Skip to main content
Annals of Oncology logoLink to Annals of Oncology
. 2009 Aug;20(Suppl 6):vi35–vi40. doi: 10.1093/annonc/mdp254

Small molecules and targeted therapies in distant metastatic disease

P Hersey 1,*, L Bastholt 2, V Chiarion-Sileni 3, G Cinat 4, R Dummer 5, A M M Eggermont 6, E Espinosa 7, A Hauschild 8, I Quirt 9, C Robert 10, D Schadendorf 11
PMCID: PMC2712592  PMID: 19617296

Abstract

Chemotherapy, biological agents or combinations of both have had little impact on survival of patients with metastatic melanoma. Advances in understanding the genetic changes associated with the development of melanoma resulted in availability of promising new agents that inhibit specific proteins up-regulated in signal cell pathways or inhibit anti-apoptotic proteins. Sorafenib, a multikinase inhibitor of the RAF/RAS/MEK pathway, elesclomol (STA-4783) and oblimersen (G3139), an antisense oligonucleotide targeting anti-apoptotic BCl-2, are in phase III clinical studies in combination with chemotherapy. Agents targeting mutant B-Raf (RAF265 and PLX4032), MEK (PD0325901, AZD6244), heat-shock protein 90 (tanespimycin), mTOR (everolimus, deforolimus, temsirolimus) and VEGFR (axitinib) showed some promise in earlier stages of clinical development. Receptor tyrosine-kinase inhibitors (imatinib, dasatinib, sunitinib) may have a role in treatment of patients with melanoma harbouring c-Kit mutations. Although often studied as single agents with disappointing results, new targeted drugs should be more thoroughly evaluated in combination therapies. The future of rational use of new targeted agents also depends on successful application of analytical techniques enabling molecular profiling of patients and leading to selection of likely therapy responders.

Keywords: B-Raf, c-Kit, inhibitor, melanoma, mTOR, multikinase

introduction

Treatment of melanoma once it has metastasised beyond locoregional sites remains unsatisfactory. A range of different treatments based on chemotherapy, biological agents or a combination of both (reviewed elsewhere) has had little impact on survival [13].

Single-agent chemotherapy produces responses in 10–20% of patients with advanced melanoma, although there is no evidence that this translates into a survival advantage. Complete responses occur in ∼2% of the cases; median survival associated with chemotherapy is 9 months and 13% of patients are alive at 2 years [4]. Commonly used compounds include dacarbazine (DTIC), temozolomide, fotemustine, cisplatin, carboplatin, vinblastine, paclitaxel and docetaxel [5].

As a single agent, DTIC has been most commonly used even when it has not been formally compared with other agents or with observation alone. The usual dose is 1000 mg/m2 every 3–4 weeks (given either in 1 day or at five daily doses of 200 mg/m2). Some centres substitute DTIC with temozolomide for its convenience of administration [2]. Temozolomide demonstrated efficacy equal to that of DTIC in a phase III trial at a dose of 200 mg/m2/day for 5 days every 28 days [6]. Recent results from a large, randomised phase III trial (EORTC 18032), which examined the efficacy of an extended schedule of temozolomide (week on-week off, 150 mg/m2/day for 7 days repeated every 14 days) compared with standard dose single-agent DTIC, showed no difference in overall survival (OS), progression-free survival (PFS) and overall response rate (ORR) between the two arms [7].

Combination chemotherapy is associated with a response rate of 30–50%, but with <5% complete responses and a median survival of 9 months [2, 5]. The better-known combinations are cisplatin–vinblastine–dacarbazine (CVD) and cisplatin–dacarbazine–BCNU–tamoxifen (Dartmouth regimen) [8, 9]. Although preliminary reports favoured the use of these regimens over single-agent chemotherapy, further comparisons with DTIC alone did not show differences in survival or even in response rate [10]. In addition, combination chemotherapy produces significant toxicity. Poor results with single-agent or combination chemotherapy regimens have remained unchanged for decades and underscore the need for application of fundamentally different strategies in treatment of advanced melanoma.

Substantial advances have been made in understanding the genetic changes associated with the onset of this malignancy. As reviewed elsewhere [11], consensus is emerging about primary events involved in the development of melanoma largely from comparative genome hybridisation (CGH) [12]. These include (a) up-regulation of the RAS/RAF/MEK pathway [12, 13]; (b) down-regulation of the retinoblastoma protein (RB) by increased cyclin D1 or CDK4 activity [12, 14]; and (c) inactivation of the CDKN2A p16 suppressor of CDK4 and 6, in >50% of melanoma [12, 15]. Activating mutations in c-Kit or FGF may occur in some melanoma [16]. Inactivation of the CDKN2A gene may also affect production of p14 ARF (alternate reading frame), which is important in maintenance of p53 protein levels by inhibiting the HDM2-mediated ubiquitination of p53 that leads to its degradation.

signal pathway inhibitors

Numerous studies have shown that several signal pathways related to survival of cancer cells are frequently up-regulated in melanoma. Arguably the most important of these is the RAF/RAS/MEK pathway, which is involved in proliferation and resistance to apoptosis. The pathway can be turned on by activating mutations in NRAS or BRAF or by endogenous receptor–ligand interactions. Importantly, activation of the pathway was shown to be related to progression of the disease [17] and resistance to apoptosis [18, 19]. Another survival pathway is the PI3K/Akt3 pathway, activated in 5–10% of melanomas by a mutation in the phosphatase and tensin (PTEN) protein, receptor–ligand interactions or activating mutations in NRAS [2022]. The Src/Stat3 pathway was reported to be variably activated in some melanoma lines and metastatic melanoma in vivo [23]. Expression of c-Met/HGF receptors was also associated with progression of melanoma [2426].

A number of new drugs have been developed that target members of these pathways. These are summarised in Tables 1 and 2. Many of these agents are still being evaluated in preclinical studies, and very few have been evaluated in randomised phase III studies. Sorafenib is a multikinase inhibitor with selectivity for B-Raf, C-Raf, VEGFR-2 and -3, platelet-derived growth factor receptor (PDGFR) and c-Kit. When used as a single agent, it stabilised the disease in 19% of stage IV patients, and when given with carboplatin and paclitaxel, it induced promising objective responses and PFS [27]. A randomised phase II trial comparing DTIC with or without sorafenib at twice-daily, 400 mg doses was conducted in 101 patients. The group receiving sorafenib had a PFS of 21.1 weeks compared with 11.7 weeks in the DTIC-alone group. Response rates were 24% and 12%, respectively [28]. Another phase II trial with a complex design investigated sorafenib in combination with temozolomide. Again, encouraging response rates were reported [29]. Skin rashes and haematologic toxicity were the main side-effects reported. A phase III trial recruited 270 patients into a second-line study and compared carboplatin plus paclitaxel with or without sorafenib. The median PFS was 17.9 and 17.4 weeks in the placebo and sorafenib groups, respectively. The ORR was 11% in both groups [30]. Although the addition of sorafenib did not improve PFS or ORR in this second-line patient population, the utility of carboplatin plus paclitaxel with sorafenib in chemotherapy-naïve advanced melanoma patients remains to be determined. The Eastern Cooperative Oncology Group (ECOG) is conducting a similar trial in previously untreated patients that is now closed to accrual (ECOG 2603). The results of the trial and further studies on sorafenib plus DTIC are awaited with interest.

Table 1.

RAS/RAF/MEK signal pathway inhibitors

Agent Class of inhibitor Target protein(s) Reference
Sorafenib Multikinase inhibitor C-Raf; B-Raf; VEGF-2, -3; PDGF; Flt-3; c-Kit [2730]
Tanespimycin (KOS-953, 17-AGG) Hsp90 inhibitor Hsp90 (client proteins, B-Raf, Akt, others) [31]
RAF265 Multikinase inhibitor Mutant B-Raf, VEGFR-2 [32]
PLX4032, PLX4720 Selective B-Raf kinase inhibitor Mutant B-Raf [33]
PD0325901 Non-ATP-competitive specific MEK inhibitor MEK1, 2 [32]
AZD6244 Non-ATP-competitive specific MEK inhibitor MEK1, 2 [34]
Tipifarnib (R115777) Farnesyl transferase inhibitor Prenylated proteins [35, 36]

Table 2.

Akt, receptor tyrosine kinase (RTK) and Stat signal pathway inhibitors

Agent(s) Target protein Reference
PI 103 PI3K/mTOR [37]
SF1126 (LY294002-prodrug) PI3K [38]
Perifosine, PX-866 Akt [22]
CMEP Akt [39]
Temsirolimus (CCI-779) mTOR [40]
Everolimus (RAD001) mTOR [40]
Deforolimus (AP23573) mTOR [41]
XL765 P13K/mTOR [42]
SB216763, DW1/2 GSK3β [43]
Imatinib, dasatinib, sunitinib, erlotinib RTKs [44]
Dasatinib Src [45]
S31-M2001 Stat3 [46]
SUI1274 c-Met/HGF [24, 47]

The specific MEK inhibitor AZD6244 was evaluated in a randomised phase II trial of 200 patients with stage IV melanoma. Patients were randomised to AZD6244 or temozolomide; recently reported results indicate that there was no significant difference in PFS between those arms [34]. However, AZD6244 monotherapy resulted in lasting remissions mainly in patients with documented B-Raf mutations. Combinations with other agents, such as a taxanes, are being planned. Taxanes are known to activate the anti-apoptotic MEK pathway [48], and combination therapy with inhibitors of this pathway may be beneficial.

The other two B-Raf inhibitors, RAF265 and PLX4032 (Table 1), have high affinity for the mutated B-Raf and are in dose-finding and early phase II studies. The MEK-specific inhibitor PD0325901 was associated with some retinal disturbances, and its further evaluation was halted. Tanespimycin (KOS-953), an inhibitor of heat shock protein 90 (Hsp90), targets proteins protected (chaperoned) by Hsp90. This includes RAF, Akt and other signal pathway proteins. The drug was tested in a phase II study in previously treated stage IV melanoma patients and administered twice weekly for 2 weeks out of 3 weeks. Results from a treatment of 14 patients met the criteria for further evaluation in the second stage of the trial [31]. Another group, farnesyl transferase inhibitors, should in theory inhibit activation of RAS. However, when used as a single agent this group of drugs has been disappointing [35, 36].

Most of the agents in Table 2 are at early stages of investigation and are listed to indicate the rich supply of agents that remain to be evaluated in treatment of melanoma. Given that activation of the Akt pathway has been implicated in resistance to chemotherapy [22], trials with inhibitors of this pathway or downstream targets such as mTOR, GSK3β or HDM2 are awaited with much interest.

A key downstream target of Akt—mTOR exists in two complexes, mTORC1 and mTORC2. The latter is not inhibited by rapamycin or its analogues and is believed to be responsible for rapamycin-induced activation of Akt and PKC-α [49]. Newer inhibitors, which target the mTORC2 complex or those that inhibit both PI3K and mTOR (XL765), should avoid this problem. XL765 has proved to be well-tolerated in phase I studies [42].

A rich supply of inhibitors of receptor tyrosine kinases (RTKs), such as those against Bcr-Ab1, c-Kit, PDGFR, epidermal growth factor receptor (EGFR), c-Met and Src, may have a role in the treatment of some melanomas. For example, a high percentage of mucosal or acral melanoma and some cutaneous melanoma have amplified and mutated c-Kit and may respond to imatinib, sunitinib or dasatinib [44, 45]. Four phase II trials with sunitinib or imatinib in patients with c-Kit melanoma mutations are ongoing. A subgroup of melanoma with overexpression of phosphorylated c-Kit and CDK4 were resistant to B-Raf inhibitors but sensitive to imatinib [50].

Several agents of indeterminate action such as histamine [51] and lenalidomide (CC-5013) [52] are no longer under investigation. Elesclomol (STA-4783) is a new agent that appears to increase reactive oxygen species. Results from a recent randomised phase II study of elesclomol in combination with paclitaxel indicated a significant benefit for chemotherapy-naïve patients in PFS (HR = 0.315, P = 0.02) [53]. Evaluation of this combination therapy has progressed to a phase III trial [54]. Axitinib (AG-013736), an oral inhibitor of VEGFR-1, -2 and -3, c-Kit, PDGR-α and PDGR-β, is also at an early stage of evaluation in melanoma [55], and recent results from a phase II study demonstrated its single-agent activity in a subgroup of melanoma patients [56].

inhibitors of anti-apoptotic proteins

Another group of new drugs targets the anti-apoptotic proteins. Mitochondria-dependent apoptotic pathways are regulated mainly by the Bcl-2 family of proteins, which, as reviewed elsewhere [5760], consists of a family of BH3-only pro-apoptotic proteins, two multi-domain pro-apoptotic proteins (Bax and Bak) and several multi-domain anti-apoptotic proteins (Bcl-2, Bcl-XL, Bcl-W, Mcl-1 and A1). In one model, binding the anti-apoptotic proteins to the BH3 proteins displaces Bax or Bak from the anti-apoptotic proteins, allowing them to bind to mitochondria and induce mitochondrial outer membrane permeabilisation (MOMP) [58, 61]. Certain BH3 proteins have selectivity for different anti-apoptotic proteins. In particular, Noxa binds selectively to Mcl-1. The latter also binds Bak, and hence Noxa may displace Bak from Mcl-1, allowing it to bind to mitochondria [62, 63].

It is of particular interest that immunohistological studies on tissue sections from melanoma have shown that Mcl-1 and Bcl-XL increase in expression with progression of the disease whereas Bcl-2 decreased during progression of the disease [64]. Further studies are needed to define the regulators of these proteins more closely, particularly Mcl-1, as current studies suggest it is up-regulated as part of the unfolded protein response to endoplasmic reticulum stress [65].

These findings are important in the design of treatment strategies in melanoma. As shown in Table 3, a number of new agents can be used clinically to target the anti-apoptotic proteins. Oblimersen is an antisense agent targeted to mitochondrial Bcl-2. Results from a randomised phase III trial comparing DTIC combined with oblimersen with DTIC alone in 771 patients showed improved PFS (2.6 months compared with 1.6 months, P < 0.01) and response rate (13.5% compared with 7.5%, P = 0.007) but no statistical difference in overall survival (9.0 months compared with 7.8 months, P = 0.077) [66]. Problems with study design and failure to measure tumour Bcl-2 expression made these results difficult to interpret. A significant interaction between baseline serum lactate dehydrogenase (LDH) and treatment was noted, with oblimersen significantly increasing survival in patients with normal LDH (11.4 months compared with 9.7 months, P = 0.02). Another agent, ABT-737, has high affinity for Bcl-2, Bcl-XL and Bcl-W, but not Mcl-1. Preclinical studies have shown that many tumours are resistant to this agent due to its failure to inhibit Mcl-1 in cancer cells; down-regulation of Mcl-1 resulted in sensitivity to ABT-737 [68, 69]. ABT-263 is an orally active form of ABT-737 [70]. Sorafenib or MEK inhibitors may down-regulate Mcl-1 and may thus be useful in combination studies. As shown in Table 3, however, a number of protein inhibitors have selectivity for all the anti-apoptotic proteins. One of these inhibitors, obatoclax, is now in preliminary trials in patients with haematological malignancies [75] and was shown to overcome Mcl-1 resistance to apoptosis [76]. At this stage, we would expect that these broad-spectrum inhibitors would be more effective when given in combination with a treatment that induces apoptosis, such as immunotherapy or chemotherapy.

Table 3.

Targeting anti-apoptotic proteins

Agent Target protein(s) Reference
Oblimersen (G3139) Bcl-2 (specific) [66]
YM155 Survivin [67]
ABT-737 (ABT-263) BH3-mimetic (inhibits Bcl-2 group: Bcl-2, Bcl-XL, Bcl-W, not Mcl-1) [6870]
Gossypol (AT-101) BH3-mimetic (inhibits Bcl-2 group) [71]
Obatoclax (GX015-070) BH3-mimetic (inhibits Bcl-2 group) [72]
TW37 Bim-mimetic (inhibits Bcl-2 group) [73]
Smac mimetic Inhibitor of IAP1,2, XIAP [74]

conclusions

We may already have agents that would control the disease if targeted to patient subgroups or if given in appropriate combinations. A number of new agents are in various stages of clinical evaluation. The current strategy of testing new targeted drugs as single agents is necessary, but should be regarded as the first step in evaluation of the agent for future combination with other agents. For example, agents that induce apoptosis, such as taxanes, platinum compounds or immunotherapy, can be combined with agents that inhibit anti-apoptotic proteins. Failure of a drug as a single agent is probably no guide to the ultimate effectiveness of the drug when given in combination and planning for trials of combined agents would appear to be an important part of drug evaluation (e.g. as carried out in the evaluation of the elesclomol–paclitaxel combination). Future research will also need to develop approaches that help to select subgroups of patients that are more likely to respond to particular agents. Combining high-density single-nucleotide polymorphism arrays and the mutation analysis of relevant oncogenes might provide the rational basis for a sophisticated use of new agents in the treatment of melanoma [77]. To improve the outcome of melanoma treatments and to determine the biological mechanisms of efficacy or failure, future studies with small molecules and targeted therapies will also require strict monitoring of biological end points.

conflict of interest disclosures

L. Bastholt has had an advisory role with Schering Plough, Celgene, Pfizer, Genmab and AstraZeneca, and has received honoraria for educational lectures from Schering-Plough and AstraZeneca; V. Chiarion-Sileni has received research funding from Bristol-Myers Squibb and Synta and provided an expert testimony for Schering-Plough; G. Cinat has served as advisor for Pfizer and received lecture honoraria from Pfizer; R. Dummer has received research funding from AstraZeneca, Novartis, Cephalon, Merck Sharp & Dohme, Transgene, and Bayer, and has served as a consultant or on advisory boards with AstraZeneca, Novartis, Cephalon, Merck Sharp & Dohme, Transgene, Genta, Bayer and Schering Plough; A. M. M. Eggermont has been consultant for Schering-Plough; E. Espinosa has received honoraria from Schering-Plough; A. Hauschild has served on advisory boards with Onyx Pharmaceuticals/Bayer (USA/Germany), AstraZeneca (Germany), Synta Pharmaceuticals/GlaxoSmithKline and Genta Pharmaceuticals (USA); C. Robert has had an advisory role with Bayer, Pfizer, Bristol-Myers Squibb, Johnson & Johnson and Novartis; D. Schadendorf has had an advisory role with Bristol-Myers Squibb, AstraZeneca, GlaxoSmithKline, Schering-Plough, Synta Pharmaceuticals, Bayer and Altona, and has received research support from Schering-Plough.

Acknowledgments

The authors thank Pavel Kramata, ScienceFirst, LLC, Cedar Knolls, NJ 07927, USA for writing support and coordination during preparation of the manuscript.

References

  • 1.Khayat D, Meric J-B, Rixie O. Systemic chemotherapy and biochemotherapy for non-resected and metastatic melanoma. In: Thomson JF, Morton DL, Kroon BBL, editors. Textbook of Melanoma. London: Martin Dunitz; 2004. pp. 586–601. [Google Scholar]
  • 2.Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med. 2004;351:998–1012. doi: 10.1056/NEJMra041245. [DOI] [PubMed] [Google Scholar]
  • 3.Tawbi HA, Kirkwood JM. Management of metastatic melanoma. Semin Oncol. 2007;34:532–545. doi: 10.1053/j.seminoncol.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 4.Lee ML, Tomsu K, Von Eschen KB. Duration of survival for disseminated malignant melanoma: results of a meta-analysis. Melanoma Res. 2000;10:81–92. [PubMed] [Google Scholar]
  • 5.Balch CM, et al. Cutaneous melanoma. In: De Vita VT, Hellmann S, Rosenberg SA, editors. Cancer: Principles and Practice of Oncology. Philadelphia: Lippincott Williams & Wilkins; 2005. pp. 1797–1801. [Google Scholar]
  • 6.Middleton MR, Grob JJ, Aaronson N, et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol. 2000;18:158–166. doi: 10.1200/JCO.2000.18.1.158. [DOI] [PubMed] [Google Scholar]
  • 7.Patel PM, Suciu S, Mortier L, et al. Extended schedule, escalated dose temozolomide versus dacarbazine in stage IV malignant melanoma: final results of the randomised phase III study (EORTC 18032) Ann Oncol. 2008:19. doi: 10.1016/j.ejca.2011.04.030. Abstr LBA8. [DOI] [PubMed] [Google Scholar]
  • 8.Legha SS, Ring S, Papadopoulos N, et al. A prospective evaluation of a triple-drug regimen containing cisplatin, vinblastine, and dacarbazine (CVD) for metastatic melanoma. Cancer. 1989;64:2024–2029. doi: 10.1002/1097-0142(19891115)64:10<2024::aid-cncr2820641010>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  • 9.Del Prete SA, Maurer LH, O'Donnell J, et al. Combination chemotherapy with cisplatin, carmustine, dacarbazine, and tamoxifen in metastatic melanoma. Cancer Treat Rep. 1984;68:1403–1405. [PubMed] [Google Scholar]
  • 10.Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17:2745–2751. doi: 10.1200/JCO.1999.17.9.2745. [DOI] [PubMed] [Google Scholar]
  • 11.Bennett DC. How to make a melanoma: what do we know of the primary clonal events? Pigment Cell Melanoma Res. 2008;21:27–38. doi: 10.1111/j.1755-148X.2007.00433.x. [DOI] [PubMed] [Google Scholar]
  • 12.Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med. 2005;353:2135–2147. doi: 10.1056/NEJMoa050092. [DOI] [PubMed] [Google Scholar]
  • 13.Meier F, Schittek B, Busch S, et al. The RAS/RAF/MEK/ERK and PI3K/AKT signaling pathways present molecular targets for the effective treatment of advanced melanoma. Front Biosci. 2005;10:2986–3001. doi: 10.2741/1755. [DOI] [PubMed] [Google Scholar]
  • 14.Halaban R. Rb/E2F: a two-edged sword in the melanocytic system. Cancer Metastasis Rev. 2005;24:339–356. doi: 10.1007/s10555-005-1582-z. [DOI] [PubMed] [Google Scholar]
  • 15.Sharpless E, Chin L. The INK4a/ARF locus and melanoma. Oncogene. 2003;22:3092–3098. doi: 10.1038/sj.onc.1206461. [DOI] [PubMed] [Google Scholar]
  • 16.Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–4346. doi: 10.1200/JCO.2006.06.2984. [DOI] [PubMed] [Google Scholar]
  • 17.Zhuang L, Lee CS, Scolyer RA, et al. Activation of the extracellular signal regulated kinase (ERK) pathway in human melanoma. J Clin Pathol. 2005;58:1163–1169. doi: 10.1136/jcp.2005.025957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhang XD, Borrow JM, Zhang XY, et al. Activation of ERK1/2 protects melanoma cells from TRAIL-induced apoptosis by inhibiting Smac/DIABLO release from mitochondria. Oncogene. 2003;22:2869–2881. doi: 10.1038/sj.onc.1206427. [DOI] [PubMed] [Google Scholar]
  • 19.Wang YF, Jiang CC, Kiejda KA, et al. Apoptosis induction in human melanoma cells by inhibition of MEK is caspase-independent and mediated by the Bcl-2 family members PUMA, Bim, and Mcl-1. Clin Cancer Res. 2007;13:4934–4942. doi: 10.1158/1078-0432.CCR-07-0665. [DOI] [PubMed] [Google Scholar]
  • 20.Haluska FG, Tsao H, Wu H, et al. Genetic alterations in signaling pathways in melanoma. Clin Cancer Res. 2006;12:2301s–2307s. doi: 10.1158/1078-0432.CCR-05-2518. [DOI] [PubMed] [Google Scholar]
  • 21.Kalinsky K, Haluska FG. Novel inhibitors in the treatment of metastatic melanoma. Expert Rev Anticancer Ther. 2007;7:715–724. doi: 10.1586/14737140.7.5.715. [DOI] [PubMed] [Google Scholar]
  • 22.LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat. 2008;11:32–50. doi: 10.1016/j.drup.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Homsi J, Cubitt C, Daud A. The Src signaling pathway: a potential target in melanoma and other malignancies. Expert Opin Ther Targets. 2007;11:91–100. doi: 10.1517/14728222.11.1.91. [DOI] [PubMed] [Google Scholar]
  • 24.Natali PG, Nicotra MR, Di Renzo MF, et al. Expression of the c-Met/HGF receptor in human melanocytic neoplasms: demonstration of the relationship to malignant melanoma tumour progression. Br J Cancer. 1993;68:746–750. doi: 10.1038/bjc.1993.422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.McGill GG, Haq R, Nishimura EK. Fisher DE. c-Met expression is regulated by Mitf in the melanocyte lineage. J Biol Chem. 2006;281:10365–10373. doi: 10.1074/jbc.M513094200. [DOI] [PubMed] [Google Scholar]
  • 26.Christensen JG, Burrows J, Salgia R. c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Lett. 2005;225:1–26. doi: 10.1016/j.canlet.2004.09.044. [DOI] [PubMed] [Google Scholar]
  • 27.Flaherty KT. Chemotherapy and targeted therapy combinations in advanced melanoma. Clin Cancer Res. 2006;12:2366s–2370s. doi: 10.1158/1078-0432.CCR-05-2505. [DOI] [PubMed] [Google Scholar]
  • 28.McDermott DF, Sosman JA, Hodi FS, et al. Randomized phase II study of dacarbazine with or without sorafenib in patients with advanced melanoma. 2007 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2007;25 Abstr 8511. [Google Scholar]
  • 29.Amaravadi R, Schuchter LM, McDermott DF, et al. Updated results of a randomized phase II study comparing two schedules of temozolomide in combination with sorafenib in patients with advanced melanoma. 2007 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2007;25 Abstr 8527. [Google Scholar]
  • 30.Hauschild A, Agarwala SS, Trefzer U, et al. Phase III randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel in second-line treatment in patients with unresectable stage III or stage IV melanoma. J Clin Oncol. 2009 doi: 10.1200/JCO.2007.15.7636. (in press) [DOI] [PubMed] [Google Scholar]
  • 31.Kefford R, Millward M, Hersey P, et al. Phase II trial of tanespimycin (KOS-953), a heat shock protein-90 (Hsp90) inhibitor in patients with metastatic melanoma. 2007 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2007;25 Abstr 8558. [Google Scholar]
  • 32.Fecher LA, Cummings SD, Keefe MJ, Alani RM. Toward a molecular classification of melanoma. J Clin Oncol. 2007;25:1606–1620. doi: 10.1200/JCO.2006.06.0442. [DOI] [PubMed] [Google Scholar]
  • 33.Tsai J, Lee JT, Wang W, et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA. 2008;105:3041–3046. doi: 10.1073/pnas.0711741105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dummer R, Robert C, Chapman PB, et al. AZD6244 (ARRY-142886) vs temozolomide (TMZ) in patients (pts) with advanced melanoma: an open-label, randomized, multicenter, phase II study. 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008;26 Abstr 9033. [Google Scholar]
  • 35.Haluska P, Dy GK, Adjei AA. Farnesyl transferase inhibitors as anticancer agents. Eur J Cancer. 2002;38:1685–1700. doi: 10.1016/s0959-8049(02)00166-1. [DOI] [PubMed] [Google Scholar]
  • 36.Fouladi M, Nicholson HS, Zhou T, et al. A phase II study of the farnesyl transferase inhibitor, tipifarnib, in children with recurrent or progressive high-grade glioma, medulloblastoma/primitive neuroectodermal tumor, or brainstem glioma: a Children's Oncology Group study. Cancer. 2007;110:2535–2541. doi: 10.1002/cncr.23078. [DOI] [PubMed] [Google Scholar]
  • 37.Raynaud FI, Eccles S, Clarke PA, et al. Pharmacologic characterization of a potent inhibitor of class I phosphatidylinositide 3-kinases. Cancer Res. 2007;67:5840–5850. doi: 10.1158/0008-5472.CAN-06-4615. [DOI] [PubMed] [Google Scholar]
  • 38.Garlich JR, De P, Dey N, et al. A vascular targeted pan phosphoinositide 3-kinase inhibitor prodrug, SF1126, with antitumor and antiangiogenic activity. Cancer Res. 2008;68:206–215. doi: 10.1158/0008-5472.CAN-07-0669. [DOI] [PubMed] [Google Scholar]
  • 39.Zhang M, Fang X, Liu H, et al. Bioinformatics-based discovery and characterization of an AKT-selective inhibitor 9-chloro-2-methylellipticinium acetate (CMEP) in breast cancer cells. Cancer Lett. 2007;252:244–258. doi: 10.1016/j.canlet.2006.12.035. [DOI] [PubMed] [Google Scholar]
  • 40.Abraham RT, Eng CH. Mammalian target of rapamycin as a therapeutic target in oncology. Expert Opin Ther Targets. 2008;12:209–222. doi: 10.1517/14728222.12.2.209. [DOI] [PubMed] [Google Scholar]
  • 41.Mita MM, Britten CD, Poplin E, et al. Deforolimus trial 106—a phase I trial evaluating 7 regimens of oral deforolimus (AP23573, MK-8669). 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008;26 Abstr 3509. [Google Scholar]
  • 42.Papadopoulos KP, Markman B, Tabernero J, et al. A phase I dose-escalation study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of a novel PI3K inhibitor, XL765, administered orally to patients (pts) with advanced solid tumors. 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008;26 Abstr 3510. [Google Scholar]
  • 43.Smalley KS, Contractor R, Haass NK, et al. An organometallic protein kinase inhibitor pharmacologically activates p53 and induces apoptosis in human melanoma cells. Cancer Res. 2007;67:209–217. doi: 10.1158/0008-5472.CAN-06-1538. [DOI] [PubMed] [Google Scholar]
  • 44.Hodi FS, Friedlander P, Corless CL, et al. Major response to imatinib mesylate in KIT-mutated melanoma. J Clin Oncol. 2008;26:2046–2051. doi: 10.1200/JCO.2007.14.0707. [DOI] [PubMed] [Google Scholar]
  • 45.Guilhot F, Apperley J, Kim DW, et al. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase. Blood. 2007;109:4143–4150. doi: 10.1182/blood-2006-09-046839. [DOI] [PubMed] [Google Scholar]
  • 46.Siddiquee KA, Gunning PT, Glenn M, et al. An oxazole-based small-molecule Stat3 inhibitor modulates Stat3 stability and processing and induces antitumor cell effects. ACS Chem Biol. 2007;2:787–798. doi: 10.1021/cb7001973. [DOI] [PubMed] [Google Scholar]
  • 47.Puri N, Ahmed S, Janamanchi V, et al. c-Met is a potentially new therapeutic target for treatment of human melanoma. Clin Cancer Res. 2007;13:2246–2253. doi: 10.1158/1078-0432.CCR-06-0776. [DOI] [PubMed] [Google Scholar]
  • 48.Mhaidat NM, Zhang XD, Jiang CC, Hersey P. Docetaxel-induced apoptosis of human melanoma is mediated by activation of c-Jun NH2-terminal kinase and inhibited by the mitogen-activated protein kinase extracellular signal-regulated kinase 1/2 pathway. Clin Cancer Res. 2007;13:1308–1314. doi: 10.1158/1078-0432.CCR-06-2216. [DOI] [PubMed] [Google Scholar]
  • 49.Roforth MM, Tan C. Combination of rapamycin and 17-allylamino-17-demethoxygeldanamycin abrogates Akt activation and potentiates mTOR blockade in breast cancer cells. Anticancer Drugs. 2008;19:681–688. doi: 10.1097/CAD.0b013e3283067681. [DOI] [PubMed] [Google Scholar]
  • 50.Smalley KS, Contractor R, Nguyen TK, et al. Identification of a novel subgroup of melanomas with KIT/cyclin-dependent kinase-4 overexpression. Cancer Res. 2008;68:5743–5752. doi: 10.1158/0008-5472.CAN-08-0235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Middleton M, Hauschild A, Thomson D, et al. Results of a multicenter randomized study to evaluate the safety and efficacy of combined immunotherapy with interleukin-2, interferon-{alpha}2b and histamine dihydrochloride versus dacarbazine in patients with stage IV melanoma. Ann Oncol. 2007;18:1691–1697. doi: 10.1093/annonc/mdm331. [DOI] [PubMed] [Google Scholar]
  • 52.Galustian C, Labarthe MC, Bartlett JB, Dalgleish AG. Thalidomide-derived immunomodulatory drugs as therapeutic agents. Expert Opin Biol Ther. 2004;4:1963–1970. doi: 10.1517/14712598.4.12.1963. [DOI] [PubMed] [Google Scholar]
  • 53.Gonzalez R, Lawson DH, Weber RW, et al. Phase II trial of elesclomol (formerly STA-4783) and paclitaxel in stage IV metastatic melanoma (MM): Subgroup analysis by prior chemotherapy. 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008 Abstr 9036. [Google Scholar]
  • 54.Tuma RS. Reactive oxygen species may have antitumor activity in metastatic melanoma. J Natl Cancer Inst. 2008;100:11–12. doi: 10.1093/jnci/djm299. [DOI] [PubMed] [Google Scholar]
  • 55.Rixe O, Bukowski RM, Michaelson MD, et al. Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study. Lancet Oncol. 2007;8:975–984. doi: 10.1016/S1470-2045(07)70285-1. [DOI] [PubMed] [Google Scholar]
  • 56.Fruehauf JP, Lutzky J, McDermott DF, et al. Axitinib (AG-013736) in patients with metastatic melanoma: a phase II study. 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008;26 Abstr 9006. [Google Scholar]
  • 57.Cory S, Adams JM. The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer. 2002;2:647–656. doi: 10.1038/nrc883. [DOI] [PubMed] [Google Scholar]
  • 58.Willis SN, Adams JM. Life in the balance: how BH3-only proteins induce apoptosis. Curr Opin Cell Biol. 2005;17:617–625. doi: 10.1016/j.ceb.2005.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Green DR. At the gates of death. Cancer Cell. 2006;9:328–330. doi: 10.1016/j.ccr.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 60.Hetz CA. ER stress signaling and the BCL-2 family of proteins: from adaptation to irreversible cellular damage. Antioxid Redox Signal. 2007;9:2345–2355. doi: 10.1089/ars.2007.1793. [DOI] [PubMed] [Google Scholar]
  • 61.Certo M, Del Gaizo Moore V, Nishino M, et al. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006;9:351–365. doi: 10.1016/j.ccr.2006.03.027. [DOI] [PubMed] [Google Scholar]
  • 62.Chen L, Willis SN, Wei A, et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403. doi: 10.1016/j.molcel.2004.12.030. [DOI] [PubMed] [Google Scholar]
  • 63.Willis SN, Chen L, Dewson G, et al. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes Dev. 2005;19:1294–1305. doi: 10.1101/gad.1304105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhuang L, Lee CS, Scolyer RA, et al. Mcl-1, Bcl-XL and Stat3 expression are associated with progression of melanoma whereas Bcl-2, AP-2 and MITF levels decrease during progression of melanoma. Mod Pathol. 2007;20:416–426. doi: 10.1038/modpathol.3800750. [DOI] [PubMed] [Google Scholar]
  • 65.Jiang CC, Lucas K, Avery-Kiejda KA, et al. Up-regulation of Mcl-1 is critical for survival of human melanoma cells upon endoplasmic reticulum stress. Cancer Res. 2008;68:6708–6717. doi: 10.1158/0008-5472.CAN-08-0349. [DOI] [PubMed] [Google Scholar]
  • 66.Bedikian AY, Millward M, Pehamberger H, et al. Bcl-2 antisense (oblimersen sodium) plus dacarbazine in patients with advanced melanoma: the Oblimersen Melanoma Study Group. J Clin Oncol. 2006;24:4738–4745. doi: 10.1200/JCO.2006.06.0483. [DOI] [PubMed] [Google Scholar]
  • 67.Iwasa T, Okamoto I, Suzuki M, et al. Radiosensitizing effect of YM155, a novel small-molecule survivin suppressant, in non-small cell lung cancer cell lines. Clin Cancer Res. 2008;14:6496–6504. doi: 10.1158/1078-0432.CCR-08-0468. [DOI] [PubMed] [Google Scholar]
  • 68.van Delft MF, Wei AH, Mason KD, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10:389–399. doi: 10.1016/j.ccr.2006.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen S, Dai Y, Harada H, et al. Mcl-1 down-regulation potentiates ABT-737 lethality by cooperatively inducing Bak activation and Bax translocation. Cancer Res. 2007;67:782–791. doi: 10.1158/0008-5472.CAN-06-3964. [DOI] [PubMed] [Google Scholar]
  • 70.Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–3428. doi: 10.1158/0008-5472.CAN-07-5836. [DOI] [PubMed] [Google Scholar]
  • 71.MacVicar GR, Kuzel TM, Curti BD, et al. An open-label, multicenter, phase I/II study of AT-101 in combination with docetaxel (D) and prednisone (P) in men with hormone refractory prostate cancer (HRPC). 2008 ASCO Annual Meeting Proceedings Part I. J Clin Oncol. 2008;26 Abstr 16043. [Google Scholar]
  • 72.Schimmer AD, O'Brien S, Kantarjian H, et al. A phase I study of the pan bcl-2 family inhibitor obatoclax mesylate in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14:8295–8301. doi: 10.1158/1078-0432.CCR-08-0999. [DOI] [PubMed] [Google Scholar]
  • 73.Mohammad RM, Goustin AS, Aboukameel A, et al. Preclinical studies of TW-37, a new nonpeptidic small-molecule inhibitor of Bcl-2, in diffuse large cell lymphoma xenograft model reveal drug action on both Bcl-2 and Mcl-1. Clin Cancer Res. 2007;13:2226–2235. doi: 10.1158/1078-0432.CCR-06-1574. [DOI] [PubMed] [Google Scholar]
  • 74.Zobel K, Wang L, Varfolomeev E, et al. Design, synthesis, and biological activity of a potent Smac mimetic that sensitizes cancer cells to apoptosis by antagonizing IAPs. ACS Chem Biol. 2006;1:525–533. doi: 10.1021/cb600276q. [DOI] [PubMed] [Google Scholar]
  • 75.Trudel S, Li ZH, Rauw J, et al. Preclinical studies of the pan-Bcl inhibitor obatoclax (GX015–070) in multiple myeloma. Blood. 2007;109:5430–5438. doi: 10.1182/blood-2006-10-047951. [DOI] [PubMed] [Google Scholar]
  • 76.Nguyen M, Marcellus RC, Roulston A, et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci USA. 2007;104:19512–19517. doi: 10.1073/pnas.0709443104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lin WM, Baker AC, Beroukhim R, et al. Modeling genomic diversity and tumor dependency in malignant melanoma. Cancer Res. 2008;68:664–673. doi: 10.1158/0008-5472.CAN-07-2615. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Annals of Oncology are provided here courtesy of Oxford University Press

RESOURCES