Non-BRAF-targeted therapy, immunotherapy, and ...

2 downloads 23 Views 712KB Size Report
Mar 21, 2014 - The more prom- ising ones as single agents are axitinib and lenvatinib and their investigations alone or in combination are moving forward. In.
Review

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma 1.

Introduction

2.

FDA-approved treatments for metastatic melanoma

3.

Targeted therapy

4.

Immunotherapy

5.

Combination therapy

6.

Expert opinion

Sara Tomei†, Ena Wang, Lucia Gemma Delogu, Francesco M Marincola & Davide Bedognetti †

National Institutes of Health, Clinical Center and trans-NIH Center for Human Immunology (CHI), Department of Transfusion Medicine, Infectious Disease and Immunogenetics Section (IDIS), Bethesda, MD, USA

Introduction: Melanoma is an aggressive disease characterized by a complex etiology. The discovery of key driving mutations (primarily BRAF mutations) led to the development of specific molecular inhibitors providing clinical benefit. Areas covered: Although BRAF-specific drugs have perhaps yielded the best results in melanoma-targeted therapy, there still remain several limitations, mostly due to the emergence of resistance and the lack of efficacy in patients without BRAF mutation. Novel drugs are currently being tested in clinical trials and showed encouraging results. Such drugs can specifically target molecular pathways aberrantly activated or repressed during melanoma development (targeted therapy) or act in a way to enhance the host immune system to fight cancer (immunotherapy). Here we provide a detailed overview of the current clinical strategies, which lay beyond BRAF-targeted therapy, spanning from molecular-targeted therapy to immunotherapy and to combination therapy. Expert opinion: Major advances in our understanding of the mechanisms behind melanoma development have led to the implementation of novel therapeutic drugs. Unfortunately, tools allowing prediction of responsiveness to a given treatment are not available yet. The increasing availability of highthroughput technologies will allow the elucidation of molecular mechanisms underlying responsiveness to cancer therapy and unveil an increased number of potential therapeutic targets. Keywords: adoptive T-cell transfer, combination therapy, immunotherapy, melanoma, targeted therapy Expert Opin. Biol. Ther. [Early Online]

1.

Introduction

Cutaneous melanoma is a highly heterogeneous disease characterized by a complex etiology. If diagnosed early, it can be potentially cured by surgical resection; however, once metastasized, it is highly refractory to conventional antineoplastic treatments, urging the identification of novel effective therapeutic solutions. High-throughput experimental strategies, such as gene mutation analysis and comparative genomic hybridization, allowed the identification of crucial signaling pathways essential for melanoma development and progression [1]. The recognition of molecular aberrations occurring during melanoma development facilitated the application of new therapeutic strategies, which have improved the clinical management of melanoma patients [2,3]. The best results have been obtained after the introduction of BRAF inhibitors. BRAF is a serine-threonine kinase, member of the MAPK pathway; when activated, 10.1517/14712598.2014.890586 © 2014 Informa UK, Ltd. ISSN 1471-2598, e-ISSN 1744-7682 All rights reserved: reproduction in whole or in part not permitted

1

S. Tomei et al.

Article highlights. .

.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

.

.

.

A huge amount of novel therapeutic strategies are being developed for the treatment of metastatic melanoma, including targeted therapies, immunotherapies, and combinatorial strategies. FDA recently approved two new drugs for metastatic melanoma, namely: ipilimumab (anti-cytotoxic T-lymphocyte antigen 4 [anti-CTLA]-4), dabrafenib (BRAF inhibitor), and trametinib (MAPK extracellular signalregulated kinase [MEK] inhibitor). Several additional drugs are being tested and Phase I/II clinical trials are providing encouraging results. MAPK and Phosphatidylinositol 3-Kinase (PI3K) pathways are the most frequently altered pathways in melanoma. Pharmaceutical inhibitors, specifically blocking altered oncogene proteins within these pathways, have been proved to counteract melanoma development (e.g., BRAF inhibitors, MEK inhibitors, AKT inhibitors). Steps forward have been made in the field of melanoma immunotherapy, considering the exceptional immunogenicity of melanoma. These advances refer to high-dose IL-2, vaccination strategies, immune checkpoint blockade, and adoptive T-cell transfer. Albeit targeted therapy and immunotherapy strategies have revolutionized the clinical management of melanoma patients, it is becoming clear that combinatorial strategies are most likely to result in even more significant advances. Examples include BRAF inhibitors plus mTOR inhibitors, BRAF inhibitors plus MEK inhibitors, BRAF inhibitors plus PI3K inhibitor, combination of immune-checkpoint inhibitors and also targeted therapy plus immunotherapy.

This box summarizes key points contained in the article.

it transfers growth signals to the nucleus of the cells. The high success of BRAF inhibitors is mainly due to the fact that melanoma development often relies on the activation of MAPK pathway. BRAF mutations occur in about 50 -- 60% of melanoma [4-7]. More than 90% of BRAF mutations result in a single nucleotide mutation in the 600 codon (V600) [5,6]. Among them, about 90% of mutation are represented by the valine to glutamic acid substitution (V600E) [5,6], associated with a 400-fold increased activity of the protein [8,9]. Overall, approximately 50% of melanoma patients carry the V600E mutation. Vemurafenib, a kinase inhibitor acting by blocking V600E mutated BRAF, have been extensively investigated in Phase II and III trials [4,10]. It has been approved by FDA in August 2011 for the treatment of metastatic or unresectable melanoma patients carrying the V600E mutation. In the interim analysis of the Phase III trial (BRIM-3), vemurafenib has been shown to induce an objective response (OR) in 48% of the patients, significantly higher than that obtained by dacarbazine (5%) [4]. Beyond tumor shrinkage, administration of vemurafenib resulted in a significantly prolonged progression-free survival (PFS; 6.9 vs 1.6 months; vemurafenib vs dacarbazine, respectively) and 2

overall survival (OS; 13.6 vs 9.7 months; vemurafenib vs dacarbazine), according to a recent update of the BRIM-3 trial [11]. In the same setting, similar results in term of OR and PFS have been obtained by the BRAF inhibitor dabrafenib, which has been approved by FDA in May 2013. However, the trial allowed crossover and was not powered enough to detect differences in OS [12]. Even though Phase III trials focused on patients bearing V600E mutation, BRAF inhibitors may be similarly active in patients carrying other V600 mutations (i.e., V600K and V600D) [4,10,13,14]. These observations led the European Medical Agency (EMA) to express favorable opinion on the use of vemurafenib and dabrafenib in metastatic or unresectable melanoma patients carrying any kind of BRAF V600 mutation. However, despite BRAF inhibition has shown the most promising results in the field of melanoma targeted therapy, several challenges remain. The first important obstacle is that half of melanoma patients do not carry V600 mutations and therefore cannot be treated with BRAF inhibitors. Moreover, a complete response (CR) only occurs in a very few cases (3 -- 6%), the duration of response rate is relatively short (5 -- 7 months) and all but few patients relapse within 1 or 2 years through developing secondary resistance [4,10,12,13]. Thus, much effort needs to be put into developing novel, more effective therapeutic approaches. The purpose of this review is to discuss the most significant advances in the treatment of metastatic melanoma, highlighting the novel therapeutic strategies (currently being tested in clinical trials [15]), which preclude BRAF treatment, encompassing single molecular-targeted therapy, immunotherapy, and combinatorial therapeutic approaches.

FDA-approved treatments for metastatic melanoma

2.

To date there are four FDA-approved treatments for metastatic melanoma besides the two aforementioned BRAF inhibitors: recombinant IL-2, ipilimumab (approved, respectively, in 1998 and 2011; discussed in the following sections), dacarbazine, and trametinib [4,16-18]. Dacarbazine is an alkylating agent, which was accepted as standard treatment for metastatic melanoma in the 1970s [19,20], and it remains the only cytotoxic chemotherapy drug approved for the treatment of metastatic melanoma. In preliminary studies, OR was reported in up to 25% of patients [21]. However, in more rigorous randomized trials, dacarbazine has shown to induce an OR in about 10% of patients (range 5 -- 12%) [4,12,21,22]. CR is exceptionally rare (1 -- 2%) [4,12,21,22], remission is in general transient [21], and advantage in OS has not been conclusively demonstrated in randomized trials. Although other cytotoxic agents (e.g., fotemustine and temozolomide) have been shown to be at least not inferior to dacarbazine, none of them produces superior benefit to dacarbazine in terms of OS [21]. For this reason, dacarbazine is often chosen as control arm in randomized trials assessing new therapeutics. Trametinib, a MAPK extracellular

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

signal–regulated kinase (MEK) inhibitor (see following section), was approved by FDA on 29 May 2013 (concurrently with the approval of dabrafenib) for the treatment of patients with unresectable or metastatic melanoma harboring BRAF V600E or V600K mutation [23]. Currently, several drugs are being tested and several encouraging results have been recently published. Therapeutic strategies for the treatment of melanoma can be systematically divided in three broad categories: targeted therapy, immunotherapy, and combination therapy. Targeted treatments act by directly inhibiting molecules (e.g., proteins encoded by protoncogenes or growth factors), which are essential for the tumor growth. Immunotherapy treatments aim at stimulating the immune system of the host to reject cancer cells. Combination therapy refers to the use of multiple approaches to achieve a stronger anticancer response and overcome potential mechanisms of resistance. Examples of the most effective treatments belonging to each of the above categories will be given in the following sections of this review. 3.

Targeted therapy

Biologic targeted therapy usually refers to a treatment that specifically targets molecules involved in neoplastic process with less harm to normal cells. Targeted treatments act by blocking the activity of specific molecules essential for cancer growth and development. The majority of targeted therapies include either small molecules or monoclonal antibodies. The testing and implementation of possible therapeutic drugs in melanoma is growing up at an exponential rate as our understanding of the genetic heterogeneity of this disease improves. An interesting recent study from Henary et al. [24] showed that the administration of molecularly matched therapies predicted better outcomes, emphasizing the relevance of targeted therapy in the context of personalized medicine. Melanoma cells can acquire a proliferative advantage or protection against cell death by deregulating molecular pathways relevant to cell biology. The understanding of cell growth and molecular signaling pathways in melanoma is essential to develop and implement new therapeutic drugs. In the following, we provide an overview of the most commonly deregulated signaling pathways in melanoma responsible for cell proliferation and cell survival, and the most relevant advance in clinical setting. MAPK pathway MAPK pathway is probably the best-studied signaling pathway in cancer, most likely because of its essential role in melanoma onset and development [25]. A simplified representation of MAPK pathway is given in Figure 1. Signaling through MAPK pathway occurs upon the binding of extracellular growth factors to a variety of tyrosine kinase receptors (TKRs), including c-KIT and c-MER, which in turn lead to the activation of RAS. RAS is a small GTP-binding protein, which can exist in three isoforms: HRAS (Harvey-RAS), KRAS (Kirsten-RAS), and NRAS 3.1

(neuroblastoma-RAS). NRAS is the most prevalent form in melanoma being constitutively activated by point mutation in about 15% of cases [26]. Once activated, NRAS binds BRAF, which further phosphorylates and activates MEK. MEK finally signals to ERK, which translocates into the nucleus and enhances the transcription of a bunch of transcription factors resulting in increased cell proliferation and increased cell survival. Activating mutations within MAPK pathway account for 90% of melanomas [27]. Studies have reported a constitutive activation of MAPK pathway through mutation and/or gene amplification of c-KIT. C-KIT gene encodes a cell-membrane tyrosine kinase receptor, which plays an essential role in cellular differentiation [28]. The role of c-KIT in chronic myelogenous leukemia (CML) and gastrointestinal stromal tumors (GIST) has been well established and the use of imatinib, an oral c-KIT inhibitor, has revolutionized the treatment of these diseases [29]. The use of c-KIT inhibitors in melanoma represents an evolving matter of investigation. C-KIT mutations account for about 2% of cutaneous melanoma, albeit mutation and/or amplification of c-KIT are found in about 20 -- 25% in some melanoma subtypes, such as mucosal melanoma, acral melanoma, or melanoma arising from chronically sun-damaged skin [30,31]. Consistent with the essential role of c-KIT in melanocytes differentiation and migration, several melanomas express c-KIT. However, experimental evidence from studies in thyroid and melanoma showed that the loss of this growth factor receptor is strangely related to a worse malignant phenotype [32-34]. This may be explained by the acquisition of a less-differentiated phenotype during tumor progression. A number of drugs targeting c-KIT have been developed and their effectiveness has been proved in GISTs. These drugs include sunitinib, nilotinib, sorafenib, dasatinib, and imatinib, and their therapeutic activity has been shown to be dependent on the c-KIT specific type of molecular alteration [35-37]. Results of c-KIT inhibitor trials in metastatic melanoma were initially disappointing. Three Phase II trials did not show clinical activity of imatinib in unselected melanoma patients [38-40]. The scenario substantially changed when the studies focused on melanoma patients carrying c-KIT mutations and/ or amplifications, therefore highlighting the importance of patient selection in early targeted-therapy development. According to two Phase II trials, indeed, an OR is achieved in almost 25% of patients, and temporary stabilization occurs in about 50 -- 70% of subjects [31,41]. Although results in terms of OS seem encouraging (median OS from 11 to 14 months), the median PFS appears to be low, ranging from 3 to 3.5 months [31,41]. Importantly, responses seem to be restricted to patients carrying hot spot mutations either in exon 11, 13, or multiple mutations [31,41]. There is a great interest for c-KIT inhibition in mucosal and acral melanoma and other drugs, such as sunitinib, nilotinib, and dasatinib, are currently investigated in Phase II trials [42]. In view of the growing enthusiasm in this field, the Society for Immunotherapy of Cancer (SITC) melanoma expert panels recommend to routine testing of

Expert Opin. Biol. Ther. (2014) 14(5)

3

S. Tomei et al.

c-KIT

GPCR

Cytosol

PIP2

GNAQ/GNA11

PKC

PI3K

NRAS

PTEN PIP3

PDK1

BRAF

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

ERK1/2 ELK

MITF

AKT

BAD

MEK1/2

BCL2

JUN

mTOR

BAX

Proliferation, differentiation

IKK

NFκB

MDM2

GSK3β

p53

β-catenin

Figure 1. Simplified representation of MAPK and PI3K pathways. Red bolts indicate targets of molecular alterations in melanoma. AKT: V-Akt murine thymoma Viral Oncogene Homolog; BAD: BCL2-associated agonist of cell death; BAX: BCL2-associated X protein; BCL2: B cell lymphoma-2; BRAF: V-raf murine sarcoma viral oncogene homolog B; c-KIT: V-Kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog; ELK: ETS-domain protein; ERK1/ 2: Extracellular signal--regulated kinase 1 and 2; GPCR: G protein--coupled receptor; GSK3b: Glycogen synthase kinase 3 b; IKK: IkB kinase; JUN: V-Jun avian sarcoma virus 17 oncogene homolog; MDM2: Mouse double minute-2; MEK1/2: MAPK extracellular signal–regulated kinase 1 and 2; MITF: Microphthalmia transcription factor; mTOR: Mammalian target of rapamycin; NFkB: nuclear factor-kB; NRAS: Neuroblastoma RAS; p53: Tumor protein 53; PDK1: Phosphoinositidedependent kinase-1; PI3K: Phosphoinositide-3 kinase; PIP: Phosphatidylinositol phosphate; PKC: Protein kinase C; PTEN: Phosphatase and tensin homolog.

c-KIT mutations in clinical practice [43]. Whether acquisition of resistances to imatinib in melanoma is due to the acquisition of additional c-KIT mutations, as demonstrated in GIST [44], remains to be elucidated [42]. Similarly to c-KIT, another important RTK in melanoma is c-MER tyrosine kinase (MERTK); c-MER is a member of the TAM (TYRO, AXL, MERTK) family of RTKs. A recent study from Schlegel and colleagues [45] has pointed c-MER as a potential therapeutic target in melanoma. In this work, they were able to show that treatment of melanoma cell lines with UNC1062, a novel c-MER inhibitor, induced apoptosis and inhibited invasion of melanoma cells, encouraging a deeper testing of such drug. In addition to the deregulation of RTKs, several MAPK downstream molecules are often altered in melanoma. The first oncogene to be discovered was NRAS [46]. As mentioned earlier, NRAS is a member of the rat sarcoma (RAS) protein family, together with HRAS and KRAS. RAS are GTPbinding proteins, which, once activated by RTKs, can signal a complex network of downstream molecular pathways. Although evidence shows KRAS and HRAS to be implicated in melanoma [47,48], NRAS is the most active form. Most common mutations in NRAS gene have been found located at codon 61 [49], causing the impairment of NRAS enzymatic activity to switch GTP to GDP. Albeit several attempts have been made to directly target NRAS, no clinical success has been reached yet. One probable explanation is the low 4

concentration range (picomolar) required for the affinity of GTP to bind NRAS, which made the development of GTPspecific antagonists impossible so far [50]. Nevertheless, significant effort has been spent to indirectly inhibit NRAS by targeting key post-translational modifications, such as farnesylation. Farnesylation consists in a lipid modification necessary for NRAS membrane association. The inhibition of farnesylation process results in the blockage of NRAS translocation to the plasma membrane. A study from Niessner and colleagues [51] has shown that the farnesyltransferase inhibitor (FTI) lonafarnib was also able to inhibit mTOR signaling pathway enhancing apoptosis induced by sorafenib in melanoma cell lines. However, testing FTI tipifarnib (also called R115777) in a Phase II clinical trial failed to obtain encouraging results [52], most likely due to aspecific responses as farnesylation occurs in the maturation process of several proteins other than RAS. Although in this trial patients were unselected for NRAS mutation, the intrinsic difficulty to develop NRAS inhibitors and the scarce activity of this approach in other tumors have shifted the efforts to inhibit MAPK pathway toward more effective targets. Another important protein kinase belonging to MAPK pathway is MEK, and its inhibition is currently under investigation. Considering that MEK activation has been shown to be a resistance mechanism by which melanoma treated with BRAF inhibitors relapse, MEK inhibition was tested in combination with BRAF inhibitors (see combination therapy

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

section subsequently), yielding extremely promising results. MEK proteins include MEK1 and MEK2, which are the only substrate of BRAF. Mutations on MEK genes are very rare, although they have been reported in a few cases [49]. However, the acquisition of MEK1 mutations P124L and Q56P have been demonstrated to confer resistance to BRAF and MEK inhibitors [53]. A recent review by Salama and Kim [54] lists a broad variety of MEK inhibitors currently under investigation. Worthy of note, selumetinib is an oral potent inhibitor of MEK1 and MEK2 kinases. The efficacy of this drug was evaluated in a large Phase II clinical trial in advanced melanoma patients. Unfortunately, the study failed to show a difference of the OR rate and PFS between the treatment arm and the control group (temozolomide) [55]. Interestingly, five out of six responders were BRAF mutated. Combination of selumetinib and dacarbazine was reported to be clinically active in a Phase II randomized trial (PFS 5.6 vs 3.0 months, selumetinb plus dacarbazine vs dacarbazine alone), although nonsignificant benefit in terms of OS was observed [56]. Several clinical trials are still testing MEK inhibitors in melanoma, given the fact that MAPK reactivation is often driven by MEK signaling. Among the MEK inhibitors tested, trametinib has shown so far the most promising results. In a Phase III study, 322 patients carrying V600E or V600K mutation were randomized to receive either trametinib or chemotherapy (either decarbazine or paclitaxel) [23]. A statistical significant prolongation of PFS was shown for patients treated with trametinib compared to the control group (4.8 vs 1.5 months). OR rate was also higher in trametinib arm than in chemotherapy arm (22 vs 8%). Despite the crossover to trametinib at progression, the 6 months OS was significantly higher in the trametinib arm (81 vs 67%) [23]. On the basis of these results, FDA approved trametinib for the treatment of metastatic melanoma harboring V600E or V600K mutations. Importantly, tremetinib is not active if administered to patients progressing on a BRAF inhibitor, stressing the need to better identify the window of efficacy of trametinib in BRAF mutant melanoma [57]. However, patients carrying NRAS mutations cannot benefit from such treatment, urging the need to find other additional treatment options. Recently, Ascierto and colleagues [58] in an open-label, nonrandomized, Phase II trial assessed the efficacy of MEK162, a potent MEK inhibitor, in patients carrying NRAS or V600E BRAF mutations. In addition to an OR rate of 20% in BRAF-mutated patients, they were able to show that MEK162 has activity in NRAS mutant patients (20% of OR rate), although no CR was observed. However, all patients relapsed within 9 months. A similar OR rate (18%) in patients bearing NRAS mutation has been observed with the use of pimasertib [59], another MEK inhibitor under clinical development. These proof-of-principle studies pinpoint MEK inhibition as potential new approach for NRAS mutant melanoma, which till now has few treatment options. Once again, the

main limitation of MAPK-targeted therapy is the rapid development of resistance. However, while melanoma can take different roads to counteract to MAPK-targeted inhibition, the reactivation of the extracellular-signal-regulated kinases (ERK) appears to be a common feature [60,61]. Two isoforms of ERK, ERK1 and ERK2, transmit proliferative signals once phosphorylated by the upstream MEK proteins. Quite interestingly, Morris and colleagues [62] recently identified a potent ATP-competitive ERK inhibitor by screening a library of 5 million compounds. Such inhibitor, called SCH772984, showed promising clinically attractive results as it was able to inhibit ERK phosphorylation in a dose-dependent manner. Testing this compound in vivo is warranted to prove its efficacy and its potential relevance in clinical settings. Phosphatidylinositol 3-kinase pathway Phosphatidylinositol 3-kinase (PI3K) pathway is the second most frequently deregulated pathway in melanoma after MAPK pathway (Figure 1). Similarly to MAPK, PI3K cascade is triggered by RTKs and RAS protein. Once activated by NRAS, PI3K can catalyze the phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-trisphosphate (PIP3). PIP3 functions as a second messenger, and its production is antagonized by the tumor suppressor phosphatase and tensin homolog (PTEN). PIP3 can activate AKT (through the phosphatidylinositoldependent kinase 1, PDK1), which, in turn, phosphorylates mTOR and multiple additional targets determining increased oncogenic transformation, survival, proliferation, and cellcycle regulation [63]. The deregulation of PI3K pathway in melanoma occurs through activation of PI3K (encoded by PI3KCA gene), PTEN loss, and AKT activation. The independent therapeutic relevance of inhibiting PI3K pathway itself has not been well established yet, however, preclinical evidence suggests that PI3K pathway inhibition may be a relevant adjunct to MAPK-targeted therapy. Several small molecule inhibitors targeting AKT and mTOR activity have been developed and tested in melanoma cell lines and xenografted animal models [64]. However, the number of clinical studies currently evaluating their clinical efficacy is very low. The AKT inhibitors wortmannin and LY294002 have been shown to block AKT activity in a variety of functional studies, but their application in clinical settings is limited by their offtarget activities [63,65]. Several derivatives of rapamycin, such as temsirolimus, have also been used as mTOR inhibitors and have demonstrated clinical activity in other malignancies such as renal cell carcinoma. Nevertheless, the administration of temsirolimus (either used alone or in combination with the multiple kinase inhibitor sorafenib) failed to show significant clinical activity in Phase II trials [66,67]. Even though several additional AKT inhibitors, such as API-2, SR13668, BI-69A11, GSK690693, and MEK-2206 are under investigation, it is widely believed that PI3K pathway inhibition works better when combined to other targeted therapies. An interesting study by Werzowa and colleagues 3.2

Expert Opin. Biol. Ther. (2014) 14(5)

5

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

S. Tomei et al.

assessed the potential efficacy of PI3K pathway vertical inhibition in melanoma cell lines and xenograft models [68]. The concurrent treatment of rapamycin and the mTOR inhibitor PI-103 had a potent antitumor activity both in vitro and in vivo. Although in general PI3K and mTOR inhibition seems to be a promising strategy against cancer development and some drugs have been already approved for the treatment of renal cancer [69], such drugs did not obtain the same success in melanoma. The reason behind this phenomenon is not clear yet albeit it has been hypothesized that the ability of rapamycin derivatives to phosphorylate AKT may impede their clinical success [70]. Nevertheless, MEK and PI3K combination seems promising. An interesting in vitro study by Greger et al. showed that combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to dabrafenib, mediated by NRAS or MEK mutations [71]. Another study showed that combined MEK and PI3K/ mTOR pathway inhibition induces regression of NRASmutant melanoma cell lines [72]. An increasing number of clinical trials combining PI3K and MAPK inhibitions in melanoma are currently undergoing or in development. As a note, inhibition of Bcl-2, a key anti-apoptotic protein expressed in cancer cells, through the antisense oligonucleotide oblimersen seemed to increase OS in a subset of patients (i.e., those with normal or low level of LDH) [73], but these findings were not confirmed in a recent randomized Phase III trial [74]. In light of recent in vitro or in vivo studies, modulation cellcycle checkpoints (which are de-regulated in melanoma) and inhibition of G protein-coupled receptor signaling (found to be activated in up to 80% of uveal melanoma) have recently gained the attention of clinical investigators [42,72]. Remarkably, two very recent investigations have found that > 70% of melanoma bears somatic mutation of telomerase reverse transcriptase (TERT) promoter, therefore highlighting another important oncogenic mechanism [75,76]. Angiogenesis Because melanoma is a highly vascularized cancer, several attempts to therapeutically target angiogenic mechanisms in melanoma have been explored. Anti-angiogenic drugs have been developed and demonstrated to be useful in different types of cancer. Still, their clinical benefit in melanoma is not clear. Among the anti-angiogenic drugs, bevacizumab, a humanized monoclonal antibody targeting the VEGF A (VEGF-A), has been approved for use in certain malignancies, such as colon (in combination with chemotherapy), kidney (in combination with IFN-a), lung (in combination with chemotherapy), and brain cancers. In metastatic melanoma, the use of bevacizumab as single agent, or in combination with IFN-a failed to show significant clinical activity [77]. According to a large randomized Phase II trial (BEAM) enrolling 314 treatment naı¨ve patients, bevacizumab, in combination with chemotherapy enhances, though not significantly, PFS and OR (4.2 vs 5.6 month; 3.3

6

16 vs 26%; PFS and OR, bevacizumab vs bevacizumab plus carboplatin and paclitaxel). OS advantage was initially significant (8.6 vs 12.3 months; p = 0.04), but it was reduced in a subsequent analysis with longer follow-up, (9.2 vs 12.3 months; p = 0.19) [78]. Although it is not clear if this combination will move forward, the BEAM study, which is often considered a positive Phase II trial, still represents one of the best evidence of the activity of anti-angiogenic therapy in advanced melanoma [79]. Interestingly, Del Vecchio and co-workers [80] evaluated the clinical and biological activity of the use of bevacizumab in combination with fotemustine, a chemotherapeutic drug widely used in Europe. The authors were able to show that such regimen had important clinical activity as first-line treatment of metastatic melanoma, obtaining an encouraging OS of 20.5 months (PFS = 8.8 months, OR 15%), although grade 3 -- 4 hematological toxicity was quite high. Attempts to use bevacizumab in clinical trials employing other combinatorial setting are currently underway and results are awaited. Recently, a preliminary analysis of 20 patients enrolled in a Phase I study assessing combination of the cytotoxic T-lymphocyte antigen 4 (CTLA-4) mAb ipilimumab and bevacizumab showed an intriguing OR of 38%; all the responses lasted > 6 months [81]. Similarly to VEGF-A mAbs, other drugs targeting the VEGF receptors (VEGFRs) have been tested in melanoma. Among them, sorafenib, which inhibits tyrosine kinases associated with VEGF and plateled-derived growth factor, failed to demonstrate significant clinical activity in combination with chemotherapy in Phase III trials [82,83]. The more promising ones as single agents are axitinib and lenvatinib and their investigations alone or in combination are moving forward. In a multicenter, Phase II study, axitinib monotherapy resulted in a 19% OR rate [84]. Similarly, in a Phase I evaluation of patients with solid malignancies, including melanoma, lenvatinib (also called E7080) demonstrated an OR of 21% [85]. Based on this evidence, it is likely that anti-angiogenic therapies might be helpful in treating melanoma especially when combined with other drugs and much effort should be spent in identifying the more beneficial anti-angiogenic combination therapies. Moreover, other novel molecules, as those mimicking natural anti-angiogenic agent inhibitors (e.g., endostatin), inhibitors of placental growth factor, and agents blocking HIFa, a key molecule in hypoxia signaling, are currently under development [86]. 4.

Immunotherapy

Cancer immunotherapy is meant as the treatment aiming at annealing tumors by inducing, enhancing, or suppressing specific immune responses. Aside from the molecular alterations within proliferative pathways described earlier, several immune alterations have been reported in melanoma, including decreased number of peripheral B and T cells, increased number of regulatory T cells, impaired activation of natural

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

killer (NK) cells (due to downregulation of activating ligands), acquisition of tolerance by CD8+ T cells (due to increased expression of inhibitory receptors), increased level of pro-tumorigenic cytokines such as TGF-b, TNF-a, IL-6, IL-8, and IL-1, and altered expression of immune-related genes [87,88]. Immunotherapy strategies for melanoma treatment carry a high clinical relevance considering the exceptional immunogenicity of such disease. The immunogenicity of melanoma has been established based on the high number of tumor-infiltrating lymphocytes (TIL) found in resected melanoma samples, and several strategies have been proposed to conquer melanoma taking advantage of this peculiar feature. A diverse set of melanoma antigens have been identified as target of cellular immune response, which can be grouped into three broad categories: tumor differentiation antigens (TDA), including melanoma antigen recognized by T cells 1 (MART-1), glycoprotein 100 (gp100), tyrosinase (TYR), tyrosinase-related protein 1 (TYRP1), and 2 (TYRP2), which are found expressed in normal melanocytes and are overexpressed in melanoma; tumor-specific antigen (TSA), including the melanoma antigen (MAGE), the B melanoma antigen (BAGE), and G antigen (GAGE) gene families, the NY-ESO-1 (also known as CTAG1), which are found in normal gametic cells in the testes and in cancers in addition to melanoma but are not expressed by normal melanocytes; and mutated antigens, including b-catenin, which can be found in different tissues. Additionally, several melanoma antigens have been identified as targets of humoral immune responses. The expression of melanoma antigens is of great importance as it allows the innate and adaptive immune system to track and destroy nascent tumor cells, through a mechanism known as ‘immune surveillance’. However, cancer cells can escape immune surveillance by becoming poorly immunogenic. This phenomenon has been commonly referred to as ‘immune editing’ [89]. Several mechanisms of immune editing have been described, including the downregulation of antigen presentation MHC molecules by the tumor cells themselves, the secretion of soluble factors and cytokines, such as TGF-b and IL-10, which inhibit effective immune response, the creation of an immune suppressive environment by recruiting inhibitory immune cells, such as T-reg, NKs, and macrophages, and the expression of negative regulators of the immune system, including programmed death ligand 1 (PD-L1) and the CTLA-4 [90]. We report subsequently the best-characterized immunotherapy strategies for the treatment of melanoma.

Nonspecific immunotherapy: IL-2 IL-2 is a pro-inflammatory cytokine regulating T cells and NK cells homeostasis. Studies in animal models and humans have shown that IL-2 has strong antitumor activity [91,92] 4.1

and high-dose IL-2 (HD-IL-2) was approved by FDA in 1998 for the treatment of metastatic melanoma. HD-IL-2 was initially shown to have dose-dependent anticancer effect by the group of Steven Rosenberg at the National Cancer Institute [93]. FDA approved HD-IL-2 in 1998 based on the observation that it can induce long-term remissions. In large retrospective analyses, administration of HD-IL-2 induce an OR in about 15% of patients (range 13 -- 19%) of patients, and CR in 4 -- 6% [92,94-96]. However, in a recent randomized Phase III trial, these percentages dropped from 10 to 6% for OR and from 2 (two patients) to 1% (one patient) for CR when evaluation was assessed by central review. Even though the real rate of OR needs to be determined, complete-remission under HD-IL-2 appear to be extremely durable (median CR duration > 14 years, according to a long follow-up analysis of a retrospective casuistic) [95]. However, HD-IL-2-related adverse events are severe (e.g., hemodynamic shock, respiratory insufficiency, and neurotoxicity) and require intensive inpatient care [21]. The opinions of European and US specialists about the risk--benefit ratio of this treatment largely diverge. In light of the potentially curative role of HD-IL-2, the US National Comprehensive Cancer Network US guidelines still list HD-IL-2 as treatment option in a subset of (clinically fit) patients, if the center has a considerable experience in the management of this regimen. Even more, the (US) melanoma panelists of the Society for Immunotherapy of Cancer (SITC) recommend the administration of HD-IL-2 as first-line treatment in good performance status patients without central nervous system metastases, even in presence of BRAF mutation [43]. Conversely, the European Society of Medical Oncology (ESMO) melanoma panelists do not encourage its routine use in clinical practice, in view of the high toxicity and the lack of randomized trials demonstrating survival advantage in the overall population [97]. Considering the relatively low benefit--risk ratio, one of the main limitations of HD-IL-2 is the lack of pretreatment tools to predict patients who are likely to respond to the treatment. Recently, in a proteomic study, we showed that low levels of fibronectin and VEGF, an angiogenetic factor with immunosuppressive functions, predict response to HD-IL-2 [98] in metastatic melanoma patients. Although these findings need to be prospectively validated, Hodi and co-workers recently reported that low levels of VEGF also predict OS and clinical response to ipilimumab in metastatic melanoma [99] Noteworthy, an interesting study from Joseph and colleagues has recently shown that patients harboring NRAS mutation carry a higher likelihood to respond to therapy [96], linking, for the first time, alterations of the MAPK pathway to the responsiveness to HD-IL-2. Although IL-2 has been used in oncology since > 20 years, its mechanism of action is still not completely understood. Only few studies have attempted to comprehensively understand the mechanisms behind the cancer rejection mediated by IL-2.

Expert Opin. Biol. Ther. (2014) 14(5)

7

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

S. Tomei et al.

Our group performed a series of proteomic and transcriptomic analyses of peripheral mononuclear cells (PBMCs) and pre- and post-melanoma metastasis using fine-needle aspiration biopsies (FNA) obtained from patients undergoing systemic IL-2 administration [98,100-103]. We observed that administration of IL-2 induces a cytokine storm already after the first administration, where most of the cytokines investigated (e.g., IFN-g, IL-10, and TNF-a) increased significantly compared with the baseline value. CXCR3 ligands (i.e., CXCL9, 10 and 11) and CCR5 ligands (i.e., CCL3 and CCL4) were found among the chemokines that significantly increased early after treatment [100,101]. Even when investigated by whole genome gene-expression analysis, the effects of HD-IL-2 were dramatic within PBMCs, whereas effects within the tumor microenvironment were lesion-specific and limited [103]. Altogether these findings suggest that the main effect of IL-2 in the tumor is the activation of innate mechanisms driven by monocytes and eventually sustained by NK, and the tumor infiltrating lymphocytes. These cells in concert can initiate an inflammatory chemotactic cascade, orchestrated by the activation of the IRF-1/ IFN-g pathway and inducing release of related specific chemotactic ligands (i.e., CXCR3 and CCR5 ligands) with consequent polarized (Th1) recruitment of other immune cells later on [104]. Importantly, the degree of inflammation induced by HD-IL-2 correlates with the development of tumor rejection [102,103]. Vaccination strategies Cancer vaccines aim at treating cancer by stimulating the host’s immune reaction against cancer cells. The implementation of therapeutic vaccines to treat melanoma remains a challenge [105-107]. Broadly, melanoma vaccines are classified into the following main categories: whole-cell vaccines (either autologous or allogeneic, prepared using melanoma cells or subcellular fraction); dendritic-cell (DC) vaccines (which take advantage of the highly specialization of DCs to present antigens and induce primary cellular immune responses); peptide vaccines (which use melanoma antigens to stimulate cytotoxic T lymphocytes response against tumor cells); ganglioside vaccines (which take advantage of the high expression of gangliosides on the surface of melanoma cells to generate an immune response against them); DNA vaccines (composed of naked DNA expression plasmids that include a gene encoding the target antigens from tumor cells); viral vector vaccines (in which a virus is used as vector of melanoma antigens); and oncolytic vaccine (in which an oncolytic virus replicating in cancer cells is used to mediate tumor cell lysis) [107-110]. The best-characterized vaccines for melanoma treatment are the peptide-based ones directed to activate T-cell response [111]. The recognition of the critical role of T cells in the immune-mediated treatment of melanoma dates back to 1988, when Rosenberg and colleagues first showed the regression of metastatic melanoma through the autologous transfer 4.2

8

of tumor infiltrating lymphocytes combined with IL-2 [112]. In 1991, van der Bruggen et al. described for the first time a gene encoding an antigen recognized by T cells (MAGE-1) [113]. This study, followed by the description of the first cancer-specific epitope HLA-A1 restricted [114], gave molecular accuracy to this rather disregarded field and provided the opportunity to investigate with scientific precision the phenomenon of immune-mediated cancer rejection. This led, in turn, to the characterization of a myriad of tumor antigens and their immunodominant peptides, with consequent implementation of active immunization strategies [115]. The study of the tumor-antigen-specific immunization restricted to an individual epitopic determinant offered the opportunity to study the dynamic of the immune response in humans by reducing the algorithm of tumor--host crosstalk to a specific HLA/epitope interaction with the complementary T-cell receptor [116]. These studies have shown that, in order to develop clinical active immunotherapies, factors beyond T-cell/HLA/epitope interactions must be considered. In fact, the large majority of the studies failed to report an association between specific response against the administered antigens and clinical benefit [104]. Humoral and/or cellular immune response against vaccine antigens are necessary for the development of an effective antitumor response but they are certainly not sufficient [117]. Other factors, as the presence of co-stimulatory or inhibitory stimuli in tumor microenvironment, genetic polymorphisms of the host, genetic of the tumors, and environmental factors (i.e., intestinal microbiota) [118], can certainly modify the algorithm of immune response, making the evaluation of a single parameter unlike to predict the clinical benefit [119-121]. Besides local toxicity, vaccines are in general extremely well tolerated. However, in melanoma, as well as in many other solid tumors, vaccination alone has failed to induce significant tumor regression, the OR rate being around 3 -- 4% [122]. Nevertheless, recent randomized Phase III trials in other malignancies (i.e., prostate cancer) have shown that vaccine administration can prolong survival even in absence of detectable tumor shrinkage [123]. The convincing detection of a remarkable survival benefit dissociated with measurable changes in the tumor size forced us to reconsider the mechanism of actions of vaccinations. An interesting interpretation of these paradigmatic results comes from the analysis of tumor kinetic following different treatments. By applying a rather novel mathematical model to Phase II trials in prostate cancer patients receiving chemotherapy or PROSTVAC vaccine (consisting in recombinant poxviral vectors containing transgenes for prostate specific antigen (PSA) and the costimulatory molecules B7), Fojo and Schlom showed that chemotherapy determines an initial reduction of tumor burden followed, at relapse, by a tumor-growth rate similar to that before chemotherapy [124,125]. Vaccination, however, modifies clinical outcome (OS) by reducing tumor-growth rate. The same mechanisms likely explain differences between survival curves in BRAF and ipilimumab trials (Figure 2).

Expert Opin. Biol. Ther. (2014) 14(5)

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

OS (%)

Targeted therapy (BRAFi)

Immunotherapy (anti-CTLA-4 mAb)

100

100

100

80

80

80

60

60

60

40

40

40

20

20

20

0

12

24

36

48

0

PFS (%)

Time (months) Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Combination therapy

12

24

36

Time (months)

100

80

80

80

60

60

60

40

40

40

20

20

20

24

Time (months)

36

48

0

12

24

12

24

36

48

Time (months)

100

12

> Comb-TT

0

48

100

0

> Comb-TT

36

Time (months)

48

> Comb-TT > Comb-TT

0

12

24

36

48

Time (months)

Figure 2. Survival curves of BRAF inhibitors, CTLA-4 mAb (ipilimumab) and combination therapy. In each panel blue line represents the control treatment (dacarbazine). Orange line represents the alternative treatment (BRAF inhibitors, ipilimumab, or combination therapy), as indicated. The BRAF inhibitor curves were approximated from the vemurafenib and dabrafenib randomized Phase III trials. Immunotherapy curves were approximated from ipilimumab randomized trials. Dotted line represents survival projection when no data are available. PFS of combination therapy was derived from the dabrafenib plus trametinib Phase I/II randomized trial. BRAF inhibition induces response in most of the patients, however the response is transient, and all but few patients relapse within 1 or 2 years through developing secondary resistance. The long-term benefit in OS is expected to be very low. Vice versa, immunotherapy (i.e., ipilimumab) determines a delayed response in a small proportion of patients. Response is however durable, with impact on long-term survival. Combination immuno-targeted therapy could act sinergically therefore switching the curve on the right (targeted therapy) and up (immunotherapy). Although results from such combinations are not available yet, certain combination of immunotherapy (i.e., ipilimumab and IL-2 or IL-2 and vaccine) seems to increase survival, therefore shifting the curve up. However, it seems that combination of BRAF and MAPK extracellular signalregulated kinase inhibitors as well as certain immunotherapy combination (i.e., ipilimumab and the anti-PD1 nivolumab) are able to shift the curve on right and up by inducing deep, frequent and durable antitumor response. Even though preliminary data suggest that survival curve of anti-PD1 and anti-PD1 ligand will differ as compared with those of ipilimumab, they are not represented here because no formal survival curves of PD-1/PD-1 ligand trials are available yet. BRAFi: BRAF inhibitors; CTLA-4: Cytotoxic T-lymphocyte antigen 4; comb: Combination; IT: Immunotherapy; OS: Overall survival; PFS: Progression-free survival; TT: Targeted therapy.

Even though several scientific progresses in the field of vaccinology have been accomplished during the past decade, it is clear that active immunization alone would unlikely represent a curative option for patients affected by advanced melanoma. The rather disappointing results of cancer vaccines could be due to a mixture of the following factors: i) the presence of a highly immunosuppressive microenvironment with consequent inability of T cells to overcome environmental immunosuppression in absence of co-stimulatory or anti-inhibitory

stimuli [126]; ii) the scant ability of antitumor T cells to localize at tumor site [127-129]; iii) the rapid senescence of tumor-specific T cells or T-cell repertoire [122,130]; iv) the highly mutable tumor target capable of immune-evasion and antigen loss [122]; and v) the presence of unfavorable genetic condition (host polymorphisms or cancer mutations) [129,131,132] or unfavorable environmental factors (unintact commensale gut microbiota) [118] that can negatively switch the balance between tolerance and acute (therapeutic) inflammation.

Expert Opin. Biol. Ther. (2014) 14(5)

9

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

S. Tomei et al.

Therefore, novel generation of vaccines [133] could be effective when combined with other therapies (e.g., inflammatory cytokines, immune-checkpoint inhibitors, chemotherapy, or radiation therapy, see combination therapy section), with the goal to overcome the barriers listed earlier. Vaccination could be even more successful when used in patients with low tumor burden (i.e., adjuvant setting), or in selected patients particularly sensitive to immune-manipulation (i.e., those bearing specific polymorphisms). Only very few vaccination strategies have shown promising results in Phase II trials and are currently being tested in Phase III randomized trials (in addition to gp100 vaccine plus IL-2 [134], see combination therapy). In a Phase II trial on stage III/IV M1a melanoma, MAGE-A3 vaccine combined with AS15 immunostimulant (a mixture of QS21 saponin, TLR4, and TLR9 agonist) induced an OR in 11% of patients (including 2% of CR) and a median OS of 33 months [135]. The advantage of vaccination was even higher in patients bearing an inflammatory gene-signature centered on IRF-1/IFN-g-related transcripts [117,136]. This adjuvant--vaccine combination was selected for the Phase III trial. Unfortunately, it has been recently announced that the study did not meet its first co-primary end point as it did not significantly extend disease-free survival (DFS) when compared to placebo [137]. The result of the other co-primary end point (DFS in patients bearing the inflammatory gene signature identified in Phase II trial) is expected in 2015. In the OPTIM Phase III trials by Kaufman et al. T-VEC, an oncolytic immunotherapy derived from herpes simplex virus type-1 designed to selectively replicate within tumors and to produce GM-CSF to enhance systemic antitumor immune responses, induced OR in 26% of patients, including 11% of CR. According to the interim analysis, there was also a trend in OS favoring vaccination (HR: 0.79; vaccination vs GM-CSF alone) [138,139]. Allovectin-7 (a plasmid--lipid complex containing the DNA sequences encoding HLA-B7 and b2 microglobulin) in combination with dacarbazine [140], and M-VAX (an autologous hapten-modified vaccine), alone or in combination with IL-2 (NCT00477906), represent the other two vaccination strategies that are currently being evaluated in Phase III trials [141]. Notably, there is an increasing interest for testing nanoparticle as vaccine adjuvant [142]. Functionalizing carbon nanotubes are particularly promising in view of their theranostic (i.e., both diagnostic and therapeutic) potential. In fact, functionalized carbon nanotubes can stimulate monocytes to release pro-inflammatory cytokines and chemokines associated with T helper 1 polarization and immune-mediated tumor rejection [143]. Moreover, because of their high echogenicity these nanotubes could be potentially used as ultrasound contrast agents [144]. An emerging and promising approach to generate tumorspecific T cells is represented by chimeric antigen receptor (CAR) or T-cell receptor (TCR) engineered T cells. Studies in B-cell hematological malignancies have shown that engineered T cells against CD19+ can mediate deep tumor 10

remission [145]. In melanoma, the administration of modified T cells expressing high avidity anti-gp100 or anti-MART-1 TCR had induced a considerable OR rate (19 -- 30%), with few complete remissions (one patient in the MART-1 trial) [146]. However, patients exhibited destruction of melanocytes in skin, eye, and ear, which also express, although at lower level, these antigens. Similarly, administration of engineered T cells against MAGE-A3 is associated with high incidence of neurotoxicity because of cross reactivity with MAGE proteins expressed by brain [146]. Therefore, it is imperative to select antigens that are not expressed by normal cells and to generate T cells that do not cross-react with epitope within self-antigens [146]. The most promising results have been obtained by targeting NY-ESO (OR in 5/10 melanoma patients, including 2 CR) [146]. Immune checkpoint blockade An extremely promising approach used in clinical settings to counteract the immune escape mechanisms mounted by cancer cells is the inhibition of the CTLA-4 (Figure 3) [147]. CTLA-4 is a key player in establishing immune tolerance and one of the main regulators of T-cell-mediated antitumor immune responses. The major function of CTLA-4 is to modulate T cells at the time of their initial response to antigen [148]. In fact, although CTLA-4 is expressed by activated CD8+ effector T cells, its key role relies in the regulation of T CD4+ populations through down modulation of helper T-cell activity and enhancement of regulatory T-cell immunosuppressive function [147,148]. The possible role of CTLA-4 blockage in clinical setting was proposed in 1995 by Allison and Krummel [149,150]. It is now clear that antibodies binding the extracellular domain of CTLA-4 can, therefore, block its inhibitory signals and enhance the immune system to fight against cancer cells. Two anti-CTLA-4 monoclonal antibodies, that is, ipilimumab and tremelimumab, are currently being applied in metastatic melanoma setting. The former has been approved by FDA in March 2011. Ipilimumab is a fully human IgG1 monoclonal antibody. Results of ipilimumab trials have boldly increased the enthusiasm in the field of cancer immunotherapy and deserve detailed description. Two Phase III trials have conclusively demonstrated its efficacy in previously treated (ipilimumab monotherapy, MDX010-020 trial) [17], or naive (ipilimumab+dacrabazine, CA184-024 trial) [22] advanced melanoma patients. In the MDX010-20, ipilimumab showed to increase OS, as compared with gp100 vaccine alone (median OS: 10.1 vs 6.4 months, ipilimumab vs gp100 vaccine, p = 0.003) [17]. The differences in terms of OS increased with the time. The 1- and 2-year OS were 46 and 24% in ipilimumab arm, and 25 and 14% in the vaccine arm, respectively [17]. In the CA184-024 study median OS was 11.1 months in the dacarbazine-ipilimumab arm, and 9.1 months in dacarbazine alone arm [22]. A recent follow-up analysis of this latter trial highlights the progressive benefit in OS associated with 4.3

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

ipilimumab. The 1-, 2-, 3- and 4-year OS were, respectively, 48, 29, 21, and 19% in the ipilimumab-dacrabazine arm and 36, 18, 12 and 10% in the dacarbazine arm [151]. Therefore, ipilimumab almost doubles the 4-year survival rate. Although responses were few in number, they were more likely to occur in patients treated with ipilimumab. As for the MDX010-20the OR rate was 11% (including 1.5% of CR) in the ipilimumab arm, and 1.5% in the gp100 arm. No CRs were observed in the gp100 alone arm [17]. As for CA184-024, the OR rate was 15% (including 1.5% of CR) in the ipilimumabdacarbazine arm and 10% (including 1% of CR) in the dacarbazine alone arm [22]. Even though 15% of OR rate is not particularly high, the responses were more durable in patients treated with ipilimumab (OR duration: 19 months vs 8 months, dacarbazine plus ipilimumab vs dacarbazine) [22]. Similar (although less pronounced) differences were observed in term of PFS in the two studies. The continuous follow-up of patients enrolled in three Phase II trials also confirm the long-term benefit of ipilimumab, with a 5-year OS ranging from 12 to 28% and 38 to 50% for previously treated and naı¨ve patients [152,153]. These results should, however, be taken with caution, being derived from early Phase trials. However, the US ipilimumab expanded access program, which prospectively surveyed the activity and safety of ipilimumab in > 800 (pre-treated) patients, showed a 2- and 3-year OS rate of 22 and 17%, respectively [154]. Remarkably, these results are very close to those obtained in Phase III trials. According to Phase II trials, the combinations of ipilimumab plus fotemustine (NIBIT-M1 trial) [155], and ipilimumab plus temozolomide [156] seem also to be promising. To evaluate the response to treatment, the NIBIT-M1 trial was the first to employ the immune-related criteria, instead than the classic WHO or RECIST criteria for solid tumors. In fact, the onset of clinical response is often delayed, leading to problems in applying classic criteria of response. Response can be achieved after stabilization or even after progression, and CR can occur years after an initial remission. These observations led to define new criteria (immune-related response criteria; irRC) for the definition of response in immunotherapeutic trials [157]. Tremelimumab is a fully human IgG2 monoclonal antibody. The G2 isotype was chosen to diminish complement activation and cytokine storm. A recent Phase III trials failed to show a significant advantage in terms of OS when compared standard chemotherapy (dacarbazine or temozolamide) in treatment naı¨ve advanced melanoma patients (median OS: 12.6 vs 10.7 months, p = 0.13) [158]. Response rate was also similar in the two arms (11 vs 10%, tremelimumab vs chemotherapy, respectively) although duration was significantly longer after tremelimumab (35.8 vs 13.7 months, p = 0.001). However, tremelimumab survival curves are similar to those obtained in the Phase III ipilimumab trials in the same patient setting. The 2- and 3-year OS were 26 and 21% in patients treated with tremelimumab and 23 and 17% in patients treated with chemotherapy. The failure to detect significant differences could be due, at least in part, to the enrichment in patients

more responsive to chemotherapy or to study contaminations. Moreover, although the trial did not allow crossover, a proportion of patients randomized to receive chemotherapy likely received ipilimumab at progression. During trial conduct, in fact, ipilimumab became widely available for patients randomized in the control arm [158]. Dosage and schedules could also have influenced the results. Nevertheless, promising results are obtained in Phase I/II trial, in pretreated melanoma [159,160] and mesothelioma patients [161]. Overall, we believe there is still room for this drug in metastatic melanoma setting. One of the main limitations of the CTLA-4 blockades is that immune response takes time to be effective and about 70% of patients progress within the first 6 months [17,22]. Common immunerelated adverse event include colitis, hepatitis, and endochrinopaties, and can be life threatening in a small but considerable proportion of patients. In view of the toxic profile, another limitation of this approach is that so far there are no tools allowing to define with confidence the patients who will benefit from this approach. Another critical immune checkpoint is represented by PD-1. Four anti-PD1 and 3 anti-PD1 ligands mAbs are currently in clinical development (Figure 3, Table 1). PD-1 is induced once T cells become activated. The major function of PD1 is to repress the activity of effector T cells in peripheral tissues to limit autoimmunity or collateral damages in response to infections [148]. PD1 has two known ligands, PD-L1 (B7-H1) and PDL-2 (B7-DC) [162,163], which are expressed by tumor cells to avoid T-cell mediated lysis. Results of large Phase I/II trials of two anti-PD1 mAbs (nivolumab/BMS-936558 and lambrolizumab/MK-3475) and anti-PD-1 ligand mAbs (BMS-936559 and MPDL3280A) are extremely encouraging. If paradigmatic pattern of response of CTLA-4 (delayed and slow effect on tumor shrinkage and prolonged OS) in a small proportion of patients have forced us to define (or redefine) mechanism of action of immunotherapy, recent results of Phase I/II anti-PD-1 and anti-PD-1 ligand trials have shown once again that pattern of response among different immune manipulations could be extremely different. Surprisingly, anti-PD-1 or anti-PD-1 mAb induce tumor shrinkage in most patients with advanced melanoma, lung, and kidney cancers [164-166]. OR rate in melanoma cohorts were 31, 38, 17, and 29% for nivolumab [165,167], lambrolizumab [166], BMS-936559 [164], and MPDL3280A [168], respectively. Tumor shrinkage was already evident at the time of the first assessment (at 6 or 12 weeks), and was extremely durable (1 year or beyond in most cases). Importantly, toxicity (most immunomediated) was much lower as compared with that observed in ipilimumab trials. Although the magnitude of the clinical benefit needs to be assessed through (undergoing) randomized trials and longer follow-up, the pattern of response and the (favorable) safety profile of anti-PD1/PD-1 ligands appear to be quite unique. Importantly, PD-1/PD-1 blockage was effective in patients already previously treated with ipilimumab, therefore,

Expert Opin. Biol. Ther. (2014) 14(5)

11

S. Tomei et al.

Macrophage

Clinical development status T cell

Dendritic cell MHC-II

BMS-986016 LAG-3 IMP-321 Ipilimumab CTLA-4 Tremelimumab

CD80/CD86

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

R R

Phase III trials

CD40L

Phase II trials R

CD137 Urelumab

CD137L

R

R

FDA approved

R CP-870,893 Dacetuzumab CD40 Chi Lob 7/4 Lucatumumab

GITRL

GITR

B7-H3

?

BMS-936559 AMP-224 R MEDI4736

R

R R

OX40 Anti-OX40

OX40L

R MGA271

R

TRX518

Nivolumab PD-1 Lambrolizumab MPDL3280A CT-011

PD-L1/PD-L2

R

R R

R

Phase I trials Recruiting trials

Stimulatory signal Inhibitory signal

R R R

Tumor

Figure 3. Immune-checkpoints, co-stimulatory signaling, and clinical development of immune-targeted therapy. Stimulatory signaling is in green, inhibitory signaling is in red. The clinical development status is represented by colored rectangles and refers to trials that have recruited (or are currently recruiting) patients with advanced or metastatic melanoma. The symbol ‘R’ highlights trials currently recruiting melanoma patients. Name of the drug is next to the corresponding targeted molecule (either receptor or ligand). Status of clinical trials is update as of January 2014 (source: clinicaltrials.gov; more details in Table 1). Represented expression of ligands and receptors should not be considered exhaustive: for example, PD1L-PD2L are also expressed by antigen presenting cells as macrophages and dendritic cells (not shown in the figure); MHC-II is also expressed by dendritic cells and can be expressed by melanoma cells as well. CTLA-4: Cytotoxic T-lymphocyte antigen 4.

confirming that CTLA-4 and PD-1 immune-suppressive pathways are not redundant. Besides the co-inhibitory interactions described earlier, costimulatory signaling between antigen presenting cells (APC) and T cells could also be targeted to enhance antitumor immunity. A schematic representation of the clinical development of immune-checkpoint inhibitors and co-stimulatory molecules in melanoma is provided in Figure 3. The status of the corresponding clinical trials (updated as of January 2014) is provided in Table 1. Agonist antibodies aimed at enhancing T and NK functions (e.g., CD40, CD137 (4-1BB), anti-CD134 (OX40), and glucocorticoid-induced TNF receptor (GITR)) are being evaluated in early phase trials [169]. Perhaps, the most clinically relevant examples in this regard come from the 12

interactions between OX40 and OX40L and between CD40 and CD40L. OX40 (also known as CD134) is a TNF-receptor family member, which is transiently expressed by T cells upon the binding to the TCR. The engagement of OX40 promotes a vast spectrum of T-cell responses, including increased proliferation, cytokine production, and increased differentiation and survival. The interaction between OX40 and its ligand, OX40L, occurs in 48 h after antigen recognition by T cells [170]. Targeting of OX40/OX40L has been proposed as a relevant approach for autoimmunity and cancer [171]. Preclinical studies have shown that ligation of OX40 via agonist antiOX40 mAB or OX40L-Ig fusion proteins can induce robust T-cell-mediated antitumor immunity [172,173]. In a recent Phase I trial [174], Curti and colleagues showed that the

Expert Opin. Biol. Ther. (2014) 14(5)

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

Table 1. Agents targeting immune-checkpoints and co-stimulatory signaling*. Agent

Clinical trials

BMS-986016 (anti-LAG3)

Phase I trial in solid tumors as monotherapy and in combination with nivolumab recruiting (NCT01968109) Phase I/II trial in combination with vaccine in melanoma recruiting (NCT01308294) FDA approved for melanoma; two positive Phase III trials (published) in previously treated melanoma patients as monotherapy versus combination with gp100 vaccine versus gp100 vaccine (NCT00094653) and in naı¨ve patients in combination with dacarbazine versus dacarbazine completed (NCT00324155); Phase III trials in melanoma as monotherapy versus combination with nivolumab versus nivolumab (NCT01844505) or versus lambrolizumab (NCT01866319) recruiting; Phase III trials in small-cell lung cancer (NCT0145076) and non-small-cell lung cancer (NCT01285609) in combination with chemotherapy versus chemotherapy alone recruiting; enrollment completed in a Phase III trial in prostate cancer versus placebo (NCT01057810); > 60 Phase I/II trials evaluating combinations with targeted therapy, radiotherapy, or various immunotherapies in melanoma and in other cancers are currently recruiting Negative Phase III trial in melanoma versus chemotherapy published (NCT00257205); Phase I trial in melanoma in combination with the anti-CD40 mAb CP-870,893 (NCT01103635) recruiting; Phase II trial in mesothelioma recruiting (NCT01843374); Phase I trial in non-small-cell lung cancer in combination with gefitinib (NCT02040064) or the anti-PD-L1 mAb MEDI4736 (NCT02000947) and in solid tumors in combination with MEDI4736 (NCT01975831) recruiting; Phase I trial in combination with chemoembolization or ablation in liver cancer recruiting (NCT01853618) Encouraging results from a Phase I trial in multiple cancers recently published (NCT01644968); Phase II trial in melanoma as monotherapy (NCT01416844) withdrawn prior to enrollment; Phase I/II in combination with ipilimumab (NCT01689870) recently suspended; Phase I/II trials in breast cancer patients with lung metastases in combination with radiotherapy (NCT01862900) and in prostate cancer in combination with radiotherapy and cyclophosphamide recruiting (NCT01303705) Phase I trial in melanoma in combination with tremelimumab, recruiting (NCT01103635); Phase I trial in melanoma in combination with vaccination ongoing but not recruiting (NCT01008527); Phase I trial in pancreatic cancer in combination with chemotherapy recruiting (NCT01456585); two Phase I trials in solid tumors (NCT00607048) and in pancreatic cancer (NCT00711191) in combination with chemotherapy completed and listed as ‘having results’ Phase I/II trial completed in leukemia patients (NCT00283101); 6 Phase I and 2 Phase II trials completed in multiple myeloma and non-Hodgkin’s lymphoma A Phase I trial in multiple cancers ongoing but not recruiting (NCT01561911) Phase I trial for follicular lymphoma patients in combination with bendamustine completed (NCT01275209) Phase II trial in melanoma completed (NCT00612664), Phase I trial in melanoma in combination with ipilimumab withdrawn prior to enrollment (NCT00803374); Phase I trials in non-Hodgkin’s lymphoma recruiting (NCT01471210; NCT01775631); Phase I trials in advanced tumors (NCT00351325, NCT00309023) and non-small-cell lung cancer (NCT00461110) terminated Phase I trial in melanoma and other solid tumors recruiting (NCT01239134) Phase I trials in melanoma (NCT01918930) and in melanoma and other solid tumors (NCT01391143) recruiting Positive results of a Phase I trials in patients with melanoma and other solid tumors (NCT00730639) or in melanoma in combination with ipilimumab (NCT01024231) published; Phase I trials in melanoma in combination with vaccination recruiting (NCT01176474, NCT01176461); Phase III trial in melanoma as monotherapy versus dacarbazine (NCT01721772) or versus combination with ipilimumab versus ipilimumab (NCT01844505) recruiting; Phase II trials in melanoma as monotherapy versus combination with ipilimumab versus ipilimumab (NCT01927419) or given sequentially with ipilimumab recruiting (NCT01783938); Phase I trial in solid tumors in combination with anti-KIR (NCT01714739), IL-21 (NCT01629758), and the anti-LAG1 mAb BMS-986016 (NCT01968109) recruiting; Phase I trials assessing combination with ipilimumab in melanoma (NCT01024231) and other combinations in other solid or liquid tumors recruiting

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

IMP321 (anti-LAG3) IPILIMUMAB (anti-CTLA-4)

TREMELIMUMAB (anti-CTLA-4)

Anti-OX40 (OX40 agonist)

CP-870,893 (CD40 agonist)

Dacetuzumab (SGN-40 or huS2C6; anti-CD-40) Chi Lob 7/4 (anti-CD40) Lucatumumab (HCD122;anti-CD40) Urelumab (BMS-663513; anti-CD137)

TRX518 (anti-GITR) MGA271 (anti-BT-H3) Nivolumab (BMS-936558/MDX-1106; Anti-PD1)

*Updated as of January 2014 (source: clinicaltrial.gov). CTLA-4: Cytotoxic T-lymphocyte antigen 4; LAG3: Lymphocyte activation genes 3; PD-L1: Programmed death ligand 1.

Expert Opin. Biol. Ther. (2014) 14(5)

13

S. Tomei et al.

Table 1. Agents targeting immune-checkpoints and co-stimulatory signaling* (continued). Agent

Clinical trials

Lambrolizumab (MK-3475 anti-PD1)

Phase III trial in melanoma as monotherapy versus ipilimumab (NCT01866319) recruiting; Phase II trial in melanoma ongoing but not recruiting (NCT01704287); Phase I trials in melanoma, solid tumors or non-small-cell lung cancer recruiting (NCT01295827, NCT01848834, NCT01840579); Phase II/III trials in non-small-cell lung cancer versus docetaxel recruiting (NCT01905657); Phase II trial in tumor with microsatellite instability recruiting (NCT01876511); positive results of the melanoma cohort of the NCT01295827 study recently published Phase I trial in melanoma or hematologic malignancies (NCT01375842), melanoma (combination with vemurafenib, NCT01656642), or in solid tumors in combination with bevacizumab or chemotherapy (NCT01633970) recruiting; several Phase I-II trials and one Phase III trial in lung cancer recruiting (NCT02008227) Phase II trial in melanoma completed (NCT01435369); Phase II trials in prostate, renal, pancreatic cancer and hematological malignancies in combination with chemotherapy or vaccine recruiting; other Phase I and Phase II trials in gastrointestinal tumors and hematological malignancies recently concluded Phase I trials in melanoma (NCT01455103) and hematological malignancies (NCT01452334) withdrawn before enrollment; a Phase I trial in solid cancer is ongoing but not recruiting (NCT00729664) Phase I trial in solid cancers ongoing but not recruiting (NCT01352884) Phase I/II trial in melanoma in combination with dabrafenib or dabrafenib and trametinib recruiting (NCT02027961); Phase I trial in japanese patients with solid tumors recruiting (NCT01938612); Phase I trial in solid tumors recruiting (NCT01693562); Phase I trial in combination with tremelimumab in solid tumors (NCT01975831) or in non-small-lung cancer recruiting (NCT02000947)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

MPDL3280A (anti-PD1)

CT-011 (anti-PD1)

BMS-936559 (MDX-1105; anti-PD-L1)

AMP-224 (anti-PD-L2) MEDI4736 (anti-PD-L1)

*Updated as of January 2014 (source: clinicaltrial.gov). CTLA-4: Cytotoxic T-lymphocyte antigen 4; LAG3: Lymphocyte activation genes 3; PD-L1: Programmed death ligand 1.

administration of an anti-OX40 mouse mAB in patients with advanced cancer had an acceptable toxicity profile and was able to induce regression of at least one metastatic lesion in 12 out of 30 patients. The therapy also increased CD8 T-cell IFN-g production in two of three patients with melanoma. Moreover, in another recent study, Redmond and colleagues [175] demonstrated that the targeting of OX40 in combination with IL2 therapy increased tumor regression in preclinical tumor model and restored the function of anergic tumor-reactive CD8 T cells in mice, leading to enhanced survival. CD40 is a cell-surface molecule, member of the TNFreceptor family, expressed by APC, such as dendritic cells, B cells, and monocytes, but also by many nonimmune cells and a wide range of tumors [169]. The natural ligand of CD40 is CD145, which is expressed on T cells. It has been shown that in tumor-bearing hosts, CD40 agonist trigger effective immune response against tumor-associated antigens [176-178]. Four CD40 agonist mABs have been investigated in clinical trials so far, namely: CP-870,893, dacetuzumab, Chi Lob 7/4, and lucatumumab [169,179]. However, these treatments showed wide heterogeneity ranging from strong agonism (CP-870,893) to antagonism (lucatumumab), suggesting that their mechanism of action might be different. A study employing CP-870,893 as CD40 agonist demonstrated that a single intravenous infusion of CP-870,893 was well tolerated and an objective PR was observed in four patients with metastatic melanoma (14% 14

of all patients and 27% of melanoma patients) [180]. Additional clinical trials employing CD40 agonists are ongoing and results are strongly awaited. Blockage of other inhibitory pathways as the immunosuppressive enzyme indoleamine 2,3-dioxygenas (IDO) seems promising in preclinical study [181], and clinical studies are ongoing (NCT01961115; NCT00739609). Drugs blocking other immune-checkpoints such as T-cell membrane protein 3 (TIM3; inhibitor receptor) and B7-H4 (inhibitory ligand), TGF-b, and KIR inhibitory receptors are under development [148,169]. Notably, trials evaluating blockage of the inhibitory receptor lymphocyte activation genes 3 (LAG3), the inhibitory ligand B7-H3 are ongoing [148,182,183] . Finally, the recent discovery that the manipulation of BACH2 gene can shift the balance between effector and regulatory T-cell response also represents a novel starting point for the development of next-generation immunotherapeutic approaches [184]. Adoptive cell therapy Adoptive cell therapy (ACT) transfer is a form of passive immunotherapy, which consists in the ex vivo expansion of autologous antitumor reactive lymphocytes and their reinfusion into lymphodepleted patients, accompanied by the exogenous administration of IL-2 to further boost their antitumor activity. Lymphodepletion, in fact, seems to favor the antitumor activity of ACT by suppressing regulatory T cells and limiting host tumor immunosuppressive factors. ACT was developed by the group of Steven Rosenberg at the National 4.4

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

Cancer Institute and has shown encouraging results in Phase II clinical trials. In a recent analysis evaluating 93 heavily pretreated metastatic melanoma patients enrolled in three Phase II trials, ACT was able to induce OR in 56% of patients. Complete remission was obtained in 20 patients (21.5%), with 19 patients potentially cured as they had ongoing regression beyond 3 years [185]. A similar OR rate (48%, including 6.5% of CR), measured using immune-related response criteria (irRC), had been obtained by Radvanyi et al. in a Phase II trial enrolling 31 patients [186]. Beyond the toxicity related to HD-IL-2 administration, the main limitation associated to ACT is that TIL expansion requires highly specialized personnel. Using the traditional approach, TILs are generally successfully grown from only 50 to 60% of the patients and require a long period of time (about 5 weeks). Simplified protocols using short-term cultures derive TILs from up to 90% of patients in a shorter period of time (10 -- 18 days) [187]. Rosenberg’s group, however, recently reported a global OR of 28% (CR rate 7%), in randomized Phase II trials comparing two different TIL protocols (young TILs vs CD8 enriched TILs) [188]. By using young TILs Besser et al., in a Phase II trial enrolling 70 patients, reported an encouraging OR of 40% in the evaluated population, including 5 CR [187]. In all the ACT trials responses were durable and patients in CR very rarely relapsed. The results of these Phase II trials need to be taken with caution, and the true response rate needs to be evaluated in randomized Phase III trials. Nevertheless, ACT seems to represent a potential curative therapy for patients with metastatic melanoma [189]. Randomized multicenter trials aimed at evaluating the true response rate and OS advantage of ACT represent the reasonable next step. Despite these encouraging results, the dynamic of TIL migration at tumor site is still not completely clear. Studies where TILs were labeled with Indium 111 showed that TILs migration in the tumor site does not follow a linear kinetic. After re-infusion, which is followed by IL-2 administration, TIL localize in lung, spleen, and liver but not at tumor sites [190,191]. The migration of TILs at tumor site is detectable only after 24 to 48 h. It is possible that this early compartmentalization is mediated by the release (following HD-IL-2 administration) of specific chemokines (i.e., CXCR3 and CCR5 ligands) from resident immune cells and stromal cell in peripheral oragans (e.g., spleen, lung, and liver) [129,192]. Intriguingly, by analyzing a large cohort of patients treated with adoptive therapy and HDIL-2, we recently showed that the downregulation of CXCR3 and CCR5 receptors in TILs correlates with the frequency and the degree of response [129]. This receptor down-modulation is due to the transcript downregulation and/or to the presence of common polymorphism (CCR5 D32, which codify for a nonfunctioning receptor) [129]. It is tempting to hypothesize that a reduced, but not absent, expression of these receptors might prevent the earlier sequestration of TIL by extra-tumoral sites and paradoxically allow their subsequent localization to the tumor

when the cytokine storm has subsided and the tumor represents the only tissue maintaining chemokine secretion [129]. 5.

Combination therapy

Combination therapy is intended as the use of multiple therapies to treat a single disease with the aim to overcome the hurdles of mono-therapeutic strategies [193]. Combining chemotherapy agents represents a pillar of the medical treatment of virtually all the major liquid and solid malignancies, with exception of melanoma and renal cell carcinoma. In these two tumors combination of cytotoxic agents failed to show increase efficacy as compared with monotherapy. In melanoma, > 40 randomized trials have evaluated combination of chemotherapy +/- cytokines (IFN-a and/or IL-2) [21]. It is now clear that although polychemotherapy +/IFN-a and/or IL-2 in general enhance the OR rate, they do not increase the OS [21]. The failure of polychemotherapy is in general attributed to the intrinsic chemo-resistance of melanoma. As for first generation immunochemotherapy, it should be noted that, because of safety issues, IFN-a and or IL-2 were administered at low dose, while their maximal antitumor effect is in general achieved using high dosages. Nevertheless, prospective trials have shown that combination of HD-IL-2 and IFN-a does not increase OS, as compared with HD-IL-2 alone [194,195]. This depressing scenario dramatically changed after advent of targeted therapy immune checkpoint inhibitors. Although the mono-targeted therapy and immunotherapy strategies have revolutionized the clinical management of melanoma patients, it is becoming clear that combination strategies are most likely to result in even more significant advances. Building upon the early success of mono-therapy agents in melanoma, trials involving new combination therapy are underway. It has been thought that combining vaccines with anti-CTLA-4 could produce a synergic effect. When ipilimumab was administered together with peptide vaccination (gp100), the combined treatment was more effective (both in term of PFS and OS) than gp100 alone [17]. However, there were no significant differences in both PFS and OS between ipilimumab alone and ipilimumab combined with vaccine [30]. Conversely, gp100 vaccine and IL-2 can act in a synergistic way. In a Phase III randomized trials in metastatic melanoma patients, Schwartzentruber et al. showed that the addition of the gp100 vaccine to HDIL-2 can significantly increase the number of responses (CR: 7 vs 1%, and OR: 16 vs 6%, IL-2 + gp100 vs IL-2, respectively, p = 0.03) and lengthen PFS survival (median PFS 2.2 vs 1.6 months, p = 0.008), with similar trend in OS [134]. Two recent studies showed that efficacy of CTLA-4 can be enhanced by the concomitant administration of immune-stimulant. In a recent randomized Phase II trial Hodi et al. showed that the addition of GM-CSF to ipilimumab increase OS (1 year OS: 68 vs 51%, GM-CSF plus ipilimumab vs ipilimumab alone, p = 0.03) and was also

Expert Opin. Biol. Ther. (2014) 14(5)

15

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

S. Tomei et al.

associate with less toxicity [196]. Remarkably, long-term follow-up Phase I/II trials in metastatic melanoma patients have suggested that the addition of IL-2 to ipilimumab could dramatically improve the rate of durable CR (CR rate: 17 vs 7%, ipilimumab + IL-2 vs ipilimumab +/- gp 100 vaccine) and extend five overall survival up to 25% [197]. Overall, these apparently clashing results underline once again the need to understand the mechanisms of action of simultaneous administration by designing clinical or translation trial aimed at evaluating the effect of immunotherapy in vivo in humans [198]. However, in view of the huge number of potential combinations, novel generations of clinical trials using factorial design are needed [199] to select the more promising combinations. Another possible combination approach is the use of targeted therapy together with immunotherapy [200]. The rationale supporting this type of combination approach relies on the possibility to achieve a deeper and long lasting antitumor effect (Figure 2C), as pointed out by Ribas et al. [201]. BRAF inhibitors, for example, induce almost immediate tumor shrinkage in most patients (median time to response 1.5 months). Responses are, however, transient, and the effect of long-term survival is predicted to be very limited (Figure 2A). Conversely, CTLA-4 blockades induce less number of clinical responses (usually delayed), but they are much more durable (Figure 2B). A number of observations support the target-immunotherapy combination. Several cytotoxic agents have been shown to induce antitumor immune responses by promoting cell death with consequent release of tumor-associated antigens [202]. In melanoma, this could be achieved through targeted therapy [203], rather than with conventional chemotherapy. Moreover, constitutive activation of oncogenic pathways, as the BRAFMAPK pathway, can induce several immunosuppressive mechanisms [204,205], including promotion of stromal cell-mediated immunosuppression via induction of IL-1 [7,206]. In addition, BRAF inhibition can enhance melanoma antigens expression and create a more favorable immune environment, facilitating the effectiveness of immunotherapy [89,207-209]. A study evaluating combination of ipilimumab and vemurafenib was object of a Phase I study by Ribas et al. [210]. Although this combination is supported by a strong rationale, investigators noticed an unacceptable hepatotoxicity, which prompted them to close prematurely the study. This example reinforces the need to carefully evaluate the safety of combination approach in appropriate early trials [210]. Very recently, two additional prospective studies in metastatic melanoma have highlighted the potentiality of combining highly effective agents. In a first Phase I/II randomized trial, combination of BRAF inhibitor dabrafenib and trametinib resulted superior to dabrafenib alone [23]. The two drugs combined at full monotherapy dose resulted in 76% of OR (including 9% of CR), while 54% of patients achieved OR in the dabrafenib group (including 4% of CR; p = 0.03). Progression-free survival in the combination was 9.4 months, 16

as compared with 5.8 months in the monotherapy group (p < 0.001). Responses were also more durable with the combination (10.2 vs 5.6). Importantly, > 40% of patients were alive and progression-free at 1 year in the combination arm, as compared with 10% in the monotherapy arm. Combination was also feasible in term of safety [211]. This landmark study demonstrated that the inhibition of the same pathways through highly effective targeted therapy agents (vertical inhibition) could dramatically impact on the course of this aggressive disease, overcoming (at least in part) mechanisms of resistance occurring during monotherapy. Several trials also employing therapies targeting different pathways are underway; examples include vemurafenib (BRAF inhibitor) plus everolimus or temsirolmus (mTOR inhibitors, identifier NCT01596140), MEK162 plus AMG479 (insulin-like growth factor1 receptor inhibitor, identifier NCT015 62899), vemurafenib plus BKM120 (a PI3K inhibitor, identifier NCT01512251). In a second Phase I trial, combination of PD-1 and CTLA-4 blockers (nivolumab and ipilimumab) resulted, at the maximum tolerated dose, in 53% of OR rate, with tumor regression of 80% or more in every patient who had response [212]. The profound effect of concurrent combination was already evident at the first imaging assessment (3 months after treatment initiation). Responses were extremely durable and ongoing in 90% patients who had a response (duration ranging from 6 to 72 weeks at the time of data analysis). This pattern of response (rapid, deep, and durable) appeared to be distinct of that observed in previous immune-checkpoint blockade trials. Importantly, toxicity was qualitatively similar to that of monotherapy [212].

6.

Expert opinion

Although major advances have been made for the treatment of metastatic melanoma, with combination therapies paving the road toward clinical success, much effort needs to be spent in identifying patients who are more likely to respond to a given treatment. It is becoming now clear that patients who are most likely to reject tumors following immunotherapy are the ones who display a constitutive activation of the IFN-g/signal transducers and activators of transcription (STAT)-1/IRF-1 axis (Th1 phenotype) [81,117,121] with IRF-1 playing a central role in this regard [1,117,121,192,213]. Gene signatures predictive of response to immunotherapy (i.e., IL-2-based therapy, vaccines) invariably included genes belonging to the following functional modules: 1) IFN-g/T helper 1 module (STAT-1/IRF-1/IFN-gstimulated genes pathway); 2) The specific cytotoxic recruitment module (CXCR3/ CXCR3 ligand and CCR5/CCR5 ligand pathway); 3) The immuno-effector function module (Granzyme/ Perforin/Granulisin/TIA-1 pathway).

Expert Opin. Biol. Ther. (2014) 14(5)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

This phenotype is also characterized by the counteractivation of suppressive mechanisms. Therefore, it is not surprising that high expression of IDO and PD-1 ligands, which are both IFN-g inducible genes, have been found to be associated with responsiveness to ipilimumab [214] and anti-PD1 treatment [165]. The determinants of this responsive immune phenotype are object of investigations. Coordinate enhancement of these modules following treatment is associated with responsiveness to IL-2 [103], ipilimumab [215], and other immunotherapies, including vaccines [216] and Toll-like receptor agonists [217]. These observations suggest that, although distinct immunotherapies clearly act in very different ways, their final therapeutic relies on the activation of similar and redundant downstream mechanisms, as recently reviewed elsewhere [121]. It is currently becoming accepted that immune-mediated tumor rejection is mediated by both the genetic background of the host and the genetic make-up of individual cancer cells carrying their own specific mutations [119,120,218]. Yet, the relative weight played by each of those factors is not completely understood as well as their contribution to mediate responsiveness to a given cancer therapy. The relevance of the genetic background of the host in mediating tumor rejection has been recently witnessed by two studies from our group. In the first one, we hypothesized that polymorphisms of IRF5 gene, known to be implicated in autoimmune diseases and in particular to systemic lupus erythematosus, might play a role in the immune-mediated rejection of the tumor. Concordant to our hypothesis, we were able to show that the IRF5 genetic variants protecting against the development of autoimmunity were highly predictive of nonresponsiveness to adoptive therapy in melanoma [131]. These results support the link between autoimmunity and immune responsiveness. In the second study, we assessed the prognostic relevance of genetic variations of CTLA-4 and HLA polymorphisms in melanoma patients who received adjuvant IFN-a [132]. Multivariate analysis revealed that a five-marker genotyping signature based on HLA-B38, HLA-C15, HLA-C3 DRB1*15, and CTLA-4 CT60* polymorphisms, was predictive of OS in 284 melanoma patients. The findings of these two studies strengthen the relevance of inherited genetic variations in mediating tumor rejection induced by immunotherapy regimens.

Aside from inherited genetic variations, somatic mutations acquired during tumor development are likely to influence tumor rejection. The most evident proof that the genetic background of individual cancer cells plays a role in mediating cancer rejection comes from the observation of mixed responses, a rare phenomenon where multiple metastases from the same patient respond differently to a given therapy. By comparing mixed metastases from two melanoma patients who received immunotherapy, we were able to show that metastases who responded to the treatment were the ones who had upregulated genes associated with antigen presentation and IFN-mediated rejection [216]. These findings demonstrate that tumors sharing the same host’s genetic background can behave differently depending on the differential expression of genes involved in triggering an IFN-mediated immune signaling. It should be also noted that cancer cells share the same genetic background of the host, thus, tumor behavior is concurrently influenced by inherited variations other than somatic mutations. Environmental factors also play a role in this regard, and evidence accumulated in the recent years show that it is a combination of all the three categories that affect tumor rejection rather than each single one alone [218]. Understanding the mechanisms underlying tumor rejection is essential to implement personalized therapies. We believe that this goal is reachable only by understanding the complex interplay between the genetic background of the host, the genetic make-up of individual cancer cells, and environmental factors. The availability of highthroughput technologies allowed the investigators to start the race toward cancer-personalized medicine. We believe that, in the future, the integration of the ‘omic’ research approaches will allow a more potent understanding of the algorithm governing tumor rejection and responsiveness to a given therapy and lead to improved clinical management of melanoma patients.

Declaration of interest The authors have no competing interests to declare and have received no funding in preparation of the manuscript.

Expert Opin. Biol. Ther. (2014) 14(5)

17

S. Tomei et al.

Bibliography

metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012;380:358-65

Papers of special note have been highlighted as either of interest (!) or of considerable interest (!!) to readers. 1.

Spivey TL, De Giorgi V, Zhao Y, et al. The stable traits of melanoma genetics: an alternate approach to target discovery. BMC Genomics 2012;13:156

2.

Ibrahim N, Haluska FG. Molecular pathogenesis of cutaneous melanocytic neoplasms. Annu Rev Pathol 2009;4:551-79

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

18

Palmieri G, Capone M, Ascierto ML, et al. Main roads to melanoma. J Transl Med 2009;7:86 Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364:2507-16 Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949-54 Colombino M, Lissia A, Capone M, et al. Heterogeneous distribution of BRAF/NRAS mutations among Italian patients with advanced melanoma. J Transl Med 2013;11:202 Tomei S, Civini S, Bedognetti D, et al. The immune-related role of BRAF mutation in melanoma [abstract p788]. J Immuno Therapy of Cancer 2012;23:9 Pollock PM, Harper UL, Hansen KS, et al. High frequency of BRAF mutations in nevi. Nat Genet 2003;33:19-20

13.

14.

Ascierto PA, Kirkwood JM, Grob JJ, et al. The role of BRAF V600 mutation in melanoma. J Transl Med 2012;10:85

15.

Clinical Trials. Avaialable from http:// clinicaltrial.gov/)

16.

Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med 2004;351:998-1012

17.

Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23 First Phase III study showing an advantage of overall survival in metastatic melanoma using immunotherapy.

..

18.

19.

Eigentler TK, Meier F, Garbe C. Protein kinase inhibitors in melanoma. Expert Opin Pharmacother 2013;14(16):2195-201 Benjamin RS. Chemotherapy of malignant melanoma. World J Surg 1979;3:321-8

25.

Smalley KS. A pivotal role for ERK in the oncogenic behaviour of malignant melanoma? Int J Cancer 2003;104:527-32

26.

Colombino M, Capone M, Lissia A, et al. BRAF/NRAS mutation frequencies among primary tumors and metastases in patients with melanoma. J Clin Oncol 2012;30:2522-9

27.

Nikolaou VA, Stratigos AJ, Flaherty KT, Tsao H. Melanoma: new insights and new therapies. J Invest Dermatol 2012;132:854-63

28.

Yoshida H, Kunisada T, Grimm T, et al. Review: melanocyte migration and survival controlled by SCF/c-kit expression. J Investig Dermatol Symp Proc 2001;6:1-5

29.

Hochhaus A. Imatinib mesylate (Glivec, Gleevec) in the treatment of chronic myelogenous leukemia (CML) and gastrointestinal stromal tumors (GIST). Ann Hematol 2004;83(Suppl 1):S65-6

30.

Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res 2008;14:6821-8

31.

Carvajal RD, Antonescu CR, Wolchok JD, et al. KIT as a therapeutic target in metastatic melanoma. JAMA 2011;305:2327-34 First study showing that imatinib mesylate is active in a subset of metastatic melanoma patients carrying c-KIT mutations and amplification.

20.

Luce JK. Chemotherapy of malignant melanoma. Cancer 1972;30:1604-15

21.

Garbe C, Eigentler TK, Keilholz U, et al. Systematic review of medical treatment in melanoma: current status and future prospects. Oncologist 2011;16:5-24

32.

Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517-26

Natali PG, Nicotra MR, Winkler AB, et al. Progression of human cutaneous melanoma is associated with loss of expression of c-kit proto-oncogene receptor. Int J Cancer 1992;52:197-201

33.

Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med 2012;367:107-14 First Phase III study showing an advantage of overall survival in metastatic melanoma using non-BRAf target therapy MEK inhibitor.

Shen SS, Zhang PS, Eton O, Prieto VG. Analysis of protein tyrosine kinase expression in melanocytic lesions by tissue array. J Cutan Pathol 2003;30:539-47

34.

Henary H, Hong DS, Falchook GS, et al. Melanoma patients in a phase I clinic: molecular aberrations, targeted

Tomei S, Mazzanti C, Marchetti I, et al. c-KIT receptor expression is strictly associated with the biological behaviour of thyroid nodules. J Transl Med 2012;10:7

35.

Guo T, Agaram NP, Wong GC, et al. Sorafenib inhibits the imatinib-resistant KITT670I gatekeeper mutation in

Kumar R, Angelini S, Snellman E, Hemminki K. BRAF mutations are common somatic events in melanocytic nevi. J Invest Dermatol 2004;122:342-8 Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med 2012;366:707-14

22.

Chapman PB, Hauschild A, Robert C, et al. Updated overall survival (OS) results for BRIM-3, a phase III randomized, open-label, multicenter trial comparing BRAF inhibitor vemurafenib (vem) with dacarbazine (DTIC) in previously untreated patients with BRAFV600E-mutated melanoma. J Clin Oncol 2012:30(Suppl): abstract 8502

23.

Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated

Ascierto PA, Minor D, Ribas A, et al. Phase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanoma. J Clin Oncol 2013;31:3205-11

therapy and outcomes. Ann Oncol 2013;24(8):2158-65

..

24.

Expert Opin. Biol. Ther. (2014) 14(5)

.

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

gastrointestinal stromal tumor. Clin Cancer Res 2007;13:4874-81 36.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

37.

38.

39.

40.

41.

42.

43.

..

Prenen H, Cools J, Mentens N, et al. Efficacy of the kinase inhibitor SU11248 against gastrointestinal stromal tumor mutants refractory to imatinib mesylate. Clin Cancer Res 2006;12:2622-7 Schittenhelm MM, Shiraga S, Schroeder A, et al. Dasatinib (BMS-354825), a dual SRC/ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies. Cancer Res 2006;66:473-81 Ugurel S, Hildenbrand R, Zimpfer A, et al. Lack of clinical efficacy of imatinib in metastatic melanoma. Br J Cancer 2005;92:1398-405 Kim KB, Eton O, Davis DW, et al. Phase II trial of imatinib mesylate in patients with metastatic melanoma. Br J Cancer 2008;99:734-40 Wyman K, Atkins MB, Prieto V, et al. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy. Cancer 2006;106:2005-11 Guo J, Si L, Kong Y, et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol 2011;29:2904-9 Flaherty KT, Hodi FS, Fisher DE. From genes to drugs: targeted strategies for melanoma. Nat Rev Cancer 2012;12:349-61 Kaufman HL, Kirkwood JM, Hodi FS, et al. The Society for Immunotherapy of Cancer consensus statement on tumour immunotherapy for the treatment of cutaneous melanoma. Nat Rev Clin Oncol 2013;10:588-98 This manuscript provides the first guidelines on the use of immunotherapy in melanoma patients.

44.

Antonescu CR. The GIST paradigm: lessons for other kinase-driven cancers. J Pathol 2011;223:251-61

45.

Schlegel J, Sambade MJ, Sather S, et al. MERTK receptor tyrosine kinase is a therapeutic target in melanoma. J Clin Invest 2013;123:2257-67

46.

Albino AP, Le Strange R, Oliff AI, et al. Transforming ras genes from human melanoma: a manifestation of tumour heterogeneity? Nature 1984;308:69-72

47.

Milagre C, Dhomen N, Geyer FC, et al. A mouse model of melanoma driven by oncogenic KRAS. Cancer Res 2010;70:5549-57

48.

Tomei S, Adams S, Uccellini L, et al. Association between HRAS rs12628 and rs112587690 polymorphisms with the risk of melanoma in the North American population. Med Oncol 2012;29:3456-61

49.

COSMIC (Catalogue Of Somatic Mutations In Cancer. Available from: http://cancer.sanger.ac.uk/cancergenome/ projects/cosmic

50.

Posch C, Ortiz-Urda S. NRAS mutant melanoma--undrugable? Oncotarget 2013;4:494-5

51.

Niessner H, Beck D, Sinnberg T, et al. The farnesyl transferase inhibitor lonafarnib inhibits mTOR signaling and enforces sorafenib-induced apoptosis in melanoma cells. J Invest Dermatol 2011;131:468-79

52.

53.

Gajewski TF, Salama AK, Niedzwiecki D, et al. Phase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma (CALGB 500104). J Transl Med 2012;10:246 Emery CM, Vijayendran KG, Zipser MC, et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci USA 2009;106:20411-16

MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol 2013;31:482-9 58.

Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a nonrandomised, open-label phase 2 study. Lancet Oncol 2013;14:249-56

59.

Lebbe C, Lesimple T, Raymond E, et al. Pimasertib (MSC1936369B/AS703026), a selective oral MEK1/2 inhibitor, shows clinical activity in cutaneous and uveal metastatic melanoma in the phase I program [abstract C07]. EADO 2012 Annual Meeting; 2012

60.

Basile KJ, Abel EV, Dadpey N, et al. In vivo MAPK reporting reveals the heterogeneity in tumoral selection of resistance to RAF inhibitors. Cancer Res 2013;73(23):7101-10

61.

Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance to B-RAF (V600E) inhibition by RTK or N-RAS upregulation. Nature 2010;468:973-7

62.

Morris EJ, Jha S, Restaino CR, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov 2013;3:742-50

63.

Wangari-Talbot J, Chen S. Genetics of melanoma. Front Genet 2012;3:330

64.

Madhunapantula SV, Robertson GP. Therapeutic Implications of Targeting AKT Signaling in Melanoma. Enzyme Res 2011;2011:327923

54.

Salama AK, Kim KB. MEK Inhibition in the treatment of advanced melanoma. Curr Oncol Rep 2013;15:473-82

65.

Knight ZA, Shokat KM. Chemically targeting the PI3K family. Biochem Soc Trans 2007;35:245-9

55.

Kirkwood JM, Bastholt L, Robert C, et al. Phase II, open-label, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanoma. Clin Cancer Res 2012;18:555-67

66.

Margolin K, Longmate J, Baratta T, et al. CCI-779 in metastatic melanoma: a phase II trial of the California Cancer Consortium. Cancer 2005;104:1045-8

67.

Margolin KA, Moon J, Flaherty LE, et al. Randomized phase II trial of sorafenib with temsirolimus or tipifarnib in untreated metastatic melanoma (S0438). Clin Cancer Res 2012;18:1129-37

68.

Werzowa J, Koehrer S, Strommer S, et al. Vertical inhibition of the mTORC1/mTORC2/PI3K pathway shows synergistic effects against melanoma in vitro and in vivo. J Invest Dermatol 2011;131:495-503

56.

57.

Robert C, Dummer R, Gutzmer R, et al. Selumetinib plus dacarbazine versus placebo plus dacarbazine as first-line treatment for BRAF-mutant metastatic melanoma: a phase 2 double-blind randomised study. Lancet Oncol 2013;14:733-40 Kim KB, Kefford R, Pavlick AC, et al. Phase II study of the MEK1/

Expert Opin. Biol. Ther. (2014) 14(5)

19

S. Tomei et al.

69.

70.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

71.

72.

73.

74.

75.

Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271-81 Hay N. The Akt-mTOR tango and its relevance to cancer. Cancer Cell 2005;8:179-83 Greger JG, Eastman SD, Zhang V, et al. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol Cancer Ther 2012;11:909-20 Posch C, Moslehi H, Feeney L, et al. Combined targeting of MEK and PI3K/ mTOR effector pathways is necessary to effectively inhibit NRAS mutant melanoma in vitro and in vivo. Proc Natl Acad Sci USA 2013;110:4015-20 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;24:4738-45 Bedikian AY, Lebbe C, Robert C, et al. Survival in a phase III, randomized, double-blind study of dacarbazine with or without oblimersen (Bcl-2 antisense) in patients with advanced melanoma and low-normal serum lactate dehydrogenase (LDH; AGENDA). J Clin Oncol 2011;29(Suppl):abstract 8531 Horn S, Figl A, Rachakonda PS, et al. TERT promoter mutations in familial and sporadic melanoma. Science 2013;339:959-61

76.

Huang FW, Hodis E, Xu MJ, et al. Highly recurrent TERT promoter mutations in human melanoma. Science 2013;339:957-9

77.

Varker KA, Biber JE, Kefauver C, et al. A randomized phase 2 trial of bevacizumab with or without daily lowdose interferon alfa-2b in metastatic malignant melanoma. Ann Surg Oncol 2007;14:2367-76

78.

20

Kim KB, Sosman JA, Fruehauf JP, et al. BEAM: a randomized phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma. J Clin Oncol 2012;30:34-41

79.

.

80.

..

81.

82.

83.

84.

85.

86.

Minor DR. Bevacizumab advanced melanoma (BEAM) was a positive trial. J Clin Oncol 2012;30:2023; author reply 2023-2024 This is an interesting comment on the BEAM trial, describing why inhibiting angiogenesis represents an approach that should not be dismissed. Del Vecchio M, Mortarini R, Canova S, et al. Bevacizumab plus fotemustine as first-line treatment in metastatic melanoma patients: clinical activity and modulation of angiogenesis and lymphangiogenesis factors. Clin Cancer Res 2010;16:5862-72 An interesting Phase II study evaluating combination of bevacizumab and chemotherapy and assessing biological correlates. Hodi FS, Friedlander PA, Atkins MB, et al. A phase I trial of ipilimumab plus bevacizumab in patients with unresectable stage III or stage IV melanoma. J Clin Oncol 2011;29(Suppl):abstract 8511 Flaherty KT, Lee SJ, Zhao F, et al. Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma. J Clin Oncol 2013;31:373-9 Hauschild A, Agarwala SS, Trefzer U, et al. Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma. J Clin Oncol 2009;27:2823-30 Fruehauf J, Lutzky J, McDermott D, et al. Multicenter, phase II study of axitinib, a selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, in patients with metastatic melanoma. Clin Cancer Res 2011;17:7462-9 Hong DS, Koetz BS, Kurzrock R, et al. Phase I dose-escalation study of E7080, a selective tyrosine kinase inhibitor, administered orally to patients with solid tumors. J Clin Oncol 2010;28:15s(Suppl):abstract 2540 Corrie PG, Basu B, Zaki KA. Targeting angiogenesis in melanoma: prospects for the future. Ther Adv Med Oncol 2010;2:367-80

Expert Opin. Biol. Ther. (2014) 14(5)

87.

.

Ascierto ML, De Giorgi V, Liu Q, et al. An immunologic portrait of cancer. J Transl Med 2011;9:146 A comprehensive review proposing an immunological interpretation of cancer.

88.

Shimanovsky A, Jethava A, Dasanu CA. Immune alterations in malignant melanoma and current immunotherapy concepts. Expert Opin Biol Ther 2013;13:1413-27

89.

Wilmott JS, Long GV, Howle JR, et al. Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin Cancer Res 2012;18:1386-94

90.

Gajewski TF, Meng Y, Harlin H. Immune Suppression in the tumor microenvironment. J Immunother 2006;29:233-40

91.

Krieg C, Letourneau S, Pantaleo G, Boyman O. Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells. Proc Natl Acad Sci USA 2010;107:11906-11

92.

Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999;17:2105-16

93.

Rosenberg SA, Mule JJ, Spiess PJ, et al. Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2. J Exp Med 1985;161:1169-88

94.

Phan GQ, Attia P, Steinberg SM, et al. Factors associated with response to high-dose interleukin-2 in patients with metastatic melanoma. J Clin Oncol 2001;19:3477-82

95.

Smith FO, Downey SG, Klapper JA, et al. Treatment of metastatic melanoma using interleukin-2 alone or in conjunction with vaccines. Clin Cancer Res 2008;14:5610-18

96.

Joseph RW, Sullivan RJ, Harrell R, et al. Correlation of NRAS mutations with clinical response to high-dose IL-2 in patients with advanced melanoma. J Immunother 2012;35:66-72

97.

Dummer R, Hauschild A, Guggenheim M, et al. Melanoma: ESMO Clinical Practice Guidelines for

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

diagnosis, treatment and follow-up. Ann Oncol 2010;21(Suppl 5):v194-7 98.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

99.

100.

101.

102.

103.

.

104.

Sabatino M, Kim-Schulze S, Panelli MC, et al. Serum vascular endothelial growth factor and fibronectin predict clinical response to high-dose interleukin2 therapy. J Clin Oncol 2009;27:2645-52 Yuan J, Zhou J, Dong Z, et al. Pre-treatment serum vascular endothelial growth factor is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. J Immuno Ther Cancer 2013;1(Suppl 1):P247 Panelli MC, Wang E, Phan G, et al. Gene-expression profiling of the response of peripheral blood mononuclear cells and melanoma metastases to systemic IL-2 administration. Genome Biol 2002;3:RESEARCH0035 Panelli MC, White R, Foster M, et al. Forecasting the cytokine storm following systemic interleukin (IL)2 administration. J Transl Med 2004;2:17 Wang E, Miller LD, Ohnmacht GA, et al. Prospective molecular profiling of melanoma metastases suggests classifiers of immune responsiveness. Cancer Res 2002;62:3581-6 Weiss GR, Grosh WW, Chianese-Bullock KA, et al. Molecular insights on the peripheral and intratumoral effects of systemic high-dose rIL-2 (aldesleukin) administration for the treatment of metastatic melanoma. Clin Cancer Res 2011;17:7440-50 An interesting prospective study on gene expression in tumor and PBMC following IL-2 administration. Bedognetti D, Wang E, Sertoli MR, Marincola FM. Gene-expression profiling in vaccine therapy and immunotherapy for cancer. Expert Rev Vaccines 2010;9:555-65

105.

Blanchard T, Srivastava PK, Duan F. Vaccines against advanced melanoma. Clin Dermatol 2013;31:179-90

106.

Mocellin S, Rossi CR, Nitti D, et al. Dissecting tumor responsiveness to immunotherapy: the experience of peptide-based melanoma vaccines. Biochim Biophys Acta 2003;1653:61-71

107.

Lens M. The role of vaccine therapy in the treatment of melanoma. Expert Opin Biol Ther 2008;8:315-23

108.

109.

110.

Terando AM, Faries MB, Morton DL. Vaccine therapy for melanoma: current status and future directions. Vaccine 2007;25(Suppl 2):B4-16 Ascierto ML, Worschech A, Yu Z, et al. Permissivity of the NCI-60 cancer cell lines to oncolytic Vaccinia Virus GLV-1h68. BMC Cancer 2011;11:451 Reinboth J, Ascierto ML, Chen NG, et al. Correlates between host and viral transcriptional program associated with different oncolytic vaccinia virus isolates. Hum Gene Ther Methods 2012;23:285-96

111.

Wang E, Phan GQ, Marincola FM. T-cell-directed cancer vaccines: the melanoma model. Expert Opin Biol Ther 2001;1:277-90

112.

Rosenberg SA, Packard BS, Aebersold PM, et al. Use of tumorinfiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 1988;319:1676-80

113.

114.

van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. J Immunol 2007;178:2617-21 Traversari C, van der Bruggen P, Luescher IF, et al. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E. J Exp Med 1992;176:1453-7

115.

Alexandrescu DT, Ichim TE, Riordan NH, et al. Immunotherapy for melanoma: current status and perspectives. J Immunother 2010;33:570-90

116.

Wang E, Selleri S, Sabatino M, et al. Spontaneous and treatment-induced cancer rejection in humans. Expert Opin Biol Ther 2008;8:337-49

117.

.

118.

Wang E, Bedognetti D, Marincola FM. Prediction of response to anticancer immunotherapy using gene signatures. J Clin Oncol 2013;31:2369-71 An interesting mini-review on the role of gene-expression in predicting outcome to immunotherapy. Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the

Expert Opin. Biol. Ther. (2014) 14(5)

.

tumor microenvironment. Science 2013;342:967-70 An interesting study showing the role of gut microbiota in modulating the tumor microenvironment and thereby influencing response to immunotherapy.

119. Wang E, Bedognetti D, Tomei S, Marincola FM. Common pathways to tumor rejection. Ann NY Acad Sci 2013;1284:75-9 120. Wang E, Tomei S, Marincola FM. Reflections upon human cancer immune responsiveness to T cell-based therapy. Cancer Immunol Immunother 2012;61:761-70 . This manuscript reflects on the mechanisms behind patients’ individual tumor rejection and immune responsiveness. 121. Galon J, Angell HK, Bedognetti D, Marincola FM. The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures. Immunity 2013;39:11-26 .. An updated review summarizing and explaining striking analogies between prognostic and predictive gene-expression signatures. 122. Klebanoff CA, Acquavella N, Yu Z, Restifo NP. Therapeutic cancer vaccines: are we there yet? Immunol Rev 2011;239:27-44 123. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22 124. Stein WD, Gulley JL, Schlom J, et al. Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: the growth rate constant as an indicator of therapeutic efficacy. Clin Cancer Res 2011;17:907-17 125. Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst 2012;104:599-613 126. Motz GT, Coukos G. Deciphering and reversing tumor immune Suppression. Immunity 2013;39:61-73 127. Salerno EP, Shea SM, Olson WC, et al. Activation, dysfunction and retention of T cells in vaccine sites after injection of incomplete Freund’s adjuvant, with or without peptide. Cancer Immunol Immunother 2013;62:1149-59

21

S. Tomei et al.

128. Hailemichael Y, Dai Z, Jaffarzad N, et al. Persistent antigen at vaccination sites induces tumor-specific CD8(+) T cell sequestration, dysfunction and deletion. Nat Med 2013;19:465-72

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

129. Bedognetti D, Spivey TL, Zhao Y, et al. CXCR3/CCR5 pathways in metastatic melanoma patients treated with adoptive therapy and interleukin-2. Br J Cancer 2013;109:2412-23 . An interesting study evaluating a combinatorial approach to biomarker discovery in cancer immunotherapy. 130. Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with stem cell-like properties. Nat Med 2011;17:1290-7 131. Uccellini L, De Giorgi V, Zhao Y, et al. IRF5 gene polymorphisms in melanoma. J Transl Med 2012;10:170 132. Wang E, Zhao Y, Monaco A, et al. A multi-factorial genetic model for prognostic assessment of high risk melanoma patients receiving adjuvant interferon. PLoS One 2012;7:e40805 133. Parmiani G, Cimminiello C, Maccalli C. Increasing immunogenicity of cancer vaccines to improve their clinical outcome. Expert Rev Vaccines 2013;12:1111-13 134. Schwartzentruber DJ, Lawson DH, Richards JM, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med 2011;364:2119-27 .. The first randomized trial showing that vaccination could be effective in metastatic melanoma when combined to other modalities. 135. Kruit WH, Suciu S, Dreno B, et al. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma. J Clin Oncol 2013;31:2413-20 136. Ulloa-Montoya F, Louahed J, Dizier B, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol 2013;31:2388-95 137. Brichard VG. Selection of patients for cancer immunotherapy. Society for immunotherapy of cancer 2013 Annual Meeting, oral presentation

22

138. Kaufman HL. Vaccines for melanoma and renal cell carcinoma. Semin Oncol 2012;39:263-75 139. Andtbacka R, Collichio FA, Amatruda T, et al. OPTiM: a randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colonystimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. J Clin Oncol 2013;31(Suppl):abstract LBA9008

150. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 1995;182:459-65 151. Maio M, Bondarenko I, Robert C, et al. Four-year survival update for metastatic melanoma (MM) patients (pts) treated with ipilimumab (IPI) + dacarbazine (DTIC) on Phase 3 study CA184-024. Ann Oncol 23(9s Suppl): abstract 1127P

140. Chowdhery R, Gonzalez R. Immunologic therapy targeting metastatic melanoma: allovectin-7. Immunotherapy 2011;3:17-21

152. Wolchok JD, Weber JS, Maio M, et al. Four-year survival rates for patients with metastatic melanoma who received ipilimumab in phase II clinical trials. Ann Oncol 2013;24:2174-80

141. Berd D, Sato T, Maguire HC Jr, et al. Immunopharmacologic analysis of an autologous, hapten-modified human melanoma vaccine. J Clin Oncol 2004;22:403-15

153. Maio M, Di Giacomo AM, Robert C, Eggermont AM. Update on the role of ipilimumab in melanoma and first data on new combination therapies. Curr Opin Oncol 2013;25:166-72

142. Gottardi R, Douradinha B. Carbon nanotubes as a novel tool for vaccination against infectious diseases and cancer. J Nanobiotechnology 2013;11:30

154. Hamid O, Hwu W, Richards J, et al. Ipilimumab (Ipi) expanded access program (EAP) for patients (pts) with stage III/IV melanoma: 10 mg/kg cohort interim results. J Clin Oncol 2012;30(Suppl):abstract 8508

143. Pescatori M, Bedognetti D, Venturelli E, et al. Functionalized carbon nanotubes as immunomodulator systems. Biomaterials 2013;34:4395-403 144. Delogu LG, Vidili G, Venturelli E, et al. Functionalized multiwalled carbon nanotubes as ultrasound contrast agents. Proc Natl Acad Sci USA 2012;109:16612-17 145. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 2012;12:269-81 .. A comprehensive review on the advances in adoptive therapy.

155. Di Giacomo AM, Ascierto PA, Pilla L, et al. Ipilimumab and fotemustine in patients with advanced melanoma (NIBIT-M1): an open-label, single-arm phase 2 trial. Lancet Oncol 2012;13:879-86 156. Patel SP, Hwu W, Kim KB, et al. Phase II study of the frontline combination of ipilimumab and temozolomide in patients with metastatic melanoma. J Clin Oncol 2012;30(Suppl): abstract 8514

146. Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol 2013;31:999-1008

157. Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 2009;15:7412-20

147. Wang XY, Zuo D, Sarkar D, Fisher PB. Blockade of cytotoxic T-lymphocyte antigen-4 as a new therapeutic approach for advanced melanoma. Expert Opin Pharmacother 2011;12:2695-706

158. Ribas A, Kefford R, Marshall MA, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol 2013;31:616-22

148. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64

159. Ribas A. Clinical development of the anti-CTLA-4 antibody tremelimumab. Semin Oncol 2010;37:450-4

149. Allison JP, Krummel MF. The Yin and Yang of T cell costimulation. Science 1995;270:932-3

160. Tarhini AA, Cherian J, Moschos SJ, et al. Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage

Expert Opin. Biol. Ther. (2014) 14(5)

Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma

IV melanoma. J Clin Oncol 2012;30:322-8 161.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

162.

163.

164.

Calabro L, Morra A, Fonsatti E, et al. Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial. Lancet Oncol 2013;14:1104-11

Redmond WL, Weinberg AD. Targeting OX40 and OX40L for the treatment of autoimmunity and cancer. Crit Rev Immunol 2007;27:415-36

172.

Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol 2005;23:23-68

173.

Croft M. Control of immunity by the TNFR-related molecule OX40 (CD134). Annu Rev Immunol 2010;28:57-78

Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027-34

174.

Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2001;2:261-8

Curti BD, Kovacsovics-Bankowski M, Morris N, et al. OX40 Is a Potent Immune-Stimulating Target in LateStage Cancer Patients. Cancer Res 2013;73:7189-98

175.

Redmond WL, Triplett T, Floyd K, Weinberg AD. Dual anti-OX40/IL2 therapy augments tumor immunotherapy via IL-2R-mediated regulation of OX40 expression. PLoS One 2012;7:e34467

176.

French RR, Chan HT, Tutt AL, Glennie MJ. CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help. Nat Med 1999;5:548-53

177.

Diehl L, den Boer AT, Schoenberger SP, et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy. Nat Med 1999;5:774-9

Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455-65

165.

Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443-54

166.

Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134-44

167.

Sznol M, Kluger H, Hodi FS, et al. Survival and long-term follow-up of safety and response in patients (pts) with advanced melanoma (MEL) in a phase I trial of nivolumab (anti-PD-1; BMS936558; ONO-4538). J Clin Oncol 2013;31(Suppl):abstract CRA9006

168.

171.

Hamid O, Sosman J, Lawrence D, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). J Clin Oncol 2013;31(Suppl): abstract 9010

178.

Sotomayor EM, Borrello I, Tubb E, et al. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40. Nat Med 1999;5:780-7

179.

Vonderheide RH, Glennie MJ. Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res 2013;19:1035-43

180.

Vonderheide RH, Flaherty KT, Khalil M, et al. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin Oncol 2007;25:876-83

169.

Melero I, Grimaldi AM, Perez-Gracia JL, Ascierto PA. Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination. Clin Cancer Res 2013;19:997-1008

181.

Spranger S, Gajewski T. Therapeutic efficacy of combined blockade of CTLA4 +/- PD-L1 +/- IDO is associated with re-activation of T cells directly within the tumor microenvironment. J Immuno Ther Cancer 2013;1(Suppl 1):O8

170.

Sugamura K, Ishii N, Weinberg AD. Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40. Nat Rev Immunol 2004;4:420-31

182.

Brignone C, Gutierrez M, Mefti F, et al. First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and

Expert Opin. Biol. Ther. (2014) 14(5)

antitumor activity. J Transl Med 2010;8:71 183. Poirier N, Haudebourg T, Brignone C, et al. Antibody-mediated depletion of lymphocyte-activation gene-3 (LAG-3(+)) -activated T lymphocytes prevents delayed-type hypersensitivity in nonhuman primates. Clin Exp Immunol 2011;164:265-74 184. Roychoudhuri R, Hirahara K, Mousavi K, et al. BACH2 represses effector programs to stabilize T(reg)mediated immune homeostasis. Nature 2013;498:506-10 185. Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011;17:4550-7 186. Radvanyi LG, Bernatchez C, Zhang M, et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 2012;18:6758-70 187. Besser MJ, Shapira-Frommer R, Itzhaki O, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 2013;19:4792-800 188. Dudley ME, Gross CA, Somerville RP, et al. Randomized selection design trial evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma. J Clin Oncol 2013;31:2152-9 189. Phan GQ, Rosenberg SA. Adoptive cell transfer for patients with metastatic melanoma: the potential and promise of cancer immunotherapy. Cancer Control 2013;20:289-97 190. Bedognetti D, Wang E, Sato-Matsushita M, et al. Molecular profiling of immunotherapeutic resistance. In: Prendergsat GC, Jaffee EM, editors, Cancer immunotherapy, immune suppression and tumor growth. 2nd edition. Elsevier, 2013. 191. Fisher B, Packard BS, Read EJ, et al. Tumor localization of adoptively transferred indium-111 labeled tumor infiltrating lymphocytes in patients with

23

S. Tomei et al.

metastatic melanoma. J Clin Oncol 1989;7:250-61 192. Spivey TL, Uccellini L, Ascierto ML, et al. Gene expression profiling in acute allograft rejection: challenging the immunologic constant of rejection hypothesis. J Transl Med 2011;9:174

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Cornell University on 03/21/14 For personal use only.

193. Ascierto PA, Marincola FM. Combination therapy: the next opportunity and challenge of medicine. J Transl Med 2011;9:115

203. Ribas A, Wolchok JD. Combining cancer immunotherapy and targeted therapy. Curr Opin Immunol 2013;25:291-6 204. Castelli C, Sensi M, Lupetti R, et al. Expression of interleukin 1 alpha, interleukin 6, and tumor necrosis factor alpha genes in human melanoma clones is associated with that of mutated N-RAS oncogene. Cancer Res 1994;54:4785-90

microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med 2011;9:204 215. Ji RR, Chasalow SD, Wang L, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother 2012;61:1019-31

205. Sumimoto H, Imabayashi F, Iwata T, Kawakami Y. The BRAF-MAPK signaling pathway is essential for cancerimmune evasion in human melanoma cells. J Exp Med 2006;203:1651-6

216. Carretero R, Wang E, Rodriguez AI, et al. Regression of melanoma metastases after immunotherapy is associated with activation of antigen presentation and interferon-mediated rejection genes. Int J Cancer 2012;131:387-95

206. Khalili JS, Liu S, Rodriguez-Cruz TG, et al. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin Cancer Res 2012;18:5329-40

217. Panelli MC, Stashower ME, Slade HB, et al. Sequential gene profiling of basal cell carcinomas treated with imiquimod in a placebo-controlled study defines the requirements for tissue rejection. Genome Biol 2007;8:R8

196. Hodi FS, Lee SJ, McDermott DF, et al. Multicenter, randomized phase II trial of GM-CSF plus ipilimumab versus ipi alone in metastatic melanoma. J Clin Oncol 2013;31(Suppl): abstract CRA9007

207. Frederick DT, Piris A, Cogdill AP, et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin Cancer Res 2013;19:1225-31

218. Wang E, Uccellini L, Marincola FM. A genetic inference on cancer immune responsiveness. Oncoimmunology 2012;1:520-5

197. Prieto PA, Yang JC, Sherry RM, et al. CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res 2012;18:2039-47

208. Liu C, Peng W, Xu C, et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin Cancer Res 2013;19:393-403

198. Marincola FM. The trouble with translational medicine. J Intern Med 2011;270:123-7

209. Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res 2010;70:5213-19

194. Marincola FM, White DE, Wise AP, Rosenberg SA. Combination therapy with interferon alfa-2a and interleukin-2 for the treatment of metastatic cancer. J Clin Oncol 1995;13:1110-22 195. Daponte A, Signoriello S, Maiorino L, et al. Phase III randomized study of fotemustine and dacarbazine versus dacarbazine with or without interferonalpha in advanced malignant melanoma. J Transl Med 2013;11:38

199. Simon R. Novel clinical trial designs for development of immunotherapy combinations. SITC Cancer Immunotherapy Clinical Trials: Concepts and Challenges Meeting 2013, oral presentation 200. Yang Y, Lizee G, Hwu P. Strong emerging rationale for combining oncogene-targeted agents with immunotherapy. Oncoimmunology 2013;2:e22730 201. Ribas A, Hersey P, Middleton MR, et al. New challenges in endpoints for drug development in advanced melanoma. Clin Cancer Res 2012;18:336-41 202. Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013;39:74-88

24

210. Ribas A, Hodi FS, Callahan M, et al. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med 2013;368:1365-6 211. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 2012;367:1694-703 212. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122-33 213. Murtas D, Maric D, De Giorgi V, et al. IRF-1 responsiveness to IFN-gamma predicts different cancer immune phenotypes. Br J Cancer 2013;109:76-82 214. Hamid O, Schmidt H, Nissan A, et al. A prospective phase II trial exploring the association between tumor

Expert Opin. Biol. Ther. (2014) 14(5)

Affiliation Sara Tomei†1,2, Ena Wang1,3, Lucia Gemma Delogu4, Francesco M Marincola1,3 & Davide Bedognetti*1 †,* Authors for correspondence 1 National Institutes of Health, Clinical Center and trans-NIH Center for Human Immunology (CHI), Department of Transfusion Medicine, Infectious Disease and Immunogenetics Section (IDIS), Bethesda, MD 20892, USA E-mail: [email protected] 2 Weill Cornell Medical College in Qatar, Department of Genetic Medicine, P.O. Box 24144, Doha, Qatar E-mail: [email protected] 3 Sidra Medical and Research Center, Research Branch, P.O. Box 26999, Doha, Qatar 4 Universita` degli Studi di Sassari, Dipartimento di Chimica e Farmacia, 07100 Sassari, Italy