Abstract
With the FDA approval of six novel targeted agents since December 2005 and limited comparative trials to discern relative efficacy, the treatment of metastatic renal cell carcinoma (RCC) has become immensely complex. The research community must look to novel ways in which to identify appropriate candidates for selected targeted therapies; one potential strategy is the use of clinical and molecular biomarkers. A growing body of knowledge related VHL-driven pathways in this disease has highlighted the potential role of hypoxia-inducible factor (HIF) subtypes in distinguishing RCC patients clinically. Techniques applied in other malignancies, such as gene expression and proteomic profiling, may also ultimately allow for clinical stratification. An emerging understanding of immunologic phenomena that may affect cancer progression (i.e., tumor infiltration by CD68 lymphocytes, memory T-cells, etc.) has unveiled a number of other potential biomarkers of response. Several vascular endothelial growth factor (VEGF) receptor-directed therapies classically thought to function as antiangiogenics may also have complex effects upon the tumor microenvironment, including the associated immune cell milieu. As such, immunologic parameters could potentially predict response to current therapies. Finally, clinical biomarkers such as hypertension may predict the efficacy of several currently available targeted agents, although implementation of such biomarkers remains challenging. Herein, the clinical relevance of putative RCC biomarkers is examined in detail.
INTRODUCTION
Over the past decade, the clinical and scientific community has witnessed unprecedented developments in targeted therapies for metastatic renal cell carcinoma (mRCC). Whereas the oncologist was previously equipped with a limited number of immunotherapeutic agents such as interferon-α (IFN-α) and interleukin-2 (IL-2), a total of 6 targeted agents have been approved by the US Food and Drug Administration (FDA) within the past 5 years (1). These agents can be broadly divided into 2 categories: 1) agents that directly counteract vascular endothelial growth factor (VEGF)-mediated signaling, and 2) agents that act on the mammalian target of rapamycin (mTOR) pathway. Agents in the former category supported by randomized, phase III data include sunitinib, sorafenib, pazopanib and bevacizumab (2–6). Similarly, mTOR inhibitors supported by phase III data in mRCC include temsirolimus and everolimus (7, 8).
Oncologists are now charged with determining which targeted agent to select for the patient with mRCC. Limited comparative trials are available to juxtapose efficacy; thus, the decision is typically based on patient preference after a thorough discussion of benefits, adverse effects associated with each agent, and consideration of co-morbidities. Borrowing paradigms from other diseases, it is likely that targeted agents are more apt to work in circumstances in which the biological target is more abundant. For instance, in breast cancer, human epidermal growth factor 2 (HER2)-directed therapies have shown profound clinical benefit specifically in the subset of patients with HER2 overexpression (9–12). As a more recent example, the oral anaplastic lymphoma kinase (ALK) inhibitor crizotinib appears to have significant activity in patients with non-small cell lung cancer (NSCLC) who bear the EML4-ALK fusion oncogene (13). Similarly, certain patients with melanoma may be exquisitely sensitive to the agent PLX4032, targeting a frequently mutated form of the serine-threonine kinase B-RAF (BRAF V600E) (14). In a similar fashion, identification of relevant biomarkers in RCC may refine current algorithms for treatment selection. Herein, we review the current status of biomarkers in RCC in hopes of developing a framework for their use as prognostic and predictive tools.
HYPOXIA INDUCIBLE FACTOR-α (HIF-α) AND RELATED PATHWAYS
A key driver in RCC tumorigenesis is the von Hippel Lindau (VHL) gene mutation. VHL encodes a tumor suppressor protein (pVHL) with a molecular weight between 24–30 kDa (15). In its native form, pVHL typically forms multimeric complex with several other moieties (Elongin B, Elongin C, Cul2 and Rbx1) and binds to HIF-α in the setting of hypoxia (16–18). Purified pVHL appears to have ubiquitin ligase activity, directing HIF-α towards proteasomal degradation (19). Mutation or hypermethylation of VHL occurs in up to 80% of sporadic clear cell RCC (ccRCC) cases; these phenomena may interrupt formation of the pVHL complex and thereby stabilize HIF-α (20, 21). In addition, HIF-1α levels may also be upregulated by oncogenic signaling through signal transducer and activator of transcription 3 (STAT3), commonly activated in human tumors such as RCC (22, 23). The resultant effect is increased binding of HIF-α to hypoxia response elements, leading to transcription of HIF-target genes such as VEGF (24) or carbonic anhydrase IX (CAIX) (25). These observations offer mechanistic rationale for agents that target the VEGF-signaling axis in this disease.
Recognition of two subtypes of HIF-α (HIF-1α and HIF-2α) has led to the development of a potential biomarker for ccRCC. These two subunits can form complexes with a β subunit, allowing for subsequent binding to HIF response elements (HREs). HIF-1α and HIF-2α differ with respect to sites of expression. While the former is ubiquitously expressed, the latter is expressed principally in endothelium, heart, lungs, kidney and small intestine (26–28). In in vivo assays, only HIF-2α was able to overcome pVHL induced suppression (29, 30). This phenomenon may be a consequence of HIF-2α-mediated increases in c-myc activity, documented in recent in vitro studies (31).
A comprehensive analysis of 57 sporadic human ccRCC specimens by Gordan et al has elicited a potential classification schema on the basis of HIF subtype (32). In this cohort of patients, only 12% were VHL wild type (WT) with no detectable level of HIF-α. The remainder bore VHL mutations, and had either increased expression of both HIF-1α and HIF-2α (termed the H1H2 phenotype, 61%), or HIF-2α alone (termed the H2 phenotype, 27%). Consistent with previously noted observations, H2 tumors exhibited greater expression of c-myc-activated targets, such as cyclin D2 and E2F. Furthermore, H2 tumors exhibited increased expression of homologous recombination (HR) mediators BRCA1, BARD21 and XRCC2 (notably, HR allows DNA-damaged cells to avoid checkpoint activation).
The expression profile of H1H2 tumors appears to be markedly different than the other subtypes (31, 32). In H1H2 tumors, increased expression of the growth factor signaling molecules Akt2 and RhoC was observed. Furthermore, increased phospho-S6 and phospho-ERK was seen in both WT and H1H2 tumors. The findings of Gordan et al. thus suggest several new targets for ccRCC therapy. Furthermore, they suggest a potential stratification schema for determining the most appropriate candidates for specific therapies. Albeit speculative, increased Akt2 and phospho-S6 activation in H1H2 tumors may suggest a potential role for the mTOR inhibitors everolimus and temsirolimus, and perhaps a benefit from VEGF-TKIs which abrogate upstream signaling. Furthermore, the unique expression profile of H2 tumors may lead to an intrinsic resistance to VEGF-directed therapies – for this subset of patients, further assessment of agents that interrupt DNA repair pathways (i.e., PARP inhibitors) and c-myc-mediated signaling may be prudent.
GENE EXPRESSION PROFILING
Rini et al have recently presented the largest and most comprehensive gene profiling effort in RCC to date (33). Utilizing tissue specimens derived from a cohort of 931 patients with stage I-III ccRCC, RT-PCR was used to quantify RNA expression of 732 candidate genes. Clinical follow-up was available for a median of 5.6 years in this cohort, and Cox models were used to determine which genes were associated with recurrence-free interval (RFI). Notably, several clinicopathologic variables were associated with RFI, including clinical stage, Fuhrman grade, size and lymph node involvement (P<0.001 for each) (34). Of the 732 genes assessed, 448 genes were associated with RFI (P<0.05). On further multivariate analysis accounting for clinicopathologic covariates and false discovery rates, a total of 16 genes remained associated with RFI. Amongst these genes are several with a plausible relationship to RCC tumorigenesis – for instance, EMCN and NOS3 are associated with angiogenic pathways, while CCL5 and CXCL9 are associated with immune related pathways. The 16 candidate genes (along with 5 reference genes) will be evaluated in distinct validation cohorts.
Dalgliesh et al have utilized a broader sequencing approach in a smaller cohort of RCC patients to identify several moieties intricately linked to RCC biology (35). In 101 ccRCC specimens, the group sequenced 3,544 protein-encoding genes. A total of 5 genes with relevance to cancer development were identified that exhibited clustering of somatic mutations (SETD2, JARIDI1C, NF2, UTX, and MLL2). Subsequent focused screening of a larger cohort including 407 ccRCC specimens demosnrated inactivating mutations in the genes encoding SETD2 (a histone H3lysine 36 methyltransferase) and JARID1C (a histone H3 lysine 4 demethylase). These thorough experiments strongly implicate the role of these histone modifying genes in RCC pathogenesis. NF2 gene mutations were also found in secondary screening; importantly, mutations in this gene were found independently of VHL mutation or expression of a hypoxia expression phenotype. This suggests that NF2 mutation (associated with neurofibromatosis II and the formation of acoustic neuromas and meningiomas) may also define a distinct subset of ccRCC with unique pathogenesis.
The information rendered from traditional gene profiling techniques can be augmented through the additional knowledge provided by broad transcriptomic profiles. Cifola et al have performed a detailed analysis of 27 RCC samples (36). DNA copy number alterations (CNAs) and loss of heterozygosity (LOH) events were determined using single-nucleotide polymorphism (SNP) array technology. Outside of previously recognized areas of DNA alteration, the genome-wide map highlighted several new sites of deletion, amplification and LOH (36). A simultaneous assessment of the transcriptomic profile of these specimens suggested that 27 differentially expressed genes were present in DNA-amplified regions. For instance, the transcripts encoding lysyl oxidase (LOX, 5q) and chemokine C-X-C receptor 4 (CXCR4, 2q) were noted to be upregulated – notably, the associated regions of DNA were concomitantly amplified. Both LOX and CXCR4 may play a critical role in the metastatic potential of tumor, possibly promoting tumor cell trafficking into premetastatic niches (as suggested in murine studies) (37–40). Ultimately, paired genomic and transcriptomic profiling may render unique targets for RCC therapy, and may offer individualized prognostic information.
A greater understanding of epigenetic phenomena may also allow for distinct characterization of renal tumors. A panel of 18 cancer-related genes was assessed for methylation status in a pool of 85 resected RCC specimens and 62 paired normal tissue samples (41). Notably, the genes fell into one of several groups, including those involved in cellular communication, nucleic acid metabolism, signal transduction, energy regulation, and cell cycle progression. Altered methylation patterns were observed in CDH1 and RASSF1A, involved in cellular communication and cell cycle progression, respectively. Differences in methylation patterns were also noted between clear cell and non-clear cell RCC specimens – for instance, PTGS2 methylation (involved in metabolic pathways) was increased in clear cell as opposed to papillary RCC. In a similar but separate series of experiments, 38 specimens derived from nephrectomy in RCC patients (and paired normal tissue) were assessed for methylation of 19 genes that were noted to be underexpressed in RCC (42). A total of 5 genes were found to be hypermethylated, and amongst these were 2 genes encoding tumor suppressor proteins (TFAP2A and MT1G). These epigenetic phenomena may therefore highlight characteristic features of RCC specimens, and if validated in larger cohorts, could serve as critical prognostic or predictive tools.
Broader approaches across multiple cancer subtypes may also genetic alterations that more uniformly affect cancer growth and proliferation across histologies. As one example, Beroukhim et al have assessed 3,131 cancer specimens across 26 histological subtypes (including RCC) (43). Through high-resolution analyses of somatic copy-number alterations (SCNAs), it was found that BCL2-related genes (functioning in apoptosis regulation) and genes encoding moieties along the NF-κB signaling pathway were enriched. Two amplifications peaks were seen for the BCL2-related genes MCL1 and BCL2L1. Subsequent experiments comparing MCL1-amplified and non-amplified cell lines showed greater growth inhibition in amplified cell lines treated with MCL-1 shRNA. Similar results were yielded in experiments assessing BCL2L1 inhibition in several models.
PROTEOMIC PROFILING
In comparison to gene expression profiling, the amount of published data on renal tumor proteomic profiles is less abundant. Nonetheless, several emerging datasets suggest the potential utility of this tool. In one series of experiments, serum samples from 65 patients with renal cell carcinoma was compared to 34 patients with benign renal tumors and 69 normal controls (44). Using surface enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS), a total of 29 proteins with differential expression were identified between patients with RCC and normal controls. A lesser number of proteins—18—were identified that differed between RCC patients and those with benign tumors. Using a diagnostic decision tree based on the protein expression profile, this study suggested that SELDI-TOF-MS could separate RCC patients from normal controls with an appreciable sensitivity and specificity of 81.8% and 100%, respectively. A separate report from the same group suggested the utility of adding CT scans to proteomic evaluation, enhancing the diagnostic capability of the test (45).
The VHL-driven biology of RCC may ultimately be assessable through proteomic techniques. In a recent report, Aggelis et al utilized the VHL-defective UMRC2 RCC cell line (46). Cells were transfected with either vector control or VHL cDNA. Membrane proteins were enriched and assessed using SDS-PAGE coupled to liquid chromatography mass spectrometry (GeLC-MS); analysis showed 19 differentially expressed proteins. These proteins included several moieties relevant to VHL-dependent pathways, including transferrin receptor-1 and α3β1 integrin (47, 48). Other novel protein markers were defined, including CD166 (or Activated Leukocyte Cell Adhesion Molecule) and CD147 (or Extracellular Matrix Metalloproteinase Inducer), both of which were upregulated in VHL-defective cells. In a small cohort of matched patient specimens (n=8), CD166 expression was observed to be higher in tumor tissue as compared to normal tissue.
Pairing genomic and proteomic profiles may provide the most extensive pool of information for the individual patient, although combining these technologies is complex. In one representative study, malignant and matched normal tissue was obtained from 37 patients with RCC who had undergone nephrectomy (49). Standard 2D-PAGE proteomic techniques identified 334 differentially expressed proteins. In contrast, PROTEOMEX assessment (proteomic techniques paired with serologic testing to identify immunogenic biomarkers) identified 50 proteins, while transcription profiling identified 110 differentially transcribed genes. In combination, only three candidate biomarkers were elicited (ANXA4, tubulin α-1A chain and UCHL1). Nonetheless, with greater refinement of each technique and larger scale validation, combined genetic and proteomic profiling may represent a novel approach to characterizing patients with RCC. Larger scale validation of these techniques will improve the signal:noise ratio observed in these early studies. As one example of this, The Cancer Genome Atlas (TCGA) is a 5-year NIH funded effort currently devoting attention to comprehensive genomic assessment of 20 cancer types (50). Amongst the urologic tumors under study, the TCGA will focus on both conventional and papillary RCC.
IMMUNOLOGIC MARKERS
Tumors contain a number of immune cells which can positively or negatively influence their progression. Tumor infiltration by CD8 lymphocytes and especially memory T cells are considered a strong indicator of disease-free survival as shown by large cohort studies of various solid cancers including RCC (51, 52). In contrast, the presence of tumor-infiltrating regulatory T cells (Tregs) and myeloid cells, such as tumor-associated macrophages (TAMs) and myeloid derived suppressor cells (MDSCs), is known to be associated with tumor immune evasion and reduced survival (51, 53, 54). The elimination of immunosuppressive activity by antibody-mediated Tregs depletion or blockade of T cell-imparing molecules expressed on tumor cells, such as B7-H1 and B7-H4, are potential strategies for RCC therapy (55, 56). In addition, while the putative mechanism of most antiangiogenic agents is principally antagonism of the VEGF-signaling axis, there is an emerging understanding that these agents may also elicit their antitumor effect through complex immunologic phenomena (Figure 1). As one example, sunitinib has been shown to reduce recruitment of MDSCs and tumor-infiltrating Tregs at the site of tumor, thereby augmenting the antitumor immune response (57). The effect on MDSC recruitment appears to be mediated by direct inhibition of Stat3 in MDSCs by sunitinib.
Figure 1.

Agents such as sunitinib may act through a multitude of mechanisms, yielding numerous potential biomarkers of response.
Clinical support for these findings is provided by Finke et al (58). In a series of 42 patients with mRCC treated with sunitinib monotherapy, serial blood collections were performed on day 1 and day 28 of therapy. Sunitinib was dosed on a standard 4 week on, 2 week off schedule at a dose of 50 mg oral daily. In comparison to age-matched blood from normal donors, patients treated with sunitinib had an improved type-1 T-cell cytokine response (increased IFN-γ producing T-cells and reduced IL-4 production) with sunitinib therapy, but had a decrease in circulating Tregs. The prognostic value of Tregs in RCC has been established in a separate study. When assessed alongside clinicopathologic criteria in a 125 patients with RCC, multivariate analysis indicated that an increase in peritumoral Tregs along with higher TNM stage, higher tumor size and higher nuclear grade were each independent predictors for shorter overall survival (OS) and RFI. A predictive role of Tregs in the context of sunitinib therapy has yet to be established.
Other immune effector cells have been explored as predictors of sunitinib response. Myeloid differentiation was assessed in a cohort of 26 patients with advanced RCC treated with sunitinib therapy on a standard schedule, as in the previous study (59). With measurements at baseline, at 28 days and at 42 days in the first treatment cycle, it was observed that higher levels of CD1c+ myeloid dendritic cells (MDCs) predicted improved progression-free survival (PFS) with sunitinib. The effect of sunitinib on cells of the innate immune system has been assessed in a similar fashion; however, preliminary reports suggest no change in natural killer (NK) cell populations with sunitinib therapy (60).
Do the sunitinib-induced changes in immune phenotype represent a class effect amongst VEGF-directed therapies? Albeit limited, there is evidence to the contrary. One such study juxtaposed the activity of sorafenib and sunitinib in C57BL/6 mice immunized with OVA257–264 peptide (61). While the proportion of Tregs decreased over the duration of therapy with sunitinib, this phenomenon was not observed with sorafenib. Furthermore, sorafenib (unlike sunitinib) inhibited the ability of mice to mount an antigen-specific T-cell response. Thus, in moving forward, it will be important to discern the immunologic effect of each targeted agent independently. Monitoring these effects may have a prognostic or predictive role.
CLINICAL BIOMARKERS
Several prognostic schema (i.e, the UCLA Integrated Staging System (UISS) and Memorial Sloane Kettering Cancer Center (MSKCC) criteria) have been devised and validated in large cohorts of patients with both advanced and localized mRCC (62–69). However, these were largely formulated during the era of cytokine therapies. Given the rapid shift towards newer targeted therapies, a revised model has been suggested. Using clinical data from 645 patients with mRCC who received first-line with VEGF-directed agents, Heng et al employed Cox proportional hazards regression and bootstrap validation to identify independent prognostic factors for OS (70). Incorporating several of the original MSKCC criteria, the revised model adds platelet and neutrophil counts to establish favorable-, intermediate- and poor-risk groups.
Amassing data for targeted therapies has also led to several potential clinical biomarkers of response. Perhaps most intriguingly, hypertension appears to be strongly correlated with clinical outcome in the context of several VEGF-directed agents. Using pooled efficacy data for 544 and safety data for 4,541 patients treated with sunitinib, Kaplan-Meier methods were employed to compare survival in patients with and without hypertension (71). In this study, blood pressure measurements were mandated on days 1 and 28 of each 42 day cycle. Distinguishing groups of patients based on the maximally achieved blood pressure, those patients who achieved a maximum systolic blood pressure (SBP) ≥ 140 mmHg had a profound improvement in both PFS (12.5 v 2.5 months, P<0.0001) and OS (30.5 v 7.8 months, P<0.0001), as compared to patients who achieved a maximum SBP < 140 mmHg. A similar, albeit more subtle, difference in PFS and OS was observed using a diastolic blood pressure (DBP) threshold of 90 mmHg. Paralleling the data for sunitinib, Cancer and Leukemia Group B (CALGB) trial 90206 suggests the role of hypertension as a predictor of bevacizumab activity (6, 72). This study compared bevacizumab with IFN-α to IFN-α alone in patients with treatment-naïve mRCC. Patients treated with bevacizumab who incurred ≥ grade 2 hypertension had both an improved PFS (13.2 v 8.0 months, P<0.001) and OS (41.6 v 16.2 months, P<0.001). These data are akin to that for use of bevacizumab in advanced breast cancer (73).
Complicating the use of hypertension as a predictive tool is the post hoc nature of this biomarker. To this end, VEGF and VEGFR SNPs that predict the occurrence of hypertension have been proposed (74). In a cohort of 64 patients with clear cell mRCC receiving sunitinib, several candidate VEGF and VEGFR SNPs were assessed. Notably, VEGF SNP 2578 was associated with a higher frequency of sunitinib induced hypertension (C/C: 100%, A/C: 89%, A/A: 69%, P=0.03). Another manner in which to incorporate biomarkers such as hypertension is through dose titration. A trial of the novel VEGF-TKI axitinib is employing a schema that allows for modification of dosing to elicit hypertension (75). With these studies underway, attention should also be devoted to understanding the mechanism of hypertension in this setting.
Outside of hypertension, several other clinical predictors of VEGF-directed therapy have been proposed. Most recently, Chouieri et al presented a comprehensive analysis of 475 patients treated at US and Canadian centers with first-line VEGF-directed therapy (76). In this cohort, the impact of body-mass index (BMI) and body surface area (BSA) on survival was assessed. Interestingly, obese patients (BMI > 30 kg/m2) had a superior median OS as compared to non-obese patients (BMI ≤ 30 kg/m2; 32.5 v 20.6 months, P=0.0001). Importantly, these differences persisted even after multivariate analysis accounting for adapted MSKCC prognostic grouping.
EXAMINING THE VEGF-SIGNALING AXIS
Moieties along the VEGF-signaling axis have been examined in detail to determine any correlation with the activity of VEGF-directed agents and mTOR inhibitors. As described previously, VEGF SNPs has been correlated with the frequency of hypertension; the same studies seem to suggest that SNPs in VEGFR2 may predict clinical outcome (74). Specifically, VEGFR SNP genotypes 889 (G/A) and 1416 (A/A) had significantly improved OS as compared to other genotypes (P=0.03). DePrimo et al have characterized circulating levels of VEGF, VEGFR2 and VEGFR3 in 63 patients with mRCC receiving sunitinib after failure of prior cytokine therapy (77). Notably, larger changes in each of these moieties were observed in patients who achieved an objective tumor response as compared to patients who exhibited stable disease or progression (P<0.05 for each). In a similar fashion, Rini et al assessed patients receiving sunitinib for bevacizumab refractory mRCC (78). In a study of 62 patients receiving sunitinib on a standard schedule, VEGF-C and sVEGFR-3 levels decreased during the course of therapy, and lower baseline levels were associated with higher response rates and a longer PFS. A similar study by Porta et al points to the potential predictive role of the cumulative baseline VEGF titer (79). In a study including 85 patients treated with sunitinib therapy, an above-threshold VEGF titer was associated with a median PFS of 4.7 months (95%CI: 2.8–8.3), whereas a below-threshold titer was associated with a median PFS of 11.2 months (95%CI: 6.5–15).
Downstream of VEGFR, the serine-threonine kinase p70S6K and pAKT have been proposed as potential predictors of mTOR activity in RCC (80). It is important to recognize that outside of the VEGF-signaling axis, multiple other moieties have been suggested as either potential prognostic factors (Table 1) or predictive factors for currently existing therapies (Table 2). Although the majority constitute relatively small, hypothesis-generating efforts, several are derived from some of the largest randomized trials in RCC to date. For instance, correlative studies associated with the phase III study comparing sorafenib to placebo (n=903) have identified TIMP-3 as an independent prognostic factor in RCC, correlating with survival even after adjustment for VEGF-axis moieties and MSKCC risk criteria (81). In a similarly sized cohort of mRCC patients (n=716), the insulin-like growth factor-II (IGF-II) mRNA binding protein IMP-3 has been associated (on multivariate analysis) with the 5-year risk of distant metastasis and OS (82). Finally, studies accompanying the randomized discontinuation trial of pazopanib have identified several potential markers of drug activity and further outline a strategy for further biomarker assessment (21). Using a platform assessing various cytokines and angiogenic factors, Heymach et al have identified a potential association between lower baseline levels of HGF, IL-6 and IL-8 and increased tumor shrinkage in patients treated with pazopanib. It would be of interest to see if a similar platform could be utilized to predict the activity of other VEGF-TKIs, as well.
Table 1.
Studies examining potential molecular prognostic markers in RCC.
| Marker | Reference | Description |
|---|---|---|
| TIMP-3 | Pena et al (81) | In the phase III TARGET trial comparing sorafenib to placebo in largely cytokine-refractory patients, baseline blood was assessed for VEGF, sVEGFR-2, CAIX, TIMP-1, Ras p21 and VHL mutation. In a multivariate analysis that included these biomarkers and MSKCC risk criteria, only TIMP-1 remained prognostic for OS (P=0.002). |
| S100A4 | Bandiera et al(98) | In a series of 32 primary tumor specimens derived from patients with RCC, 5-year survival was lower in patients with high S100A4 expression as compared to weak expression (41% v 78%; P<0.05). |
| IMP3 | Hoffman et al(82) | In a series 716 primary tumor specimens derived from patients with unilateral ccRCC, IMP3 expression was associated with advanced stage and grade, as well as sarcomatoid differentiation. Positive IMP3 expression was associated with a significant increase in the risk of distant metastasis (HR 4.71, P<0.001) in patients with localized disease. On multivariate analysis, IMP3 expression was associated with a 42% increase in the risk of death from RCC (P=0.024). |
| MMP-9 | Kawata et al(99) | In a series of 120 primary tumor specimens derived from patients with localized RCC, MMP-9 was associated with high nuclear grade (P-0.017). MMP-9 was also an independent prognostic factor (P=0.003). |
| PI3K | Merseburger et al(100) | In a series of 176 primary tumor specimens derived from patients with RCC, increased PI3K expression was associated with reduced survival (P=0.030). |
| Thrombospondin-1 | Zuback et al(101) | In 172 consecutive patients with ccRCC who received nephrectomy, thrombospondin-1 expression was associated with high nuclear grade (P,0.001), advanced stage (P<0.001), and tumor progression (P=0.006). |
| Circulating tumor cells (CTCs) | Bluemke et al(102) | In blood samples derived from 154 patients with RCC, CTCs were found 81 specimens (53%). Detection of CTCs was correlated with lymph node status (P<0.0001), presence of synchronous metastases at the time of resection (P=0.014), and poor overall survival (RR 2.3; P=0.048). |
Table 2.
Studies describing potential predictive markers associated with a response to currently available targeted therapies.
| Marker | Drug | Reference | Description |
|---|---|---|---|
| HGF, IL-6, IL-8 (Panel) | Pazopanib | Heymach et al(21, 103) | In 129 blood samples derived from patients enrolled in a randomized discontinuation study of pazopanib for mRCC (n=217), elevated HGF, IL-8 and IL-6 at baseline correlated with less tumor shrinkage. |
| CAIX | Sorafenib | Choueiri et al(104) | Tumor tissue derived from 118 mRCC patients initiating VEGF-directed therapy was assessed for CAIX expression. For patients receiving sorafenib, mean shrinkage with high CAIX was −13% v 9% with low CAIX (P=0.05 for interaction). |
| TNF-α, MMP-9 | Sunitinib | Perez-Garcia et al(105) | In serum from 31 patients treated with sunitinib for mRCC, 174 cytokines were assessed. TNF-α and MMP-9 baseline levels were significantly associated with OS and TTP (P<0.05). |
| NGAL | Sunitinib | Porta et al(79) | In 85 patients receiving sunitinib for mRCC, VEGF and NGAL were significant predictors of OS on multivariate analysis. The RR for NGAL was 1.91 (95%CI 1.39–2.42). |
| bFGF | Sunitinib | Tsimafeyeu et al(106) | In 38 patients receiving sunitinib for mRCC, levels of bFGF significantly increased with disease progression (P=0.001). A non-significant decline in bFGF accompanied tumor stabilization or response. |
| LDH | Temsirolimus | Armstrong et al(107) | In the pivotal phase III evaluation of temsirolimus (n=404), survival was significantly improved in 140 patients who received temsirolimus and had an elevated LDH (P<0.002). In 264 patients with a normal LDH, survival was not improved with temsirolimus as compared to IFN-α (P=0.514). |
CONCLUSIONS
The numerous candidate biomarkers identified herein suggest numerous potential strategies to optimize patient selection for specific targeted therapies. Validation and implementation, however, represent a major obstacle. Sargent et al have proposed several potential trial designs for biomarker validation (83). Each requires an immense allocation of patient resources. For example, consider a marker-based strategy design’ (Figure 2) employed to assess a biomarker with 50% prevalence and an ability to predict a 20% benefit from a specified agent. Assuming an acceptable power of 80% and type I error rate of 5%, approximately 2,700 patients would need to be randomized. This immense allocation of patient resources would have to be resolved with other demands of the research community. For instance, an ever-growing pipeline of agents awaits clinical evaluation. These agents include distinct VEGF-TKIs (i.e., axitinib, tivozanib, and linifanib), as well agents targeting novel signal transduction mediators such as fibroblast growth factor (dovitinib), Akt (MK-2206), and PI3K (BEZ235) (84–90). With each agent warranting clinical trials in hundreds of patients prior to clinical implementation, the research community will be forced to decide between either assessing novel drugs or optimizing use of existing agents through detailed biomarker analyses.
Figure 2.

A proposed marker-based strategy design to evaluate predictive biomarkers.
In the interim, novel trial designs can be used to foster the identification of potential biomarkers. Several neoadjuvant studies have recently been reported, which provide an ideal mechanism for target validation and biomarker assessment in association with targeted therapies. For instance, a neoadjuvant trial of sorafenib in RCC included 17 patients with localized disease and 13 patients with metastatic disease (91). Radiographic correlates showed a decrease in the size of the primary tumor with loss of intratumoral enhancement; other biologic correlates are pending. Bex et al have reported correlative data from patients receiving pre-operative monotherapy with either sunitinib (n=15) or bevacizumab (n=18) (92). Correlative data from these efforts suggest alterations in the CD4+CD25+Foxp3+ cellular population (Tregs), and further suggest variations in the CD8+/Foxp3+ ratio (both assessed by immunohistochemical methods in tumor tissue). As noted previously, modulation of the former population can modulate the adaptive immune system and enhance the anti-tumor immune response (93, 94). As these studies demonstrate, neoadjuvant and pre-operative studies are safe and feasible, and offer a prime opportunity for “proof-of-principle” of preclinical observations.
Efforts in other malignancies can offer insight into potential approaches for efficient biomarker-based studies in RCC. A prime example is the Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) trial. In this study, patients are randomized to one of several agents (erlotinib/bexarotene, sorafenib, vandetanib or erlotinib) with consideration of relevant targets/predictors (such as EGFR mutation, BRAF mutation, KRAS mutation, etc.) (95). Looking ahead, a panel of salient biological alterations in RCC could be used to direct the design of upcoming clinical trials. At present, it is critical that biomarker studies be incorporated into the comparative trials that are ongoing. For example, RECORD-3 is a randomized, phase II switching study that will juxtapose sunitinib against everolimus as first-line therapy for mRCC (96). A separate randomized, phase II study comparing sorafenib to temsirolimus as second line therapy is ongoing (97). Aside from the obvious clinical merits of these studies, both offer a prime opportunity to identify biomarkers that may predict a superior response to either mTOR inhibitors or VEGF-TKIs. With a limited allocation of patient resources and multiple clinical and preclinical hypotheses to validate, creativity in clinical trial design is of prime importance in the research community.
Acknowledgments
Dr. Pal’s efforts are supported by the NIH Loan Repayment Plan (LRP), the CBCRP 15IB-0140 (California Breast Cancer Research Program Junior IDEA Award) and NIH K12 2K12CA001727-16A1. The City of Hope Renal Cell Carcinoma Program is also supported by Kure It, Nancy and Ira Norris, the Hoeven family and the Richard and Nancy Bloch Kidney Cancer Research Funds.
Contributor Information
Sumanta Kumar Pal, Email: spal@coh.org, Division of Genitourinary Malignancies, Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673, Fax: (626) 301-8233.
Marcin Kortylewski, Email: mkortylewski@coh.org, Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673, Fax: (626) 301-8233.
Hua Yu, Email: hyu@coh.org, Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673, Fax: (626) 301-8233.
Robert A. Figlin, Email: Robert.Figlin@cshs.org, Hematology-Oncology, Department of Medicine, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Phone: (310) 423-1331, Fax: (310) 659-3928.
References
- 1.Pal SK, Figlin RA. Targeted therapies: Pazopanib: carving a niche in a crowded therapeutic landscape. Nat Rev Clin Oncol. 2010;7(7):362–3. doi: 10.1038/nrclinonc.2010.83. [DOI] [PubMed] [Google Scholar]
- 2.Motzer RJ, Hutson TE, Tomczak P, et al. Overall Survival and Updated Results for Sunitinib Compared With Interferon Alfa in Patients With Metastatic Renal Cell Carcinoma. J Clin Oncol. 2009 August 1;27(22):3584–90. doi: 10.1200/JCO.2008.20.1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Escudier BJ, Bellmunt J, Negrier S, et al. Final results of the phase III, randomized, double-blind AVOREN trial of first-line bevacizumab (BEV) + interferon-{alpha}2a (IFN) in metastatic renal cell carcinoma (mRCC) J Clin Oncol (Meeting Abstracts) 2009 May 20;27(15S):5020. [Google Scholar]
- 4.Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in Locally Advanced or Metastatic Renal Cell Carcinoma: Results of a Randomized Phase III Trial. J Clin Oncol. 2010 February 20;28(6):1061–8. doi: 10.1200/JCO.2009.23.9764. [DOI] [PubMed] [Google Scholar]
- 5.Escudier B, Bellmunt J, Negrier S, et al. Phase III Trial of Bevacizumab Plus Interferon Alfa-2a in Patients With Metastatic Renal Cell Carcinoma (AVOREN): Final Analysis of Overall Survival. J Clin Oncol. 2010 May 1;28(13):2144–50. doi: 10.1200/JCO.2009.26.7849. [DOI] [PubMed] [Google Scholar]
- 6.Rini BI, Halabi S, Rosenberg JE, et al. Phase III Trial of Bevacizumab Plus Interferon Alfa Versus Interferon Alfa Monotherapy in PatientsWith Metastatic Renal Cell Carcinoma: Final Results of CALGB 90206. J Clin Oncol. 2010 May 1;28(13):2137–43. doi: 10.1200/JCO.2009.26.5561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kay A, Motzer R, Figlin R, et al. Updated data from a phase III randomized trial of everolimus (RAD001) versus PBO in metastatic renal cell carcinoma (mRCC). Presented at the 2009 Genitourinary Cancers Symposium (Abstr 278). [Google Scholar]
- 8.Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, Interferon Alfa, or Both for Advanced Renal-Cell Carcinoma. N Engl J Med. 2007 May 31;356(22):2271–81. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
- 9.Slamon D, Leyland-Jones B, Shak S. Addition of Herceptin (humanized anti-HER2 antibody) to first line chemotherapy for HER2 overexpressing metastatic breast cancer (HER21/MBC) markedly increases anticancer activity: A randomized multinational controlled phase III trial. ProcAm Soc Clin Oncol. 1998;17(98a) abstr 377. [Google Scholar]
- 10.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987 January 9;235(4785):177–82. doi: 10.1126/science.3798106. [DOI] [PubMed] [Google Scholar]
- 11.Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989 May 12;244(4905):707–12. doi: 10.1126/science.2470152. [DOI] [PubMed] [Google Scholar]
- 12.Slamon DJ, Leyland-Jones B, Shak S, et al. Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2. N Engl J Med. 2001 March 15;344(11):783–92. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
- 13.Bang Y, Kwak EL, Shaw AT, et al. Clinical activity of the oral ALK inhibitor PF-02341066 in ALK-positive patients with non-small cell lung cancer (NSCLC) J Clin Oncol (Meeting Abstracts) 2010 June 20;28(18_suppl):3. [Google Scholar]
- 14.Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. New England Journal of Medicine. 363(9):809–19. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Iliopoulos O, Kibel A, Gray S, Kaelin WG. Tumour suppression by the human von Hippel-Lindau gene product. Nat Med. 1995;1(8):822–6. doi: 10.1038/nm0895-822. [DOI] [PubMed] [Google Scholar]
- 16.Kishida T, Stackhouse TM, Chen F, Lerman MI, Zbar B. Cellular Proteins That Bind the von Hippel-Lindau Disease Gene Product: Mapping of Binding Domains and the Effect of Missense Mutations. Cancer Research. 1995 October 15;55(20):4544–8. [PubMed] [Google Scholar]
- 17.Kibel A, Iliopoulos O, DeCaprio J, Kaelin W., Jr Binding of the von Hippel-Lindau tumor suppressor protein to Elongin B and C. Science. 1995 September 8;269(5229):1444–6. doi: 10.1126/science.7660130. [DOI] [PubMed] [Google Scholar]
- 18.Lonergan KM, Iliopoulos O, Ohh M, et al. Regulation of Hypoxia-Inducible mRNAs by the von Hippel-Lindau Tumor Suppressor Protein Requires Binding to Complexes Containing Elongins B/C and Cul2. Mol Cell Biol. 1998 February 1;18(2):732–41. doi: 10.1128/mcb.18.2.732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Iwai K, Yamanaka K, Kamura T, et al. Identification of the von Hippel – Lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proceedings of the National Academy of Sciences of the United States of America. 1999 October 26;96(22):12436–41. doi: 10.1073/pnas.96.22.12436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kim WY, Kaelin WG. Role of VHL Gene Mutation in Human Cancer. J Clin Oncol. 2004 December 15;22(24):4991–5004. doi: 10.1200/JCO.2004.05.061. [DOI] [PubMed] [Google Scholar]
- 21.Heymach J, Tran HT, Fritsche HA, et al. Lower baseline levels of plasma hepatocyte growth factor (HGF), IL-6 and IL-8 are correlated with tumor shrinkage in renal cell carcinoma patients treated with pazopanib. Mol Cancer Ther. 2009;8(12 Suppl) Abstr A11. [Google Scholar]
- 22.Xu Q, Briggs J, Park S, et al. Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene. 2005 Aug 25;24(36):5552–60. doi: 10.1038/sj.onc.1208719. [DOI] [PubMed] [Google Scholar]
- 23.Horiguchi A, Oya M, Shimada T, Uchida A, Marumo K, Murai M. Activation of signal transducer and activator of transcription 3 in renal cell carcinoma: a study of incidence and its association with pathological features and clinical outcome. J Urol. 2002 Aug;168(2):762–5. [PubMed] [Google Scholar]
- 24.Lubensky I, Gnarra J, Bertheau P, Walther M, Linehan W, Zhuang Z. Allelic deletions of the VHL gene detected in multiple microscopic clear cell renal lesions in von Hippel-Lindau disease patients. Am J Pathol. 1996 December 1;149(6):2089–94. [PMC free article] [PubMed] [Google Scholar]
- 25.Grabmaier K, AdW MC, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 2004 Jul 22;23(33):5624–31. doi: 10.1038/sj.onc.1207764. [DOI] [PubMed] [Google Scholar]
- 26.Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fujii-Kuriyama Y. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1α regulates the VEGF expression and is potentially involved in lung and vascular development. Proceedings of the National Academy of Sciences of the United States of America. 1997 April 29;94(9):4273–8. doi: 10.1073/pnas.94.9.4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tian H, McKnight SL, Russell DW. Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes & Development. 1997 January 1;11(1):72–82. doi: 10.1101/gad.11.1.72. [DOI] [PubMed] [Google Scholar]
- 28.Wiesener MS, Jürgensen JS, Rosenberger C, et al. Widespread hypoxia-inducible expression of HIF-2alpha in distinct cell populations of different organs. The FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2003;17(2):271–3. doi: 10.1096/fj.02-0445fje. [DOI] [PubMed] [Google Scholar]
- 29.Kondo K, Klco J, Nakamura E, Lechpammer M, Kaelin WG., Jr Inhibition of HIF is necessary for tumor suppression by the von Hippel-Lindau protein. Cancer Cell. 2002;1(3):237–46. doi: 10.1016/s1535-6108(02)00043-0. [DOI] [PubMed] [Google Scholar]
- 30.Maranchie JK, Vasselli JR, Riss J, Bonifacino JS, Linehan WM, Klausner RD. The contribution of VHL substrate binding and HIF1-[alpha] to the phenotype of VHL loss in renal cell carcinoma. Cancer Cell. 2002;1(3):247–55. doi: 10.1016/s1535-6108(02)00044-2. [DOI] [PubMed] [Google Scholar]
- 31.Gordan JD, Bertout JA, Hu C-J, Diehl JA, Simon MC. HIF-2[alpha] Promotes Hypoxic Cell Proliferation by Enhancing c-Myc Transcriptional Activity. Cancer Cell. 2007;11(4):335–47. doi: 10.1016/j.ccr.2007.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gordan JD, Lal P, Dondeti VR, et al. HIF-[alpha] Effects on c-Myc Distinguish Two Subtypes of Sporadic VHL-Deficient Clear Cell Renal Carcinoma. Cancer Cell. 2008;14(6):435–46. doi: 10.1016/j.ccr.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rini BI, Zhou M, Aydin H, et al. Identification of prognostic genomic markers in patients with localized clear cell renal cell carcinoma (ccRCC) J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4501. [Google Scholar]
- 34.Fuhrman SA, Lasky LC, Limas C. Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol. 1982 Oct;6(7):655–63. doi: 10.1097/00000478-198210000-00007. [DOI] [PubMed] [Google Scholar]
- 35.Dalgliesh GL, Furge K, Greenman C, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. 463(7279):360–3. doi: 10.1038/nature08672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Cifola I, Spinelli R, Beltrame L, et al. Genome-wide screening of copy number alterations and LOH events in renal cell carcinomas and integration with gene expression profile. Molecular Cancer. 2008;7(1):6. doi: 10.1186/1476-4598-7-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chi J-T, Wang Z, Nuyten DSA, et al. Gene Expression Programs in Response to Hypoxia: Cell Type Specificity and Prognostic Significance in Human Cancers. PLoS Med. 2006;3(3):e47. doi: 10.1371/journal.pmed.0030047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhao H, Ljungberg Br, Grankvist K, Rasmuson T, Tibshirani R, Brooks JD. Gene Expression Profiling Predicts Survival in Conventional Renal Cell Carcinoma. PLoS Med. 2005;3(1):e13. doi: 10.1371/journal.pmed.0030013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Burger JA, Kipps TJ. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006 Mar 1;107(5):1761–7. doi: 10.1182/blood-2005-08-3182. [DOI] [PubMed] [Google Scholar]
- 40.Erler JT, Bennewith KL, Cox TR, et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009 Jan 6;15(1):35–44. doi: 10.1016/j.ccr.2008.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Costa V, Henrique R, Ribeiro F, et al. Quantitative promoter methylation analysis of multiple cancer-related genes in renal cell tumors. BMC Cancer. 2007;7(1):133. doi: 10.1186/1471-2407-7-133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dalgin GS, Drever M, Williams T, King T, DeLisi C, Liou LS. Identification of Novel Epigenetic Markers for Clear Cell Renal Cell Carcinoma. The Journal of Urology. 2008;180(3):1126–30. doi: 10.1016/j.juro.2008.04.137. [DOI] [PubMed] [Google Scholar]
- 43.Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 463(7283):899–905. doi: 10.1038/nature08822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xu G, Xiang CQ, Lu Y, et al. Application of SELDI-TOF-MS to identify serum biomarkers for renal cell carcinoma. Cancer Letters. 2009;282(2):205–13. doi: 10.1016/j.canlet.2009.03.017. [DOI] [PubMed] [Google Scholar]
- 45.Xu G, Xiang CQ, Lu Y, et al. SELDI-TOF-MS-based serum proteomic screening in combination with CT scan distinguishes renal cell carcinoma from benign renal tumors and healthy persons. Technol Cancer Res Treat. 2009 Jun;8(3):225–30. doi: 10.1177/153303460900800307. [DOI] [PubMed] [Google Scholar]
- 46.Aggelis V, Craven RA, Peng J, et al. Proteomic identification of differentially expressed plasma membrane proteins in renal cell carcinoma by stable isotope labelling of a von Hippel-Lindau transfectant cell line model. Proteomics. 2009;9(8):2118–30. doi: 10.1002/pmic.200800756. [DOI] [PubMed] [Google Scholar]
- 47.Alberghini A, Recalcati S, Tacchini L, Santambrogio P, Campanella A, Cairo G. Loss of the von Hippel Lindau tumor suppressor disrupts iron homeostasis in renal carcinoma cells. J Biol Chem. 2005 Aug 26;280(34):30120–8. doi: 10.1074/jbc.M500971200. [DOI] [PubMed] [Google Scholar]
- 48.Davidowitz EJ, Schoenfeld AR, Burk RD. VHL induces renal cell differentiation and growth arrest through integration of cell-cell and cell-extracellular matrix signaling. Mol Cell Biol. 2001 Feb;21(3):865–74. doi: 10.1128/MCB.21.3.865-874.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Seliger B, Dressler SP, Wang E, et al. Combined analysis of transcriptome and proteome data as a tool for the identification of candidate biomarkers in renal cell carcinoma. PROTEOMICS. 2009;9(6):1567–81. doi: 10.1002/pmic.200700288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. [last accessed October 7, 2010.).];The Cancer Genome Atlas (TCGA) Pilot Project: Cancers Under Study. (Available at: http://cancergenome.nih.gov/wwd/cancers_studied_by_tcga.asp.
- 51.Pages F, Galon J, Dieu-Nosjean MC, Tartour E, Sautes-Fridman C, Fridman WH. Immune infiltration in human tumors: a prognostic factor that should not be ignored. Oncogene. 2010;29(8):1093–102. doi: 10.1038/onc.2009.416. [DOI] [PubMed] [Google Scholar]
- 52.Nakano O, Sato M, Naito Y, et al. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res. 2001 Jul 1;61(13):5132–6. [PubMed] [Google Scholar]
- 53.Siddiqui SA, Frigola X, Bonne-Annee S, et al. Tumor-infiltrating Foxp3-CD4+CD25+ T cells predict poor survival in renal cell carcinoma. Clin Cancer Res. 2007 Apr 1;13(7):2075–81. doi: 10.1158/1078-0432.CCR-06-2139. [DOI] [PubMed] [Google Scholar]
- 54.Kusmartsev S, Su Z, Heiser A, et al. Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2008 Dec 15;14(24):8270–8. doi: 10.1158/1078-0432.CCR-08-0165. [DOI] [PubMed] [Google Scholar]
- 55.Yang JC, Hughes M, Kammula U, et al. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother. 2007 Nov-Dec;30(8):825–30. doi: 10.1097/CJI.0b013e318156e47e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Thompson RH, Webster WS, Cheville JC, et al. B7-H1 glycoprotein blockade: a novel strategy to enhance immunotherapy in patients with renal cell carcinoma. Urology. 2005 Nov;66(5 Suppl):10–4. doi: 10.1016/j.urology.2005.06.010. [DOI] [PubMed] [Google Scholar]
- 57.Xin H, Zhang C, Herrmann A, Du Y, Figlin R, Yu H. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 2009 Mar 15;69(6):2506–13. doi: 10.1158/0008-5472.CAN-08-4323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Finke JH, Rini B, Ireland J, et al. Sunitinib Reverses Type-1 Immune Suppression and Decreases T-Regulatory Cells in Renal Cell Carcinoma Patients. Clinical Cancer Research. 2008 October 15;14(20):6674–82. doi: 10.1158/1078-0432.CCR-07-5212. [DOI] [PubMed] [Google Scholar]
- 59.van Cruijsen H, van der Veldt AAM, Vroling L, et al. Sunitinib-Induced Myeloid Lineage Redistribution in Renal Cell Cancer Patients: CD1c+ Dendritic Cell Frequency Predicts Progression-Free Survival. Clinical Cancer Research. 2008 September 15;14(18):5884–92. doi: 10.1158/1078-0432.CCR-08-0656. [DOI] [PubMed] [Google Scholar]
- 60.Chowdhury S, O’Brien TS, Sarwar N, et al. The effect of sunitinib on immune parameters and haemopoetic stem cell markers in patients with untreated clear cell renal cancer. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4620. [Google Scholar]
- 61.Hipp MM, Hilf N, Walter S, et al. Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses. Blood. 2008 Jun 15;111(12):5610–20. doi: 10.1182/blood-2007-02-075945. [DOI] [PubMed] [Google Scholar]
- 62.Cindolo L, Chiodini P, Gallo C, et al. Validation by calibration of the UCLA integrated staging system prognostic model for nonmetastatic renal cell carcinoma after nephrectomy. Cancer. 2008 Jul 1;113(1):65–71. doi: 10.1002/cncr.23517. [DOI] [PubMed] [Google Scholar]
- 63.Zisman A, Pantuck AJ, Figlin RA, Belldegrun AS. Validation of the ucla integrated staging system for patients with renal cell carcinoma. J Clin Oncol. 2001 Sep 1;19(17):3792–3. doi: 10.1200/JCO.2001.19.17.3792. [DOI] [PubMed] [Google Scholar]
- 64.Onea C, Mouracade P, Roman H, et al. External validation of the University of Los Angeles Integrated Staging System (UISS) in localized renal cell carcinoma: single center study. Prog Urol Jan. 20(1):35–9. doi: 10.1016/j.purol.2009.07.007. [DOI] [PubMed] [Google Scholar]
- 65.Ng CF, Wan SH, Wong A, Lai FM, Hui P, Cheng CW. Use of the University of California Los Angeles Integrated Staging System (UISS) to predict survival in localized renal cell carcinoma in an Asian population. Int Urol Nephrol. 2007;39(3):699–703. doi: 10.1007/s11255-006-9134-1. [DOI] [PubMed] [Google Scholar]
- 66.Belldegrun AS, Klatte T, Shuch B, et al. Cancer-specific survival outcomes among patients treated during the cytokine era of kidney cancer (1989–2005): a benchmark for emerging targeted cancer therapies. Cancer. 2008 Nov 1;113(9):2457–63. doi: 10.1002/cncr.23851. [DOI] [PubMed] [Google Scholar]
- 67.Lam JS, Shvarts O, Leppert JT, Pantuck AJ, Figlin RA, Belldegrun AS. Postoperative surveillance protocol for patients with localized and locally advanced renal cell carcinoma based on a validated prognostic nomogram and risk group stratification system. J Urol. 2005 Aug;174(2):466–72. doi: 10.1097/01.ju.0000165572.38887.da. discussion 72; quiz 801. [DOI] [PubMed] [Google Scholar]
- 68.Bellmunt J, Flodgren P, Roigas J, Oudard S. Optimal management of metastatic renal cell carcinoma: an algorithm for treatment. BJU Int. 2009 Jul;104(1):10–8. doi: 10.1111/j.1464-410X.2009.08563.x. [DOI] [PubMed] [Google Scholar]
- 69.Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol. 2002 Jan 1;20(1):289–96. doi: 10.1200/JCO.2002.20.1.289. [DOI] [PubMed] [Google Scholar]
- 70.Heng DYC, Xie W, Regan MM, et al. Prognostic Factors for Overall Survival in Patients With Metastatic Renal Cell Carcinoma Treated With Vascular Endothelial Growth Factor–Targeted Agents: Results From a Large, Multicenter Study. Journal of Clinical Oncology. 2009 December 1;27(34):5794–9. doi: 10.1200/JCO.2008.21.4809. [DOI] [PubMed] [Google Scholar]
- 71.Rini BI, Cohen DP, Lu D, et al. Hypertension (HTN) as a biomarker of efficacy in patients (pts) with metastatic renal cell carcinoma (mRCC) treated with sunitinib. Presented at the 2010 Genitourinary Cancers Symposium; Abstr 312. [Google Scholar]
- 72.Harzstark AL, Halabi S, Stadler WM, et al. Hypertension is associated with clinical outcome for patients (pts) with metastatic renal cell carcinoma (RCC) treated with interferon and bevacizumab on CALGB 90206. Presented at the 2010 Genitourinary Cancers Symposium; Abstr 351. [Google Scholar]
- 73.Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007 Dec 27;357(26):2666–76. doi: 10.1056/NEJMoa072113. [DOI] [PubMed] [Google Scholar]
- 74.Kim JJ, Vaziri SA, Elson P, et al. Role of VEGF and VEGFR2 single nucleotide polymorphisms (SNPs) in predicting treatment-induced hypertension (HTN) and clinical outcome (CO) in metastatic clear cell RCC (mccRCC) patients (pts) treated with sunitinib. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4629. [Google Scholar]
- 75. [last accessed August 26, 2010]; NCT00835978: Randomized, Double-Blind Phase 2 Study of Axitinib (AG-013736) With Or Without Dose Titration In Patients With Metastatic Renal Cell Carcinoma. (Available on-line at http://www.clinicaltrials.gov.
- 76.Choueiri TK, Xie W, Kollmannsberger CK, et al. The impact of body mass index (BMI) and body surface area (BSA) on treatment outcome to vascular endothelial growth factor (VEGF)-targeted therapy in metastatic renal cell carcinoma: Results from a large international collaboration. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4524. [Google Scholar]
- 77.DePrimo S, Bello C, Smeraglia J, et al. Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. Journal of Translational Medicine. 2007;5(1):32. doi: 10.1186/1479-5876-5-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rini BI, Michaelson MD, Rosenberg JE, et al. Antitumor Activity and Biomarker Analysis of Sunitinib in Patients With Bevacizumab-Refractory Metastatic Renal Cell Carcinoma. Journal of Clinical Oncology. 2008;26(22):3743–8. doi: 10.1200/JCO.2007.15.5416. [DOI] [PubMed] [Google Scholar]
- 79.Porta C, Paglino C, De Amici M, et al. Predictive value of baseline serum vascular endothelial growth factor and neutrophil gelatinase-associated lipocalin in advanced kidney cancer patients receiving sunitinib. Kidney Int. 2010;77(9):809–15. doi: 10.1038/ki.2009.552. [DOI] [PubMed] [Google Scholar]
- 80.O’Reilly T, McSheehy PM. Biomarker Development for the Clinical Activity of the mTOR Inhibitor Everolimus (RAD001): Processes, Limitations, and Further Proposals. Transl Oncol. 2010 Apr;3(2):65–79. doi: 10.1593/tlo.09277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pena C, Lathia C, Shan M, Escudier B, Bukowski RM. Biomarkers Predicting Outcome in Patients with Advanced Renal Cell Carcinoma: Results from Sorafenib Phase III Treatment Approaches in Renal Cancer Global Evaluation Trial. Clinical Cancer Research. 16(19):4853–63. doi: 10.1158/1078-0432.CCR-09-3343. [DOI] [PubMed] [Google Scholar]
- 82.Hoffmann NE, Sheinin Y, Lohse CM, et al. External validation of IMP3 expression as an independent prognostic marker for metastatic progression and death for patients with clear cell renal cell carcinoma. Cancer. 2008;112(7):1471–9. doi: 10.1002/cncr.23296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sargent DJ, Conley BA, Allegra C, Collette L. Clinical Trial Designs for Predictive Marker Validation in Cancer Treatment Trials. Journal of Clinical Oncology. 2005 March 20;23(9):2020–7. doi: 10.1200/JCO.2005.01.112. [DOI] [PubMed] [Google Scholar]
- 84.Rini BI, Wilding G, Hudes G, et al. Phase II Study of Axitinib in Sorafenib-Refractory Metastatic Renal Cell Carcinoma. J Clin Oncol. 2009 September 20;27(27):4462–8. doi: 10.1200/JCO.2008.21.7034. [DOI] [PubMed] [Google Scholar]
- 85.Rixe O, Bukowski RM, Michaelson MD, et al. Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study. Lancet Oncol. 2007 Nov;8(11):975–84. doi: 10.1016/S1470-2045(07)70285-1. [DOI] [PubMed] [Google Scholar]
- 86.Bhargava P, Esteves B, Al-Adhami M, et al. Activity of tivozanib (AV-951) in patients with renal cell carcinoma (RCC): Subgroup analysis from a phase II randomized discontinuation trial (RDT) J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4599. [Google Scholar]
- 87.Tannir NM, Wong Y, Kollmannsberger CK, et al. Phase II trial of linifanib in patients with advanced renal cell cancer (RCC) after sunitinib failure. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4527. [Google Scholar]
- 88.Angevin E, Lin C, Pande AU, et al. A phase I/II study of dovitinib (TKI258), a FGFR and VEGFR inhibitor, in patients (pts) with advanced or metastatic renal cell cancer: Phase I results. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):3057. [Google Scholar]
- 89.Tolcher AW, Yap TA, Fearen I, et al. A phase I study of MK-2206, an oral potent allosteric Akt inhibitor (Akti), in patients (pts) with advanced solid tumor (ST) J Clin Oncol (Meeting Abstracts) 2009 May 20;27(15S):3503. [Google Scholar]
- 90.Burris H, Rodon J, Sharma S, et al. First-in-human phase I study of the oral PI3K inhibitor BEZ235 in patients (pts) with advanced solid tumors. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):3005. [Google Scholar]
- 91.Cowey CL, Amin C, Pruthi RS, et al. Neoadjuvant Clinical Trial With Sorafenib for Patients With Stage II or Higher Renal Cell Carcinoma. J Clin Oncol. 2010 March 20;28(9):1502–7. doi: 10.1200/JCO.2009.24.7759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Bex A, Jonasch E, Vyth-Dreese F, de Hoog C, Dellemijn T, Griffioen AW. Presurgical bevacizumab and sunitinib induced changes in primary clear cell renal tumor tissue. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4607. [Google Scholar]
- 93.Stephens GL, Andersson J, Shevach EM. Distinct Subsets of FoxP3+ Regulatory T Cells Participate in the Control of Immune Responses. J Immunol. 2007 June 1;178(11):6901–11. doi: 10.4049/jimmunol.178.11.6901. [DOI] [PubMed] [Google Scholar]
- 94.Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJC, John S, Taams LS. CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proceedings of the National Academy of Sciences. 2007 December 4;104(49):19446–51. doi: 10.1073/pnas.0706832104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kim ES, Herbst RS, Lee JJ, et al. Phase II randomized study of biomarker-directed treatment for non-small cell lung cancer (NSCLC): The BATTLE (Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination) clinical trial program. J Clin Oncol. 2009;27(15s) (suppl; abstr 8024) [Google Scholar]
- 96. [last accessed June 23, 2009]; NCT00903175: Efficacy and Safety Comparison of RAD001 Versus Sunitinib in the First-Line and Second-Line Treatment of Patients With Metastatic Renal Cell Carcinoma. (Available at http://www.ClinicalTrials.gov.
- 97. [last accessed March 12, 2010]; NCT00474786: A Randomized Trial of Temsirolimus Versus Sorafenib as Second-Line Therapy in Patients With Advanced Renal Cell Carcinoma Who Have Failed First-Line Sunitinib Therapy. (Available at http://www.clinicaltrials.gov.
- 98.Bandiera A, Melloni G, Freschi M, et al. Prognostic Factors and Analysis of S100a4 Protein in Resected Pulmonary Metastases from Renal Cell Carcinoma. World Journal of Surgery. 2009;33(7):1414–20. doi: 10.1007/s00268-009-0023-5. [DOI] [PubMed] [Google Scholar]
- 99.Kawata N, Nagane Y, Hirakata H, et al. Significant Relationship of Matrix Metalloproteinase 9 with Nuclear Grade and Prognostic Impact of Tissue Inhibitor of Metalloproteinase 2 for Incidental Clear Cell Renal Cell Carcinoma. Urology. 2007;69(6):1049–53. doi: 10.1016/j.urology.2007.02.044. [DOI] [PubMed] [Google Scholar]
- 100.Merseburger AS, Hennenlotter J, Kuehs U, et al. Activation of PI3K is associated with reduced survival in renal cell carcinoma. Urol Int. 2008;80(4):372–7. doi: 10.1159/000132694. [DOI] [PubMed] [Google Scholar]
- 101.Zubac DP, Bostad L, Kihl B, Seidal T, Wentzel-Larsen T, Haukaas SA. The Expression of Thrombospondin-1 and p53 in Clear Cell Renal Cell Carcinoma: Its Relationship to Angiogenesis, Cell Proliferation and Cancer Specific Survival. The Journal of Urology. 2009;182(5):2144–9. doi: 10.1016/j.juro.2009.07.015. [DOI] [PubMed] [Google Scholar]
- 102.Bluemke K, Bilkenroth U, Meye A, et al. Detection of Circulating Tumor Cells in Peripheral Blood of Patients with Renal Cell Carcinoma Correlates with Prognosis. Cancer Epidemiology Biomarkers & Prevention. 2009 August;18(8):2190–4. doi: 10.1158/1055-9965.EPI-08-1178. [DOI] [PubMed] [Google Scholar]
- 103.Heymach J, Tran HT, Fritsche HA, et al. Lower baseline levels of plasma hepatocyte growth factor (HGF), IL-6 and IL-8 are correlated with tumor shrinkage in renal cell carcinoma patients treated with pazopanib. Presented at the AACR-NCI-EORTC International Conference on Molecular Targets adn Cancer Therapeutics; November 15–17, 2009; Boston, MA. Abstr A11. [Google Scholar]
- 104.Choueiri TK, Regan MM, Rosenberg JE, et al. Carbonic anhydrase IX and pathological features as predictors of outcome in patients with metastatic clear-cell renal cell carcinoma receiving vascular endothelial growth factor-targeted therapy. BJU International. 2010:9999–9999. doi: 10.1111/j.1464-410X.2010.09218.x. [DOI] [PubMed] [Google Scholar]
- 105.Perez-Gracia JL, Prior C, Guillen-Grima F, et al. Identification of TNF-[alpha] and MMP-9 as potential baseline predictive serum markers of sunitinib activity in patients with renal cell carcinoma using a human cytokine array. Br J Cancer. 2009;101(11):1876–83. doi: 10.1038/sj.bjc.6605409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Tsimafeyeu I, Demidov L, Ta H, Stepanova E, Wynn N. Fibroblast growth factor pathway in renal cell carcinoma. J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4621. [Google Scholar]
- 107.Armstrong AJ, George DJ, Halabi S. Serum lactate dehydrogenase (LDH) as a biomarker for survival with mTOR inhibition in patients with metastatic renal cell carcinoma (RCC) J Clin Oncol (Meeting Abstracts) 2010 May 20;28(15_suppl):4631. [Google Scholar]
