM, Johnson MD, Weinstein IB. Cells [267] Paillard F. Induction of apoptosis with I-kB, the inhibitor of
that overproduce protein kinase C are more susceptible to NF-kB. Human Gene Ther 1999;10:1– 3.
transformation by an activated H-ras oncogene. Mol Cell Biol [268] Bodnar AG, Kim NW, Effros RB, Chiu CP. Mechanism of
1989;9:2641– 7. telomerase induction during T cell activation. Exp Cell Res
[251] Chiarugi V, Bruni P, Pasquali F, Magnelli L, Basi G, Ruggiero 1996;228:58– 64.
M, Farnarraro M. Synthesis of diacylglycerol de novo is re- [269] Ku WC, Cheng AJ, Wang TC. Inhibition of telomerase activity
sponsible for permanent activation and down-regulation of by PKC inhibitors in human nasopharyngeal cancer cells in
protein kinase C in transformed cells. Biochem Biophys Res culture. Biochem Biophys Res Commun 1997;241:730–6.
Commun 1989;164:816–23. [270] Blackburn EH. Telomerasaes. Annu Rev Biochem
[252] Diaz-Laviada I, Larrodera P, Diaz-Meco MT, Cornet ME, 1992;61:113– 29.
Guddal PH, Johannsen T, Moscat J. Evidence for a role of [271] deLange T. Activation of telomerase in a human tumor. Proc
phosphatidylcholine-hydrolysing phospholipase C in the regula- Natl Acad Sci USA 1994;91:2882– 5.
tion of protein kinase C by ras and src oncogenes. EMBO J [272] Lundblad V, Wright WE. Telomeres and telomerase: a simple
1990;9:3907– 12. picture becomes complex. Cell 1996;87:369– 75.
[253] Huang M, Chida K, Kamata N, Nose K, Kato M, Homma Y, [273] Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD,
Takenawa T, Kuroki T. Enhancement of inositol phospholipid Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW.
metabolism and activation of protein kinase C in ras-trans- Specific association of human telomerase activity with immortal
formed rat fibroblasts. J Biol Chem 1988;263:17975–80. cells and cancer. Science 1994;266:2011– 5.
[254] Preiss J, Loomis CR, Bishop WR, Stein R, Neidel JE, Bell RM. [274] DePaoli-Roach A, Roach PJ, Zucker KE, Smith SS. Selective
Quantitative measurement of sn-1,2-diacylglycerols present in phosphorylation of human DNA methyltransferase by protein
platelets, hepatocytes and ras- and sis-transformed normal rat kinase C. FEBS Lett 1986;197:149– 53.
kidney cells. J Biol Chem 1986;261:8597–600. [275] Koenig A, Schulz-Jander M, Schwartz GK, Al-Katib A,
[255] Price BD, Morris JD, Marshall CJ, Hall A. Stimulation of Gabrilove JL. Inhibition of protein kinase C (PKC) arrests cell
phosphatidylcholine hydrolysis, diacylglycerol release and growth, induced apoptosis, and alters the effect of basic fibrob-
arachidonic acid production by oncogenic ras is a consequence last growth factor (bFGF) on bcl-2 expression in chronic
of protein kinase C activation. J Biol Chem 1989;264:16638– 43. lymphoid malignancies. Blood 1996;15:67a.
[256] Wolfman A, Macara IG. Elevated levels of diacylglycerol and [276] Basu A. The potential of protein kinase C as a target for
decreased phorbol ester sensitivity in ras-transformed fibrob- anticancer treatment. Pharmacol Ther 1993;59:257– 80.
lasts. Nature 1987;325:359–61. [277] Basu A, Miura A, Akkaraju GR, Johnson DE. Regulation of
[257] Borner C, Guadagno SN, Hsieh LL, Hsiao WLW, Weinstein cell death by protein kinase C. Proc Am Assoc Cancer Res
IB. Transformation by a ras oncogene causes increased expres- 1999;40:165.
sion of protein kinase C-a and decreased expression of protein [278] Reddy DS. Development of multidrug resistance protein in-
kinase C-o. Cell Growth Diff 1990;1:653–60. hibitors. Drugs Future 1997;22:653– 60.
H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170 169

[279] Gottesman MM, Pastan I. Biochemistry of multidrug resistance [300] Blobe GC, Sachs CW, Khan WA, Fabbro D, Stabel S, Wetsel
mediated by the multidrug transporter. Annu Rev Biochem WC, Obeid LM, Fine RL, Hannun YA. Selective regulation of
1993;62:385– 427. expression of protein kinase C (PKC) isoenzymes in multidrug
[280] Cole SPC, Deeley RG. Multidrug resistance mediated by the resistant MCF-7 cells. J Biol Chem 1993;268:658– 64.
ATP-binding cassette transporter protein MRP. Bioessays [301] Fine RL, Patel J, Chabner BA. Phorbol esters induce multidrug
1998;20:931– 40. resistance in human breast cancer cells. Proc Natl Acad Sci
[281] Cole SPC, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE, USA 1988;85:582– 6.
Almquist KC, Stewar AJ, Kurz EU, Duncan AM, Deeley RG. [302] Gupta KP, Ward NE, Gravitt KR, Bergman PJ, O’Brian CA.
Overexpression of a transporter gene in a multidrug-resistant Partial reversal of multidrug resistance in human breast cancer
human lung cancer cell line. Science 1992;258:1650–4. cells by an N-myristoylated protein kinase C-a pseudosubstrate
[282] Krishnamachary N, Center MS. The MRP gene associated with peptide. J Biol Chem 1996;271:2102– 11.
a non-P-glycoprotein multidrug resistance encodes a 190-kDa [303] Sato W, Yusa K, Naito M, Tsuruo T. Staurosporin, a potent
membrane bound glycoprotein. Cancer Res 1993;53:3658– 61. inhibitor of C-kinase, enhances drug accumulation in multidrug
[283] Hickman JA. Apoptosis induced by anticancer drugs. Cancer resistant cells. Biochem Biophys Res Commun 1990;173:1252–
Metastas Rev 1992;11:121–39. 7.
[284] Lowe SW, Rule HE, Jacks T, Housman DE. p53-dependent [304] Ohno S, Kawaski H, Imajoh S, Suzuki K. Tissue-specific
apoptosis modulates the cytoxicity of anticancer agents. Cell expression of three distinct types of rabbit protein kinase C.
1993;74:957– 67. Nature 1987;325:161– 6.
[285] Singh G, Dorward A, Moorehead R, Sweet S. Novel mecha- [305] Yu G, Ahmad S, Aquino A, Fairchild CR, Trepel J, Ohno S,
nisms of resistance to cancer chemotherapy. Cancer J Ohno K, Suzuki T, Tsuruo T, Cowan K, Glazer R. Transfec-
1995;8:304. tion with protein kinase Ca confers increased multidrug resis-
[286] Preisler HD. Multidrug resistance is more than MDR1 activity. tance to MCF-7 cells expressing P-glycoprotein. Cancer
Leuk Res 1995;19:429–31. Commun 1991;3:181– 9.
[287] Higgins CF. The multidrug resistance P-glycoprotein. Curr [306] Hamada H, Hagiwara K, Nakajima T, Tsuruo T. Phosphoryla-
Opin Cell Biol 1993;5:684–7. tion of the M 170 000 to 180 000 glycoprotein specific to
[288] Müller M, Meijer C, Zaman GJR, Borst P, Scheper RJ, Mulder multidrug-resistant tumor cells: effects of verapamil, trifluoper-
azine and phorbol esters. Cancer Res 1987;47:2860– 5.
NH, de Vries EGE, Jansen PLM. Overexpression of the gene
[307] Smith V, Workman P, Kelland LR. Drug resistance to the
encoding the multidrug resistance-associated protein results in
cyclin-dependent kinase (cdk) inhibitor, flavopiridol. Proc Am
increased ATP-dependent glutatione S-conjugate transport.
Assoc Cancer Res 2000;41:32.
Proc Natl Acad Sci USA 1994;91:13033–7.
[308] Bible KC, Boerner SA, Kirkland K, Anderl KL, Bartelt D,
[289] Higgins CF. ABC transporters: from micro-organisms to man.
Svingen PA, Kottke TJ, Lee YK, Eckdahl S, Stalboerger PG,
Annu Rev Cell Biol 1992;8:67–113.
Jenkins RB, Kaufmann SH. Characterization of an ovarian
[290] Jedlitschky G, Leier I, Buchholz U, Center M, Keppler D.
carcinoma cell line resistant to cisplatin and flavopiridol. Clin
ATP-dependent transport of glutathione S-conjugates by the
Cancer Res 2000;6:661– 70.
multidrug resistance-associated protein. Cancer Res
[309] Hooijberg JH, Broxterman HJ, Scheffer GL, Vrasdonk C,
1994;54:4833– 6.
Heijn M, de Jong MC, Scheper RJ, Lankelma J, Pinedo HM.
[291] Broxterman HJ, Giaccone G, Lankelma J. Multidrug resistance
Potent interaction of flavopiridol with MRP1. Br J Cancer
proteins and other drug transport-related resistance to natural
1999;81:269– 76.
product agents. Curr Opin Oncol 1995;7:532–40.
[310] Hooijberg JH, Broxterman HJ, Heijn M, Flex DL, Lankelma J,
[292] Chin KV, Ueda K, Pastan I, Gottesman MM. Modulation of Pinedo HM. Modulation by (iso)flavonoids of the ATPase
activity of the promoter of the human MDR1 gene by Ras and activity of the multidrug resistance protein. FEBS Lett
p53. Science 1992;255:459–62. 1997;413:344– 8.
[293] Scanlon KJ, Kashani-Sabet M, Tone T, Funato T. Cisplatin [311] Ciomei M, Pastori W, Albanese C, Isacchi A. Induction of
resistance in human cancers. Pharmacol Ther 1991;52:385– 406. apoptosis on tumor or normal cells: comparison between
[294] Delaporte C, Larsen AK, Dautry F, Sablon AJ. Influence of Flavopiridol and camptothecin. Proc Am Assoc Cancer Res
myc overexpression on the phenotypic properties of Chinese 1999;40:580.
hamster lung cells resistant to antitumor agents. Exp Cell Res [312] Marconcini L, Marchio S, Morbidelli L, Cartocci E, Albini A,
1991;197:176– 82. Ziche M, Bussolino F, Oliviero S. c-fos-induced growth factor/
[295] Kim SH, Tung YT, Heo K, Kim DW, Kang CD, Chung BS. vascular endothelial growth factor D induces angiogenesis in
Presence of the Raf-1 responsive element in human MDR1 vivo and in vitro. Proc Natl Acad Sci USA 1999;96:9671–6.
promoter. Korean J Biochem 1994;26:137–43. [313] Mukhopadhyay D, Tsiokas L, Sukhatme VP. Wild-type p53
[296] Chambers TC, Pohl J, Raynor RL, Kuo JF. Identification of and v-Src exert opposing influences on human vascular en-
specific sites in human P-glycoprotein phosphorylated by dothelial growth factor gene expression. Cancer Res
protein kinase C. J Biol Chem 1993;268:4592–5. 1995;55:6161– 5.
[297] Chambers TC, Zheng B, Kuo JF. Regulation of phorbol ester [314] Robinson CS, Slee AM, Kerr JS. Flavopiridol inhibits angio-
and protein kinase C inhibitors of P-glycoprotein phosphoryla- genesis. FASEB J 1997;11:A63.
tion and relationship of drug accumulation in multidrug-resis- [315] Brooks PC, Silletti S, von Schalscha TL, Friedlander M,
tant human KB cells. Mol Pharmacol 1992;41:1008–15. Cheresh DA. Integrins, proteases in angiogenesis. In: Proceed-
[298] Posada J, McKeegan E, Worthington K, Morin M, Jaken S, ings Angiogenesis and Cancer, Orlando, FL, January, 1998:24–
Tritton T. Human multidrug resistant KB cells overexpress 8.
protein kinase C: involvement in drug resistance. Cancer Com- [316] Griffioen AW, Tromp SC, Hillen HFP. Angiogenesis modulates
mun 1989;1:285– 92. the tumour immune response. Int J Exp Path 1998;79:363–8.
[299] Ward NE, O’Brian C. Distinct patterns of phorbol ester-in- [317] Griffioen AW, Damen CA, Mayo KH, Barendsz-Janson AF,
duced downregulation of protein kinase C activity in adri- Martinotti S, Blijham GH, Groenewegen G. Angiogenesis in-
amycin-selected multidrug resistant and parental murine hibitors overcome tumor induced endothelial cell anergy. Int J
fibrosarcoma cells. Cancer Lett 1991;58:189–93. Cancer 1999;80:315– 9.
170 H.H. Sedlacek / Critical Re6iews in Oncology/Hematology 38 (2001) 139–170

[318] Melder RJ, Koenig GC, Witwer BP, Safabakshs N, Munn LL, [326] König A, Schwartz GK, Mohammad RM, Al-Katib A,
Jain RK. During angiogenesis, vascular endothelial growth Gabrilove JL. The novel cyclin-dependent kinase inhibitor
factor and basic fibroblast growth factor regulate natural killer Flavopiridol downregulates Bcl-2 and induces growth arrest
cell adhesion to tumor endothelium. Nature Med 1996;2:992– 7. and apoptosis in chronic B-cell leukemia lines. Blood
[319] Piali L, Fichtel A, Terpe HJ, Imhof BA, Gisler RH. Endothelial 1997;90:4307– 12.
vascular cell adhesion molecule 1 expression is suppressed by [327] Sugiyama K, Akiyama T, Shimizu M, Courge C, Gescher A,
melanoma and carcinoma. J Exp Med 1995;181:811–6. Tamaoki T, Akinaga S. Differences between human wild-type
[320] Webster KR, Mulheron JG, Quian L, Wautlet BS, Chao ST, and UCN-01 resistant A549 lung carcinoma cells in susceptibil-
ity towards apoptosis induction and changes in G1 checkpoint
Stratton L, Kelly YF, Misra RN, Sack JS, Hunt JT, Kimball
function by UCN-01. Proc Am Assoc Cancer Res 1999;40:305.
D, Kim KS. Thio-flavopiridol, a selective inhibitor of Cdk1.
Proc Am Assoc Cancer Res 1999;119:789.
[321] Shapiro GI, Edwards CD, Sausville EA, Rollins BJ. Flavopiri-
dol produces growth arrest and apoptosis in NSCLC cell lines. Biography
Proc Am Assoc Cancer Res 1997;38:472.
[322] Shapiro GI, Koestner DA, Sausville EA, Rollins BJ. Flavopiri- H.H. Sedlacek — Education: Veterinary Medicine
dol-mediated cdk inhibition results in p53-independent apopto- (University of Giessen); DVM, Endocrine Pharmacol-
sis in non-small cell lung cancer cell lines. Proc Am Assoc ogy (1968). Awards: Adjunct Professor for Tumor Biol-
Cancer Res 1998;39:70. ogy, Medical School, University of Marburg (1994).
[323] Shapiro GI, Koestner DA, Matranga CB, Rollins BJ.
Occupational activities: Schering AG; Berlin/Bergka-
Flavopiridol induces cell cycle arrest and p53-independent
apoptosis in non-small cell lung cancer cell lines. Clin Cancer men 1969 –1971 Immunotoxicology; Behringwerke,
Res 1999;5:2925– 38. Marburg; 1972 –1983, Head of Immunopharmacology
[324] Werner JL, Greaney C, Spriggs D, Kelsen DK, Schwartz GK. and Tumor Pharmacology; 1984 –1991 Director of On-
The enhancement of chemotherapy-induced apoptosis by cology Research; 1991 –1996, Head of Strategic Plan-
flavopiridol (FLAVO) in gastric cancer cells is sequence-depen- ning of R&D; Hoechst Marion Roussel/Aventis
dent (meeting abstract). Proc Annu Meet Am Soc Clin Oncol
Pharma Germany, 1997 –2000, Coordinator of Gene
1997;16:A1972.
[325] Jung CP, Motwani MV, Schwartz GK. Flavopiridol (F) poten-
Therapy R&D.
tiates gemcitabine (G) induced apoptosis in association with Major achievement: Essential Contribution to Dis-
downregulation of ribonucleotide reductase (RR). Proc Am covery and Development of Flavopiridol (Innovation
Assoc Cancer Res 2000;41:32. Award of German Industry in 1998).

You might also like