Melanoma is entering into an era of combinatorial approaches to build upon recent clinical breakthroughs achieved by novel single-agent therapies. One of the leading targets to emerge from the growing understanding of the molecular pathogenesis, heterogeneity, and resistance mechanisms of melanomas is the phosphoinositide 3-kinase (PI3K)–AKT pathway. Multiple genetic and epigenetic aberrations that activate this pathway have been identified in melanomas de novo and in acquired resistance models. These developments have been paralleled by the establishment of models for preclinical testing and the availability of compounds that target various effectors in the pathway. Thus, in addition to having a strong rationale for targeting, the PI3K–AKT pathway presents an immediate clinical opportunity. However, the development of effective strategies against this pathway must overcome several key challenges, including optimizing patient selection, overcoming feedback loops, and pathway cross-talk that can mediate resistance. This review discusses the current understanding and ongoing research about the PI3K–AKT pathway in melanoma and emerging strategies to achieve clinical benefit in patients by targeting it. Clin Cancer Res; 19(19); 5310–9. ©2013 AACR.

The phosphoinositide 3-kinase (PI3K)–AKT cascade is one of the most studied pathways in cancer. The pathway is a critical regulator of many essential physiologic processes that are critical to the aggressive nature and behavior of malignant cells. Previous studies have shown that the pathway is among the most frequent targets of genetic aberrations across many types of cancer (1). These alterations include mutations and copy number changes within the core components of the pathway, as well as alterations in genes that use that pathway as a critical effector [i.e., receptor tyrosine kinases (RTK)]. For all of these reasons, the PI3K–AKT pathway has also been the focus of aggressive pharmacologic development and testing (2, 3).

The high prevalence of activating mutations in BRAF and NRAS in cutaneous melanomas supports a critical role for activation of the RAS–RAF–MEK–ERK pathway in the pathogenesis of this disease (4). However, multiple lines of evidence have also shown a significant role for the PI3K–AKT pathway. This review highlights some of the key findings about the PI3K–AKT pathway in melanoma, and the rationale, approaches, and challenges to the development of effective therapeutic approaches against it.

The physiologic regulation of the PI3K–AKT cascade is shown in Fig. 1 (5). PI3K, which consists of a dimer of catalytic (i.e., p110) and regulatory (i.e., p85) subunits, can be activated by multiple signals, including RTKs, RAS proteins, and cell–cell contacts, among others. Activated PI3K phosphorylates phosphatidylinositols in the plasma membrane at the 3′-OH group. These 3′-phospholipids attract proteins that contain a pleckstrin homology domain to the cell membrane, including the serine-threonine kinase AKT. AKT, which has three isoforms (AKT1/2/3), is phosphorylated at two critical and conserved residues, Thr308 (by PDK1) and Ser473 (by the mTORC2 complex), which fully activate its catalytic activity. Activated AKT then phosphorylates a number of effector proteins, thereby regulating multiple key cellular processes, including proliferation, survival, motility, metabolism, angiogenesis, and more. PTEN regulates the activity of the pathway by dephosphorylating phosphatidylinositols at the 3′-position, thereby antagonizing the activity of PI3K (6). Multiple other lipid and protein phosphatases also regulate various steps and effectors in the pathway (7).

Figure 1.

Regulators, effectors, and somatic alterations in the PI3K–AKT pathway in melanoma.

Figure 1.

Regulators, effectors, and somatic alterations in the PI3K–AKT pathway in melanoma.

Close modal

The PI3K–AKT pathway is activated multiple ways in melanoma. The two most common and studied events are activating mutations in the oncogene NRAS (15–20%) and loss of expression and/or function of the tumor suppressor PTEN (20–30%; ref. 4). Similar to BRAF and NRAS mutations in the RAS–RAF–MEK–ERK signaling pathway, NRAS mutations and PTEN mutations/deletions are largely mutually exclusive. In contrast, PTEN loss commonly occurs in melanomas with activating BRAF mutations, resulting in concurrent activation of the RAS–RAF–MEK–ERK and PI3K–AKT pathways (8–10). The general mutual exclusivity of NRAS mutations and PTEN loss in melanoma is thought by many to be attributable to the fact that both events activate the PI3K–AKT pathway, thus rendering the presence of both alterations in the same tumor functionally redundant. However, similar to findings in other tumor types, quantitative analysis of melanoma cell lines and clinical specimens has shown that melanomas with PTEN loss consistently have higher levels of AKT activation than those with NRAS mutations (11–13). Furthermore, experiments in a RAS-mutant melanoma genetically engineered mouse model (GEMM) showed that loss of PTEN increased invasiveness and metastatic potential (14). Although rare, deletions and mutations of PTEN have been detected in some melanomas with activating NRAS mutations, including in two recent whole exome sequencing studies of >100 melanomas, which also detected PTEN alterations in melanomas with wild-type BRAF and NRAS (15, 16). However, these data should be interpreted with caution, as no standardized protocol has been established for defining PTEN deletions. Preliminary analysis of The Cancer Genome Atlas (TCGA) data suggests that such a standard should take into account both copy number and focality, and would decrease the discrepancies between studies. Additional studies support that PTEN expression can be regulated epigenetically, including by microRNAs and the PTENP1 pseudogene (17–20). A more complete understanding of the prevalence, pattern, molecular causes, and clinical associations of PTEN loss will likely be possible with the completion of the ongoing melanoma TCGA effort, which will include DNA-, RNA-, and protein-based analyses of up to 500 clinically annotated melanoma specimens.

The functional significance of PTEN loss has been studied extensively in the setting of melanomas with activating BRAF mutations. To date, nearly all published patient-derived melanoma cell lines with complete loss of PTEN have concurrent BRAF mutations (11, 21–24). This strong association with BRAF mutations has also been shown functionally in GEMM. Although expression of the BRAF V600E protein in murine melanocytes results in increased proliferation of melanocytes, concurrent PTEN loss results in 100% penetrance of invasive, metastatic tumors, thus establishing the first such model for this disease (25). BRAF-mutant human melanoma cell lines with loss of PTEN are generally sensitive to growth inhibition by BRAF and MEK inhibitors, but they are significantly resistant to apoptosis induction by these treatments (26–29). Supporting the clinical relevance of these findings, two independent analyses of PTEN status, one genetic and one immunohistochemical, identified decreased clinical benefit with selective BRAF inhibitors (vemurafenib, dabrafenib) in patients with loss of PTEN in pretreatment (including archival) tumor specimens (30, 31). Although these results support the potential value for evaluating PTEN function in BRAF-mutant melanomas in future studies, it is not yet clear what methodology of PTEN testing (i.e., DNA-, RNA- or protein-based) will prove most informative. Furthermore, very little information is available at this time about the concordance of PTEN among different tumors in individual patients (32).

Both broad and focused sequencing studies have identified additional genetic events that can activate the PI3K–AKT pathway in melanoma. Point mutations in PIK3CA, which encodes the p110α catalytic subunit of PI3K, are detected in 2% to 6% of melanomas (15, 16, 33, 34). Notably, although some of these mutations are recurrent hotspots reported in other tumor types, others are novel and of unclear functional significance. Mutations producing the activating substitution E17K in AKT1, which are detected as rare events in several tumor types, have also been detected as rare events in melanoma (1–2%; refs. 35, 36). However, melanoma is the only disease in which the analogous mutation in AKT3 has been detected (1–2%). The identification of AKT3 mutations builds upon previous studies reporting increased expression and activation of AKT3 in melanoma progression, potentially implicating it as a novel therapeutic target (37, 38). Recently, amplification of a 5 Mbp locus including RICTOR, which encodes a component of the multiprotein TORC2 complex that phosphorylates AKT at the Ser473 residue, has been reported in up to 5% of melanomas, particularly those that are relatively protected from ultraviolet radiation (16). Temporally, PI3K activation seems to be a secondary event. In an immunohistochemical survey, PTEN protein loss was observed in melanoma but not in nevi (39), in contrast with the uniformly high mutation rate of BRAF across all stages (40). Similarly, phosphorylated AKT was found to be high in melanoma but not in nevi (41). The findings are also consistent with lack of the melanocytic phenotype in the PTEN−/− mice in the absence of the mutant BRAF allele (25).

The PI3K–AKT pathway is also implicated as a critical effector of alterations that activate RTKs. Activating mutations in c-Kit are rare in cutaneous melanomas, but they are relatively common in acral and mucosal melanomas (42). Although the low prevalence of BRAF and NRAS mutations in these subtypes, and their general mutual exclusivity with c-Kit mutations, suggested that signaling by mutant KIT proteins might activate the RAS–RAF–MEK–ERK pathway, functional studies in cell lines have shown activation of, and in some studies dependence upon, the PI3K–AKT pathway (43–45). One study has also reported frequent (∼20%) somatic mutations in the ERBB4 gene (46). Although these mutations do not cluster in any functional domain, preclinical studies suggested that multiple mutant forms of the encoded ERBB4 protein activated the PI3K–AKT pathway. However, recent whole exome-sequencing studies did not identify ERBB4 as a significantly mutated gene by the algorithms used in those analyses (15, 16). In addition to genetic events, it seems that epigenetically mediated activation of RTKs plays a role in melanoma, specifically in resistance to BRAF inhibitors. Two different groups identified increased expression and activation of different RTKs [platelet-derived growth factor β (PDGFβ) and insulin-like growth factor receptor (IGF1R), respectively] in progressing tumors and melanoma cell lines with acquired resistance to BRAF inhibitors (47–49). Both groups showed that the activation of these RTKs did not rescue the activity of the mitogen-activated protein kinase (MAPK) pathway but instead caused compensatory activation of the PI3K–AKT pathway. Importantly, no mutations or amplification in the genes encoding the RTKs were detected in the cell lines. Similar compensatory activation of this pathway via IGF1R was also reported in human melanoma cells with de novo resistance to killing by MEK inhibitors (29). More recently, two different groups showed that secretion of hepatocyte growth factor by nontransformed cells in the tumor microenvironment results in PI3K–AKT pathway activation in melanoma cells (50, 51). This interaction caused resistance to BRAF inhibitors in vitro and was correlated with inferior clinical outcomes in patients.

The multiple ways in which the PI3K–AKT pathway is activated in melanoma, and existing evidence for a functional role in progression and resistance, support the rationale to target it therapeutically. Indeed, similar evidence in multiple tumor types has led to the development of multiple classes of inhibitors against this pathway. Classes of agents include inhibitors of PI3K (pan-isoform and isoform-specific), dual PI3K–mTOR, AKT, and mTOR (mTORC1 and dual mTORC1/2 inhibitors; Table 1). Multiple agents are available in each class, many of which are currently undergoing clinical evaluation in patients. Although the availability of this spectrum of agents presents a tremendous opportunity, a key challenge for a relatively rare disease such as metastatic melanoma is to rationally use and prioritize these agents to determine their clinical value effectively and efficiently.

Table 1.

PI3K pathway inhibitors currently in clinical trials for any cancer

TargetInhibitorAlternative nameCompany
AKT AZD5363  AstraZeneca 
 GDC-0068  Genentech 
 GSK2110183  GlaxoSmithKline 
 GSK2141795  GlaxoSmithKline 
 GSK690693  GlaxoSmithKline 
 KRX-0401 Perifosine Keryx 
 MK2206  Merck 
 SR13668  SRI 
mTORC1 Rapamycin Sirolimus Pfizer 
 CCI779 Temsirolimus Pfizer 
 MK-8669 Ridaforolimus Ariad 
 RAD001 Everolimus Novartis 
Dual mTORC1/2 AZD2014  AstraZeneca 
 AZD8055  AstraZeneca 
 CC-223  Celgene 
 MLN0128 INK-128 Millenium 
 OSI-027  Astellas 
 Palomid 529  Paloma 
PI3K p110α-selective GDC-0032  Genentech 
 MLN1117 INK-1117 Millenium 
 NVP-BYL719  Novartis 
PI3K p110β-selective GSK2636771  GlaxoSmithKline 
 SAR260301  Sanofi-Aventis 
PI3K p110δ-selective CAL101  Gilead 
 GSK2269557  GlaxoSmithKline 
Pan PI3K BAY80-6946  Bayer 
 GDC-0941  Genentech 
 NVP-BKM120  Novartis 
 PX866  Oncothyreon 
 SF1126  Semafore 
 XL147 SAR245408 Exelixis 
 ZSTK474  Zenyaku Kogyo 
Dual PI3K/mTOR DS-7423  Daiichi Sankyo 
 GDC-0980  Genentech 
 GSK2126458  GlaxoSmithKline 
 NVP-BEZ235  Novartis 
 NVP-BGT226  Novartis 
 P7170  Piramal 
 PF-05212384  Pfizer 
 PF-4691502  Pfizer 
 XL765 SAR245409 Exelixis 
TargetInhibitorAlternative nameCompany
AKT AZD5363  AstraZeneca 
 GDC-0068  Genentech 
 GSK2110183  GlaxoSmithKline 
 GSK2141795  GlaxoSmithKline 
 GSK690693  GlaxoSmithKline 
 KRX-0401 Perifosine Keryx 
 MK2206  Merck 
 SR13668  SRI 
mTORC1 Rapamycin Sirolimus Pfizer 
 CCI779 Temsirolimus Pfizer 
 MK-8669 Ridaforolimus Ariad 
 RAD001 Everolimus Novartis 
Dual mTORC1/2 AZD2014  AstraZeneca 
 AZD8055  AstraZeneca 
 CC-223  Celgene 
 MLN0128 INK-128 Millenium 
 OSI-027  Astellas 
 Palomid 529  Paloma 
PI3K p110α-selective GDC-0032  Genentech 
 MLN1117 INK-1117 Millenium 
 NVP-BYL719  Novartis 
PI3K p110β-selective GSK2636771  GlaxoSmithKline 
 SAR260301  Sanofi-Aventis 
PI3K p110δ-selective CAL101  Gilead 
 GSK2269557  GlaxoSmithKline 
Pan PI3K BAY80-6946  Bayer 
 GDC-0941  Genentech 
 NVP-BKM120  Novartis 
 PX866  Oncothyreon 
 SF1126  Semafore 
 XL147 SAR245408 Exelixis 
 ZSTK474  Zenyaku Kogyo 
Dual PI3K/mTOR DS-7423  Daiichi Sankyo 
 GDC-0980  Genentech 
 GSK2126458  GlaxoSmithKline 
 NVP-BEZ235  Novartis 
 NVP-BGT226  Novartis 
 P7170  Piramal 
 PF-05212384  Pfizer 
 PF-4691502  Pfizer 
 XL765 SAR245409 Exelixis 

Experimental evidence from other tumor types supports that different ways of activating the PI3K–AKT pathway result in functional dependence upon different effectors, and thus sensitivity to different classes of therapeutic agents. Melanomas with loss of PTEN represent a high-priority opportunity, due to the high prevalence of this alteration de novo, the availability of models for functional testing, and the evidence for a role in resistance to MAPK pathway inhibitors. Previous studies in multiple tumor types showed that loss of PTEN correlates with marked dependence on AKT and sensitivity to AKT inhibition in gene knockdown experiments (13). However, recently reported experiments using the BRAF-mutant, PTEN-null melanoma GEMM suggest superior in vivo tumor growth inhibition with PI3K inhibitors than with AKT inhibitors (52, 53). Although these results are interesting, it remains unclear how well this model will reflect results in BRAF-mutant, PTEN-null melanomas in patients, which will likely have significant heterogeneity and additional molecular alterations that cannot be modeled easily in GEMM systems. Testing of dual PI3K–mTOR inhibitors in melanoma cell lines has shown that these agents are broadly inhibitory, and often superior to the inhibition achieved by PI3K or mTOR inhibition alone (54, 55). Although improved antitumor activity is preferred, a key question is whether this will translate into an acceptable therapeutic index in patients due to the broad physiologic functions of PI3K and mTOR. Recently reported experiments in other models have suggested that the efficacy of AKT inhibitors is relatively selective for tumors with PTEN loss (56). It is possible that this selectivity for cells with loss of PTEN will translate into selective killing of tumor cells in patients with AKT inhibitors at clinically tolerated doses, even if they are less potent. Rapamycin and its analogues, which inhibit mTORC1, are reasonably well tolerated clinically, as shown by long-standing use in patients who have undergone organ transplantation. However, mTORC1 inhibitors have not shown significant clinical activity as single agents in metastatic melanoma patients, or in combination with RAF inhibitors (57–59). As discussed below, this lack of activity may be due to compensatory hyperactivation of AKT due to inhibition of an mTORC1-mediated negative feedback loop within the PI3K pathway. In contrast, dual mTORC1/2 inhibitors block this upregulation through the additional blockade of mTORC2-mediated phosphorylation/activation of AKT, and thus may represent a more effective strategy to test the effects of mTOR inhibition (29). A recent study has also shown that genetic inhibition of PDK1 can induce melanoma regression (60), supporting the rationale for testing of PDK1 inhibitors in melanoma as they are developed clinically.

One strategy to achieve significant pathway inhibition clinically with an acceptable therapeutic index is the use of isoform-specific PI3K inhibitors. Genetic studies in mouse models have shown that the PI3K catalytic subunit p110α is predominantly responsible for mediating growth factor signaling from RTKs, but it is largely dispensable for pathway activation in tumors with PTEN loss. Cells with PTEN loss instead seem to depend largely on p110β to activate the pathway, drive proliferation, and mediate tumorigenesis in vivo (61, 62). Testing of a p110β-selective inhibitor in a panel of >400 cancer cell lines showed significantly greater activity in lines with loss of PTEN than in those with PTEN intact (63). However, despite the overall trend, some PTEN-intact cell lines were sensitive, and a number of PTEN-null cell lines were resistant. Clinical testing of two different p110β-selective inhibitors (GSK2636771, SAR260301) is currently ongoing, with planned analysis of PTEN built into both studies (www.clinicaltrials.gov). Other PI3K isoform-specific inhibitors, particularly BYL719 (p110α) and CAL-101 (p110δ), have been well tolerated and showed clinical efficacy in other cancer types (64). The use of a p110α-selective inhibitor may be a rational approach, in particular, for tumors with PI3K–AKT pathway activation mediated by RTKs, but to date there are no published experimental data testing this hypothesis in melanoma.

Another strategy to optimize the therapeutic index of PI3K–AKT pathway inhibitors is the use of alternative dosing schedules. Multiple studies have shown that induction of apoptosis by BRAF or MEK inhibitors in melanoma cell lines with activating BRAF mutations generally is not observed until the MAPK pathway has been suppressed for 48 to 72 hours (29, 65). In contrast, when PI3K pathway inhibitors are combined with those agents not only is apoptosis increased but it is also generally induced at much earlier timepoints (i.e., 24 hours or less; refs. 29, 55). This suggests that relatively short-term exposure to PI3K–AKT pathway inhibitors may be effective clinically. This strategy is similar to that used conventionally with chemotherapy agents, in which dosing regimens have been developed to deliver the maximally tolerated doses of agents intermittently (i.e., every 7, 14, or 21 days). The clinical development of targeted therapies instead has generally used continuous dosing regimens. As one of the concerns about the clinical development of PI3K–AKT pathway inhibitors has been whether sufficient pathway inhibition is being achieved, the use of high, intermittent dosing may overcome this hurdle. Indeed, intermittent dosing of the combination of a MEK and a PI3K inhibitor exhibited marked antitumor activity in vivo in multiple xenograft models, including melanoma (66). Although this strategy can be explored empirically in mouse models, one of the critical challenges to the rational development of this strategy is the identification of pharmacodynamic markers that correlate with the achievement of clinically effective pathway inhibition by PI3K–AKT inhibitors (67). Notably, PI3K inhibitors generally produce marked inhibition of AKT activation at doses that are much lower than those that correlate with antiproliferative and/or proapoptotic effects. The identification of targets, and/or the degree of target modulation, that will correspond to clinical benefit, similar to what has been shown for P-ERK and BRAF inhibitors (68), will facilitate the preclinical development and clinical evaluation of candidate agents and dosing regimens.

Growing experience with effective targeted therapies, particularly in melanoma with selective BRAF inhibitors, has shown that compensatory signaling within and between signaling pathways can be critical to both clinical activity and the emergence of resistance (69–71). Consistent with this experience, effective clinical targeting of the PI3K–AKT pathway will likely also need to account for and overcome complex feedback loops that blunt the activity of single-target inhibitors against it. The seminal example is the feedback induction of AKT phosphorylation by mTORC1 inhibitors, such as rapamycin and RAD001 (Fig. 2). In these studies (72, 73), the mTORC1 complexes were found to negatively regulate IRS1 at baseline, a critical second messenger from the IGF1R to PI3K. mTORC1 inhibitor–mediated relief of this negative loop activates PI3K, AKT, and sometimes ERK (74, 75), promoting cell survival (Fig. 2B). This particular feedback has been shown in a variety of cancers (72–76) including melanoma (28, 77).

Figure 2.

Feedback signaling following mTORC1 inhibition. A, the baseline status of the PI3K signaling cascade, indicating negative feedback from p70S6K to IRS1. B, inhibition of mTORC1 blocks the negative feedback loop, activating IRS1, and leading to PI3K and AKT activation. C, paradoxical activation of PI3K and AKT in the setting of mTORC1 inhibition can be overcome by dual PI3K–mTOR inhibitors, which also inhibit PI3K, or dual mTORC1/2 inhibitors, which block mTORC2-mediated AKT activation.

Figure 2.

Feedback signaling following mTORC1 inhibition. A, the baseline status of the PI3K signaling cascade, indicating negative feedback from p70S6K to IRS1. B, inhibition of mTORC1 blocks the negative feedback loop, activating IRS1, and leading to PI3K and AKT activation. C, paradoxical activation of PI3K and AKT in the setting of mTORC1 inhibition can be overcome by dual PI3K–mTOR inhibitors, which also inhibit PI3K, or dual mTORC1/2 inhibitors, which block mTORC2-mediated AKT activation.

Close modal

More complex feedback perturbations are generated by PI3K and AKT inhibitors (Fig. 3). In breast, lung, and prostate cancer cell lines, these inhibitors induced the FOXO-mediated transcription of multiple RTKs, most commonly HER3 and IGF1R [78–80 (Fig. 3B)]. Independent studies showed that these RTKs are capable of transducing signals to both the PI3K–AKT (78) and the RAS–RAF–MEK–ERK pathways (80), potentially reinforcing mutual oncogenic cross-talk. Only when these RTKs were targeted by RNA interference knockdown or by small-molecule inhibitors (lapatinib, NVP-AEW541) were these feedback activations extinguished. Indeed, combinations of the PI3K and RTK inhibitors displayed synergy in xenograft models (79–81), supporting their therapeutic value. However, one point of contention is whether dual mTORC1/2 inhibitors (also referred to as mTORC catalytic inhibitors) can induce RTKs, as some studies explicitly observe this (72, 80, 81) whereas others do not (78, 79), even in the same cell type. These varied results may be due to differences in whether the readout is mRNA or protein, as posttranslational modifications and/or protein turnover rates can result in discordant levels (82, 83), or whether the inhibitor hits both the mTORC1 and mTORC2 complexes. Regardless, these overall results serve as an important caution for the future development of PI3K inhibition in melanoma and reveal potential cotargets to suppress the feedback activity (Figs. 2 and 3).

Figure 3.

Feedback signaling following PI3K, AKT, or dual mTORC1/2 inhibition. A, the baseline status of the PI3K signaling cascade, indicating negative feedback to RTKs such as HER3 and IGF1R, via inactivation of the FOXO transcription factors by AKT. B, PI3K, AKT, or dual mTORC1/2 inhibitor inactivate AKT, releasing the inhibition of FOXO transcription factors, leading to expression and activation of HER3, IGF1R, and other RTKs, subsequent activation of PI3K and AKT activation, and potentially other pathways (i.e., RAS–RAF–MEK–ERK). This effect is delayed in vitro by 24 to 72 hours or more and represents a re-equilibration of the pathway over time. C, the addition of RTK inhibitors can block the compensatory signaling and induce synergy with PI3K, AKT, and/or dual mTORC1/2 inhibitors.

Figure 3.

Feedback signaling following PI3K, AKT, or dual mTORC1/2 inhibition. A, the baseline status of the PI3K signaling cascade, indicating negative feedback to RTKs such as HER3 and IGF1R, via inactivation of the FOXO transcription factors by AKT. B, PI3K, AKT, or dual mTORC1/2 inhibitor inactivate AKT, releasing the inhibition of FOXO transcription factors, leading to expression and activation of HER3, IGF1R, and other RTKs, subsequent activation of PI3K and AKT activation, and potentially other pathways (i.e., RAS–RAF–MEK–ERK). This effect is delayed in vitro by 24 to 72 hours or more and represents a re-equilibration of the pathway over time. C, the addition of RTK inhibitors can block the compensatory signaling and induce synergy with PI3K, AKT, and/or dual mTORC1/2 inhibitors.

Close modal

Dual PI3K–mTOR inhibitors (84) improve on these single-target agents by providing a built-in inhibition of the mTOR feedback loops. Characterization of multiple dual inhibitors in melanoma cell lines has shown a potent and durable extinction of pAKT and its downstream targets, matching or even exceeding the effects of combining single PI3K and mTOR inhibitors (54, 77). Indeed, BEZ235 has shown preliminary success in various preclinical models, particularly in combination with MEK inhibitors (77, 85, 86). Relevantly, in a mouse model of melanoma, the combination of BEZ235 with the MEK inhibitor AZD6244 produced a 37% partial response rate (>30% decrease in tumor volume; ref. 87). Various clinical trials are currently in progress with this class of drugs, although whether pharmacokinetic and toxicity issues can be optimized remains to be seen (88).

The existence of these and other feedback loops suggests that pharmacodynamic, mechanistic, and resistance tissue-based studies of PI3K–AKT pathway inhibitors in patients should optimally allow for the evaluation of multiple markers and/or pathways. Emerging proteomic technologies including phospho-RTK and reverse phase protein arrays (RPPA) facilitate such analyses by analyzing a large number of proteins in individual samples concurrently. Notably, although most experimental work to date examining markers and mechanisms of efficacy and resistance with PI3K–AKT pathway inhibitors has focused on the effects and changes observed in tumor cells, it is becoming clear that anticancer treatments also have marked effects on the host, including the immune system and the tumor microenvironment. In melanoma, the durable efficacy of immunotherapies (89) and a growing appreciation of the effects of MAPK-pathway inhibitors on the antitumor response (90–93) mandate examination of the immunologic effects of PI3K–AKT pathway inhibitors in this disease. The marked activity of p110δ-selective inhibitors in hematologic malignancies, and the long-standing use of mTORC1 inhibitors (rapamycin) as immunosuppressants in transplant patients, raises the possibility that strategies that target the PI3K–AKT pathway could actually inhibit the antitumor immune response and thus blunt long-term clinical benefit. However, an improved understanding of antitumor immunology and the differential effects of various PI3K–AKT pathway inhibitors on different immune cell populations (94), coupled with strategies (i.e., isoform-specific inhibitors) that are designed to achieve selective pathway inhibition in tumors, suggests that this challenge will not be insurmountable.

The PI3K–AKT pathway remains an attractive combinatorial target to improve clinical outcomes in patients with melanoma. As described, emerging understanding and models for this pathway are facilitating the development of rational strategies. However, critical challenges remain, including matching patients to the appropriate agents; developing appropriate markers to facilitate efficient and meaningful evaluation of doses that are achieved safely in patients; and, ultimately, identifying strategies that achieve acceptable therapeutic indices.

M.A. Davies has commercial research grants from GlaxoSmithKline, AstraZeneca, Genentech, Merck, Oncothyreon, and Myriad. Also, M.A. Davies is a consultant/advisory board member for GlaxoSmithKline, Genentech, and Novartis. No potential conflicts of interest were disclosed by the other author.

Conception and design: L. Kwong, M.A. Davies

Writing, review, and/or revision of the manuscript: L. Kwong, M.A. Davies

M.A. Davies is supported by NIH 1R01CA154710-01, a Melanoma Research Alliance Young Investigator Award, an American Society of Clinical Oncology Career Development Award, and Cancer Prevention Institute of Texas RP120505.

1.
Yuan
TL
,
Cantley
LC
. 
PI3K pathway alterations in cancer: variations on a theme
.
Oncogene
2008
;
27
:
5497
510
.
2.
Hennessy
BT
,
Smith
DL
,
Ram
PT
,
Lu
Y
,
Mills
GB
. 
Exploiting the PI3K/AKT pathway for cancer drug discovery
.
Nat Rev Drug Discov
2005
;
4
:
988
1004
.
3.
Courtney
KD
,
Corcoran
RB
,
Engelman
JA
. 
The PI3K pathway as drug target in human cancer
.
J Clin Oncol
2010
;
28
:
1075
0183
.
4.
Hocker
T
,
Tsao
H
. 
Ultraviolet radiation and melanoma: a systematic review and analysis of reported sequence variants
.
Hum Mutat
2007
;
28
:
578
88
.
5.
Cantley
LC
. 
The phosphoinositide 3-kinase pathway
.
Science
2002
;
296
:
1655
7
.
6.
Maehama
T
,
Dixon
JE
. 
The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate
.
J Biol Chem
1998
;
273
:
13375
8
.
7.
Vasudevan
KM
,
Garraway
LA
. 
AKT signaling in physiology and disease
.
Curr Top Microbiol Immunol
2010
;
347
:
105
33
.
8.
Goel
VK
,
Lazar
AJ
,
Warneke
CL
,
Redston
MS
,
Haluska
FG
. 
Examination of mutations in BRAF, NRAS, and PTEN in primary cutaneous melanoma
.
J Invest Dermatol
2006
;
126
:
154
60
.
9.
Tsao
H
,
Goel
V
,
Wu
H
,
Yang
G
,
Haluska
FG
. 
Genetic interaction between NRAS and BRAF mutations and PTEN//MMAC1 inactivation in melanoma
.
J Invest Dermatol
2004
;
122
:
337
41
.
10.
Tsao
H
,
Zhang
X
,
Fowlkes
K
,
Haluska
FG
. 
Relative reciprocity of NRAS and PTEN/MMAC1 alterations in cutaneous melanoma cell lines
.
Cancer Res
2000
;
60
:
1800
4
.
11.
Davies
MA
,
Stemke-Hale
K
,
Lin
E
,
Tellez
C
,
Deng
W
,
Gopal
YN
, et al
Integrated molecular and clinical analysis of AKT activation in metastatic melanoma
.
Clin Cancer Res
2009
;
15
:
7538
46
.
12.
Park
ES
,
Rabinovsky
R
,
Carey
M
,
Hennessy
BT
,
Agarwal
R
,
Liu
W
, et al
Integrative analysis of proteomic signatures, mutations, and drug responsiveness in the NCI 60 cancer cell line set
.
Mol Cancer Ther
2010
;
9
:
257
67
.
13.
Vasudevan
KM
,
Barbie
DA
,
Davies
MA
,
Rabinovsky
R
,
McNear
CJ
,
Kim
JJ
, et al
AKT-independent signaling downstream of oncogenic PIK3CA mutations in human cancer
.
Cancer Cell
2009
;
16
:
21
32
.
14.
Nogueira
C
,
Kim
KH
,
Sung
H
,
Paraiso
KHT
,
Dannenberg
JH
,
Bosenberg
M
, et al
Cooperative interactions of PTEN deficiency and RAS activation in melanoma metastasis
.
Oncogene
2010
;
29
:
6222
32
.
15.
Hodis
E
,
Watson Ian
R
,
Kryukov
Gregory V
,
Arold
Stefan T
,
Imielinski
M
,
Theurillat
J-P
, et al
A landscape of driver mutations in melanoma
.
Cell
2012
;
150
:
251
63
.
16.
Krauthammer
M
,
Kong
Y
,
Ha
BH
,
Evans
P
,
Bacchiocchi
A
,
McCusker
JP
, et al
Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma
.
Nat Genet
2012
;
44
:
1006
14
.
17.
Aguissa-Toure
AH
,
Li
G
. 
Genetic alterations of PTEN in human melanoma
.
Cell Mol Life Sci
2012
;
69
:
1475
91
.
18.
Zhou
X-P
,
Gimm
O
,
Hampel
H
,
Niemann
T
,
Walker
MJ
,
Eng
C
. 
Epigenetic PTEN silencing in malignant melanomas without PTEN mutation
.
Am J Pathol
2000
;
157
:
1123
8
.
19.
Poliseno
L
,
Salmena
L
,
Zhang
J
,
Carver
B
,
Haveman
WJ
,
Pandolfi
PP
. 
A coding-independent function of gene and pseudogene mRNAs regulates tumour biology
.
Nature
2010
;
465
:
1033
8
.
20.
Karreth
Florian A
,
Tay
Y
,
Perna
D
,
Ala
U
,
Tan
Shen M
,
Rust
Alistair G
, et al
In vivo identification of tumor- suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma
.
Cell
2011
;
147
:
382
95
.
21.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
2012
;
483
:
603
307
.
22.
Stark
M
,
Hayward
N
. 
Genome-wide loss of heterozygosity and copy number analysis in melanoma using high-density single-nucleotide polymorphism arrays
.
Cancer Res
2007
;
67
:
2632
42
.
23.
Haluska
FG
,
Tsao
H
,
Wu
H
,
Haluska
FS
,
Lazar
A
,
Goel
V
. 
Genetic alterations in signaling pathways in melanoma
.
Clin Cancer Res
2006
;
12
:
2301s
7s
.
24.
Daniotti
M
,
Oggionni
M
,
Ranzani
T
,
Vallacchi
V
,
Campi
V
,
Di Stasi
D
, et al
BRAF alterations are associated with complex mutational profiles in malignant melanoma
.
Oncogene
2004
;
23
:
5968
77
.
25.
Dankort
D
,
Curley
DP
,
Cartlidge
RA
,
Nelson
B
,
Karnezis
AN
,
Damsky
WE
 Jr
, et al
BRAF(V600E) cooperates with PTEN loss to induce metastatic melanoma
.
Nat Genet
2009
;
41
:
544
52
.
26.
Xing
F
,
Persaud
Y
,
Pratilas
CA
,
Taylor
BS
,
Janakiraman
M
,
She
QB
, et al
Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF
.
Oncogene
2012
;
31
:
248
58
.
27.
Paraiso
KH
,
Xiang
Y
,
Rebecca
VW
,
Abel
EV
,
Chen
A
,
Munko
AC
, et al
PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression
.
Cancer Res
2011
;
71
:
2750
60
.
28.
Deng
W
,
Yennu-Nanda
VG
,
Scott
A
,
Chen
G
,
Woodman
SE
,
Davies
MA
. 
Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition
.
Pigment Cell Melanoma Res
2012
;
25
:
248
58
.
29.
Gopal
YN
,
Deng
W
,
Woodman
SE
,
Komurov
K
,
Ram
P
,
Smith
PD
, et al
Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in BRAF-mutant human cutaneous melanoma cells
.
Cancer Res
2010
;
70
:
8736
47
.
30.
Nathanson
K
,
Martin
A
,
Letrero
R
,
D'Andrea
K
,
O'Day
S
,
Infante
JR
, et al
Tumor genetic analyses of patients with metastatic melanoma treated with the BRAF inhibitor GSK2118436 (GSK436)
.
J Clin Oncol
2011
;
29
:
abst 8501
.
31.
Sosman
JA
,
Pavlick
AC
,
Schuchter
LM
,
Lewis
KD
,
McArthur
GA
,
Cowey
CL
, et al
Analysis of molecular mechanisms of response and resistance to vemurafenib (vem) in BRAF V600E melanoma
.
J Clin Oncol
2012
;
30
:
8503
.
32.
Niessner
H
,
Forschner
A
,
Klumpp
B
,
Honegger
JB
,
Witte
M
,
Bornemann
A
, et al
Targeting hyperactivation of the AKT survival pathway to overcome therapy resistance of melanoma brain metastases
.
Cancer Med
2013
;
2
:
76
85
.
33.
Curtin
JA
,
Stark
MS
,
Pinkel
D
,
Hayward
NK
,
Bastian
BC
. 
PI3-kinase subunits are infrequent somatic targets in melanoma
.
J Invest Dermatol
2006
;
126
:
1660
3
.
34.
Omholt
K
,
Krockel
D
,
Ringborg
U
,
Hansson
J
. 
Mutations of PIK3CA are rare in cutaneous melanoma
.
Melanoma Res
2006
;
16
:
197
200
.
35.
Carpten
JD
,
Faber
AL
,
Horn
C
,
Donoho
GP
,
Briggs
SL
,
Robbins
CM
, et al
A transforming mutation in the pleckstrin homology domain of AKT1 in cancer
.
Nature
2007
;
448
:
439
44
.
36.
Davies
MA
,
Stemke-Hale
K
,
Tellez
C
,
Calderone
TL
,
Deng
W
,
Prieto
VG
, et al
A novel AKT3 mutation in melanoma tumours and cell lines
.
Br J Cancer
2008
;
99
:
1265
8
.
37.
Stahl
JM
,
Sharma
A
,
Cheung
M
,
Zimmerman
M
,
Cheng
JQ
,
Bosenberg
MW
, et al
Deregulated Akt3 activity promotes development of malignant melanoma
.
Cancer Res
2004
;
64
:
7002
10
.
38.
Cheung
M
,
Sharma
A
,
Madhunapantula
SV
,
Robertson
GP
. 
Akt3 and mutant V600EB-raf cooperate to promote early melanoma development
.
Cancer Res
2008
;
68
:
3429
39
.
39.
Singh
RS
,
Diwan
AH
,
Zhang
PS
,
Prieto
VG
. 
Phosphoinositide 3-kinase is not overexpressed in melanocytic lesions
.
J Cutan Pathol
2007
;
34
:
220
5
.
40.
Pollock
PM
,
Harper
UL
,
Hansen
KS
,
Yudt
LM
,
Stark
M
,
Robbins
CM
, et al
High frequency of BRAF mutations in nevi
.
Nat Genet
2003
;
33
:
19
20
.
41.
Kantrow
SM
,
Boyd
AS
,
Ellis
DL
,
Nanney
LB
,
Richmond
A
,
Shyr
Y
, et al
Expression of activated Akt in benign nevi, Spitz nevi and melanomas
.
J Cutan Pathol
2007
;
34
:
593
6
.
42.
Curtin
JA
,
Busam
K
,
Pinkel
D
,
Bastian
BC
. 
Somatic activation of KIT in distinct subtypes of melanoma
.
J Clin Oncol
2006
;
24
:
4340
6
.
43.
Liang
R
,
Wallace
AR
,
Schadendorf
D
,
Rubin
BP
. 
The phosphatidyl inositol 3-kinase pathway is central to the pathogenesis of Kit-activated melanoma
.
Pigment Cell Melanoma Res
2011
;
24
:
714
23
.
44.
Monsel
G
,
Ortonne
N
,
Bagot
M
,
Bensussan
A
,
Dumaz
N
. 
c-Kit mutants require hypoxia-inducible factor 1[alpha] to transform melanocytes
.
Oncogene
2009
;
29
:
227
36
.
45.
Todd
JR
,
Becker
TM
,
Kefford
RF
,
Rizos
H
. 
Secondary c-Kit mutations confer acquired resistance to RTK inhibitors in c-Kit mutant melanoma cells
.
Pigment Cell Melanoma Res
2013
;
26
:
518
26
.
46.
Prickett
TD
,
Agrawal
NS
,
Wei
X
,
Yates
KE
,
Lin
JC
,
Wunderlich
JR
, et al
Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4
.
Nat Genet
2009
;
41
:
1127
32
.
47.
Nazarian
R
,
Shi
H
,
Wang
Q
,
Kong
X
,
Koya
RC
,
Lee
H
, et al
Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation
.
Nature
2010
;
468
:
973
7
.
48.
Villanueva
J
,
Vultur
A
,
Lee
JT
,
Somasundaram
R
,
Fukunaga-Kalabis
M
,
Cipolla
AK
, et al
Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K
.
Cancer Cell
2010
;
18
:
683
95
.
49.
Shi
H
,
Kong
X
,
Ribas
A
,
Lo
RS
. 
Combinatorial treatments that overcome PDGFRβ-driven resistance of melanoma cells to V600EB-RAF inhibition
.
Cancer Res
2011
;
71
:
5067
74
.
50.
Straussman
R
,
Morikawa
T
,
Shee
K
,
Barzily-Rokni
M
,
Qian
ZR
,
Du
J
, et al
Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion
.
Nature
2012
;
487
:
500
4
.
51.
Wilson
TR
,
Fridlyand
J
,
Yan
Y
,
Penuel
E
,
Burton
L
,
Chan
E
, et al
Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors
.
Nature
2012
;
487
:
505
9
.
52.
Marsh
V
,
Silva
J
,
Bosenberg
M
,
Phillips
W
,
McMahon
M
. 
Elucidating the role of the PI3-kinase pathway activation in melanoma progression in vivo
.
Pigment Cell Melanoma Res
2011
;
24
:
1000
.
53.
McMahon
M
,
Thakur
MD
,
Marsh
V
,
Silva
J
,
Landman
AS
,
Deuker
M
, et al
Targeting BRAF and PI3′-kinase signaling for therapy of melanoma
. In:
Proceedings of the Annual Meeting of the American Association for Cancer Research
;
Washington, DC
; 
2013
. p.
SY17-03
.
54.
Aziz
SA
,
Jilaveanu
LB
,
Zito
C
,
Camp
RL
,
Rimm
DL
,
Conrad
P
, et al
Vertical targeting of the phosphatidylinositol-3 kinase pathway as a strategy for treating melanoma
.
Clin Cancer Res
2010
;
16
:
6029
39
.
55.
Deng
W
,
Vashisht
Gopal YN
,
Scott
A
,
Chen
G
,
Woodman
SE
,
Davies
MA
. 
Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition
.
Pigment Cell Melanoma Res
2012
;
25
:
248
58
.
56.
Lin
J
,
Sampath
D
,
Nannini
MA
,
Lee
BB
,
Degtyarev
M
,
Oeh
J
, et al
Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models
.
Clin Cancer Res
2013
;
19
:
1760
72
.
57.
Margolin
K
,
Longmate
J
,
Baratta
T
,
Synold
T
,
Christensen
S
,
Weber
J
, et al
CCI-779 in metastatic melanoma: a phase II trial of the California Cancer Consortium
.
Cancer
2005
;
104
:
1045
8
.
58.
Davies
MA
,
Fox
PS
,
Papadopoulos
NE
,
Bedikian
AY
,
Hwu
W-J
,
Lazar
AJ
, et al
Phase I study of the combination of sorafenib and temsirolimus in patients with metastatic melanoma
.
Clin Cancer Res
2012
;
18
:
1120
8
.
59.
Margolin
KA
,
Moon
J
,
Flaherty
LE
,
Lao
CD
,
Akerley
WL
 3rd
,
Othus
M
, et al
Randomized phase II trial of sorafenib with temsirolimus or tipifarnib in untreated metastatic melanoma (S0438)
.
Clin Cancer Res
2012
;
18
:
1129
37
.
60.
Kaplon
J
,
Zheng
L
,
Meissl
K
,
Chaneton
B
,
Selivanov
VA
,
Mackay
G
, et al
A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence
.
Nature
2013
;
498
:
109
12
.
61.
Jia
S
,
Liu
Z
,
Zhang
S
,
Liu
P
,
Zhang
L
,
Lee
SH
, et al
Essential roles of PI(3)K-p110[bgr] in cell growth, metabolism and tumorigenesis
.
Nature
2008
;
454
:
776
9
.
62.
Wee
S
,
Wiederschain
D
,
Maira
S-M
,
Loo
A
,
Miller
C
,
deBeaumont
R
, et al
PTEN-deficient cancers depend on PIK3CB
.
Proc Natl Acad Sci
2008
;
105
:
13057
62
.
63.
Ni
J
,
Liu
Q
,
Xie
S
,
Carlson
C
,
Von
T
,
Vogel
K
, et al
Functional characterization of an isoform-selective inhibitor of PI3K-p110β as a potential anticancer agent
.
Cancer Discov
2012
;
2
:
425
33
.
64.
Hoellenriegel
J
,
Meadows
SA
,
Sivina
M
,
Wierda
WG
,
Kantarjian
H
,
Keating
MJ
, et al
The phosphoinositide 3′-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia
.
Blood
2011
;
118
:
3603
12
.
65.
Paraiso
KHT
,
Fedorenko
IV
,
Cantini
LP
,
Munko
AC
,
Hall
M
,
Sondak
VK
, et al
Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy
.
Br J Cancer
2010
;
102
:
1724
30
.
66.
Hoeflich
KP
,
Merchant
M
,
Orr
C
,
Chan
J
,
Den Otter
D
,
Berry
L
, et al
Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition
.
Cancer Res
2012
;
72
:
210
9
.
67.
Andersen
JN
,
Sathyanarayanan
S
,
Di Bacco
A
,
Chi
A
,
Zhang
T
,
Chen
AH
, et al
Pathway-based identification of biomarkers for targeted therapeutics: personalized oncology with PI3K pathway inhibitors
.
Sci Transl Med
2010
;
2
:
43ra55
.
68.
Bollag
G
,
Hirth
P
,
Tsai
J
,
Zhang
J
,
Ibrahim
PN
,
Cho
H
, et al
Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma
.
Nature
2010
;
467
:
596
9
.
69.
Solit
DB
,
Rosen
N
. 
Resistance to BRAF inhibition in melanomas
.
N Engl J Med
2011
;
364
:
772
4
.
70.
Lu
Y
,
Muller
M
,
Smith
D
,
Dutta
B
,
Komurov
K
,
Iadevaia
S
, et al
Kinome siRNA-phosphoproteomic screen identifies networks regulating AKT signaling
.
Oncogene
2011
;
30
:
4567
77
.
71.
Aksamitiene
E
,
Kiyatkin
A
,
Kholodenko
BN
. 
Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance
.
Biochem Soc Trans
2012
;
40
:
139
46
.
72.
Shi
Y
,
Yan
H
,
Frost
P
,
Gera
J
,
Lichtenstein
A
. 
Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade
.
Mol Cancer Ther
2005
;
4
:
1533
40
.
73.
O'Reilly
KE
,
Rojo
F
,
She
QB
,
Solit
D
,
Mills
GB
,
Smith
D
, et al
mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt
.
Cancer Res
2006
;
66
:
1500
8
.
74.
Carracedo
A
,
Li
M
,
Teruya-Feldstein
J
,
Rojo
F
,
Salmena
L
,
Alimonti
A
, et al
Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer
.
J Clin Invest
2008
;118:
3065
74
.
75.
Soares
HP
,
Ni
Y
,
Kisfalvi
K
,
Sinnett-Smith
J
,
Rozengurt
E
. 
Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells
.
PLoS ONE
2013
;
8
:
e57289
.
76.
Wan
X
,
Harkavy
B
,
Shen
N
,
Grohar
P
,
Helman
LJ
. 
Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism
.
Oncogene
2006
;
26
:
1932
40
.
77.
Marone
R
,
Erhart
D
,
Mertz
AC
,
Bohnacker
T
,
Schnell
C
,
Cmiljanovic
V
, et al
Targeting melanoma with dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitors
.
Mol Cancer Res
2009
;
7
:
601
13
.
78.
Chakrabarty
A
,
Sánchez
V
,
Kuba
MG
,
Rinehart
C
,
Arteaga
CL
. 
Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors
.
Proc Natl Acad Sci
2012
;
109
:
2718
23
.
79.
Chandarlapaty
S
,
Sawai
A
,
Scaltriti
M
,
Rodrik-Outmezguine
V
,
Grbovic-Huezo
O
,
Serra
V
, et al
AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity
.
Cancer Cell
2011
;
19
:
58
71
.
80.
Serra
V
,
Scaltriti
M
,
Prudkin
L
,
Eichhorn
PJA
,
Ibrahim
YH
,
Chandarlapaty
S
, et al
PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer
.
Oncogene
2011
;
30
:
2547
57
.
81.
Rodrik-Outmezguine
VS
,
Chandarlapaty
S
,
Pagano
NC
,
Poulikakos
PI
,
Scaltriti
M
,
Moskatel
E
, et al
mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling
.
Cancer Discov
2011
;
1
:
248
59
.
82.
Shankavaram
UT
,
Reinhold
WC
,
Nishizuka
S
,
Major
S
,
Morita
D
,
Chary
KK
, et al
Transcript and protein expression profiles of the NCI-60 cancer cell panel: an integromic microarray study
.
Mol Cancer Ther
2007
;
6
:
820
32
.
83.
Varambally
S
,
Yu
J
,
Laxman
B
,
Rhodes
DR
,
Mehra
R
,
Tomlins
SA
, et al
Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression
.
Cancer Cell
2005
;
8
:
393
406
.
84.
Maira
S-M
,
Stauffer
F
,
Brueggen
J
,
Furet
P
,
Schnell
C
,
Fritsch
C
, et al
Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity
.
Mol Cancer Ther
2008
;
7
:
1851
63
.
85.
Engelman
JA
,
Chen
L
,
Tan
X
,
Crosby
K
,
Guimaraes
AR
,
Upadhyay
R
, et al
Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers
.
Nat Med
2008
;
14
:
1351
6
.
86.
Migliardi
G
,
Sassi
F
,
Torti
D
,
Galimi
F
,
Zanella
ER
,
Buscarino
M
, et al
Inhibition of MEK and PI3K/mTOR suppresses tumor growth but does not cause tumor regression in patient-derived xenografts of RAS-mutant colorectal carcinomas
.
Clin Cancer Res
2012
;
18
:
2515
25
.
87.
Roberts
PJ
,
Usary
JE
,
Darr
DB
,
Dillon
PM
,
Pfefferle
AD
,
Whittle
MC
, et al
Combined PI3K/mTOR and MEK inhibition provides broad antitumor activity in faithful murine cancer models
.
Clin Cancer Res
2012
;
18
:
5290
303
.
88.
Shimizu
T
,
Tolcher
AW
,
Papadopoulos
KP
,
Beeram
M
,
Rasco
DW
,
Smith
LS
, et al
The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer
.
Clin Cancer Res
2012
;
18
:
2316
25
.
89.
Ott
PA
,
Hodi
FS
,
Robert
C
. 
CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients
.
Clin Cancer Res
2013
;
19
:
5300
9
.
90.
Boni
A
,
Cogdill
AP
,
Dang
P
,
Udayakumar
D
,
Njauw
C-NJ
,
Sloss
CM
, et al
Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function
.
Cancer Res
2010
;
70
:
5213
9
.
91.
Frederick
DT
,
Piris
A
,
Cogdill
AP
,
Cooper
ZA
,
Lezcano
C
,
Ferrone
CR
, 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
.
92.
Wilmott
JS
,
Long
GV
,
Howle
JR
,
Haydu
LE
,
Sharma
RN
,
Thompson
JF
, et al
Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma
.
Clin Cancer Res
2012
;
18
:
1386
94
.
93.
Kwong
MM
,
Neyns
B
,
Yang
JC
. 
Adoptive T-cell transfer therapy and oncogene targeted therapy for melanoma: the search for synergy
.
Clin Cancer Res
2013
;
19
:
5292
9
.
94.
Janes
MR
,
Limon
JJ
,
So
L
,
Chen
J
,
Lim
RJ
,
Chavez
MA
, et al
Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor
.
Nat Med
2010
;
16
:
205
13
.