Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 1.
Published in final edited form as: Curr Urol Rep. 2014 Jan;15(1):375. doi: 10.1007/s11934-013-0375-0

Molecular Marker For Predicting Treatment Response in Advanced Renal Cell Carcinoma: Does the Promise Fulfill Clinical Need?

Michael Garcia-Roig 1,*, Nicolas Ortiz 1,*, Vinata Lokeshwar 1,2
PMCID: PMC3913467  NIHMSID: NIHMS549445  PMID: 24337833

Abstract

Renal cell carcinoma (RCC) is largely diagnosed incidentally on imaging taken for unrelated reasons. The management of localized lesions is primarily extirpative with excellent results. Treatment of advanced RCC has evolved over recent years with the use of targeted therapies such as tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, and antibody-mediated therapies. The treatment response to these targeted therapies is highly variable, with no clear clinical method of identifying patients who will benefit from or not tolerate therapy. The field of molecular markers has evolved significantly in the last decade, with a multitude of markers identified that predict treatment response and drug toxicity. The following review critically evaluates those molecular markers that have been assessed for their utility in predicting treatment response in patients with advanced/metastatic renal cell carcinoma (mRCC). Identifying the ideal treatment for these patients will improve responses to therapy, minimize morbidity, and save significant healthcare dollars.

Keywords: Metastatic renal cell carcinoma, Tyrosine kinase inhibitors, Prognostic markers, Sunitinib, Sorafenib

Introduction

Renal cell carcinoma (RCC) is diagnosed largely via imaging technologies such as ultrasound (US) or computed tomography (CT). Over the last decade, the ubiquity of renal imaging for various indications has led to a significant increase in the detection of incidental, often small, renal masses. The classic triad of flank pain, gross hematuria, and an abdominal mass is uncommon and suggests advanced disease 1. No effective screening tests have been devised for RCC, as its low incidence in the general population makes screening impractical.

mRCC is present at the time of diagnosis in approximately 30 % of patients. About 20–40 % of patients presenting with localized disease ultimately progress to metastasis. Advanced RCC, as defined by metastasis, carries a poor, 10-year overall survival (OS) of 5 % 2, 3. Metastases are more common in larger and/or poorly differentiated tumors. Metastatic lesions in RCC are primarily identified by imaging, and a workup for such lesions is recommended for all renal masses, regardless of size, by performing an abdominopelvic CT scan and chest X-ray 4, 5. Magnetic resonance imaging (MRI) may be used alternatively in the setting of contrast allergy or pregnancy, or to further characterize a tumor thrombus. Further workup is recommended for patients with suspicious pulmonary or bony lesions 4, 5.

In RCC, molecular markers have been described to help characterize tumor type and predict the likelihood of progression and metastasis 6. These markers can help create more accurate tumor staging and prognostication 7. Advances in more effective chemotherapy for advanced RCC make it more important to identify these patients early on in the disease process. For example, markers can be useful in stratifying patients into responders and non-responders for a variety of targeted treatments that are available for metastatic RCC. If non-responders are identified at the beginning or early stages of treatment, it would avoid any delay in administration of alternate treatment(s) and avoid unnecessary side effects due to a treatment that will not be effective.

A variety of targeted treatments directed towards molecular determinants of metastatic RCC, including tyrosine kinase inhibitors (TKIs) and vascular endothelial growth factor (VEGF), have become available for mRCC. The role of the Von Hippel Lindau (VHL) gene in the development of clear cell RCC (ccRCC) has been extensively described. Inactivation of this gene leads to overexpression of pro-angiogenic elements such as VEGF, which plays a critical role in RCC tumor development. TKIs targeting this pathway are the mainstay of treatment for advanced ccRCC, and are therefore the most widely studied. Four FDA-approved targeted drugs are currently available in the United States: Sunitinib, Sorafenib, Pazopanib, and Axitinib. Several targets of the angiogenic cascade have been evaluated as predictors of prognosis and response to TKIs; however, treatment response to targeted therapies is highly variable and predicting this response would help to guide treatment. Below, we review the current literature on tumor markers in advanced renal cell carcinoma, relative to predicting treatment response to the various options for targeted therapy.

Methods

Medline databases were searched with a combination of the following terms: renal cell carcinoma, metastasis, advanced, molecular markers, targeted therapy, systemic therapy, tyrosine kinase inhibitor (TKI), mammalian target of rapamycin (mTOR), vascular endothelial growth factor (VEGF), carbonic anhydrase IX (CA IX), Sunitinib, Sorafenib, Bevacizumab, VHL, circulating endothelial cells, serum amyloid alpha (SAA), neutrophil gelatinase-associated lipocalin (NGAL), erythrocyte sedimentation rate (ESR), metalloproteinase 9 (MMP-9) and tumor necrosis factor (TNF)-α, and lactate dehydrogenase (LDH). Articles were selected based on study size, uniqueness, and importance of contribution to the field.

Treatments for Advanced Renal Cell Carcinoma

Surgery

Surgical management of metastatic renal cell carcinoma is the primary treatment modality for localized RCC and also plays a significant role in mRCC for patients who are acceptable surgical candidates. Cytoreductive nephrectomy is the mainstay of surgical management for advanced RCC. Two randomized studies evaluated the benefits of cytoreductive surgery and interferon (IFN)-α2b. When compared to IFN-α2b treatment alone, a survival benefit of 6 months (13.6 vs. 7.8 months) was noted among patients undergoing both surgery and interferon treatment for mRCC 8. It is not yet understood how incomplete tumor removal improves survival, although some hypothesize that the effect may be due to changes in cytokines or growth factors, thereby enhancing tumor immunity, or from the reduction in tumor bulk itself. Patients with a good performance status were found to benefit most from surgical intervention, as they were at the lowest risk for operative-related complications 9. Minimally-invasive cytoreductive nephrectomy may be an option for appropriately selected patients 10. To date, the role of cytoreductive nephrectomy in the setting of targeted therapy has not been investigated. However, several groups have described the use of neoadjuvant TKIs to shrink tumor size prior to partial nephrectomy in patients with multiple tumors or a solitary kidney 11, 12.

In the setting of solitary localized metastasis or solitary tumor recurrence, primary tumor excision with complete resection of the metastasis improves survival to 30–47 % at 5 years in appropriately selected patients 13. One group identified risk factors for poor outcome in a series of 175 patients with solitary pulmonary metastasis. These factors included pleural infiltration, synchronous presence of primary RCC, metastasis > 3 cm, mediastinal and hilar node status, and completeness of metastasectomy 14. Several other groups have identified similar predictors of outcome in the setting of metastasis 15, 16. Moreover, a patient’s disease-free interval plays a significant role in predicting post-metastasectomy survival 17. No studies have reported on prospectively treated patients; however, this surgical extirpation of solitary RCC metastasis is currently offered to such patients who are surgical candidates.

Systemic Therapy

Patients with residual tumor after resection for mRCC are offered systemic therapy. Immunotherapy with IFN-α (or interleukin (IL)-2 was initially used for patients with advanced disease with significant overall response rates of 6.5–18.6 % at 1 year, either alone or in combination 18. More recently, treatments targeting the VEGF pathway have become the mainstay of systemic therapy, due to improved response to therapy and reduced toxicity compared with immunotherapy. VEGF is the end product of the VHL gene deregulation pathway in ccRCC. VHL modulates the hypoxia-inducible factor (HIF) pathway, normally activated in the setting of hypoxia to promote oxygen delivery through angiogenesis and red blood cell production via VEGF and platelet-derived growth factor (PDGF) axes 19. Both VEGF and PDGF promote endothelial cell proliferation, motility, and hence, neovascularization by binding to their respective cell surface receptors on endothelial cells and stimulating signaling cascades downstream. Several treatment options have emerged that inhibit VEGF production or signaling, and therefore, abrogate angiogenesis and tumor growth. Therapy targeting the VHL–VEGF pathway falls into three classes: mTOR inhibitors, TKIs, and monoclonal antibodies against VEGF. The class of mTOR inhibitors include Temsirolimus and Everolimus, the latter being an orally bioavailable form. The mTOR pathway plays a role in growth factor-related signaling cascades. mTOR was found to be directly involved in HIF-1α transcription and stability, in which blocking its production halts tumor growth 20.

TKIs have downstream targets in the VHL-growth factor axis, including VEGF and PDGF pathways. Four TKIs are currently available in the United States: Sunitinib, Sorafenib, Pazopanib, and Axitinib. mRCC. All of these TKIs have multiple targets, including VEGF receptor-1, 2 and 3, PDGF-receptor, c-KIT, and FLT-3 21. Their efficacy was confirmed in two trials by Motzer and colleagues that established improved outcomes. In a phase-II trial for patients with mRCC who had failed immunotherapy, Sunitinib provided progression-free survival for 8.5 months, with 35 % experiencing response 22. In a follow-up, randomized, phase-III study, Sunitinib outperformed interferon with a progression-free survival benefit of 11 months vs. 5 months, and a response rate of 31 % vs 5 % based on the guidelines of Response Evaluation Criteria In Solid Tumors (RECIST) 23.

Bevacizumab is a humanized, anti-VEGF, monoclonal antibody that inhibits angiogenesis by blocking the signaling cascade produced by the aberrant VHL activation that is common to RCC 24. This antibody has demonstrated efficacy in many tumor types because it directly targets the VEGF signaling pathway 25. Based on several randomized trials aimed at determining efficacy, this agent is used as second-line therapy in combination with IFN for patients who have failed one of the first-line TKIs, as described above 9, 2628.

Markers in Targeted Therapy

Clinical parameters have been the mainstay of management and prognosis upon disease diagnosis, which relies on imaging in the case of RCC. The recent rise in the incidental diagnosis of small renal masses is due to frequent abdominal imaging, and ultimately, the choice of surgical approach is based on imaging parameters. Upon tumor removal, follow-up imaging is recommended to rule out recurrent renal masses. RECIST is used to gauge the response to treatment in mRCC, among other advanced tumors, and relies upon imaging, among several other parameters, to track tumor size during treatment. These criteria have been used for three decades and are validated for evaluating tumor response to treatment 29.

Role of Markers in Targeted Therapy

Biomarkers are used in oncology as indicators of tumor status and may be involved in the entire management process for certain malignancies. A tumor marker may allow for efficient screening, leading to diagnosis, surveillance for recurrence after treatment, and/or prediction of response to treatment. Screening markers preferably should be noninvasive and easy to test. For RCC, a diagnostic tumor marker may distinguish between benign and malignant lesions. Such distinction and early detection of RCC is crucial since up to 1/3 of patients have metastatic disease at the time of diagnosis. However, the use of a marker for screening the general population for RCC is not feasible, since the incidence of RCC in the general population is only about 0.01 %. However, markers that detect mRCC early and predict treatment response can improve clinical outcome in terms of cancer-specific survival, while avoiding unnecessary side effects from treatments that are ineffective against a patient’s disease. To date, most prognostic tumor markers that have been evaluated for RCC are tissue-based. Markers such as those in the hyaluronic acid family and certain chemokine and cheomkine receptors have shown efficacy in predicting the development of metastasis in the future, based on the their expression in primary tumors 6, 7. Molecular markers predictive of treatment response can be used to monitor response during administration of therapy. Further, pretreatment molecular characterization can help design personalized treatment regimens based on a tumor’s molecular profile. An ideal tumor marker should have high sensitivity and specificity, and it should be easy to perform.

Evaluating the efficacy of markers in advanced RCC is challenging because the disease has a heterogeneous response to the targeted therapies available. Evaluation of the cellular architecture of tumors by histology does not account for the molecular heterogeneity inherent to advanced RCC. These molecular differences account for a heterogeneous tumor behavior and treatment response to targeted therapy. Several groups have confirmed that molecular markers may predict tumor behavior, in addition to its response to treatment. Predicting this response saves time associated with treatment failure. Moreover, targeting the most effective treatment has the potential to save significant healthcare dollars 30.

Tissue-based Markers

Immunohistochemistry is used to elucidate an unclear diagnosis or classify tumor subtype, among other uses. Certain immunohistochemical markers not only have diagnostic potential, but may also accurately predict metastasis and/or response to prognosis and treatment 6, 7. These tissue-based markers may be used at the time of initial surgery or biopsy, however, they are limited beyond the time of diagnosis, as one cannot follow treatment response by repeatedly taking tissue samples from the tumor bed or yet-to-be-discovered sites of micrometastasis.

CA-9

Carbonic anhydrase 9 (CA9) is a common immunohistochemical marker in the diagnosis of clear cell RCC (ccRCC). It is a cell-surface enzyme that is overexpressed in upwards of 90 % of ccRCC cases, and its expression is regulated by HIF-1α and the inactivation of VHL 3133. Specifically, it contributes to maintaining a neutral intracellular pH and acidic extracellular space. CA9 expression can be detected directly in tissues or in tissue sampling through fine needle aspiration by immunohistochemistry or real-time PCR, and in the serum using a CA9 ELISA. Several groups have retrospectively correlated CA9 expression to prognosis. Bui and colleagues correlated CA9-stained tumor tissue with outcome and found that a low CA9 expression was associated with a poor prognosis in high-risk patients (T stage >2 and Fuhrman grade >1). Specifically, high-risk patients with <85 % CA9 expression had a shorter survival than those with >85 % expression 34. Several groups have confirmed that low expression of CA9 on tumor specimens correlates with a lower disease-specific survival, and correlates with other prognostic molecular and clinical markers 31, 35, 36. Moreover, in one study, VHL gene status was also found to correlate with CA9 levels, demonstrating that a complete absence of the VHL gene carries a worse prognosis than a VHL mutation when combined with low CA9 expression 31.

Genetic markers

Genetic factors may play an important role in regulating treatment tolerability and efficacy in mRCC. Specific genetic abnormalities may be associated with deficiencies along a pathway specifically targeted by a treatment. The knowledge of such genetic alternations (i.e., mutations, amplifications, deletions, chromosomal rearrangements) may be exploited to identify individuals predisposed to treatment failure or toxicity, and to subsequently select an ideal regimen prior to starting treatment. Since TKIs target specific pathways within tumor cells, genetic alterations may result in variable responses. A review of the literature identifies relatively few publications related to genetic markers for TKI response. Garcia-Donas and colleagues studied 101 patients receiving Sunitinib for advanced ccRCC with the aim of characterizing single-nucleotide polymorphisms on relevant genetic targets of Sunitinib, as well as predicting treatment tolerability and response 37. Two polymorphisms of the VEGFR3 gene, rs307826 and rs307821, were identified in 8–9 % of participants and were associated with decreased progression-free survival in multivariate analysis (respective hazard ratio (HR) per polymorphism 1.75–7.30; p=0·0079 and 1.64–6.68; p=0·014). One allele of the CYP3A5*1 gene, rs776746, was identified in 6 % of participants and was associated with drug toxicity related to high metabolism (HR 1·67–8·41; p=0·022).

Choueri et al correlated VHL gene status with treatment response in patients receiving a TKI for advanced ccRCC 38. In brief, an analysis of VHL gene status in paraffin-embedded RCC tissues was correlated with treatment response, and evaluated by RECIST. The authors identified an improved response in univariate analysis among those with loss-of-function mutations in the VHL gene vs. the wild-type form (52 % vs. 31 % p = 0.04). Patients in the study received one of three TKIs (i.e., Sunitinib, Sorafenib or Axitinib) or Bevacizumab. Patients with wild-type VHL did not respond to Bevacizumab or Sorafenib. However, a significant response to Sunitinib and Axitinib was noted, regardless of VHL status, in 63 and 14 patients, respectively. VHL status did not correlate with progression-free survival or overall survival, which brings the utility of this marker into question. Further studies are necessary regarding this target as a marker for treatment response.

Enzymes related to drug metabolism and genetic features of TKI targets along the VEGF and HIF pathway may serve as potential markers for treatment response, progression, resistance, etc. Several medications exist within the TKI class, with different targets and efficacy. Therefore, potential markers of efficacy or progression are unlikely to be extrapolated across medications within the same class. Moreover, TKIs are used to treat various tumor types, including for cancers of the breast and lung. However, tumor biology varies widely, and a successful marker for TKI treatment prognosis in lung cancer may not necessarily have efficacy in RCC. Similar to TKIs for RCC, headway has been made in biomarkers for antitumor activity and in the pharmacogenetics of TKIs used in other cancers, such as lung and esophageal 39, 40. Xu and colleagues studied polymorphisms related to Pazopanib’s mechanism of action (multi-target TKI for VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-α/β, and c-kit), metabolism, and involvement with angiogenesis in 397 patients with advanced RCC. Eight polymorphisms related to cytokine metabolism were identified as related to IL-8, HIF1α, NR1|2, and VEGFA that were also significantly associated with progression-free survival (PFS) and response rate. The phenotypes IL-8 2767TT and HIF1α 1790AG were significantly associated with decreased progression-free survival compared to their respective wild types; (IL-8 276TT 27 vs. 48 weeks; HIF1α: 20 vs. 44 weeks). The authors hypothesize that since IL-8 signaling is an alternative pathway for angiogenesis, and HIF1α is a transcription factor for downstream targets that promote angiogenesis, both IL-8 and HIF1α negatively correlate with PFS. Further, patients with a more active form of these genes will experience an inferior anti-angiogenesis effect from Pazopanib. Similar studies have not been published for the other TKIs that have been approved for ccRCC. Further research in this field is warranted. Ideally, a nomogram would predict optimal survival and response to known TKIs based on genetic makeup.

Circulating endothelial cells (CECs)

One of the hallmarks of RCC is the extreme neovascularization associated with tumors, stimulated by VHL and its downstream targets. Mature endothelial cells from these new vessels, or CECs, can be found in the blood in abnormal levels in cancers, including RCC, and other abnormalities of vasculature 41, 42. In patients with localized RCC, circulating endothelial progenitors are elevated and decrease after surgery 43. This was confirmed in a murine xenograft study where CEC (CD31+CD45-) and CEP (CD31+CD45 intermediate CD117+) were significantly elevated in the xenograft model compared to controls 44. It would follow that a marker of angiogenesis would be practical in the setting of advanced RCC, as targeted therapies are anti-angiogenic in nature. Indeed, this has been found in patients treated with Sunitinib for mRCC. The pretreatment levels of CD45dimCD34+VEGFR2+7AAD- progenitor cells were associated with poor PFS and overall survival (OS). Poor PFS was more likely if the levels remained stable or decreased after starting treatment as opposed to increased levels45. A second group investigated CD146+ circulating progenitor cells in patients receiving Sunitinib for advanced RCC, of which 80 % were ccRCC cases 46. Again, baseline CEC levels were noted to be higher than normal controls. Patients with improved PFS exhibited increased CEC levels after 28 days, whereas PFS was notably poor compared to the mean when no significant change in CEC occurred at 1 month. Ultimately, PFS could not be elucidated based on pre-treatment CEC levels.

Serum Markers for Targeted Therapy

Molecules detected in serum satisfy several qualities of the ideal biomarkers. They are minimally invasive, generally cost effective, easy to assay, and hence, convenient for the patient and the clinician. Markers in general can be used to predict the aggressiveness of a disease (prognostic factors) or response to a specific therapy (predictive factors). They can also be used to monitor response to treatment. ccRCC responds in a heterogeneous fashion to the different types of targeted therapy without a clear way to predict this response. For this reason, there is a strong interest in discovering serum markers of prognosis and response to the current available medications. Below, we will review the currently available serum biomarkers for response to treatment and prognosis in advanced RCC.

TKI

The role of the VHL gene in the development of ccRCC has been extensively described. As detailed above, inactivation of the signaling pathways regulated by this gene leads to overexpression of pro-angiogenic elements, such as VEGF, which play a critical role in RCC tumor development. TKIs, which target this pathway, are the mainstay of treatment for advanced ccRCC, and therefore the most widely studied. Several targets of the angiogenic cascade have been evaluated as predictors of prognosis and response to TKI.

VHL

Rini and colleagues retrospectively studied 43 patients with mRCC, treated with either TKIs (i.e., Sunitinib or Axitinib) or IFNα + bevacizumab, and found an increase in time to tumor progression (TTP) between patients with methylation or frame-shift mutations of the VHL gene. (13.3 vs. 7.4 months, p=0.06) 47. This group also found that lower baseline levels of VEGFR-3, and VEGF-C, an isoform of VEGF, were associated with longer PFS and overall response rates in patients receiving Sunitinib or Axitinib in Bevacizumab-refractory disease 48.

VEGF

VEGF is one of the most important products overexpressed in ccRCC. Therapy that targets VEGF and its receptors, VEGF2 and 3, have shown remarkable results in the treatment of RCC. VEGF as a prognostic biomarker was studied within the scope of the TARGET trial; phase 3, randomized, placebo-controlled trial for Sorafenib treatment in patients with advanced ccRCC who were previously treated with standard therapy 49. In this study, baseline serum VEGF levels in 712 patients were shown to correlate inversely with PFS and OS. High baseline levels of VEGF were also associated with higher MSKCC score and ECOG performance score, respectively, along with markers of poor prognosis and performance 50, 51. Multivariate analysis revealed baseline VEGF to be an independent factor for PFS in placebo patients. VEGF was also evaluated as a predictive factor, however both high- and low-VEGF groups derived benefit from Sorafenib treatment and no significant difference was seen.

A follow-up paper by Pena et al. confirmed that patients with baseline VEGF levels higher than the median had a shorter OS interval than those with low baseline VEGF levels (12.7 v 18 mo., HR 1.645, P=0.0027) in the placebo group. They also demonstrated improved PFS in patients who had VEGF levels greater than 75 percentile when treated with Sorafenib compared to placebo (HR 0.33 v 069 P=0.023). VEGF increased in the Sorafenib-treated group in subsequent measurements performed at treatment weeks 3 and 12. These changes were not seen in the placebo group. However, the magnitude of change did not correlate with PFS or OS.52

VEGF has also shown promise as a marker in patients treated with other medications. Porta and colleagues evaluated baseline levels of VEGF in patients treated with Sunitinib and found a relative risk for progression of 2.14 in patients with levels above the normal threshold 53. De Primo and colleagues measured levels of VEGF, VEGFR-2, VEGFR-3 and Placental Growth Factor (PlGF) at day 1 and day 28 of each treatment cycle with Sunitinib (4 weeks treatment, 2 weeks off). VEGF decreased and VEGFR-2 and VEGFR-3 increased during treatment, and then returned to baseline levels between cycles, strongly suggesting a drug-induced response. Mean fold change was significantly larger in patients exhibiting partial response compared to those with stable or progressive disease 54.

Serum Amyloid Alpha (SAA)

SAAs are a group of apolipoproteins associated with the inflammatory response. SAA proteins have been shown to be a prognostic indicator for OS in mRCC 55. Baseline SAA was measured by conventional antibody-directed enumeration assays in a cohort of 114 patients with mRCC, mostly treated with interferon-based therapy. Elevated SAA was found to be an independent prognostic factor for decreased PFS and OS. These findings were then confirmed in a validation cohort of 151 patients treated with Sunitinib or Sorafenib. SAA alone had a similar accuracy in determining OS when compared to the MSKCC model, and improved this model’s accuracy when it was included as a risk factor. In a follow-up study, Vermaat and colleagues re-measured SAA at 6–8 weeks after initiation of treatment. Persistently low SAA was correlated with improved PFS when compared with increasing SAA, declining SAA and persistently elevated SAA 56.

Neutrophil Gelatinase-Associated Lipocalin (NGAL)

NGAL is a 25 kD protein produced by innate immunity cells, as well as tumor cells, and forms complexes with MMP-9 to protect it from degradation—thus enhancing tumor growth 57. NGAL levels were measured in 85 patients prior to the initiation of Sunitinib treatment by immunohistochemistry 57. Patients were classified as favorable (n=46) and intermediate (n=3) according to MSKCC criteria. There were no poor-risk patients. Patients with baseline NGAL levels above normal had an increased risk of progression (RR 1.86, 95% CI 1.142–3.019) when compared to patients with normal NGAL levels.

Erythrocyte Sedimentation Rate (ESR)

Inflammatory marker ESR has been previously associated with poor survival in RCC 58. Zhang and Colleagues studied ESR as a predictive factor in 83 patients who failed immunotherapy with interferon and were treated with Sorafenib 59. ESR was measured by Westergren method before initiation of treatment and every 4 weeks thereafter. Patients were divided into three groups according to ESR kinetics (increased, decreased, and stable). Patients with decreasing ESR levels had longer PFS than the stable and increasing ESR groups (27, 12 and 6 mos., respectively). Median OS was 37 months in the stable ESR group and 13 months in the increasing ESR group. Median survival in the decreasing ESR group was not reached. ESR kinetics remained an independent predictor of PFS in a multivariate analysis where Karnofsky Performance Status (KPS), method of nephrectomy, time from nephrectomy to first administration of Sorafenib, lung metastasis, anemia, LDH and baseline ESR were also evaluated. Of note, baseline ESR was not a predictor of response to Sorafenib.

MMP-9 and TNFα

MMP-9 is a member of the metalloproteinase family shown to be involved in angiogenesis and metastatic growth 60. Tumor cells secrete TNFα, acting as an autocrine growth factor. Elevated levels of TNFα have also been shown to correlate with poor prognosis (Harrison). Serum samples of 31 patients with mRCC who were treated with Sunitinib were screened for 174 cytokines as potential markers; MMP-9 and TNFα were elevated in patients who progressed (p<0.05). Furthermore, lower baseline levels of MMP-9 and TNFα were associated with longer TTP and OS. MMP-9 decreased with Sunitinib treatment, regardless of response 61.

Markers for mTOR inhibitors

LDH

LDH is a validated prognostic marker for several malignancies, including RCC 62, 63. Armstrong and colleagues compared serum LDH samples in 404 subjects with MSKCC, poor-risk, advanced RCC treated with IFNα or Temsirolimus. Among patients with LDH levels above the upper limit of normal, OS was improved in the group treated with Temsirolimus when compared to the IFNα-treated group. This OS benefit was not seen in patients with normal LDH levels. LDH levels during treatment were also evaluated but yielded conflicting results. LDH decline was associated with longer OS in the IFNα group and with worse prognosis in the Temsirolimus group 64.

Cholesterol

A recent study by Lee and colleagues evaluated changes in cholesterol levels during treatment as a marker of response. A total of 416 subjects with intermediate- to poor-risk, advanced RCC were randomized to receive either IFNα or Temsirolimus. Baseline cholesterol, triglyceride and fasting glucose levels were repeated every 2 weeks. Serum cholesterol had a mean increase of 37 mg/dL in the Temsirolimus group but no significant change was observed in the IFNα group. The reduction in risk of death was calculated via univariate analysis to be 18 % for every 39 mg/dL (1mmol/L) of increase in serum cholesterol. This association remained significant in multivariate analysis. No association was seen between OS and triglyceride or fasting glucose levels 65.

Marker combination

In an effort to improve the prognostic and predictive significance of serum markers, combinations have been studied. Tran and colleagues evaluated 17 cytokine and angiogenic factors for response in 344 subjects with Pazopanib versus placebo. Low baseline levels of four of these cytokine and angiogenic factors (Hepatocyte growth factor, IL-8, osteopontin, TIMP-1) were associated with prolonged PFS in patients treated with Pazopanib. In the placebo group, IL-6, IL-8 and OPN were prognostic for PFS. Patients with an elevated IL-6 derived a greater relative benefit than those who received Pazopanib versus placebo 66.

Zurita and colleagues measured the levels of 52 cytokine/angiogenic factors in plasma samples of 69 mRCC patients treated with Sorafenib or Sorafenib + IFNα combination. A “signature” pattern of six factors (OPN, VEGF, TNF-related apoptosis-inducing ligand (TRAIL), ColIV, and sVEGFR2) was found correlate with PFS. “Signature positive” subjects had a longer PFS when treated with Sorafenib when compared with the combination group, whereas, the opposite was true for the “signature negative” group 67.

Conclusion

Research strongly points toward the molecular characterization of individual tumors as a promising method of predicting treatment response and toxicity to targeted therapy in metastatic RCC. However, individual markers have yet to be validated. Large scale, multi-centered prospective trials are necessary to confirm marker validity, making them clinically practical in everyday patient treatment. Eventually, markers can be combined to form a panel of markers that will facilitate individualized patient treatment. Moreover, effective markers for monitoring disease recurrence would allow for earlier intervention, before tumors are visible on CT scan or MRI.

Table 1.

Summary of studies on biomarkers for predicting response to targeted therapies for mRCC

Marker Author Sensitivity/ specificity Method Medication Applicability
Carbonic anhydrase IX Bui et al. 2003 NC Immunohistoc hemistry, Rt-PCR, ELISA None Prognostic factor
VEGFR3 polymorphism: rs307826, rs307821 Garcia-Donas, et al. 2011 NC Single nucleotide polymorphism Sunitinib Predictive factor
IL-8 2767TT, HIF1α 1790AG phenotypes Xu et al. 2011 NC Sequencing Pazopanib Predictive factor
CD45dimCD34+VE GFR2+7AAD- Farace et al. 2011 NC Flow cytometry Sunitinib Clinical efficacy
CD146+ Gruenwald et al. 2010 NC Flow cytometry Sunitinib Clinical efficacy
VHL mutation/methylation Rini et al. 2006 NC PCR, DNA sequencing Interferon-α + Bevacizumab Predictive factor
VEGFR-3 Rini et al. 2008
Escudier et al, 2009
DePrimo et al, 2007
NC ELISA
ELISA
Sunitinib
Sorafenib
Sunitinib
Clinical efficacy
Prognostic factor
VEGF-C Rini et al. 2008 NC ELISA Sunitinib Clinical efficacy
VEGFR-2 Escudier et al, 2009
DePrimo et al, 2007
NC ELISA
ELISA
Sunitinib
Sunitinib
Prognostic factor
VEGF Escudier et al, 2009
Pena et al, 2010
Porta et al, 2010
DePrimo et al, 2007
NC ELISA
ELISA
ELISA
ELISA
Sunitinib
Sorafenib
Sunitinib
Sunitinib
Prognostic factor
Prognostic/Predictive factor
Prognostic factors
SAA Vermaat et al, 2012 59% / 77% ELISA Sunitinib
Sorafenib
Prognostic factor
NGAL Perrin et al, 2011 NC ELISA Sunitinib Prognostic factor
ESR Zhang et al, 2011 NC Westegren Sorafenib Predictive factor
MMP-9 Perez-Garcia et al, 2009 55.6% / 75% ELISA Sunitinib Prognostic factor
TNFα Perez-Garcia et al, 2009 88% / 75% ELISA Sunitinib Prognostic factor
Hepatocyte growth factor, IL-8, OPN, TIMP-1) Tran et al, 2012 NC ELISA Pazopanib Prognostic factor
OPN, VEGF, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), ColIV and sVEGFR2 Zurita et al, 2012 NC Multiplex bead array, ELISA Sorafenib Predictive factor
LDH Armstronge et al, 2012 NC Temsirolimus Predictive factor
Cholesterol Lee et al, 2012 NC Temsirolimus Predictive factor

Abbreviations

CA-9

Carbonic anhydrase-9

cRCC

Clear cell renal cell carcinoma

CRCs

Circulating endothelial cells

ESR

Erythrocyte sedimentation rate

HIF

Hypoxia inducible factor

IFN

interferon

IL

Interleukin

LDH

Lactate dehydrogenase

MMP-9

Matrix metalloproteinase-9

mTOR

Mammalian Target of Rapamycin

mRCC

Metastatic renal cell carcinoma

NGAL

Neutrophil Gelatinase Associated Lipocalin

OS

Overall survival

PDGF

Platelet-derived growth factor

PFS

Progression-free survival

RCC

Renal cell carcinoma

RECIS

Response Evaluation Criteria in Solid Tumors

SAA

Serum amyloid alpha

TKI

Tyrosine kinase inhibitor(s)

TNF

Tumor necrosis factor

TTP

Tumor progression rate

VEGF

Vascular endothelial growth factor

VHL

Von Hipple Lindau

Footnotes

Conflict of Interest

Dr. Michael Garcia-Roig, Dr. Nicolas Ortiz, and Dr. Vinata Lokeshwar declare that there are no conflicts of interest relevant to this article.

Compliance with Ethics Guidelines

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

◆ Of importance

◆ ◆ Of outstanding importance

  • 1.Palapattu GS, Kristo B, Rajfer J. Paraneoplastic syndromes in urologic malignancy: the many faces of renal cell carcinoma. Rev Urol. 2002;4:163. [PMC free article] [PubMed] [Google Scholar]
  • 2.Bukowski RM. Natural history and therapy of metastatic renal cell carcinoma: the role of interleukin-2. Cancer. 1997;80:1198. doi: 10.1002/(sici)1097-0142(19971001)80:7<1198::aid-cncr3>3.0.co;2-h. [DOI] [PubMed] [Google Scholar]
  • 3.Motzer RJ, Russo P. Systemic therapy for renal cell carcinoma. J Urol. 2000;163:408. [PubMed] [Google Scholar]
  • 4.Campbell SC, Novick AC, Belldegrun A, et al. Guideline for management of the clinical T1 renal mass. J Urol. 2009;182:1271. doi: 10.1016/j.juro.2009.07.004. [DOI] [PubMed] [Google Scholar]
  • ◆ ◆5.Ljungberg B, Cowan NC, Hanbury DC, et al. EAU guidelines on renal cell carcinoma: the 2010 update. Eur Urol. 2010;58:398. doi: 10.1016/j.eururo.2010.06.032. The article contains guidelines and updated recommendations for the diagnosis, treatment, and follow-up of individual patients, in order to improve clinical management of renal cell carcinoma. [DOI] [PubMed] [Google Scholar]
  • 6.Chi A, Shirodkar SP, Escudero DO, et al. Molecular characterization of kidney cancer. Cancer. 2012;118:2394. doi: 10.1002/cncr.26520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gahan JC, Gosalbez M, Yates T, et al. Chemokine and chemokine receptor expression in kidney tumors: molecular profiling of histological subtypes and association with metastasis. J Urol. 2012;187:827. doi: 10.1016/j.juro.2011.10.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Flanigan RC, Mickisch G, Sylvester R, et al. Cytoreductive nephrectomy in patients with metastatic renal cancer: a combined analysis. J Urol. 2004;171:1071. doi: 10.1097/01.ju.0000110610.61545.ae. [DOI] [PubMed] [Google Scholar]
  • 9.Rini BI, Campbell SC. The evolving role of surgery for advanced renal cell carcinoma in the era of molecular targeted therapy. J Urol. 2007;177:1978. doi: 10.1016/j.juro.2007.01.136. [DOI] [PubMed] [Google Scholar]
  • 10.Mues AC, Haramis G, Rothberg MB, et al. Contemporary experience with laparoscopic radical nephrectomy. J Laparoendosc Adv Surg Tech A. 2011;21:15. doi: 10.1089/lap.2010.0345. [DOI] [PubMed] [Google Scholar]
  • ◆11.Hellenthal NJ, Underwood W, Penetrante R, et al. Prospective clinical trial of preoperative sunitinib in patients with renal cell carcinoma. J Urol. 2010;184:859. doi: 10.1016/j.juro.2010.05.041. In a single institution, prospective clinical trial for patients with localized or metastatic, clear-cell, renal cell carcinoma, neoadjuvant treatment with Sunitinib was shown to decrease the size of the primary tumor and was well-tolerated. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gorin MA, Ekwenna O, Soloway MS, et al. Dramatic reduction in tumor burden with neoadjuvant sunitinib prior to bilateral nephron-sparing surgery. Urology. 2012;79:e11. doi: 10.1016/j.urology.2011.04.018. [DOI] [PubMed] [Google Scholar]
  • 13.Kanzaki R, Higashiyama M, Fujiwara A, et al. Long-term results of surgical resection for pulmonary metastasis from renal cell carcinoma: a 25-year single-institution experience. Eur J Cardiothorac Surg. 2011;39:167. doi: 10.1016/j.ejcts.2010.05.021. [DOI] [PubMed] [Google Scholar]
  • 14.Meimarakis G, Angele M, Staehler M, et al. Evaluation of a new prognostic score (Munich score) to predict long-term survival after resection of pulmonary renal cell carcinoma metastases. Am J Surg. 2011;202:158. doi: 10.1016/j.amjsurg.2010.06.029. [DOI] [PubMed] [Google Scholar]
  • 15.Murthy SC, Kim K, Rice TW, et al. Can we predict long-term survival after pulmonary metastasectomy for renal cell carcinoma? Ann Thorac Surg. 2005;79:996. doi: 10.1016/j.athoracsur.2004.08.034. [DOI] [PubMed] [Google Scholar]
  • 16.Pfannschmidt J, Hoffmann H, Muley T, et al. Prognostic factors for survival after pulmonary resection of metastatic renal cell carcinoma. Ann Thorac Surg. 2002;74:1653. doi: 10.1016/s0003-4975(02)03803-1. [DOI] [PubMed] [Google Scholar]
  • 17.Russo P, O’Brien MF. Surgical intervention in patients with metastatic renal cancer: metastasectomy and cytoreductive nephrectomy. Urol Clin North Am. 2008;35:679. doi: 10.1016/j.ucl.2008.07.009. [DOI] [PubMed] [Google Scholar]
  • 18.Negrier S, Escudier B, Lasset C, et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal-cell carcinoma. Groupe Francais d’Immunotherapie. N Engl J Med. 1998;338:1272. doi: 10.1056/NEJM199804303381805. [DOI] [PubMed] [Google Scholar]
  • 19.Gnarra JR, Tory K, Weng Y, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7:85. doi: 10.1038/ng0594-85. [DOI] [PubMed] [Google Scholar]
  • 20.Hudson CC, Liu M, Chiang GG, et al. Regulation of hypoxia-inducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol. 2002;22:7004. doi: 10.1128/MCB.22.20.7004-7014.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chow LQ, Eckhardt SG. Sunitinib: from rational design to clinical efficacy. J Clin Oncol. 2007;25:884. doi: 10.1200/JCO.2006.06.3602. [DOI] [PubMed] [Google Scholar]
  • 22.Motzer RJ, Rini BI, Bukowski RM, et al. Sunitinib in patients with metastatic renal cell carcinoma. JAMA: The Journal of the American Medical Association. 2006;295:2516. doi: 10.1001/jama.295.21.2516. [DOI] [PubMed] [Google Scholar]
  • 23.Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115. doi: 10.1056/NEJMoa065044. [DOI] [PubMed] [Google Scholar]
  • 24.Presta LG, Chen H, O’Connor SJ, et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 1997;57:4593. [PubMed] [Google Scholar]
  • 25.Shih T, Lindley C. Bevacizumab: an angiogenesis inhibitor for the treatment of solid malignancies. Clin Ther. 2006;28:1779. doi: 10.1016/j.clinthera.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 26.Bukowski RM, Kabbinavar FF, Figlin RA, et al. Rrrandomized phase II study of erlotinib combined with bevacizumab compared with bevacizumab alone in metastatic renal cell cancer. J Clin Oncol. 2007;25:4536. doi: 10.1200/JCO.2007.11.5154. [DOI] [PubMed] [Google Scholar]
  • 27.Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370:2103. doi: 10.1016/S0140-6736(07)61904-7. [DOI] [PubMed] [Google Scholar]
  • 28.Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 2003;349:427. doi: 10.1056/NEJMoa021491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ◆ ◆29.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1. 1) Eur J Cancer. 2009;45:228. doi: 10.1016/j.ejca.2008.10.026. This article summarizes the changes in RECIST criteria that were originally published in the year 2000. The major change in the criteria is that the number of lesions required to assess tumor burden for response determination was reduced from a maximum of 10 to a maximum of five total (and from five to two per organ, maximum). Assessment of pathological lymph nodes was also included. In addition, the definition of disease progression was classified further. [DOI] [PubMed] [Google Scholar]
  • 30.Atherly AJ, Camidge DR. The cost-effectiveness of screening lung cancer patients for targeted drug sensitivity markers. Br J Cancer. 2012;106:1100. doi: 10.1038/bjc.2012.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Patard JJ, Fergelot P, Karakiewicz PI, et al. Low CAIX expression and absence of VHL gene mutation are associated with tumor aggressiveness and poor survival of clear cell renal cell carcinoma. Int J Cancer. 2008;123:395. doi: 10.1002/ijc.23496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wykoff CC, Beasley NJ, Watson PH, et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000;60:7075. [PubMed] [Google Scholar]
  • 33.Sandlund J, Oosterwijk E, Grankvist K, et al. Prognostic impact of carbonic anhydrase IX expression in human renal cell carcinoma. BJU Int. 2007;100:556. doi: 10.1111/j.1464-410X.2007.07006.x. [DOI] [PubMed] [Google Scholar]
  • 34.Bui MH, Seligson D, Han KR, et al. Carbonic anhydrase IX is an independent predictor of survival in advanced renal clear cell carcinoma: implications for prognosis and therapy. Clin Cancer Res. 2003;9:802. [PubMed] [Google Scholar]
  • 35.Phuoc NB, Ehara H, Gotoh T, et al. Prognostic value of the co-expression of carbonic anhydrase IX and vascular endothelial growth factor in patients with clear cell renal cell carcinoma. Oncol Rep. 2008;20:525. [PubMed] [Google Scholar]
  • 36.Zhou GX, Ireland J, Rayman P, et al. Quantification of carbonic anhydrase IX expression in serum and tissue of renal cell carcinoma patients using enzyme-linked immunosorbent assay: prognostic and diagnostic potentials. Urology. 2010;75:257. doi: 10.1016/j.urology.2009.09.052. [DOI] [PubMed] [Google Scholar]
  • 37.Garcia-Donas J, Esteban E, Leandro-Garcia LJ, et al. Single nucleotide polymorphism associations with response and toxic effects in patients with advanced renal-cell carcinoma treated with first-line sunitinib: a multicentre, observational, prospective study. Lancet Oncol. 2011;12:1143. doi: 10.1016/S1470-2045(11)70266-2. [DOI] [PubMed] [Google Scholar]
  • 38.Choueiri TK, Vaziri SA, Jaeger E, et al. von Hippel-Lindau gene status and response to vascular endothelial growth factor targeted therapy for metastatic clear cell renal cell carcinoma. J Urol. 2008;180:860. doi: 10.1016/j.juro.2008.05.015. [DOI] [PubMed] [Google Scholar]
  • 39.Cusatis G, Gregorc V, Li J, et al. Pharmacogenetics of ABCG2 and adverse reactions to gefitinib. J Natl Cancer Inst. 2006;98:1739. doi: 10.1093/jnci/djj469. [DOI] [PubMed] [Google Scholar]
  • 40.Wang WP, Wang KN, Gao Q, et al. Lack of EGFR mutations benefiting gefitinib treatment in adenocarcinoma of esophagogastric junction. World J Surg Oncol. 2012;10:14. doi: 10.1186/1477-7819-10-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Boos CJ, Lip GY, Blann AD. Circulating endothelial cells in cardiovascular disease. J Am Coll Cardiol. 2006;48:1538. doi: 10.1016/j.jacc.2006.02.078. [DOI] [PubMed] [Google Scholar]
  • 42.Wu H, Chen H, Hu PC. Circulating endothelial cells and endothelial progenitors as surrogate biomarkers in vascular dysfunction. Clin Lab. 2007;53:285. [PubMed] [Google Scholar]
  • 43.Bhatt RS, Zurita AJ, O’Neill A, et al. Increased mobilisation of circulating endothelial progenitors in von Hippel-Lindau disease and renal cell carcinoma. Br J Cancer. 2011;105:112. doi: 10.1038/bjc.2011.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Namdarian B, Tan KV, Fankhauser MJ, et al. Circulating endothelial cells and progenitors: potential biomarkers of renal cell carcinoma. BJU Int. 2010;106:1081. doi: 10.1111/j.1464-410X.2010.09245.x. [DOI] [PubMed] [Google Scholar]
  • 45.Farace F, Gross-Goupil M, Tournay E, et al. Levels of circulating CD45(dim)CD34(+)VEGFR2(+) progenitor cells correlate with outcome in metastatic renal cell carcinoma patients treated with tyrosine kinase inhibitors. Br J Cancer. 2011;104:1144. doi: 10.1038/bjc.2011.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gruenwald V, Beutel G, Schuch-Jantsch S, et al. Circulating endothelial cells are an early predictor in renal cell carcinoma for tumor response to sunitinib. BMC Cancer. 2010;10:695. doi: 10.1186/1471-2407-10-695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rini BI, Jaeger E, Weinberg V, et al. Clinical response to therapy targeted at vascular endothelial growth factor in metastatic renal cell carcinoma: impact of patient characteristics and Von Hippel-Lindau gene status. BJU Int. 2006;98:756. doi: 10.1111/j.1464-410X.2006.06376.x. [DOI] [PubMed] [Google Scholar]
  • 48.Rini BI, Michaelson MD, Rosenberg JE, et al. Antitumor activity and biomarker analysis of sunitinib in patients with bevacizumab-refractory metastatic renal cell carcinoma. J Clin Oncol. 2008;26:3743. doi: 10.1200/JCO.2007.15.5416. [DOI] [PubMed] [Google Scholar]
  • 49.Escudier B, Eisen T, Stadler WM, et al. Sorafenib for treatment of renal cell carcinoma: Final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial. J Clin Oncol. 2009;27:3312. doi: 10.1200/JCO.2008.19.5511. [DOI] [PubMed] [Google Scholar]
  • 50.Motzer RJ, Bacik J, Murphy BA, et al. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol. 2002;20:289. doi: 10.1200/JCO.2002.20.1.289. [DOI] [PubMed] [Google Scholar]
  • 51.Oken MM, Creech RH, Tormey DC, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5:649. [PubMed] [Google Scholar]
  • ◆ ◆52.Pena C, Lathia C, Shan M, et al. Biomarkers predicting outcome in patients with advanced renal cell carcinoma: Results from sorafenib phase III Treatment Approaches in Renal Cancer Global Evaluation Trial. Clin Cancer Res. 2010;16:4853. doi: 10.1158/1078-0432.CCR-09-3343. In a subset of patients (n=903) enrolled in the Treatment Approaches in Renal Cancer Global Evaluation, the authors evaluated levels of plasma proteins, VEGF, TIMP-1, carbonic anhydrase IX, p21Ras, soluble VEGF receptor 2 and VHL gene mutation to determine responses to Sorafenib treatment. Results showed that VEGF, CAIX, TIMP-1, and Ras p21 have prognostic significance for predicting survival in RCC patients, and of these, TIMP-1 has independent prognostic significance. [DOI] [PubMed] [Google Scholar]
  • 53.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;77:809. doi: 10.1038/ki.2009.552. [DOI] [PubMed] [Google Scholar]
  • 54.Deprimo SE, Bello CL, 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. J Transl Med. 2007;5:32. doi: 10.1186/1479-5876-5-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Vermaat JS, van der Tweel I, Mehra N, et al. Two-protein signature of novel serological markers apolipoprotein-A2 and serum amyloid alpha predicts prognosis in patients with metastatic renal cell cancer and improves the currently used prognostic survival models. Ann Oncol. 2010;21:1472. doi: 10.1093/annonc/mdp559. [DOI] [PubMed] [Google Scholar]
  • 56.Vermaat JS, Gerritse FL, van der Veldt AA, et al. Validation of serum amyloid alpha as an independent biomarker for progression-free and overall survival in metastatic renal cell cancer patients. Eur Urol. 2012;62:685. doi: 10.1016/j.eururo.2012.01.020. [DOI] [PubMed] [Google Scholar]
  • 57.Perrin C, Patard JJ, Jouan F, et al. The neutrophil gelatinase-associated lipocalin, or LCN 2, marker of aggressiveness in clear cell renal cell carcinoma. Prog Urol. 2011;21:851. doi: 10.1016/j.purol.2011.06.005. [DOI] [PubMed] [Google Scholar]
  • 58.Sengupta S, Lohse CM, Cheville JC, et al. The preoperative erythrocyte sedimentation rate is an independent prognostic factor in renal cell carcinoma. Cancer. 2006;106:304. doi: 10.1002/cncr.21617. [DOI] [PubMed] [Google Scholar]
  • 59.Zhang HL, Zhu Y, Wang CF, et al. Erythrocyte sedimentation rate kinetics as a marker of treatment response and predictor of prognosis in Chinese metastatic renal cell carcinoma patients treated with sorafenib. Int J Urol. 2011;18:422. doi: 10.1111/j.1442-2042.2011.02761.x. [DOI] [PubMed] [Google Scholar]
  • 60.Bergers G, Brekken R, McMahon G, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol. 2000;2:737. doi: 10.1038/35036374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.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;101:1876. doi: 10.1038/sj.bjc.6605409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Halabi S, Small EJ, Kantoff PW, et al. Prognostic model for predicting survival in men with hormone-refractory metastatic prostate cancer. J Clin Oncol. 2003;21:1232. doi: 10.1200/JCO.2003.06.100. [DOI] [PubMed] [Google Scholar]
  • 63.von Eyben FE. A systematic review of lactate dehydrogenase isoenzyme 1 and germ cell tumors. Clin Biochem. 2001;34:441. doi: 10.1016/s0009-9120(01)00236-3. [DOI] [PubMed] [Google Scholar]
  • 64.Armstrong AJ, George DJ, Halabi S. Serum lactate dehydrogenase predicts for overall survival benefit in patients with metastatic renal cell carcinoma treated with inhibition of mammalian target of rapamycin. J Clin Oncol. 2012;30:3402. doi: 10.1200/JCO.2011.40.9631. [DOI] [PubMed] [Google Scholar]
  • 65.Lee CK, Marschner IC, Simes RJ, et al. Increase in cholesterol predicts survival advantage in renal cell carcinoma patients treated with temsirolimus. Clin Cancer Res. 2012;18:3188. doi: 10.1158/1078-0432.CCR-11-3137. [DOI] [PubMed] [Google Scholar]
  • 66.Tran HT, Liu Y, Zurita AJ, et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 2012;13:827. doi: 10.1016/S1470-2045(12)70241-3. [DOI] [PubMed] [Google Scholar]
  • 67.Zurita AJ, Jonasch E, Wang X, et al. A cytokine and angiogenic factor (CAF) analysis in plasma for selection of sorafenib therapy in patients with metastatic renal cell carcinoma. Ann Oncol. 2012;23:46. doi: 10.1093/annonc/mdr047. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES