• Non ci sono risultati.

34 Apoptosis in Melanoma

N/A
N/A
Protected

Academic year: 2022

Condividi "34 Apoptosis in Melanoma"

Copied!
13
0
0

Testo completo

(1)

605

Apoptosis in Melanoma

Approaches to Therapy

Heike Röckmann and Dirk Schadendorf

C

ONTENTS

I

NTRODUCTION

P

ROGRAMMED

C

ELL

-D

EATH

P

ATHWAYS

A

POPTOSIS

D

EFICIENCY AND

M

ELANOMA

R

EFERENCES

Summary

Apoptosis deficiency seems to be involved in the high resistance of melanoma to therapeutic treat- ment. This has come into focus because the cytotoxic effects of chemotherapeutic agents via apoptosis are known. Extensive investigations have been made analyzing the role of alterations in the apoptotic pathway in melanoma. The molecular changes affect antiapoptotic, as well as proapoptotic, processes and survival signals and involve various molecules. These mechanisms are also discussed in light of their use in further therapeutical strategies. Actually, a number of these findings have already been employed to test their therapeutical applicability in melanoma treatment. Furthermore, two concepts have been translated from the cell system via animal models into clinical trials.

Key Words: Melanoma; apoptosis; apoptosis deficiency; death receptors; caspases; Bcl-2-family; p53.

INTRODUCTION

Most chemotherapeutic drugs act through induction of apoptosis (programmed cell death). In 1972, Kerr et al. described an experimentally induced killing of tumor cells that involved a coordinated cell disintegration following typical morphological changes, coining the term apoptosis (1). The cytotoxic effect of chemotherapeutic agents appears mainly contributed by apoptosis (2,3). The weak response of metastatic melanoma to anticancer agents gives rise to the hypothesis that the chemoresistance of this malig- nancy is caused by raising its apoptotic threshold (2). Inactivation of apoptosis is a

“hallmark of cancer,” an obligate ritual in the malignant transformation of benign cells (4). As a result, these cells enhance their chances of survival and increase their resistance to chemotherapy (3). Indeed, Staunton et al. (5) demonstrated a constitutive low level of spontaneous apoptosis in melanoma cells compared with other malignant cell types. An intensive search for cell death factors altered in melanoma has been made. It has been

34

From: From Melanocytes to Melanoma: The Progression to Malignancy Edited by: V. J. Hearing and S. P. L. Leong © Humana Press Inc., Totowa, NJ

(2)

shown that, indeed, various molecular changes in cell-death control in melanoma are present, and three types can be distinguished: activation of antiapoptotic processes, inactivation of proapoptotic effectors, and reinforcement of survival signals.

PROGRAMMED CELL-DEATH PATHWAYS

Apoptosis (programmed cell death) represents a complex genetic program consisting of several pathways that are summarized in Fig. 1. Tremendous efforts have been made to discover and describe the molecular mechanisms of apoptosis, which are discussed elsewhere in detail (6). Briefly, depending on cell type and stimulus, a complex net of sensor and regulator proteins is activated and balanced and there is no unique linear or defined pathway. However, to simplify, two main well-characterized caspase-activating cascades that regulate apoptosis are currently known.

One cascade is triggered from the cell surface by death receptors and the other is initiated by changes of mitochondrial membrane integrity (7). Oligomerization of sur- face receptors is followed by recruitment of adapter molecules, such as Fas-associated protein with death domain (FADD) and the initiator caspases-8 and -10 (8) into the death-inducing signaling complex (DISC). The subsequent autocatalytic cleavage of procaspase-8 or -10 is followed by activation of effector caspases (e.g., caspase-3) (9) and induction of specific endonucleases, resulting in DNA fragmentation (10,11). These two pathways converge with the activation of effector caspases and induction of specific endonucleases, resulting in DNA fragmentation and cleavage of nuclear proteins essen- tial for nuclear and cellular structure, DNA-repair, and DNA-replication (12,13).

The family of death receptors include CD95 (Fas/APO-1), tumor necrosis factor (TNF)-R1, TNF-receptor apoptosis-inducing ligand receptor (TRAIL-R)1 and TRAIL-R2, DR3 (death receptor 3), and DR6 (reviewed in Locksley et al., ref. 14). Among them, the CD95 receptor, TRAIL-R1 and TRAIL-R2 are the most efficient mediators of apoptosis.

Several studies suggest that death receptor–ligand interaction is involved in tumor sen- sitivity toward chemotherapeutic drugs (15–17). The extrinsic pathway is regulated on multiple levels, whereas the death-inducing signaling complex can also recruit negative and positive regulators of caspase-8 (18,19). Furthermore, in type II cells, the initial activation of caspase-8 is not sufficient. Here, caspase-8 cleaves the Bcl-2 family mem- ber, BID, which translocates to the mitochondrial membrane and activates the intrinsic pathway (20). By this mechanism, the death receptor and the mitochondrial pathway is connected.

The intrinsic pathway involves mitochondrial release of cytochrome-c, which binds apoptotic protease activating factor (Apaf)-1, and, in the presence of adenosine triphos- phate (ATP), coordinates a series of conformational changes that allow the oligomeriza- tion of Apaf-1 into a ring-like complex, referred to as the “apoptosome” (21). The apoptosome binds and activates caspase-9 into the complex (9,22,23), which, in turn, recruits and activates effector caspases (e.g., caspase-3). These are considered as execu- tors of apoptosis.

There are various additional points of control that can modulate apoptosis after

cytochrome-c release. The family of inhibitors of apoptosis (IAP), containing X-chro-

mosome-linked IAP (XIAP), neuronal IAP (NIAP), melanoma (ML)-IAP, and survivin,

can interfere with the formation of the apoptosome and activation of the downstream

caspases (Figs. 1 and 2). IAPs are inhibited by other proapoptotic factors released by the

(3)

607

Fig. 1. The apoptotic pathway. In this simplified scheme, the extrinsic pathway is initiated by oligomerization of death receptors after binding of the ligand and recruitment of initiator caspases (caspase-8 and caspase-10). The intrinsic pathway involves the translocation of proapoptotic Bcl-2 family members to mitochondria and release of cytochrome-c into the cytosol, oligomerization of apoptotic protease activating factor-1 (Apaf-1) in a complex with caspase-9, and the subsequent activation of caspase-3. Receptor-initiated signals can be transduced through the mitochondrial pathway, for example, through cleavage of Bid (for details see page 608).

(4)

Fig. 2. Caspase regulatory proteins. The inhibitor of apoptosis (IAP) gene family encodes a group of structurally related proteins (e.g., IAP-1, IAP-2, X-chromosome-linked IAP, survivin, and melanoma IAP) that are able to suppress caspase function. They are thought to directly inhibit certain caspases and are controlled by further inhibitory proteins of IAP, Smac (second mitochon- drial activator of caspases)/Diablo (direct IAP-binding protein with low pI).

mitochondria, the second mitochondrial activator of caspases (Smac)/Diablo and Omi/

OtrA (24). Furthermore, p53 can modulate the expression of apoptotic effectors, and heat-shock proteins can also regulate the formation of the apoptosome.

APOPTOSIS DEFICIENCY AND MELANOMA

Wild-type p53 promotes cell cycle arrest and apoptosis in response to DNA-damaging drugs most likely by controlling transcriptional regulation of target genes, such as Bcl-2 and Bax (25,26) (Fig. 1), and acts, therefore, as a tumor suppressor. p53 mutations are common genetic alterations in human cancer. Melanoma cell lines expressing wild-type p53 exhibit a higher response to anticancer agents than melanoma with mutant p53 (27).

Mutation of p53 was associated with metastatic potential (28). Abnormal phosphoryla- tion of p53 by Ck2 kinase was associated with melanoma resistance to radiotherapy (29).

A phase I dose-escalation study of single intratumoral injection of a replication-defec- tive adenoviral expression vector containing p53 was performed by Dummer et al. in patients with metastatic melanoma. In this study, the therapeutic approach was proven safe, feasible, and biologically effective (30).

Another very common specific gene defect in human melanoma is mutation of acti- vated ras (31). Ras proteins are regulators of multiple signal pathways that control cell growth, differentiation, and apoptosis. The assumed mechanism by which the activated ras oncogene is involved in drug resistance is an upregulation of Bcl-2 expression (32).

A decreased CD95 surface expression in ras transfectants was demonstrated in mela-

(5)

noma cell lines (33). Indeed, overexpression of activated mutants of N-ras increase cisplatin resistance in melanoma cells and in a SCID (severe combined immunodeficient) mouse model (34). Improved understanding of the molecular mechanisms of Ras processing and membrane targeting provided important tools for the development of molecules that inhibit the association of Ras with the inner cell membrane. Such molecules include inhibitors of farnesyltransferase, prenyl-CAAX protease, methyl- transferase, and inhibitors, such as trans-farnesyl thiosalicylic acid (Fig. 3). Farnesyl thiosalicylic acid, a Ras antagonist, suppresses melanoma growth in vitro and in vivo through a combination of cytostatic and proapoptotic effects (35). Furthermore, a recent study evaluated the effect of farnesyl thiosalicylic acid treatment in combination with dacarbazine on established human melanoma xenografts grown in mice. A significant tumor growth paralleled by an acceptable toxicity profile was shown in a mouse system (36) (Fig. 3).

B-Raf, a Ras effector molecule, was found to be mutated in 66% of human melanomas (37). Previous studies indicated that wild-type B-Raf may inhibit apoptosis downstream of cytochrome-c release by activating nuclear factor (NF)-NB (38).

Extensive efforts have been made in analyzing the involvement of death receptors in melanoma cell death. Downregulation, loss, and mutation of CD95/Fas receptor in melanoma have been described, resulting in triggering resistance to CD95L/Fas ligand (39,40). A respective correlation to clinical response was demonstrated by Mouawad et al. (41). They reported that melanoma patients with low clinical response to various drugs (cisplatin, recombinant interleukin-2, and interferon-D) exhibited a significant increase of soluble sCD95 and sCD95L in the plasma after drug treatment, whereas in the plasma of responders, no changes in sCD95 or sCD95L levels were observed.

Various studies demonstrated that, in contrast to the CD95 receptor, TRAIL-R2 is widely expressed in melanoma. Furthermore, melanoma cell lines were shown to undergo apoptosis after exposure with recombinant TRAIL very readily, whereas melanocytes did not demonstrate such a high TRAIL sensitivity (42). This study also showed that TRAIL-R expression is not predictive of sensitivity. There are some known agents available to upregulate TRAIL-R expression, such as cisplatin, betulinic acid, CD437 retinoid, and TNF-D (overviewed in Hersey et al., ref. 43). Furthermore, Griffith et al.

demonstrated increased apoptosis in melanoma cells after TRAIL-expressing adenovi- ral infection as a possible therapeutic approach (44). Sensitivity toward death-receptor signaling in melanoma cells was also increased using a soluble NF-NB inhibitor (45).

Bcl-2 appears to influence response to chemotherapy by inhibiting apoptosis induc- tion by many cytostatic drugs, including alkylating agents, topoisomerase inhibitors, antimetabolites, and others. The high expression of Bcl-2 in human melanoma and other tumors has been correlated with resistance to chemotherapy and decreased survival (46).

Furthermore, Raisova et al. showed that a low Bax:Bcl-2 ratio might be characteristic for

drug-resistant melanoma cells (47). Additionally, the oncogenic potential of Bcl-2 seems

to be involved in melanoma angiogenesis through vascular endothelial growth factor

(VEGF) mRNA stabilization and hypoxia-inducible factor (HIF)-mediated transcrip-

tional activity (48). C-Myc low-expressing compared with high-expressing melanoma

lines demonstrated an increased cisplatin sensitivity (49). In a human melanoma

xenograft in SCID mouse models, the improvement of chemosensitivity of dacarbazine

in combination with bcl-2 oligo–antisense (Augmerosen, Gentasense, G-3139) could be

demonstrated (50). Furthermore, Heere-Ress et al. demonstrated recently how antisense

(6)

610

Fig. 3. Ras proteins carry C-terminal lipid modifications, through which they associate with cellular membranes and which are essential for their transforming activity. After initial cytosolic farnesylation, Ras proteins translocate to intercellular membranes. Here they undergo successive removal of the terminal –AXX residues, methylation, and further modification by S-acetylation. (Reproduced with permission from ref. 86.)

(7)

oligonucleotides reduced Bcl-X

L

expression and enhanced the chemosensitivity of melanoma cell lines to cisplatin (51). A recent study showed that the simultaneous downregulation of Bcl-2 and Bcl-X

L

expression and induction of apoptosis by antisense oligonucleotides in melanoma cells of different clinical stage may provide an additional clinical benefit (52).

Antisense oligonucleotides are short, single-stranded nucleotides that bind comple- mentary to their respective target mRNA, thus, inhibiting translation and initiating deg- radation of the targeted mRNA. Antisense oligonucleotides directed against specific genes, such as Bcl-2, associated with neoplastic progression are currently being evalu- ated in several clinical trials. Therefore, mouse systems (50) have been translated into clinical phase I/IIa studies, in which 6 of 14 treated patients showed an antitumor response associated with low Bcl-2 levels and increased apoptosis in melanoma biopsies (82).

Recently, a worldwide phase III study recruited 770 patients with metastatic melanoma to compare the efficiency of the standard chemotherapy dacarbazine alone and dacarbazine plus Bcl-2 antisense. The final analysis is currently being performed.

Antisense molecules for Bcl-X

L

and c-Myc are available, and clinical application can be expected.

Caspase proteases are initially synthesized as precursor proteins with little or no enzymatic activity. The cleaved proteins are the primary apoptotic executers, which act in a cascade ultimately leading to the cleavage of substrates that produce the character- istic features of apoptosis. Caspase proteases are cleaved by upstream molecules (such as caspases, FADD ([Fas-associated protein with death domain], or Apaf-1) and are further controlled by a variety of proteins that directly interact with proteases: IAPs and FLICE (FADD-like interleukin-1-E-converting enzyme inhibitory proteins (FLIPs).

Caspase inhibition is achieved by proteins of the IAP family (Fig. 2), which are struc-

turally related by their baculovirus IAP repeat domain. In melanoma, two members of

the IAP family (survivin and ML-IAP/Livin) and FLIP have been associated with tumor

progression. One of the best-characterized members in melanoma is survivin, which

exerts its effect by directly inhibiting caspases and was found in melanoma only (cell

lines, metastatic lesions, and invasive melanoma) compared with melanocytes (54). In

contrast to other apoptotic inhibitors, survivin expression is cell-cycle dependent. In

early mitosis, survivin, linked to the microtubules of the mitotic spindle, inhibits the

activation of caspase 3 (55). A recent study by Gradilone et al. showed a significant

correlation between survivin expression and outcome of sentinel lymph node-positive

melanoma patients (56). Antisense oligonucleotides directed against this molecule were

shown to induce spontaneous apoptosis in melanoma in vitro (54). This was proven by

the same group by a phosphorylation-defective Thr34 baculoviral IAP repeat (BIR)

mutant (the relevant domain in IAPs), which prevented tumor formation and slowed the

growth of established tumors in a melanoma xenograft model (57), suggesting that

therapeutic targeting of survivin might also be beneficial in patients with recurrent or

metastatic melanoma. An additional IAP molecule has been discovered in melanoma by

two other groups. ML-IAP/livin is expressed in developed tissues, including melano-

cytes, but is predominantly overexpressed in melanoma (58,59). ML-IAP acts directly on

the mitochondrial pathway by directly inhibiting caspase-9, caspase-3, and the

proapoptotic factor, Smac/Diablo (60). Furthermore, this molecule was identified as a

possible target for immune-mediated tumor destruction in melanoma (58,61). Further

studies found that transfection of an antisense construct against livin could trigger

(8)

apoptosis specifically in melanoma cell lines expressing livin mRNA. This was associ- ated with an increase in DNA fragmentation and in DEVD-like caspase activity (58).

A mitochondria-derived activator of caspase (Smac/Diablo) construct was shown in vitro to sensitize melanoma to TRAIL-induced apoptosis as a very selective tool inhib- iting IAP (62). There are strong reasons why caspase inhibitors can be considered char- acteristic of the drug-resistant phenotype in melanoma cells; caspase inhibitors enhance the ability of the melanoma cells to resist apoptosis and present a rewarding therapeutic target.

FLIP was shown to inhibit caspase-8 and, therefore, to inhibit apoptosis induced by receptor-associated cytokines (e.g., TNF-D, CD95L/Fas ligand, TRAIL). FLIP was shown to be overexpressed in melanoma and associated with resistance (63). Neverthe- less, endogenous levels of FLIP do not necessarily correlate with drug response in melanoma patients (40,64).

Apaf-1 represents an essential downstream molecule to induce apoptosis. Soengas et al. reported cases of drug-resistant malignant melanomas in which Apaf-1 expression was impaired (65). Here, cytostatic drugs induced cytochrome-c release, but failed to induce caspase-9 activation. This could be reversed by reactivating Apaf-1 with the demethylating agent, 5-aza-2-deoxycytidine, in vitro.

Various studies suggest that increased antiapoptotic, heat-shock protein (HsP) ex- pression, such as HsP70 and HsP90, may be related to drug resistance (66,67). HsP70 antagonizes the caspase-independent apoptotic-inducing factor (AIF) (68). Hsp70 may also interfere with Apaf-1, by inhibiting the oligomerization and/or by blocking the recruitment of caspase-9 to the apoptosome (69). HsP70 was shown to be upregulated in primary melanoma and cell lines (70,71), and an association with resistance to ultra- violet radiation was seen (72). Clinical trials using 17-AAG for Hsp90 inhibition in advanced cancer are currently ongoing (73).

Neef et al. identified the human pleckstin-homology-like domain family A, member 1 (PHLDA1)/TDAG51 gene, which was shown to be downregulated in metastatic mela- noma and associated with apoptosis resistance (74). PHLDA1 expression was associated with reduced cell growth, cloning efficiency, and colony formation, and increased basal apoptosis. Chemosensitivity to doxorubicin and camptothecin was enhanced. Moreover, recently, a group of human melanoma cell populations that are heterogeneously susceptible to C2-ceramide-mediated apoptosis was identified (75). Studies with these melanoma cells revealed a correlation between ceramide-mediated apoptosis and D-NMAPPD, a ceramide analog, confirming the effect of this inhibitor on ceramide signaling in human melanoma cells. These findings suggest ceramidase inhibitors as a potential new therapeutical class of antiproliferative and cytostatic drugs.

NF-NB is a transcription factor linked at the crossroads of life and death (Fig. 4). It functions as a modulator of inflammation, angiogenesis, differentiation, cell cycle, ad- hesion, migration, and survival (76). NF-NB has been recognized as an possible potential target in cancer treatment (77). In melanoma cells, NF-NB can be affected by upregulation of the NF-NB subunit, p50 or Rel A, or downregulation of the NF-NB inhibitor, INB (78).

Subsequently, all target molecules downstream of NF-NB regulation are affected.

Therefore antiapoptotic factors, such as c-myc and TNF receptor-associated factor

(TRAF)-2, are frequently upregulated in melanoma. In melanoma, gene-transfer

approaches targeting NF- NB disruption have been used, inactivating the NF-NB subunit,

Rel A (79),and overexpressing I NB ( 80).

(9)

Table 1 Summary of Therapeutic Approaches in Melanoma From In Vitro Studies to Clinical Trialsa Target and experimental settingReference Clinical trials bcl-2

Antisense oligonucleotides combined (plus dacarbazine) Jansen et al., 2000

(82) p53

Intratumoral injection Dummer et al., 2000

(30) Mouse systems H-ras

Overexpression in SCID-mouse model Jansen et al., 1997

(34) ras

Farnesyl thiosalicylic acid combined with dacarbazine in an human mela- noma xenograft mouse model Halaschek-Wiener et al., 2003

(

Smalley et al., 2002

(35) bcl-2

Antisense oligonucleotides Jansen et al., 1998

(50) p53

Adenoviral vector Cirielli et al., 1995

(83) survivin

Transient transfection of C85A mutant Grossman et al., 2001

(57) p53

Adenoviral vector (plus cyclin D) Sauter et al., 2002

(84) In vitro melanoma cell systems bcl-XL

Antisense oligonucleotides Heere-Ress et al., 2002

(51) bcl-2/bcl-XL

Simultaneous downregulation by antisense oligonucleotides Olie et al., 2002

(52) survivin

Antisense oligonucleotides Grossman et al., 1999

(54)

Conditional expression T

34

A mutant (plus cisplatin) Olie et al., 2002

(52) p53

Adenoviral vector Cirielli et al., 1995

(83) H-ras

Ribozyme Ohta et al., 1996

(85)

Antisense Jansen et al., 1997

(34) apaf-1

Retrovirus, 5aza2dc (plus adriamycin) Soengas et al., 2001

(65) ML-Iap

Antisense oligonucleotide Kasof et al., 2001

(58) relA

Antisense oligonucleotide McNulty et al., 2001

(79) a Many studies observed single tumor cell lines or limited tissue samples. SCID, severe combined immunodeficient.

613

(10)

Fig. 4. The nuclear factor (NF)-NB pathway. In the context of cell death control, NF-NB modu- lates the expression of survival factors.

Apoptosis deficiency appears to be strongly associated with the specific drug-resis- tant phenotype (81). In this study, cisplatin resistance has been associated with reduced caspase-9 activity and cytochrome-c release paralleled by normal DNA fragmentation, whereas no apoptotic events could be induced in etoposide-resistant melanoma cells (81).

Recently, tremendous efforts have been made in identifying new strategies and drug targets inhibiting apoptosis to support melanoma treatment (Table 1). Various investi- gations in mouse systems could prove the benefit of strategies interfering with apoptotic mechanisms in melanoma. Nevertheless, until now, these strategies could only be trans- ferred in a few clinical trails.

REFERENCES

1. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging impli- cations in tissue kinetics. Br J Cancer 1972;26:239–257.

2. Kaufmann SH, Earnshaw WC. Induction of apoptosis by cancer chemotherapy. Exp Cell Res 2000:25,642–25,649.

3. Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy.

Cell 2002;108:153–164.

4. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

5. Staunton MJ, Gaffney EF. Tumor type is a determinant of susceptibility to apoptosis. Am J Clin Pathol 1995;103:300–307.

6. Leist M, Jaattela M. Triggering of apoptosis by cathepsins. Cell Death Differ 2001;8:324–326.

7. Scaffidi C, et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO J 1998;17:1675–1687.

8. Sprick MR, et al. Caspase-10 is recruited to and activated at the native TRAIL and CD95 death- inducing signalling complex in a FADD-dependent manner but can not functionally substitute caspase- 8. EMBO J 2002;21:4520–4530.

9. Zou H, Li Y, Liu X, Wang X. An APAF-1 cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biol Chem 1999;274:11,549–11,556.

10. Tewari M, et al. Yama/CPP32 beta, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase. Cell 1995;81:801–809.

(11)

11. Lazebnik YA, Kaufmann SH, Desnoyers S, Poirier GG, Earnshaw WC. Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature 1994;371:346–347.

12. Liu X, Zou H, Slaughter C, Wang X. DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis. Cell 1997;89:175–184.

13. Thornberry NA. Caspases: key mediators of apoptosis. Chem Biol 1998;5:1074–5521.

14. Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mam- malian biology. Cell 2001;104:487–501.

15. Fulda S, et al. Cell type specific involvement of death receptor and mitochondrial pathways in drug- induced apoptosis. Oncogene 2001;20:1063–1075.

16. Los M, et al. Cross-resistance of CD95- and drug-induced apoptosis as a consequence of deficient activation of caspases (ICE/Ced-3 proteases). Blood 1997;90:3118–3129.

17. Muller M, et al. Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53. J Clin Invest 1997;99:403–413.

18. Tschopp J, Martinon F, Hofmann K. Apoptosis: silencing the death receptors. Curr Biol 1999;9:

R389–R384.

19. Wajant H. The Fas signaling pathway: more than a paradigm. Science 2002;296:1635–1636.

20. Strasser A, O’Connor L, Dixit VM. Apoptosis signaling. Annu Rev Biochem 2000;69:217–245.

21. Acehan D, et al. Three-dimensional structure of the apoptosome: implications for assembly, procaspase-9 binding, and activation. Mol Cell 2002;9:423–432.

22. Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3 [see comments]. Cell 1997;90:405–413.

23. Li P, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997;91:479–489.

24. Chai JJ, et al. Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature 2000;406:855–862.

25. Petty R, Evans A, Duncan I, Kurbacher C, Cree I. Drug resistance in ovarian cancer—the role of p53.

Pathol Oncol Res 1998;4:97–102.

26. Miyake H, et al. Enhancement of chemosensitivity in human bladder cancer cells by adenoviral- mediated p53 gene transfer. Anticancer Res 1998;18:3087–3092.

27. Li G, Bush JA, Ho VC. p53-dependent apoptosis in melanoma cells after treatment with camptothecin.

J Invest Dermatol 2000;114:514–519.

28. Hartmann A, et al. Overexpression and mutations of p53 in metastatic malignant melanomas. Int J Cancer 1996;67:313–317.

29. Satyamoorthy K, et al. Aberrant regulation and function of wild-type p53 in radioresistant melanoma cells. Cell Growth Differ 2000;11:467–474.

30. Dummer R, et al. Biological activity and safety of adenoviral vector-expressed wild-type p53 after intratumoral injection in melanoma and breast cancer patients with p53-overexpressing tumors. Can- cer Gene Ther 2000;7:1069–1076.

31. Serrone L, Hersey P. The chemoresistance of human malignant melanoma: an update. Melanoma Res 1999;9:51–58.

32. Borner C, et al. Mutated N-ras upregulates Bcl-2 in human melanoma in vitro and in SCID mice.

Melanoma Res 1999;9:347–350.

33. Urquhart JL, et al. Regulation of Fas-mediated apoptosis by N-ras in melanoma. J Invest Dermatol 2002;119:556–561.

34. Jansen B, et al. Activated N-ras contributes to the chemoresistance of human melanoma in severe combined immunodeficiency (SCID) mice by blocking apoptosis. Cancer Res 1997;57:362–365.

35. Smalley KS, Eisen TG. Farnesyl thiosalicylic acid inhibits the growth of melanoma cells through a combination of cytostatic and pro-apoptotic effects. Int J Cancer 2002;98:514–522.

36. Halaschek-Wiener J, Kloog Y, Wacheck V, Jansen B. Farnesyl thiosalicylic acid chemosensitizes human melanoma in vivo. J Invest Dermatol 2003;120:109–115.

37. Davies H, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–954.

38. Erhardt P, Schremser EJ, Cooper GM. B-Raf inhibits programmed cell death downstream of cyto- chrome c release from mitochondria by activating the MEK/Erk pathway. Mol Cell Biol 1999;19:

5308–5315.

39. Shin MS, et al. Alterations of Fas (Apo-1/CD95) gene in cutaneous malignant melanoma. Am J Pathol 1999;154:1785–1791.

(12)

40. Ugurel S, et al. Heterogenous susceptibility to CD95-induced apoptosis in melanoma cells correlates with bcl-2 and bcl-x expression and is sensitive to modulation by interferon-gamma. Int J Cancer 1999;82:727–736.

41. Mouawad R, Khayat D, Soubrane C. Plasma Fas ligand, an inducer of apoptosis, and plasma soluble Fas, an inhibitor of apoptosis, in advanced melanoma. Melanoma Research 2000;10:461–467.

42. Griffith TS, Chin WA, Jackson GC, Lynch DH, Kubin MZ. Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J Immunol 1998;161:2833–2840.

43. Hersey P, Zhang XD. How melanoma cells evade trail-induced apoptosis. Nat Rev Cancer 2001;1:142–150.

44. Griffith TS, Anderson RD, Davidson BL, Williams RD, Ratliff TL. Adenoviral-mediated transfer of the TNF-related apoptosis-inducing ligand/apo-2 ligand gene induces tumor cell apoptosis. J Immunol 2000;165:2886–2894.

45. Franco AV, et al. The role of nf-kappab in tnf-related apoptosis-inducing ligand (trail)-induced apoptosis of melanoma cells. J Immunol 2001;166:5337–5345.

46. Reed JC, Paternostro G. Postmitochondrial regulation of apoptosis during heart failure [comment].

Proc Natl Acad Sci USA 1999;96:7614–7616.

47. Raisova M, et al. The Bax/Bcl-2 ratio determines the susceptibility of human melanoma cells to CD95/

Fas-mediated apoptosis. J Invest Dermatol 2001;117:333–340.

48. Iervolino A, et al. Bcl-2 overexpression in human melanoma cells increases angiogenesis through VEGF mRNA stabilization and HIF-1–mediated transcriptional activity. FASEB J 2002;16:

1453–1455.

49. Biroccio A, et al. c-Myc down-regulation increases susceptibility to cisplatin through reactive oxygen species-mediated apoptosis in M14 human melanoma cells. Mol Pharmacol 2001;60:174–182.

50. Jansen B, et al. bcl-2 antisense therapy chemosensitizes human melanoma in Scid mice. Nat Med 1998;4:232–234.

51. Heere-Ress E, et al. Bcl-X(L) is a chemoresistance factor in human melanoma cells that can be inhibited by antisense therapy. Int J Cancer 2002;99:29–34.

52. Olie RA, et al. Bcl-2 and bcl-xL antisense oligonucleotides induce apoptosis in melanoma cells of different clinical stages. J Invest Dermatol 2002;118:505–512.

53. Jansen B, et al. Bcl-2 antisense plus dacarbacine therapy for malignan melanoma. Proc Am Assoc Cancer Res Conf Programmed Cell Death Regul 2000;A95.

54. Grossman D, McNiff JM, Li F, Altieri DC. Expression and targeting of the apoptosis inhibitor, survivin, in human melanoma. J Invest Dermatol 1999;113:1076–1081.

55. Li F, et al. Control of apoptosis and mitotic spindle checkpoint by survivin. Nature 1998;396:580–584.

56. Gradilone A, et al. Survivin, bcl-2, bax, and bcl-X gene expression in sentinel lymph nodes from melanoma patients. J Clin Oncol 2003;21:306–312.

57. Grossman D, Kim PJ, Schechner JS, Altieri DC. Inhibition of melanoma tumor growth in vivo by survivin targeting. Proc Natl Acad Sci USA 2001;98:635–640.

58. Kasof GM, Gomes BC. Livin, a novel inhibitor of apoptosis protein family member. J Biol Chem 2001;276:3238–3246.

59. Vucic D, Stennicke HR, Pisabarro MT, Salvesen GS, Dixit VM. ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed in human melanomas. Curr Biol 2000;10:1359–1366.

60. Vucic D, et al. SMAC negatively regulates the anti-apoptotic activity of melanoma inhibitor of apoptosis (ML-IAP). J Biol Chem 2002;277:12,275–12,279.

61. Schmollinger JC, et al. Melanoma inhibitor of apoptosis protein (ML-IAP) is a target for immune- mediated tumor destruction. Proc Natl Acad Sci USA 2003;100:3398–3403.

62. Srinivasula SM, et al. Molecular determinants of the caspase-promoting activity of Smac/DIABLO and its role in the death receptor pathway. J Biol Chem 2000;275:36,152–36,157.

63. Bullani RR, et al. Selective expression of FLIP in malignant melanocytic skin lesions. J Invest Dermatol 2001;117:360–364.

64. Zhang XD, et al. Relation of TNF-related apoptosis-inducing ligand (TRAIL) receptor and FLICE-inhibitory protein expression to TRAIL-induced apoptosis of melanoma. Cancer Res 1999;59:2747–2753.

65. Soengas MS, et al. Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 2001;409:207–211.

66. Han W, et al. Effects of C(2)-ceramide on the Malme-3M melanoma cell line. J Dermatol Sci 2002;30:10.

67. Sinha P, et al. Identification of novel proteins associated with the development of chemoresistance in malignant melanoma using two-dimensional electrophoresis. Electrophoresis 2000;21:3048–3057.

(13)

68. Ravagnan L, et al. Heat-shock protein 70 antagonizes apoptosis-inducing factor. Nat Cell Biol 2001;3:839–843.

69. Saleh A, Srinivasula SM, Balkir L, Robbins PD, Alnemri ES. Negative regulation of the Apaf-1 apoptosome by Hsp70. Nat Cell Biol 2000;2:476–483.

70. Dressel R, Johnson JP, Gunther E. Heterogeneous patterns of constitutive and heat shock induced expression of HLA-linked HSP70-1 and HSP70-2 heat shock genes in human melanoma cell lines.

Melanoma Res 1998;8:482–492.

71. Ricaniadis N, Kataki A, Agnantis N, Androulakis G, Karakousis CP. Long-term prognostic signifi- cance of HSP-70, c-myc and HLA-DR expression in patients with malignant melanoma. Eur J Surg Oncol 2001;27:88–93.

72. Park KC, et al. Overexpression of HSP70 prevents ultraviolet B-induced apoptosis of a human mela- noma cell line. Arch Dermatol Res 2000;292:482–487.

73. Solit DB, Scher HI, Rosen N. Hsp90 as a therapeutic target in prostate cancer. Semin Oncol 2003;30:709–716.

74. Neef R, Kuske MA, Prols E, Johnson JP. Identification of the Human PHLDA1/TDAG51 Gene: down- regulation in metastatic melanoma contributes to apoptosis resistance and growth deregulation. Can- cer Res 2002;62:5920–5929.

75. Raisova M, et al. Bcl-2 overexpression prevents apoptosis induced by ceramidase inhibitors in malig- nant melanoma and HaCaT keratinocytes. FEBS Lett 2002;516:47–52.

76. Karin M, Lin A. NF-kappaB at the crossroads of life and death. Nat Immunol 2002;3:221–227.

77. Lin A, Karin M. NF-kappaB in cancer: a marked target Semin Cancer Biol 2003;13:107–114.

78. Yang J, Richmond A. Constitutive IkappaB kinase activity correlates with nuclear factor-kappaB activation in human melanoma cells. Cancer Res 2001;61:4901–4909.

79. McNulty SE, Tohidian NB, Meyskens FL Jr. RelA, p50 and inhibitor of kappa B alpha are elevated in human metastatic melanoma cells and respond aberrantly to ultraviolet light B. Pigment Cell Res 2001;14:456–465.

80. Bakker TR, Reed D, Renno T, Jongeneel CV. Efficient adenoviral transfer of NF-kappaB inhibitor sensitizes melanoma to tumor necrosis factor-mediated apoptosis. Int J Cancer 1999;80:320–323.

81. Helmbach H, et al. Drug-resistance towards etoposide and cisplatin in human melanoma cells is associated with drug-dependent apoptosis deficiency. J Invest Dermatol 2002;118:923–932.

82. Jansen B, et al. Chemosensitisation of malignant melanoma by BCL2 antisense therapy. Lancet 2000;356:1728–1733.

83. Cirielli C, et al. Adenovirus-mediated gene transfer of wild-type p53 results in melanoma cell apoptosis in vitro and in vivo. Int J Cancer 1995;63:673–679.

84. Sauter ER, Takemoto R, Litwin S, Herlyn M. p53 alone or in combination with antisense cyclin D1 induces apoptosis and reduces tumor size in human melanoma. Cancer Gene Ther 2002;9:807–812.

85. Ohta Y, Kijima H, Kashani-Sabet M, Scanlon KJ. Suppression of the malignant phenotype of mela- noma cells by anti-oncogene ribozymes. J Invest Dermatol 1996;106:275–280.

86. Silvius JR. Mechanisms of Ras protein targeting in mammilian cells. J Membr Bio 2002; 90:83–92.

Riferimenti

Documenti correlati

Therefore, the mean of the CRMSE anoma- lies is also computed, where the anomalies are computed by subtracting the mean station or network CRMSE of a number of complete

Three different methods of sedation or sedoanalgesia using remifentanil, propo- fol, or midazolam to reduce intra- and postoperative pain and neuroendocrine stress in patients who

In modo differente viene affrontata la questione dei territori dell’entroterra nel caso di Genova, qui viene esplicitato il tema delle aree interne e vengono messe in evidenze

This work makes a quantitative comparison between the results of landslide forecasting obtained using two different rainfall threshold models, one using intensity-duration

Eppure… Presumo un’anima accesa di luce, che vive sotto la limpidezza azzurra dell’immensa volta del cielo di campagna, la cui voce s’affratella col mormorio

At the level of linear perturbations we have identified modifications in the ISW effect, the gravitational lensing, the rate of growth of structure and the B-modes spectrum,

On s’attardera ici sur les cas (1) où terme et définition sont tous deux absents des corpus étudiés, (2) où le terme n’est attesté dans aucun des trois diction- naires, (3) où

When photoactivated (7.2 J/cm 2 ) in B78-H1 melanoma cells, P4 and C14, but not control P2, caused a strong inhibition of metabolic activity, clonogenic growth and cell