You are on page 1of 8

Cancer Treatment Reviews (2006) 32, 524– 531

available at www.sciencedirect.com

journal homepage: www.elsevierhealth.com/journals/ctrv

ANTI-TUMOUR TREATMENT

Targeted therapies in melanoma


Paola Queirolo *, Mirko Acquati

Department of Medical Oncology A, National Institute for Cancer Research, Largo Rosanna Benzi 10, Genova 16132, Italy

Received 18 May 2006; revised 13 July 2006; accepted 18 July 2006

KEYWORDS Summary In the last decade the incidence of melanoma has been rising. Despite this, survival
Melanoma; remains substantially constant because early diagnosis of thin lesions has increased. By con-
Monoclonal antibody; trast, metastatic melanoma continues to have a poor prognosis and it still represents a chal-
Protein-Kinase; lenge for oncologists. Response rates with single agent dacarbazine range from 10% to 25%
Targeted therapy with median survival of 8 months. The advent of new drugs with specific mechanisms of action
could help to improve the poor results of traditional therapies. In this review, we focused on
the novel agents that entered clinical trials in melanoma patients. We show the results of some
clinical trials with target-oriented drugs in melanoma patients. Moreover pre-clinical data and
rationale for use in melanoma was explained. Trials with protein-kinase inhibitors, anti-CTLA-4
agents, pro-apoptotic oligonucleotides and anti-angiogenic agents were reviewed. Combina-
tions with chemotherapeutic agents, immunotherapy and vaccine therapy were also analyzed.
c 2006 Elsevier Ltd. All rights reserved.

Introduction novel drugs such as fotemustine and temozolomide were


not better than DTIC.4,5 Therefore, new approaches are
Chemotherapeutic agents have a limited activity in meta- needed. Cytotoxic chemotherapy is not considered a tar-
static melanoma. Response rates with single agent dacar- geted therapy, because it is not specific and it is limited
bazine (DTIC) range from 10% to 25% with a median by toxicity. In the last decade researchers have focused
survival of 8 months.1 Polychemotherapy and biochemo- on the explanation of an altered signal network in cancer
therapy do not seem to improve survival. In fact, a four- cell. The identification of pathways involved in growth reg-
drug regimen like Dartmouth (cisplatin, BCNU, DTIC and ulation, which are important for cancer biology, allowed to
tamoxifen) was not superior to single agent DTIC while act with specificity at molecular level. The understanding
proving more toxic.2 On the other hand, the addition of of EGFR and angiogenesis pathways, and the development
cytokines, interleukin-2 and interferon alfa, to chemother- of agents directed on molecular targets, has led to very
apy regimens improved response rate and progression-free surprising results, in particular for breast6 and colorectal
survival but without any benefit on median survival.3 Also cancers.7 Many agents such as monoclonal antibodies
(mAb), antisense oligonucleotides and tyrosine-kinase
* Corresponding author. Tel.: +39 010 5600668; fax: +39 010 inhibitors are now available for clinical use and many more
5600850. are under investigation. Here we reviewed the progress in
E-mail address: paola.queirolo@istge.it (P. Queirolo). the field of melanoma.


0305-7372/$ - see front matter c 2006 Elsevier Ltd. All rights reserved.
doi:10.1016/j.ctrv.2006.07.009
Targeted therapies in melanoma 525

Methods (SD) were randomized to continue therapy or take placebo.


Twenty patients were recruited and results showed a
Research strategy and selection criteria modest activity (1 partial response (PR) and 3 SD), with
favorable safety profile. The association of BAY 43-9006
Data for this review were identified by searching medical may results in an enhancement of chemotherapy effect. A
databases, including PubMed, from 1990 to 2006. Only combination phase I/II study with carboplatin and paclitaxel
papers published in English were included. For the search and escalating doses of BAY 43-9006 was performed. BAY
we used terms such as ‘‘melanoma’’, ‘‘neoangiogenesis’’, 43-9006 was administered at three dose levels from day 2
‘‘apoptosis’’, ‘‘monoclonal antibodies’’, ‘‘protein-kinases’’ to 19 of a 21-day cycle, in association with carboplatin
and relevant subheadings. Also we retrieved references (AUC 6) and paclitaxel (225 mg/m2) on day 1.17 Thirty-five
cited in review articles. Moreover the past 5 years of metastatic-melanoma patients were enrolled and no
abstracts from the American Society of Clinical Oncology changes in the pharmacokinetics values of the chemothera-
were reviewed. peutic agents were seen. A promising response rate of 31%
was yielded, also with 54% of patients experiencing a SD.
Based on these results an ECOG phase-III trial (E2603) was
Protein-kinase inhibitors planned to assess the efficacy of carboplatin and paclitaxel
plus BAY 43-9006 versus the same chemotherapeutic
Protein kinases transmit signals to the nucleus through regimen plus placebo. DTIC is the most active chemothera-
intracellular cascade. The activity of kinases involved in peutic drug in melanoma. A phase-I trial was conducted to
transmission of proliferation signals in cancer cells often set the MTD in association with DTIC 1000 mg/m2 every
become more intense. Kinase inhibitors are generally small three weeks.18 Continuous administration of 400 mg bid
molecules designed to act with specificity on intracellular was safe. BAY 43-9006 exhibits very interesting activity in
protein-kinases. In particular two oral drugs, sorafenib and melanoma patients, in particular it seems to enhance
imatinib mesylate, and an intravenous drug, temsirolimus, antitumor response when added to chemotherapy with a
were tested in melanoma patients. good tolerability profile. Results of phase-III trials will
define the real impact of this agent in melanoma patients.
Sorafenib (BAY 43-9006)
Imatinib mesylate
Mitogen-activated protein-kinase pathways have been found
to be altered in various malignancies.8 Serine-threonine Imatinib mesylate (STI-571), an oral tyrosine-kinase inhibi-
kinases of the RAF family play an important role in the signal tor of c-Kit, PDGFR and BCR-ABL, has shown striking activity
transduction, resulting in the activation of ERK 1/2 and in GIST and chronic myeloid leukaemia.19,20 C-Kit is a trans-
consequently cell’s proliferation.9 Mutations of B-RAF have membrane receptor with tyrosine-kinase intracellular do-
been frequently described in melanoma cell lines.10 In main. When stem-cell factor binds c-Kit, it causes a
particular, a missense mutation, causing a valine to glu- dimerization and consequently an autophosphorylation,
tamic acid substitution in the ATP-binding site (V599E) and activating a down-stream signal that promotes prolifera-
thus constitutional activation, accounts for 90% of B-RAF tion. Imatinib blocks the ATP domain, preventing the bind-
mutations.11 About 70% of melanomas exhibit B-RAF altera- ing of ATP. Subsequent phosphorylations and signal
tions, therefore it is a suitable target for antitumor ther- transduction are avoided. Expression of c-Kit was observed
apy.12 Sorafenib (BAY 43-9006), a bi-aryl urea, is a small in nearly 50% of early stage melanomas, but c-Kit expression
molecule originally synthesized for B-RAF and C-RAF inhibi- is progressively reduced as the cells take on a more highly
tion. It targets the ATP-binding site of the kinase and, at low metastatic phenotype.21 In contrast PDGFR is highly repre-
concentrations, it inhibits in vitro both wild-type and sented on melanoma cells, also at a more advanced stage.22
mutant B-RAF. Moreover, sorafenib also inhibits other Although imatinib inhibits PDGFR phosphorylation in mice
tyrosine-kinase receptors involved in angiogenesis and model, it does not block the growth of melanoma cells.23
tumor progression, including vascular endothelial growth- In contrast Redondo et al. showed that imatinib was able
factor receptor (VEGFR)-2, VEGFR-3, c-kit and platelet- to suppress B16F10 melanoma-cell growth.24 A phase-II trial
derived growth-factor b (PDGFR-b). Wilhelm et al. demon- of 400 mg bid imatinib was performed in metastatic mela-
strated a significant inhibition of neovascularization in noma patients with at least a 25% cell expression of c-Kit.
xenograft models, showing a broad-spectrum antitumor Only one patient had high expression of c-Kit (75% of mela-
activity.13 Preclinical studies showed a significant retarda- noma cells) and in this patient a near complete response
tion in the growth of human melanoma tumor xenografts was achieved. Unfortunately the other patients pro-
with BAY 43-9006.14 A phase-I study established the maxi- gressed.25 In an attempt to improve results, a phase-I com-
mum tolerated dose (MTD) as single agent at 400 mg twice bination trial was undertaken by Schuchter et al.26
daily, the most common toxicities being gastrointestinal Temozolomide in addition to daily imatinib was adminis-
(mainly diarrhea), dermatological (skin rash/hand-foot tered at three dose levels. Therapy was well tolerated with
syndrome) and fatigue.15 A phase-II trial was conducted at no unexpected toxicities. Preliminary results showed no
the Royal Marsden hospital in patients with refractory responses, but only stabilization of disease as best result.
stage-IV melanoma.16 Single agent BAY 43-9006 was admin- The hypothesis that the benefit of treatment with imatinib
istered at 400 mg bid for 12 weeks. At tumor reassessment, could be restricted to patients whose melanoma cells have
responders continued therapy until disease progression or a high expression of c-Kit, has been tested in a multi-center
unacceptable toxicity, whereas patients with stable disease phase-II trial, involving 18 refractory melanoma patients.
526 P. Queirolo, M. Acquati

Immunohistochemical analysis revealed that 75%, 58%, and MDX-010 alone or in combination with DTIC in stage IV un-
67% of patients, whose specimens were available, had a treated melanoma patients.33 MDX-010 was administered
positive stain respectively for CD117/c-Kit, PDGFR-a at a dose of 3 mg/kg monthly for four cycles, alone (arm
and PDGFR-b. Despite these data, no patient achieved a re- A = 37 patients) or in combination (arm B = 35 patients) with
sponse. Moreover 38.9% of patients required a dose reduc- DTIC (250 mg/m2 for 5 days monthly). There were two PRs
tion for toxicity.27 Altogether, these results do not in arm A and 1 CR and 4 PRs in arm B, suggesting more activ-
support at present a role of imatinib in melanoma patients, ity for the combination arm. Toxicities were mainly colitis,
but further trials on very selected populations could explain diarrhea and skin rash. Of note, the authors also reported
which patients benefit of such therapy. two deaths possibly related to MDX-010. In the long-term
follow up phase of this study a further CR was yielded in
Temsirolimus the combination arm and durable clinical objective re-
sponses were observed.34 Another approach is the combina-
Temsirolimus (CCI-779) is a novel potent inhibitor of the so- tion of anti-CTLA-4 to vaccine therapy. The administration
called mammalian target of rapamycin (mTor). mTor is a to mice bearing tumor of anti-CTLA-4 mAb and a vaccine re-
serine-threonine kinase that promotes phosphorylation of sulted in melanoma rejection.35 In a trial by Phan et al., 14
S6K1 and the eukaryotic initiation factor 4E-binding pro- patients with metastatic melanoma were treated with a
tein-1 (4E-BP1), which are involved in G1 phase progression. combination of MDX-010 and vaccination with two modified
Temsirolimus is a rapamycin analog that specifically binds to HLA-A*0201-restricted peptides, from the gp100 melanoma-
FK506-binding protein 12 (FKBP12), forming a complex that associated antigen. Autoimmune events were reported (der-
interacts with mTor and blocking the subsequent phosphor- matitis, colitis and hepatitis), suggesting a breaking of
ylation cascade.28 A phase-II study of intravenous 250 mg peripheral tolerance against self-antigens. Anyway, cancer
temsirolimus every week in 33 patients with metastatic mel- regression was revealed for three patients and all patients
anoma was undertaken by Margolin et al.29 Toxicities were experienced T-cell reactivity against the immunizing pep-
mild including mucositis, rash and hyperlipidemia. One pa- tides.36 Moreover in a recent phase-I trial in resected
tient had a PR, but the authors concluded that as single stage-III/IV melanoma the combination of MDX-010 with
agent temsirolimus was inactive in melanoma. multiple-peptide melanoma vaccination has been shown to
be safe and has resulted in interesting immune response
to peptide vaccination.37 In this study 19 patients were trea-
Agents acting on immunological cells ted with intravenous MDX-010 at three dose levels (0.3 mg/
kg; 1 mg/kg; 3 mg/kg). The MTD was set at 1 mg/kg, be-
Melanoma is a highly immunological neoplasm, therefore cause in the highest-dose cohort (3 mg/kg) grade-3 gastroin-
target therapies directed at the immune system may be testinal toxicity was observed. Diarrhea and abdominal pain
an effective tool to enhance responses to other active were the dose-limiting toxicities. An autoimmune etiology
agents. Two interesting targets are cytotoxic T lymphocyte was suggested; in fact, a rectal biopsy revealed a CD4+ T-
antigen 4 (CTLA-4) and toll-like receptor 9 (TLR9). cell infiltrate. Furthermore, uveitis and rash were reported.
The authors noted that patients who developed autoim-
Anti-CTLA-4 mAb mune symptoms experienced longer relapse-free survival.
Similar results in terms of toxicity and activity in metastatic
T-cell activation requires two steps: stimulation via T-cell melanoma were obtained with another human anti-CTLA-4
receptor (TCR) and additional costimulatory signals. CD28 mAb, CP-675,206.38 The dose for further phase-II trials
is the main costimulatory molecule and it is expressed on was set at 10 mg/kg, but the pharmacokinetic analysis sug-
T-cells surface. CTLA-4, a homologue of CD28, is a glycopro- gested a relationship between clinical responses and plasma
tein expressed on the surface of activated T-cells.30 In par- concentration of CP-675,206. For this reason an accurate
ticular CTLA-4 seems to have an inhibitory function. It is exploration of higher doses could be suggested. Also, clini-
suggested that CTLA-4 has a role in down-regulating T-cell cal activity (5 PRs and 8 SD) and a decrease of the T-regula-
responses. In fact, a decrease in IL-2 production and an ar- tory cells that are involved in the suppression of immune
rest in cell-cycle progression has been observed. Blockade response was demonstrated by Reuben et al.39 These data
of CTLA-4 could be an effective strategy for the mainte- on anti-CTLA-4 strategy suggest a role in the enhancement
nance of T-cell antitumor responses. Administration of of antitumor response and a possible synergistic effect with
anti-CTLA-4 mAb into early-tumor-bearing mice resulted in anticancer vaccines. The relationship between autoimmu-
regression of growing tumors. Moreover, an enhancement nity phenomena and anti-tumor response requires further
of antitumor cytotoxicity in addition to chemotherapy has exploration, because it is suggested that development of
been reported.31 Some researchers focused on such therapy autoimmunity could be a surrogate endpoint for clinical
either alone or in combination. response.
MDX-010 is a fully-human IgG1 mAb that targets human
CTLA-4 resulting in augmentation and prolongation of T-cell
antitumor response. A phase-I study in metastatic mela- CpG 7909
noma patients was performed.32 A 3 mg/kg single dose of
MDX-010 was administered to 17 patients. The only toxici- A non-antisense oligodeoxynucleotide, CpG 7909, was de-
ties noted were a mild rash and pruritus. Two PRs were ob- signed to selectively target TLR9. Through agonism with
served including a 50% reduction of a lung lesion. Hersh TLR9, CpG 7909 activates plasmacytoid dendritic cells
et al. performed a phase-II study to assess the activity of (pDC) and B cells. Moreover it is a strong activator of both
Targeted therapies in melanoma 527

innate and specific immunity. Intralesional administration in administered by continuous intravenous infusion from days
melanoma metastases revealed anti-tumor activity.40 A 1 to 14 at dose levels ranging from 0.6 mg/kg/die to
phase-I/II study of single agent CpG 7909 was undertaken 6.5 mg/kg/die and DTIC was administered at a dose of
by Wagner et al.41 Weekly subcutaneous injection of 6 mg 200 mg/m2 from day 5 to day 9. In addition, in the cohorts
was administered to 20 patients until disease progression. of patients receiving 5.6 mg/kg and 6.5 mg/kg, oblimersen
Two PRs were obtained with three patients achieving a was given by subcutaneous injection from days 1 to 7 twice
SD. Moreover, the evaluation of peripheral-blood mononu- daily combined with 800 mg/m2 DTIC on day 5. Although
clear cells revealed an activation of pDC. Based on these evaluation of toxicity was the primary end-point, one com-
encouraging results, a phase-II trial with DTIC was de- plete and two partial responses were observed. Addition-
signed.42 Patients were randomized into four arms, one ally, two durable minor responses and two disease
arm with DTIC alone (850 mg/m2), two arms with CpG stabilization were noted. Toxicities included fever, liver-
7909 alone at different doses (10 mg and 40 mg) and one function abnormalities, skin rash and lymphopenia. A
arm with CpG 7909 at 40 mg in combination with DTIC. phase-III trial in chemotherapy-naı̈ve melanoma patients
CpG 7909 was administered every week, whereas DTIC was was completed.50 Seven-hundred seventy-one patients were
given every three weeks. At the preliminary analysis there randomized to receive DTIC alone (1000 mg/m2 every three
were 4 PRs in the combination arm, 2 PRs in the DTIC arm weeks) or DTIC plus oblimersen by intravenous continuous
and 1 PR in the 10-mg CpG 7909 arm. Adverse events in- infusion for five days at 7 mg/kg/die. Primary endpoint
cluded injection site reactions, fever, arthralgias and ana- was not met; in fact, overall survival (OS) was not statisti-
phylaxis. Also, CpG 7909 was tested in combination with cally significant between the two groups (9.1 months for
vaccines. The addition of CpG 7909 to a MAGE-3 vaccine the combination group vs 7.9 months for DTIC alone),
in MAGE-3 positive patients was safe.43 Immunological re- although response rate was significantly in favor of the
sponse to CpG 7909 and vaccination with Melan A/MART-1 oblimersen arm (11.7% vs. 6.8% p = 0.019) and a better pro-
was also reported to be strong in HLA-A2+ melanoma pa- gression-free survival was observed (74 vs. 49 days for both
tients. An expansion of antigen specific CD8+ T cells was p = 0.0003). Grade 3/4-neutropenia and thrombocytopenia
also shown.44 These data represent a basis for future re- were increased by the addition of oblimersen but without
searches either in combination with chemotherapy, immu- increasing the clinical consequences. In the updated analy-
notherapy or vaccine therapy. sis presented at the ASCO meeting 2005, OS had still not
reached statistical significance, but for the patient’ sub-
group with LDH value 62x ULN there was a significant ben-
Pro-apoptotic therapy efit for the combination therapy (10.2 vs. 8.7 months
p = 0.02). Moreover, the addition of oblimersen resulted in
Apoptosis is mediated by a sequence of events that result in more durable responses (7% vs. 3% p = 0.02).51 Oblimersen
cell death. The action of classic anticancer drugs on tumor- at present is the only target agent tested in a phase-III trial
cell death is dependent on induction of apoptosis. In cancer in melanoma. Results are promising but a longer follow-up
cell there is often an imbalance in favor of antiapoptotic and further trials are needed to set the efficacy of this
signals, which confers cells a resistance to chemotherapeu- agent.
tic drugs and radiation. Bcl-2, a family of proteins, play an
important role in cancer-cell survival. In fact, Bcl-2 inhibits
apoptosis by preventing the release of cytocrome c from Anti-angiogenic agents
mitochondria, thus preventing the activation of caspase-9
and caspase-3.45 Overexpression of Bcl-2 has been observed Neoangiogenesis has an important role in sustaining tumor
in many type of solid neoplasms and hematologic malignan- growth and in the process of metastatization. The develop-
cies, including breast carcinoma, small-cell lung cancer, ment of new vessels is a key step for neoplastic progression.
lymphoma, multiple myeloma and acute myeloid leukae- Several pro-angiogenic factors like vascular endothelial
mia. High levels of Bcl-2 protein were also found in mela- growth factor (VEGF), basic fibroblastic growth factor
noma. Nearly 90% of melanomas present overexpression (bFGF) and transforming growth factor b (TGF-b) are se-
of this anti-apoptotic protein.46 Strategies to down-regu- creted by cancer cells during hypoxic conditions.52 Among
late levels of Bcl-2 protein have been developed. these, the most important factor is VEGF type A. It stimu-
lates angiogenesis by binding to VEGFR-1 and VEGFR-2 ex-
Oblimersen pressed on the surface of endothelial cells.53

Oblimersen sodium (G3139) is an antisense oligonucleotide Bevacizumab


that targets Bcl-2 at mRNA level. It has an 18-nucleotide se-
quence that is complementary to the first six codons of the Bevacizumab is a recombinant humanized monoclonal anti-
Bcl-2 mRNA. When oblimersen binds to Bcl-2 mRNA, it forms body designed to specifically bind VEGF. The inhibition of
an heteroduplex, which enables RNase H to cleave the biologic activity of VEGF translates into reduction of micro-
mRNA sequence, degrading Bcl-2 message.47 In xenotrans- vascular growth and regression of existing vessels that feed
plantation models for human melanoma, a downregulation tumor.54 Bevacizumab improves the delivery of chemother-
of Bcl-2 concentration has been observed with the adminis- apy by altering tumor vasculature and reducing the elevated
tration of oblimersen and DTIC.48 Based on these preclinical interstitial pressure in tumors. Increased intratumoral
results, a phase-I-II trial was performed by Jansen et al. in microvessel density correlates to tumor aggressiveness in
patients with metastatic melanoma.49 Oblimersen was melanoma. Moreover VEGF is overexpressed in melanoma
528 P. Queirolo, M. Acquati

and strongly correlates to poor prognosis.55 Anti-angioge- Other agents


netic therapy could be beneficial in patients with mela-
noma. A phase-II trial of bevacizumab (15 mg/kg Bortezomib
intravenously every two weeks) with or without daily subcu-
taneous interferon alpha at a dose of 1 million units was car- Ubiquitination pathway is essential for maintaining protein
ried out. Sixteen patients with metastatic melanoma were homeostasis. Protein ubiquitination permits the recognition
enrolled. A complete response with disappearance of supra- by proteasome. Proteasome can proceed to the degradation
clavicular and mediastinal adenopathy was observed. More- of polyubiquitinated proteins through several peptidases. In
over, a patient achieved a PR. The responses were all in cancer cells critical proteins such as p53 and other tumor
patients receiving the two drugs. Four prolonged stabiliza- suppressing proteins could be regulated by the ubiquitin–
tion of disease were also reported.56 proteasome system.64 A constitutive activation of the nucle-
ar factor jB (NFjB) was reported in melanoma. NFjB
SU5416 prevents cells from apoptosis and promotes proliferation.
One strategy to block the activity of NFjB is by acting on
Inhibition of VEGF activity is also possible by acting at tyro- 26S subunit of proteasome. In fact, the inhibitor of NFjB,
sine-kinase receptor level. SU5416 is a potent inhibitor of IjB, is a target for the proteasome system and its degradation
VEGFR-1 tyrosine kinase (flk-1). In a phase-II trial in pre- promotes translocation of NFjB into the nucleus with subse-
treated melanoma patients, SU5416 was administered intra- quent activation of its target genes. Bortezomib (PS-341), a
venously at 145 mg/mq twice weekly after an adequate pre- low-molecular-weight dipeptide, inhibits the activity of pro-
medication against anaphylaxis. Among 31 patients, only teasome by binding to the site of chymotryptic-like en-
one patient experienced a PR. Anyway, evaluation by dy- zymes.65 A preclinical study by Katayoun et al. suggested
namic contrast magnetic resonance imaging showed a sig- good activity by combining temozolomide and bortezomib.66
nificant decrease in tumor perfusion. Toxicities included On the contrary, a clinical phase-II trial of bortezomib alone
lymphopenia and hyperglycemia.57 in metastatic melanoma showed negative results. In fact, the
study, planned to accrue 45 patients, was early stopped at
MEDI-522 the interim analysis with 27 patients enrolled, because of
lack of clinical activity. No responses were observed. The
Another target for anti-angiogenetic therapy is integrin alp- authors concluded that single agent bortezomib adminis-
havbeta3 (avb3). Its overexpression in melanoma cells is re- tered intravenously twice weekly for two weeks is ineffec-
lated to invasive growth and metastatic pattern.58 tive.67 Anyway, based on the preclinical data, combination
Moreover, integrin avb3 promotes an upregulation of bFGF, studies with chemotherapeutic agents are required for fur-
a well known pro-angiogenic factor.59 MEDI-522 is a novel ther evaluation of its activity in melanoma.
humanized monoclonal antibody designed to target integrin
avb3 and it was tested in a phase-II trial. Patients with met-
Marimastat
astatic melanoma were enrolled and randomized to DTIC
1000 mg/m2 every three weeks plus MEDI-522 8 mg/kg/
A family of zinc-dependent enzymes, matrix metallopro-
week (55 patients) or MEDI-522 alone (57 patients). In the
teinases (MMPs), are involved in the degradation of ECM.
combination arm the response rate approached 13%
Proteolysis of the components of ECM is a step required
whereas in the other arm no response was obtained. Median
for both local and metastatic spreads of the tumor. More-
survival was promising with the combination arm approach-
over, MMPs induce angiogenesis and activation of growth
ing 9.3 months and 11.8 months for single agent treatment.
factors.68 Marimastat (BB2516) is an oral inhibitor of MMPs
There were two deaths possibly attributed to MEDI-522.60
that blocks their action through mimicry of the collagen
site, thus preventing degradation and consequently tumor
PI-88 spread.69 Melanoma cells have been shown to induce an
up-regulation of MMPs.70 A phase-II study in refractory mel-
PI-88 is a new anti-angiogenetic agent. Due to heparan sul- anoma was performed by the NCIC-CTG (National Cancer
fate mimicry, PI-88 competitively binds both to angiogenic Institute of Canada Clinical Trials Group).71 Twenty-nine pa-
growth factors such as FGF and VEGF, thus inhibiting angio- tients were enrolled in this trial and the first 18 were trea-
genesis, and to heparanase, therefore preventing degrada- ted with 100 mg twice daily of oral Marimastat. Due to
tion of the extracellular matrix (ECM), a main step in the musculo-skeletal toxicities (mainly arthralgia, myalgia)
metastatization process.61 Moreover, an increase of tissue and fatigue, the subsequent eleven patients received
factor pathway inhibitor (TFPI), an antiangiogenic and 10 mg twice daily. Evaluation of disease revealed two PRs
anti-metastatic agent, was reported.62 A phase-II trial of and five SD, showing activity in melanoma. Data were inter-
250 mg/day of PI-88 was initiated in pre-treated patients. esting because only a control growth was expected whereas
PI-88 was administered subcutaneously for four days every also a tumor shrinkage was observed.
week.63 Grade-3/4 toxicities were thrombocytopenia,
thrombosis and an episode of brain hemorrhage. Disease
control rate (PRs+SD) was 36%. Conclusions
Anti-angiogenetic agents exert promising clinical activ-
ity, in particular by increasing growth control. Combination The intrinsic resistance of melanoma to conventional che-
trials with chemotherapy are warranted. motherapy has led researchers to evaluate new approaches.
Targeted therapies in melanoma 529

Table 1 Trials with target oriented drugs in melanoma


Drug Phase No. pts Responses Ref.
Sorafenib II 20 1 PR, 3 SD [16]
Sorafenib + CBDCA/paclitaxel I/II 35 11 PRs, 19 SD [17]
Imatinib mesylate II 31 1 PR [25]
Temsirolimus II 33 1 PR [29]
II [34]
MDX-010 Arm A 37 2 PRs, 4 SD
MDX-010 + DTIC Arm B 35 2 CRs, 4 PRs, 4 SD
CP-675,206 I/II 26 5 PRs, 8 SD [39]
II 184 [42]
CpG 7909 LD 0 OR
CpG 7909 HD 1 PR
CpG 7909 + DTIC 4 PRs
DTIC 2 PRs
III [51]
Oblimersen+DTIC Arm 1 386 48 ORs, 113 SD
DTIC Arm 2 385 26 ORs, 106 SD
Bevacizumab+/ IFNa II 16 2 PRs, 4 SD [56]
SU5416 II 31 1 PR, 1 SD [57]
II [60]
MEDI-522 Arm 1 57 0 PR, 26 SD
MEDI-522+DTIC Arm 2 55 7 PRs, 22 SD
PI-88 II 44 2 PRs, 3 SD [63]
Bortezomib II 27 0 PR, 6 SD [67]
Marimastat II 29 2 PRs, 5 SD [71]
Abbreviations: OR, objective response; PR, partial response, SD, stable disease.

Table 2 Future and ongoing phase III trials across USA and Europe
Drug Randomization Treatment line End-point
Sorafenib Arm 1: CBDCA/paclitaxel + sorafenib First line OS
Arm 2: CBDCA/paclitaxel + placebo
Sorafenib Arm 1: CBDCA/Paclitaxel + Sorafenib Progression after DTIC or TMZ PFS
Arm 2: CBDCA/paclitaxel + placebo
CP-675,206 Arm 1: CP-675,206 First line OS
Arm 2: DTIC or TMZ
MDX-010 Arm 1: DTIC + MDX-010 First line PFS
Arm 2: DTIC + placebo
MDX-010 Arm 1: MDX-010 Progression after DTIC, TMZ or IL-2 OS
Arm 2: MDX-1379 vaccine
Arm 3: MDX-010 + MDX-1379 vaccine
Abbreviations: TMZ, temozolomide; DTIC, dacarbazine; OS, overall survival; PFS, progression-free-survival.

Molecular biology and better knowledge of cancer cells al- such as autoimmunity for anti-CTLA-4 mAb, could be useful
lowed the development of targeted drugs. Many trials have to predict clinical response. In the near future, targeted
been performed, testing either single agent or combination therapies are going to join chemotherapy, immunotherapy
therapies (Table 1). Unfortunately most of these are phase- and vaccines as the oncologist’s tools in the treatment of
I/II studies, thus designed to evaluate safety and activity. melanoma.
Only with well-designed phase-III trials will it be possible
to assess the real efficacy of agents oriented to the molec-
References
ular target (Table 2). In fact, also biochemotherapy in
phase-II studies72,73 seemed to have an impact on mela- 1. Eggermont AMM, Kirkwood JM. Re-evaluating the role of
noma, but this effect disappeared in subsequent phase-III dacarbazine in metastatic melanoma: what have we learned
trials.74,75 Moreover, it will be important to select patients in 30 years? Eur J Cancer 2004;40:1825–36.
that will benefit from the administration of these new anti- 2. Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multi-
melanoma weapons. Novel surrogate clinical endpoints, center randomized trial of the Dartmouth regimen versus
530 P. Queirolo, M. Acquati

dacarbazine in patients with metastatic melanoma. J Clin 22. Shen SS, Zhang PS, Eton O, Prieto VG. Analysis of protein
Oncol 1999;17:2745–51. tyrosine kinase expression in melanocytic lesions by tissue
3. Eton O, Legha SS, Bedikian AY, et al. Sequential biochemo- array. J Cutan Pathol 2003;30:539–47.
therapy versus chemotherapy for metastatic melanoma: results 23. McGary EC, Onn A, Mills L, et al. Imatinib mesylate inhibits
from a phase III randomized trial. J Clin Oncol platelet-derived growth factor receptor phosphorylation of
2002;20:2045–52. melanoma cells but does not affect tumorigenicity in vivo. J
4. Avril MF, Aamdal S, Grob JJ, et al. Fotemustine compared with Invest Dermatol 2004;122:400–5.
dacarbazine in patients with disseminated malignant mela- 24. Redondo P, Lloret P, Andreu J, Inoges S. Imatinib mesylate in
noma: a phase III study. J Clin Oncol 2004;22:1118–25. cutaneous melanoma. J Invest Dermatol 2004;123:1208–9.
5. Middleton MR, Grob JJ, Aaronson N, et al. Randomized phase III 25. Eton O, Billings L, Kim K, et al. Phase II trial of imatinib
study of temozolomide versus dacarbazine in the treatment of mesylate (STI-571) in metastatic melanoma (MM). Proc Am Soc
patients with advanced metastatic malignant melanoma. J Clin Clin Oncol 2004;22. [abstr 7528].
Oncol 2000;18:158–66. 26. Schuchter L, Flaherty K, Davidson R, et al. Phase I trial of
6. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trast- imatinib and temozolomide in patients with metastatic mela-
uzumab after adjuvant chemotherapy in HER2-positive breast noma. Proc Am Soc Clin Oncol 2004;22. [abstr 7572].
cancer. N Engl J Med 2005;353:1659–72. 27. Ugurel S, Hildenbrand R, Zimpfer A, et al. Lack of clinical
7. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab efficacy of imatinib in metastatic melanoma. Br J Cancer
plus irinotecan, fluorouracil, and leucovorin for metastatic 2005;92:1398–405.
colorectal cancer. N Engl J Med 2004;350:2335–42. 28. Meric-Bernstam F, Mills GB. Mammalian target of rapamycin.
8. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene Semin Oncol 2004;31:10–7.
in human cancer. Nature 2002;417:949–54. 29. Margolin K, Longmate J, Baratta T, et al. CCI-779 in metastatic
9. Chong H, Vikis HG, Guan KL. Mechanisms of regulating the Raf melanoma. Cancer 2005;104:1045–8.
kinase family. Cell Signal 2003;15:463–9. 30. Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its
10. Satyamoorthy K, Li G, Gerrero MR, et al. Constitutive mitogen- biological function and use in tumor immunotherapy. Nat
activated protein kinase activation in melanoma is mediated by Immunol 2002;3:611–8.
both BRAF mutations and autocrine growth factor stimulation. 31. Mokyr MB, Kalinichenko T, Gorelik L, Bluestone JA. Realization
Cancer Res 2003;63:756–9. of the therapeutic potential of CTLA-4 blockade in low-dose
11. Sharma A, Trivedi NR, Zimmerman MA, Tuveson DA, Smith CD, chemotherapy-treated tumor-bearing mice. Cancer Res
Robertson GP. Mutant V599EB-Raf regulates growth and vascu- 1998;58:5301–4.
lar development of malignant melanoma tumors. Cancer Res 32. Tchekmedyian S, Glasby J, Korman A, Keler T, Deo Y, Davis TA.
2005;65:2412–21. MDX-010 (human anti-CTLA4): a phase I trial in malignant
12. Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations melanoma. Proc Am Soc Clin Oncol 2002;21. [abstr. 56].
in human lung cancer and melanoma. Cancer Res 33. Hersh EM, Weber J, Powderly J, et al. A phase II, randomized
2002;62:6997–7000. multi-center study of MDX-010 alone or in combination with
13. Wilhelm SM, Carter C, Tang L, et al. BAY 43-9006 exhibits dacarbazine (DTIC) in stage IV metastatic malignant melanoma.
broad spectrum anti-tumor activity and targets raf/MEK/ERK Proc Am Soc Clin Oncol 2004;22. [abstr. 7511].
pathway and receptor tyrosine kinases involved in tumor 34. Fischkoff SA, Hersh E, Weber J, et al. Durable responses and long-
progression and angiogenesis. Cancer Res 2004;64:7099–109. term progression-free survival observed in a phase II study of MDX-
14. Karasarides M, Chiloeches A, Hayward R, et al. B-RAF is a 010 alone or in combination with dacarbazine (DTIC) in metastatic
therapeutic target in melanoma. Oncogene 2004;23:6292–8. melanoma. Proc Am Soc Clin Oncol 2005;23. [abstr. 7525].
15. Strumberg D, Richly H, Hilger RA, et al. Phase I clinical and 35. van Elsas A, Sutmuller RP, Hurwitz AA, et al. Elucidating the
pharmacokinetic study of the Novel Raf kinase and vascular autoimmune and antitumor effector mechanisms of a treat-
endothelial growth factor receptor inhibitor BAY 43-9006 in ment based on cytotoxic T lymphocyte antigen-4 blockade in
patients with advanced refractory solid tumors. J Clin Oncol combination with a B16 melanoma vaccine: comparison of
2005;23:965–72. prophylaxis and therapy. J Exp Med 2001;194:481–9.
16. Ahmad T, Marais R, Pyle L, et al. BAY 43-9006 in patients with 36. Phan GQ, Yang JC, Sherry RM, et al. Cancer regression and
advanced melanoma: The Royal Marsden experience. Proc Am autoimmunity induced by cytotoxic T lymphocyte-associated
Soc Clin Oncol 2004;22. [abstr. 7506]. antigen 4 blockade in patients with metastatic melanoma. Proc
17. Flaherty KT, Brose M, Schucter L, et al. Phase I/II trial of BAY Natl Acad Sci USA 2003;100:8372–7.
43-9006, carboplatin (C) and paclitaxel (P) demonstrates 37. Sanderson K, Scotland R, Lee P, et al. Autoimmunity in a phase
preliminary antitumor activity in the expansion cohort of I trial of a fully human anti-cytotoxic T-lymphocyte antigen-4
patients with metastatic melanoma. Proc Am Soc Clin Oncol monoclonal antibody with multiple melanoma peptides and
2004;22. [abstr. 7507]. Montanide ISA 51 for patients with resected stages III and IV
18. Eisen T, Ahmad T, Gore ME, et al. Phase I trial of BAY 43-9006 melanoma. J Clin Oncol 2005;23:741–50.
(sorafenib) combined with dacarbazine (DTIC) in metastatic 38. Ribas A, Camacho LH, Lopez-Berestein G, et al. Antitumor
melanoma patients. Proc Am Soc Clin Oncol 2005;23. [abstr. activity in melanoma and anti-self responses in a phase I trial
7508]. with the anti-cytotoxic T lymphocyte-associated antigen 4
19. Demetri GD, von Mehren B, Blanke CD, et al. Efficacy and monoclonal antibody CP-675,206. J Clin Oncol 2005;23:8968–77.
safety of imatinib mesylate in advanced gastrointestinal stro- 39. Reuben M, Lee BN, Li C, et al. Biologic and immunomodulatory
mal tumors. N Engl J Med 2002;347:472–80. events after CTLA-4 blockade with ticilimumab in patients with
20. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a advanced malignant melanoma. Cancer 2006;106:2437–44.
specific inhibitor of the BCR-ABL tyrosine kinase in chronic 40. Trefzer U, Kors C, Pelzer K, Walden P, Sterry W, O’Keefe E.
myeloid leukemia. N Engl J Med 2001;344:1031–7. Preliminary results of a phase I trial of intralesional injection of
21. Janku F, Tomancova V, Novotny J, et al. Expression of c-Kit CPG 7909 in patients with basal cell carcinoma or melanoma.
was found in more than 50% of early stages malignant Proc Am Soc Clin Oncol 2002;21. [abstr. 1902].
melanoma. A retrospective study of 261 patients. Proc Am 41. Wagner SN, Pashenkov M, Goess G, et al. TLR9-targeted CpG
Soc Clin Oncol 2003;22. [abstr 2864]. immunostimulatory treatment of metastatic melanoma: A
Targeted therapies in melanoma 531

phase II trial with CpG 7909 (ProMune). Proc Am Soc Clin Oncol cells upregulates bFGF expression and promotes their angio-
2004;22. [abstr 7513]. genic phenotype. Int J Cancer 2005;116:27–35.
42. Wagner S, Weber J, Redman B, et al. CPG 7909, a TLR9 Agonist 60. Hersey P, Sosman J, O’Day S, et al. A phase II, randomized,
Immunomodulator in Metastatic Melanoma: A Randomized open-label study evaluating the antitumor activity of MEDI-522,
Phase II Trial Comparing Two Doses and in Combination with a humanized monoclonal antibody directed against the human
DTIC. Proc Am Soc Clin Oncol 2005;23. [abstr 7526]. alpha v beta 3 (avb3) integrin, ± dacarbazine (DTIC) in patients
43. Kruit WH, van Ojik H, Portielje J, Verloes R, Delire M, Stoter G. with metastatic melanoma (MM). Proc Am Soc Clin Oncol
Phase I/II study with CpG 7909 as adjuvant to vaccination with 2005;23. [abstr 7507].
MAGE-3 protein in patients with MAGE-3 positive tumors. Proc 61. Joyce JA, Freeman C, Meyer-Morse N, et al. A functional
Am Soc Clin Oncol 2002;21. [abstr 1854]. heparan sulfate mimetic implicates both heparanase and
44. Speiser DE, Lienard D, Rufer N, et al. Rapid and strong human heparan sulfate in tumor angiogenesis and invasion in a mouse
CD8+ T cell responses to vaccination with peptide, IFA, and CpG model of multistage cancer. Oncogene 2005;24:4037–51.
oligodeoxynucleotide 7909. J Clin Invest 2005;115:739–46. 62. Tobu M, Ma Q, Iqbal O, et al. Comparative tissue factor
45. Kim R, Emi M, Tanabe K, Toge T. Therapeutic potential of pathway inhibitor release potential of heparins. Clin Appl
antisense Bcl-2 as a chemosensitizer for cancer therapy. Cancer Thromb Hemost 2005;11:37–47.
2004;101:2491–502. 63. Thomson DB, Gonzalez R, Millward M, et al. A phase II trial of
46. Selzer E, Schlagbauer-Wadl H, Okamoto I, Pehamberger H, PI-88, a novel antiangiogenic and antimetastatic heparanase
Potter R, Jansen B. Expression of Bcl-2 family members in inhibitor, in patients with advanced melanoma. Proc Am Soc
human melanocytes, in melanoma metastases and in melanoma Clin Oncol 2005;23. [abstr 7559].
cell lines. Melanoma Res 1998;8:197–203. 64. Rajkumar SV, Richardson PG, Hideshima T, Anderson KC.
47. Klasa RJ, Gillum AM, Klem RE, Frankel SR. Oblimersen Bcl-2 Proteasome inhibition as a novel therapeutic target in human
antisense: facilitating apoptosis in anticancer treatment. Anti- cancer. J Clin Oncol 2005;23:630–9.
sense Nucleic Acid Drug Dev 2002;12:193–213. 65. Adams J, Palombella VJ, Sausville EA, et al. Proteasome
48. Jansen B, Schlagbauer-Wadl H, Brown BD, et al. Bcl-2 antisense inhibitors: a novel class of potent and effective antitumour
therapy chemosensitizes human melanoma in SCID mice. Nat agents. Cancer Res 1999;59:2615–22.
Med 1998;4:232–4. 66. Amiri KI, Horton LW, LaFleur BJ, Sosman JA, Richmond A.
49. Jansen B, Wacheck V, Heere-Ress E, et al. Chemosensitization Augmenting chemosensitivity of malignant melanoma tumors
of malignant melanoma Bcl-2 antisense therapy. Lancet via proteasome inhibition: implication for bortezomib (VEL-
2000;356:1728–33. CADE, PS-341) as a therapeutic agent for malignant melanoma.
50. Millward MJ, Bedikian AY, Conry RM, Gore ME, Pehamberger HE, Cancer Res 2004;64:4912–8.
Sterry W. Randomized multinational phase 3 trial of dacarba- 67. Markovic SN, Geyer SM, Dawkins F, et al. A phase II study of
zine (DTIC) with or without Bcl-2 antisense (oblimersen sodium) bortezomib in the treatment of metastatic malignant mela-
in patients (pts) with advanced malignant melanoma (MM): noma. Cancer 2005;103:2584–9.
Analysis of long-term survival. Proc Am Soc Clin Oncol 2004;22. 68. Brown PD, Giavazzi R. Matrix metalloproteinase inhibition: a
[abstr 7505]. review of anti-tumour activity. Ann Oncol 1995;6:967–74.
51. Kirkwood JM, Bedikian AY, Millward MJ, et al. Long-term 69. Ramnath N, Creaven PJ. Matrix metalloproteinase inhibitor.
survival results of a randomized multinational phase 3 trial of Curr Oncol Rep 2004;6:96–102.
dacarbazine (DTIC) with or without Bcl-2 antisense (oblimersen 70. Loffek S, Zigrino P, Angel P, Anwald B, Krieg T, Mauch C. High
sodium) in patients (pts) with advanced malignant melanoma invasive melanoma cells induce matrix metalloproteinase-1
(MM). Proc Am Soc Clin Oncol 2005;23. [abstr 7506]. synthesis in fibroblasts by interleukin-1 alpha and basic fibro-
52. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic blast growth factor-mediated mechanisms. J Invest Dermatol
switch. Nat Rev Cancer 2003;3:401–10. 2005;124:638–43.
53. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, 71. Quirt I, Bodurth A, Lohmann R, et al. Phase II study of
Holash J. Vascular-specific growth factors and blood vessel marimastat (BB-2516) in malignant melanoma: a clinical and
formation. Nature 2000;407:242–8. tumor biopsy study of the National Cancer Institute of Canada
54. Kerbel R, Folkman J. Clinical translation of angiogenesis Clinical Trials Group. Invest New Drugs 2002;20:431–7.
inhibitors. Nat Rev Cancer 2002;2:727–39. 72. Legha SS, Ring S, Eton O, et al. Development of a biochemo-
55. Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased serum therapy regimen with concurrent administration of cisplatin,
concentration of angiogenic factors in malignant melanoma vinblastine, dacarbazine, interferon alfa, and interleukin-2 for
patients correlates with tumor progression and survival. J Clin patients with metastatic melanoma. J Clin Oncol
Oncol 2001;19:577–83. 1998;16:1752–9.
56. Carson WE, Biber J, Shah N, et al. A phase 2 trial of a 73. Atkins MB, O’Boyle KR, Sosman JA, et al. Multiinstitutional
recombinant humanized monoclonal anti-vascular endothelial phase II trial of intensive combination chemoimmunotherapy
growth factor (VEGF) antibody in patients with malignant for metastatic melanoma. J Clin Oncol 1994;12: 1553–60.
melanoma. Proc Am Soc Clin Oncol 2003;22. [abstr 2873]. 74. Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Prospec-
57. Peterson AC, Swiger S, Stadler WM, Medved M, Karczmar G, tive randomized trial of the treatment of patients with
Gajewski TF. Phase II study of the Flk-1 tyrosine kinase inhibitor metastatic melanoma using chemotherapy with cisplatin,
SU5416 in advanced melanoma. Clin Cancer Res 2004;10: dacarbazine, and tamoxifen alone or in combination with
4048–54. interleukin-2 and interferon alfa-2b. J Clin Oncol 1999;17:
58. Albelda SM, Mette SA, Elder DE, Stewart RM, Damjanovich S, 968–75.
Herlyn M, et al. Integrin distribution in malignant melanoma: 75. Del Vecchio M, Bajetta E, Nova P, et al. Multicenter phase III
association of the beta 3 subunit with tumor progression. randomized trial of polychemotherapy (CVD regimen) versus
Cancer Res 1990;50:6757–74. the same chemotherapy (CT) plus subcutaneous interleukin-2
59. Dome B, Raso E, Dobos J, et al. Parallel expression of and interferon-{alpha}2b in metastatic melanoma. Ann Oncol
alphaIIbbeta3 and alphavbeta3 integrins in human melanoma 2006;17:571–7.

You might also like