126
Novel Biomarkers in Metastatic Renal Cell Carcinoma Patrick Penttilä, M.D Doctoral Programme in Clinical Research Department of Oncology University of Helsinki and Helsinki University Hospital Helsinki, Finland To be publicly discussed with the permission of Faculty of Medicine, University of Helsinki, in the Auditorium of the Department of Oncology, on Friday October 5 th , 2018, at 12 o’clock noon Helsinki 2018

Novel Biomarkers in Metastatic Renal Cell Carcinoma

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Novel Biomarkers in Metastatic Renal Cell Carcinoma

Novel Biomarkers in Metastatic Renal Cell Carcinoma

Patrick Penttilä, M.D

Doctoral Programme in Clinical Research

Department of Oncology

University of Helsinki and Helsinki University Hospital

Helsinki, Finland

To be publicly discussed with the permission of Faculty of Medicine, University of Helsinki, in the Auditorium of the Department of Oncology, on Friday October 5th,2018, at 12 o’clock noon

Helsinki 2018

Page 2: Novel Biomarkers in Metastatic Renal Cell Carcinoma

2

Supervised by

Adjunct Professor and Chief Medical Officer Petri Bono, M.D., Ph.D.

Helsinki University and Helsinki University Hospital

Helsinki, Finland

Reviewed by

Docent Peeter Karihtala, M.D., Ph.D.

Department of Oncology and Radiotherapy

University of Oulu and Oulu University Hospital

Oulu, Finland

and

Docent Peter Boström, M.D., Ph.D.

Department of Urology

University of Turku and Turku University Hospital

Turku, Finland

Discussed with

Professor Axel Bex, M.D., Ph.D.

Department of Urology

Netherlands Cancer Institute

Amsterdam, Netherlands

The Faculty of Medicine uses the Urkund system (plagiarism recognition) to examine all doctoral dissertations.

Page 3: Novel Biomarkers in Metastatic Renal Cell Carcinoma

3

“Let us now pause for a moment of science”

Page 4: Novel Biomarkers in Metastatic Renal Cell Carcinoma

4

Table of Contents

Table of Contents ............................................................................... 4 Original Publications ......................................................................... 7 Abbreviations ..................................................................................... 8 Abstract ............................................................................................ 11 Review of the Literature .................................................................. 13 1. Renal Cell Carcinoma ........................................................................................ 13

1.1 Epidemiology ................................................................................................. 13

1.2 Pathology ........................................................................................................ 14 1.2.1 Clear-Cell Renal Cell Carcinoma ......................................................................14

1.2.2 Papillary Renal Cell Carcinoma .......................................................................15

1.2.3 Chromophobe Renal Cell Carcinoma ...............................................................16

1.2.4 Other Defined Subtypes ....................................................................................16

1.2.5 Sarcomatoid Differentiation in Renal Cell Carcinoma .....................................17

1.3 Genetic Environment ...................................................................................... 17 1.3.1 VHL Gene Alteration .......................................................................................18

1.3.2 Met Proto-oncogene and HPRCC .....................................................................18

1.3.3 Other Hereditary Syndromes ............................................................................19

2. Tumorigenesis and the Tumor Microenvironment ........................................ 19

2.1 Tumorigenesis ................................................................................................ 19 2.1.1 Hypoxia-induced Pathway ................................................................................20

2.1.1.1 HIF- - ...............................................21

2.1.2 mTOR Pathway ................................................................................................23

2.1.3 c-Met Signaling Pathway ..................................................................................24

2.1.4 Angiogenesis ....................................................................................................25

2.2 Tumor Microenvironment ............................................................................. 27 2.2.1 Tumor Escape ...................................................................................................29

2.2.2 Tumor Heterogeneity ........................................................................................31

Page 5: Novel Biomarkers in Metastatic Renal Cell Carcinoma

5

3. Diagnosis and Management of RCC ............................................................... 32

3.1 Diagnosis ....................................................................................................... 32 3.1.1 Imaging ............................................................................................................33

3.1.2 Staging and Pathology Assessment .................................................................. 33

3.1.3 Small Renal Masses........................................................................................... 36

3.2 Prognostication ............................................................................................... 37

3.3 Treatment ........................................................................................................ 39 3.3.1 Cytoreductive Nephrectomy ............................................................................. 39

3.3.2 Metastasectomy and Other Local Therapeutic Options for Metastases ............40

3.3.3 Systemic Therapy .............................................................................................41

3.3.3.1 Immunotherapy ...........................................................................................41

3.3.3.2 Tyrosine Kinase Inhibitors .........................................................................42

3.3.3.3 mTOR Inhibitors ........................................................................................43

3.3.3.4 Novel Approaches ......................................................................................45

3.3.3.5 Optimal Sequencing ...................................................................................47

3.3.3.5 Adjuvant Therapy .......................................................................................50

4. Biomarkers in Renal Cell Carcinoma ............................................................. 53

4.1 Prognostic and Predictive Biomarkers .......................................................... 54 4.1.1 Clinical-related Biomarkers .............................................................................. 55

4.1.2 Vascular Endothelial-derived Growth Factor ................................................... 58

4.1.3 Carbonic Anhydrase IX ....................................................................................59

4.1.4 Cytokine and Angiogenic Factors ..................................................................... 60

4.1.5 Single Nucleotide Polymorphisms .................................................................... 61

4.1.6 Immune Markers .............................................................................................. 62

4.1.7 Mechanism-based Adverse Events .................................................................... 64

4.2 Incorporation of Biomarkers into Clinical Practice and Future Directions .... 66

Aims of the Study ................................................................................................... 67

Patients and Methods............................................................................................. 68

1. Patients and Treatment ..................................................................................... 68 1.1 Assessment of Tumor Response and Adverse Events ..........................................69

1.2 Assessment of Pneumonitis .................................................................................69

Page 6: Novel Biomarkers in Metastatic Renal Cell Carcinoma

6

1.3 Immunohistochemistry ........................................................................................ 70

2. Statistical Analysis ........................................................................................... 71

3. Ethical Considerations ...................................................................................... 72

Results and Discussion ........................................................................................... 73

1. The use of angiotensin system inhibitors (ASIs) in the treatment of TKI-induced hypertension (I) ....................................................................................... 73

2. c-Met expression is associated with the outcome among mRCC patients treated with sunitinib (II) ...................................................................................... 77

3. Everolimus-induced pneumonitis predicts the treatment outcome among mRCC patients treated with everolimus (III) ....................................................... 84

4. Hyponatremia associates with a poor outcome among mRCC patients treated with everolimus (IV) ............................................................................................ 89

Limitations of the Study Materials and Methods ............................................... 95

Conclusions ............................................................................................................. 97

Acknowledgements................................................................................................. 99

References ............................................................................................................. 101

Page 7: Novel Biomarkers in Metastatic Renal Cell Carcinoma

7

Original Publications

This thesis is based on the following publications, which are referred to in the text by their Roman numerals:

I Penttilä P, Rautiola J, Poussa T, Peltola K, Bono P. Angiotensin Inhibitors as Treatment of Sunitinib/Pazopanib-induced Hypertension in Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer. 2017 Jun;15(3):384-390.

II Peltola KJ, Penttilä P, Rautiola J, Joensuu H, Hänninen E, Ristimäki A, Bono P. Correlation of c-Met Expression and Outcome in Patients With Renal Cell Carcinoma Treated With Sunitinib. Clin Genitourin Cancer. 2017 Aug;15(4):487-494.

III Penttilä P, Donskov F, Rautiola J, Peltola K, Laukka M, Bono P. Everolimus-induced pneumonitis associates with favourable outcome in patients with metastatic renal cell carcinoma. Eur J Cancer 2017 Aug;81:9-16.

IV Penttilä P, Bono P, Peltola K, Donskov F. Hyponatremia associates with poor outcome in metastatic renal cell carcinoma patients treated with everolimus: Prognostic impact. Acta Oncol 2018, 2018 Jun;4:1-6

Page 8: Novel Biomarkers in Metastatic Renal Cell Carcinoma

8

Abbreviations

AE Adverse event

Ang-1 & -2 Angiopoietin 1 and -2

AS Active surveillance

ASI Angiotensin system inhibitor

BHD Birt-Hogg-Dubé

c-Met Tyrosine protein kinase Met

CAFs Cytokine and angiogenic factors

CAIX Carbonic anhydrase IX

ccRCC Clear-cell renal cell carcinoma

chRCC Chromophobe renal cell carcinoma

ColIV Collagen IV

CR Complete response

ctDNA Circulating tumor DNA

DFS Disease-free survival

EC Endothelial cells

ECOG Eastern Cooperative Oncology Group

ESMO European Society for Medical Oncology

FDA Food and Drug Administration

FH Fumarate hydratase

FLCN Folliculin

HGF Hepatocyte growth factor

HIF Hypoxia-induced factor

HLRCC Hereditary leiomyomatosis and renal cell cancer

Page 9: Novel Biomarkers in Metastatic Renal Cell Carcinoma

9

HPRC Hereditary papillary renal cancer

HTN Hypertension

IFN Interferon

IL Interleukin

IMDC International Metastatic Renal Cell Carcinoma Database Consortium

LDH Lactate dehydrogenase

MDSC Myeloid suppressor cells

MiRNA MicroRNA

MMP Matrix metalloproteinase

mRCC Metastatic renal-cell carcinoma

MSKCC Memorial Sloan Kettering Cancer Center

mTOR Mammalian target of rapamycin

NK Natural killer

ORR Objective response rate

OS Overall survival

PD Progressive disease

PD-1 Programmed death 1

PD-L1 Programmed death ligand-1

PDGF Platelet-derived growth factor

PLGF Placenta growth factor

PFS Progression-free survival

PR Partial response

pRCC Papillary renal cell carcinoma

PTP1B Protein tyrosine phosphatase 1B

PTEN Phosphatase and tensin homologue

RCC Renal cell carcinoma

Page 10: Novel Biomarkers in Metastatic Renal Cell Carcinoma

10

SD Stable disease

SDHB Succinate dehydrogenase B

SDHD Succinate dehydrogenase D

SMGs Significantly mutated genes

SNP Single nucleotide polymorphism

SRM Small renal mass

TGF- transforming growth factor

TIMP Tissue inhibitor of metalloproteinases

TKI Tyrosine kinase inhibitor

TNM Tumor-node-metastasis

TRAIL Tumor necrosis factor-related apoptosis-inducing ligand

TSP-1 Thrombospondin 1

T-regs Regulatory T-cells

VEGF Vascular endothelial growth factor

VEGFR Vascular endothelial growth factor receptor

VHL von Hippel-Lindau

Page 11: Novel Biomarkers in Metastatic Renal Cell Carcinoma

11

ABSTRACT

During the past decade, basic and translational cancer research has provided new

information regarding the mechanisms underlying tumor growth, proliferation, and

metastasis in renal cell carcinoma (RCC). Understanding of the molecular

pathogenesis of RCC has identified new targets for therapeutic intervention.

Angiogenesis, involving complex signaling pathways such as vascular endothelial

growth factor (VEGF), platelet-derived growth factor, and angiopoietins, plays a

pivotal role in tumor growth and progression, and therapies targeting angiogenic

factors from the VEGF family have become an effective strategy in the treatment of

metastatic renal-cell carcinoma (mRCC) since 2006.

Sunitinib and pazopanib are tyrosine kinase inhibitors (TKI) and they are globally

used as first-line treatments for mRCC. Although they primarily act through

inhibiting angiogenesis, they might also have an effect on the function of immune

cells such as T-regs. Despite the initial enthusiasm for these modern-era targeted

therapies, however, some patients only receive moderate benefit, and resistance to

these therapies eventually develops in all patients. At present, no biomarkers

predicting treatment efficacy are known, despite intensive translational research

during the last decade.

While the main component of the angiogenic process in RCC is VEGF, targeting of

the mammalian target of the rapamycin (mTOR) pathway is important, because

activation of the upstream PI3K/Akt/mTOR signaling pathways is one method by

which constitutive HIF- imus is an orally

available mTOR inhibitor and is commonly used after TKI treatment

failure/intolerance. As with TKI inhibitors, however, there are wide differences in

Page 12: Novel Biomarkers in Metastatic Renal Cell Carcinoma

12

responses to everolimus treatment, and it remains unclear which patients most

benefit from the treatment.

Our results confirm TKI-induced hypertension (HTN) as a strong predictor of a good

prognosis and response among patients treated with sunitinib/pazopanib. We

additionally demonstrated that the use angiotensin system inhibitors (ASIs) in the

treatment of TKI-induced HTN may have synergistic beneficial effects when used in

conjunction with sunitinib or pazopanib.

In our investigation of tumor tyrosine protein kinase Met (c-Met) expression and the

outcome, we demonstrated that high levels of c-Met expression associate with a

worse prognosis among mRCC patients treated with sunitinib. We also demonstrated

that high c-Met expression is associated with a poor outcome among patients without

bone metastases at baseline, suggesting that the prognostic role may vary depending

on the location of the metastases.

In our two studies conducted on everolimus-treated patients, we showed that

everolimus-induced pneumonitis associates with an improved outcome, suggesting

a role as a surrogate marker of the efficacy of everolimus treatment. We additionally

confirmed the prognostic value of hyponatremia in a large cohort of everolimus-

treated patients.

Page 13: Novel Biomarkers in Metastatic Renal Cell Carcinoma

13

Review of the Literature

1. Renal Cell Carcinoma

1.1 Epidemiology

Kidney cancer accounts for 5% and 3% of all adult malignancies in men and women,

respectively, making it the sixth most common cancer in men and tenth most

common in women. The reported figures, however, also include urothelial cancer of

the renal pelvis; renal cell carcinoma (RCC) accounts fo .

(1)

In Finland, the reported incidence of RCC in 2003–2007 was 9.1 per 100 000 men

and 5.8 per 100 000 women (2). Worldwide, as well as in Finland, RCC incidence

has shown signs of plateauing, with a simultaneous decrease in mortality rates (3, 4).

This phenomenon is at least partly explained by earlier detection, allowing earlier

intervention when the disease in potentially curable; currently, more than 50% of

RCCs are detected incidentally (5, 6)

Well-known risk factors for RCC include cigarette smoking, hypertension, and

obesity (7-14). Evidence also suggests that patients with end-stage renal failure,

acquired renal cystic disease, tuberous sclerosis syndrome, and patients who undergo

kidney transplantation are at increased risk of developing RCC (15-18).

Additionally, epidemiologic studies have demonstrated that a family history of RCC

is a risk factor for the disease (19, 20), and indeed, several hereditary syndromes

predisposing patients to RCC have been identified.

Page 14: Novel Biomarkers in Metastatic Renal Cell Carcinoma

14

In the United States, the overall 5-year cancer-specific survival (CSS) for patients

with histologically confirmed kidney cancer of all stages between 1975 and 2009

was 60.4%. The stage-specific 5-year CSS rates were 87.0% for the localized stage,

62.9% for the regional stage, and 9.3% for the distant stage. (21)

1.2. Pathology

RCC is an extremely heterogeneous disease and comprises several different

subclasses, the most common being clear-cell (75%), papillary (7–15%), and

chromophobe (3–5%) carcinoma (22, 23). In addition to these common histological

variants, several relatively uncommon subclasses such as collecting duct carcinoma,

renal medullary carcinoma, tubulocystic renal cell carcinoma, and acquired cystic

disease-associated renal cell carcinoma have been identified (23). Recent important

changes from existing tumor types also include classification according to molecular

alterations and familial predisposition syndromes. These subclasses, however, only

account for less than one percent of all RCCs (23).

1.2.1 Clear-cell Renal Cell Carcinoma

Clear-cell renal cell carcinoma (ccRCC) is the most common variant of all RCCs and

it originates from the epithelium of the proximal convoluted tubules (23).

Histologically, ccRCCs are characterized by cells with a lipid- and glycogen-rich

cytoplasmic content and frequently also present with an eosinophilic granular

cytoplasm. Microscopically, an alveolar, acinar, or solid architectural pattern is

commonly detected (24). Most cases of ccRCC are sporadic, with only 5% being

Page 15: Novel Biomarkers in Metastatic Renal Cell Carcinoma

15

associated with hereditary syndromes such as von Hippel-Lindau disease and

tuberous sclerosis. Both sporadic and hereditary forms of the disease are strongly

associated with deletion in chromosome 3p, the site of the von Hippel-Lindau gene,

with deletions at this site found in up to 96% of ccRCCs (25, 26).

1.2.2 Papillary Renal Cell Carcinoma

Papillary renal cell carcinoma (pRCC) is histologically characterized by a papillary

or tubulopapillary architecture. The proportion of papillae varies and they often

present with hemorrhage, necrosis, and cystic degeneration (27). Like ccRCC, pRCC

may occur sporadically or in association with hereditary syndromes, and it is

typically associated with trisomies of chromosomes 7, 16, and 17, renal cortical

adenomas, and multifocal tumors (22).

Based on histologic appearance and biological behavior, two subtypes of pRCC have

been identified: type 1 and type 2. It has been demonstrated that type 1 pRCC is

associated with prolonged survival as compared to type 2 pRCC, most likely due to

the fact that type 1 pRCC is typically detected at an earlier stage (28, 29). Clinical features differentiating pRCCs from ccRCCs and chromophobe renal cell

carcinomas (chRCCs) include spontaneous hemorrhaging as a presenting feature in

8% of cases, as well as multifocal presentation and calcification on imaging in 30%

of cases (30).

Page 16: Novel Biomarkers in Metastatic Renal Cell Carcinoma

16

1.2.3 Chromophobe Renal Cell Carcinoma

Less aggressive than other malignant renal tumors, chRCC accounts for 3–5% of all

RCCs (22). It is characterized by huge pale cells with a reticulated cytoplasm,

prominent cell membrane, and perinuclear halos (23, 31). A very close relationship

between chRCCs and oncocytomas has been suggested, with both arising from

intercalated cells of the collecting duct and showing similar alterations in

mitochondria and mitochondrial DNA (32). Although this relationship is still under

discussion, it has been hypothesized that chRCCs might represent a more aggressive

counterpart for oncocytomas (33). ChRCC is associated with the loss of several

chromosomes, as well a hereditary syndrome, Birt-Hogg-Dubé syndrome (34, 35).

1.2.4 Other Defined Subtypes

In addition to the above-mentioned subtypes, the WHO 2016 classification

recognizes several other less common renal cell tumor subtypes. These include

collecting duct carcinoma, renal medullary carcinoma, tubulocystic RCC, acquired

cystic disease-associated RCC, MiT family translocation RCC, succinate

dehydrogenase-deficient renal carcinoma, mucinous tubular and spindle cell

carcinoma, clear cell papillary RCC, hereditary leiomyomatosis and renal cell

carcinoma-associated RCC, and papillary adenoma (23). As these subclasses only

account for a small proportion of RCCs, they will not be discussed in further detail.

Page 17: Novel Biomarkers in Metastatic Renal Cell Carcinoma

17

1.2.5 Sarcomatoid Differentiation in RCC

Sarcomatoid RCC was first described as a tumor with pronounced cytologic atypia

containing enlarged pleomorphic or malignant spindle cells (36). It used to be

considered a histologic entity of its own, but it has since been demonstrated that

sarcomatoid differentiation instead represents a transformation to a higher-grade

malignancy and has been documented in numerous recognized histologic subtypes

of RCC (23, 37). It occurs in approximately 8% of RCCs, but it remains unknown

whether any specific histologic subtype has a predilection for sarcomatoid change

(38). Furthermore, research has demonstrated that sarcomatoid RCC is associated

with a worse prognosis, regardless of the underlying RCC subtype (38, 39).

1.3 Genetic Environment

The genetic environment and genetic changes associated with RCC are

heterogeneous. Several hereditary syndromes predisposing patients to RCC, among

other malignancies, have been described; von Hippel-Lindau disease (VHL),

hereditary leiomyomatosis and renal cell cancer (HLRCC), hereditary papillary renal

cancer (HPRC), and Birt-Hogg-Dubé syndrome (BHD) are the four most common

autosomal dominantly inherited syndromes with distinct histologic features and

genetic alterations (40). Hereditary RCC may account for 4–10% of all RCCs, but

many of the chromosomal changes seen in the hereditary forms of the disease are

also frequent in the sporadic forms (41, 42).

Page 18: Novel Biomarkers in Metastatic Renal Cell Carcinoma

18

1.3.1 VHL Gene Alteration

VHL gene alteration is a broad concept of genetic abnormality that includes VHL

gene mutation, promoter hypermethylation, and loss of heterozygosity. The VHL

gene is a tumor suppressor gene and its alteration is present in 50–70% of clear cell

RCCs (25, 43). Through its important role in the regulation of the hypoxia pathway,

VHL gene alteration and subsequent functional loss of VHL protein is thought to

play a significant part in tumorigenesis and tumor angiogenesis (44). In addition to

RCC, the VHL gene has also been implicated in the development of other tumors,

such as hemangioblastomas, pancreatic cysts, and pheochromocytomas (45).

Although understanding of the underlying role of VHL gene alteration in the

pathogenesis of RCC has led to the development of several novel therapeutic

approaches, most importantly vascular endothelial growth factor (VEGF)-targeted

agents, the clinical significance of VHL gene alteration in RCC is yet to be

established.

1.3.2 Met Proto-oncogene and HPRCC

Contrary to the VHL gene, the MET gene, located on chromosome 7, is a proto-

oncogene rather than a tumor suppressor gene (46). Activating mutations of the Met

proto-oncogene have been associated with hereditary papillary renal cell carcinoma

(HPRCC) (46-49) and to a lesser extent with sporadic forms of RCC. HPRCC is

characterized by multifocal, bilateral type I papillary renal cell carcinomas, but in

comparison to VHL disease, it is quite rare (50, 51). The Met proto-oncogene and its

product, c-Met, a tyrosine kinase receptor, have been implicated as promoters of

malignant transformation and tumorigenesis, and will be discussed in detail later in

this thesis.

Page 19: Novel Biomarkers in Metastatic Renal Cell Carcinoma

19

1.3.3 Other Hereditary Syndromes

Other hereditary syndromes associated with RCC are Birt-Hogg-Dubé (BHD)

syndrome and hereditary leiomyomatosis and renal cell cancer (HLRCC). BHD

syndrome is an autosomal, dominantly inherited condition characterized by the

development of benign skin tumors, pulmonary cysts, and pneumothorax (52). The

BHD-associated gene, folliculin (FLCN), is located on chromosome 17 and it acts

as a tumor suppressor gene. In BHD syndrome, however, this gene is inactivated

(35). Patients with BHD syndrome have a predisposition for RCCs, and unlike other

hereditary syndromes with this predisposition, RCCs associated with BHD are

histologically diverse. Over two-thirds of the RCCs associated with BHD are of

chromophobe or hybrid chromophobe/oncocytoma histology, however (53)

Fumarate hydratase, the gene product of the fumarate hydratase (FH) gene, is an

enzyme involved in the Krebs cycle. Mutations in the FH gene cause HLRCC. It acts

as a tumor suppressor gene, and the impaired function of the FH gene may lead to

chronic hypoxia, encouraging increased expression of VEGF and thus promoting

tumor formation. HLRCC is associated with type 2 papillary RCCs. (54, 55)

2. Tumorigenesis and the Tumor Microenvironment

2.1 Tumorigenesis

Most of our understanding of tumorigenesis in RCC is based on the work of Dr

Linehan et al. through the study of familial kidney cancer and VHL disease at the

Page 20: Novel Biomarkers in Metastatic Renal Cell Carcinoma

20

National Institutes of Health, Bethesda, US (56-58). At least twelve different genes

are known to promote the development of RCC: VHL, MET, folliculin (FLCN),

tuberous sclerosis complexes-1 and -2, (TSC-1 and -2), TFE3, TFEB, MITF, FH,

succinate dehydrogenase B (SDHB), succinate dehydrogenase D (SDHD), and

phosphatase and tensin homologue (PTEN) genes (58). All these genes are

associated with the regulation of essential mechanisms involved in tumor

development. However, to emphasize the genetic heterogeneity of RCC, an analysis

of more than 500 primary ccRCC tumor samples identified 19 significantly mutated

genes (SMGs), including VHL, PBRM1, SETD2, KDM5C, PTEN, BAP1, MTOR,

and TP53. Furthermore, in approximately 20% of cases, none of these SMGs were

present (59). Probably the most thoroughly investigated and well-described

pathways in RCC are the hypoxia-induced pathway, the mammalian target of

rapamycin (mTOR) pathway, and the c-Met signaling pathway, all promoting

angiogenesis and cell survival, which are essential events in the tumorigenesis of

RCC. These three pathways are described in more detail below.

2.1.1 Hypoxia-induced Pathway

As described earlier, the VHL gene is a tumor suppressor gene. It encodes the VHL

protein, which under normal circumstances targets hypoxia-inducible transcription

factors (HIF-1 - -mediated proteolysis (60). However, under

hypoxic conditions, as well as when the VHL complex is defective due to genetic

alterations, this cascade is interrupted, leading to the accumulation of HIFs. Guo et

al. demonstrated that alterations in genes involved in the ubiquitin-mediated

proteolysis pathway (VHL, BAP1, CUL7, BTRC) are frequent in ccRCC and are

significantly associated with overexpression of HIF- - (61). The

overexpression of the transcription factors, in turn, leads to the upregulation of

several important downstream promoters of angiogenesis such as vascular

Page 21: Novel Biomarkers in Metastatic Renal Cell Carcinoma

21

endothelial growth factor (VEGF), epidermal growth factor, transforming growth

- -derived growth factor (PDGF), erythropoietin, and glucose

transporter 1 (62). Important pathways involved in RCC biology are presented in

Figure 1.

2.1.1.1 HIF- -

Both HIF- - function as transcription factors mediating the

activation of target genes in response to hypoxic conditions. With the inactivation of

the VHL protein, VHL-associated degradation of HIF- -

leading to the maintained activity of these transcription factors irrespective of the

oxygen levels (63, 64). The mTOR pathway also leads to the accumulation of HIF-

the stimulation of ribosomal translation of mRNAs, including the

translation of HIF- , as demonstrated in Figure 1 (65).

HIFs promote tumorigenesis in a variety of ways, including angiogenesis,

metabolism, proliferation, metastasis, and differentiation. The overexpression of

HIF- s a critical factor in renal carcinogenesis in several

studies (66-70). The tumorigenic potential of HIF-

controversial, with contradicting evidence making its role in the development of

RCC less clear (71, 72). C teins as factors

clearly influencing tumor progression by direct regulation of both unique and shared

target genes, as well as by exerting distinct effects on critical oncoproteins and tumor

suppressors.

Page 22: Novel Biomarkers in Metastatic Renal Cell Carcinoma

22

Figure 1. Important pathways involved in renal cell carcinoma biology and

tumorigenesis (Adapted from Klatte et al. Molecular biology of renal cortical

tumors. Urol Clin North Am 2008) (65).

Page 23: Novel Biomarkers in Metastatic Renal Cell Carcinoma

23

2.1.2 mTOR Pathway

The mTOR pathway regulates cell growth, proliferation, survival, and metabolism

(73). Research has shown that in addition to its role in RCC, it is involved in the

development and carcinogenesis of several different malignancies (74, 75).

mTOR is a serine/threonine kinase that in response to growth factors, hormones, and

nutrients activates protein synthesis and contributes to many different cell functions,

including protein degradation and angiogenesis (76). This response is controlled and

activated by the phosphatidylinositol-3-kinase/Akt (PI3K/Akt) pathway and opposed

by the activity of PTEN and TSC-1 and -2 (77, 78). Mutations of PTEN and TSC-1

and -2 leading to the loss of their activity and abnormal activation of the PI3K/AKT

pathway have been demonstrated to play a role in RCC progression (78-80).

The mTOR protein forms two structurally and functionally distinct complexes,

mTOR complex 1 (mTORC1) and complex 2 (mTORC2). mTORC1 is involved in

the regulation of several oncogenic proteins, such as HIF- , and c-

Myc, and mTORC2 is important for cell survival (73). Genetic alterations upstream

of or at mTOR leading to abnormal activation of the mTOR pathway promote the

upregulation of these oncogenic proteins through key downstream effectors, such as

the ribosomal S6 kinase and the eukaryotic translation initiation factor 4E binding

protein (74, 81, 82). In an analysis of over 500 primary ccRCC tumor samples, the

authors demonstrated that alterations targeting multiple components of the

PI3K/AKT/mTOR pathway were present in 28% of the tumor samples (59).

Furthermore, these alterations were associated with a worse or better outcome,

depending on their respective action on the PI3K/AKT pathway, suggesting a role as

a therapeutic target in ccRCC (59).

Page 24: Novel Biomarkers in Metastatic Renal Cell Carcinoma

24

2.1.3 c-Met Signaling Pathway

The c-Met receptor tyrosine kinase is a product of the Met proto-oncogene located

on chromosome 7q21-31. The ligand for c-Met, hepatocyte growth factor (HGF),

promotes cell proliferation, survival, motility, differentiation, and morphogenesis.

Under normal physiologic conditions, the c-Met signaling pathway regulates tissue

homeostasis, but it has been found to play a role in the tumorigenesis of various

human malignancies (83-85).

c-Met signaling is a complex entity involving several molecular events. The major

downstream responses of c-Met activation include activation of the

RAS/RAF/MAPK cascade and the PI3K/Akt signaling axis (86). The mitogen

activated protein kinase (MAPK) activates a large number of genes, which in turn

results in increased cell proliferation and cell motility (87). The PI3K/Akt signaling

axis, on the other hand, is responsible for cell survival in response to c-Met activation

(88).

In rodent models, activating mutations of Met have been shown to cause a variety of

tumors, including sarcomas, lymphomas, and carcinomas, suggesting that aberrant

Met signaling can also cause human cancer (89). In RCC, activating mutations in the

c-Met kinase domain were first discovered in both sporadic and inherited forms of

papillary RCC (48, 90).

c-Met/HGF signaling has been shown to promote tumor angiogenesis by directly

acting on endothelial cells (ECs), inducing proliferation and migration (91, 92). In a

more indirect manner, c-Met/HGF plays a part in activating the angiogenic switch

by inducing VEGF-A expression and by suppressing thrombospondin 1 (TSP-1), a

negative regulator of angiogenesis (93). Indeed, evidence suggests that there is

considerable crosstalk between c-Met/HGF signaling and various other signaling

pathways involved in cancer progression, as well as resistance to therapy.

Page 25: Novel Biomarkers in Metastatic Renal Cell Carcinoma

25

Although c-Met/HGF and VEGF/VEGFR do not directly associate with each other,

they share synergistic angiogenesis-activating intermediates such as the PI3K/Akt

and MAPK cascades, described earlier. Furthermore, HIF-dependent expression of

c-Met has been documented in several types of carcinoma cells (94-96). These

studies suggest that anti-angiogenic therapy, which leads to hypoxic conditions,

induces the accumulation of HIFs, further inducing c-Met expression. This, in turn,

might promote the MET-dependent spread of cancer cells, making it an interesting

target for therapeutic intervention.

2.1.4 Angiogenesis

The supply of oxygen and nutrients is crucial for the survival of tumor cells, and

indeed, neovascularization is considered a key event for tumor progression in RCC.

Through a complex process called the ‘angiogenic switch’, tumor cells induce

angiogenesis and ensure the further supply of oxygen and nutrients, as well as the

disposal of metabolic waste products and carbon dioxide by new sprouting vessels

(97). The regulation of tumor angiogenesis is depicted in Figure 2.

HIFs activate the expression of several proangiogenic factors such as VEGF, VEGF

receptors, angiopoietins (Ang-1 and -2), fibroblast growth factor 2 (FGF-2), platelet-

derived growth factor B (PDGF-B), the Tie-2 receptor, and matrix

metalloproteinases (MMP-2 and -9) (98). The role of VEGF, particularly VEGF-A,

and its receptor, VEGFR-2, is well established regarding angiogenesis in RCC.

VEGF-A and VEGFR-2 regulate the proliferation of ECs and the development of

vasculature by their concentration and gradient, respectively (99). While VEGF-A

and VEGFR-2 represent a key signaling system in the early stages of blood vessel

growth (100), secondary stages of the process require a high level of activity of

several other growth factors and their receptors.

Page 26: Novel Biomarkers in Metastatic Renal Cell Carcinoma

26

Angiopoietins, Ang-1 and Ang-2, are endothelial cell-derived growth factors that act

by binding to their receptors, Tie-1 and Tie-2. While the role of Tie-1 in angiopoietin

signaling remains unclear, studies in Tie-2 knockout mice have demonstrated that

the initial phases of angiogenesis are able to proceed normally, but the remodeling

and hierarchical reorganization of the sprouting vessels are disturbed (101, 102).

Current understanding depicts the Ang-Tie pathway as a mediator of vessel growth,

remodeling, and maturation through the regulation of EC permeability, EC–

extracellular matrix interaction, and inflammation (103). Ang-1 stabilizes vessels

and decreases vascular permeability (104), whereas Ang-2 is involved in vessel

regression and destabilization (105). Since tumor vasculature often appears as

primitive, hemorrhagic, and disorganized, it is not surprising that elevated levels of

Ang-2 have been found in several cancers and appear to be associated with a worse

prognosis (106-108). Thus, blocking of Ang-2 and targeting of the Ang–Tie system

has been of great interest in anti-angiogenic drug development.

PDGF-B and FGF-2 are angiogenic factors frequently expressed in tumors, and their

expression has been linked to cancer progression and metastasis (109-111). PDGF-

B is involved in the recruitment of pericytes and vascular smooth muscle cells, while

FGF-2 displays a broad spectrum of biological functions. Several studies have

reported FGF-2 and PDGF-B to have angiogenic synergism (112-114). Interestingly,

murine models have shown that whereas FGF-2 and PDGF-B work in concert to

promote vessel maturation and stabilization under physiological conditions, tumor

vasculature induced by these factors is primitive and disorganized (114). It has been

suggested that this might represent cross-communication with VEGF, counteracting

the recruitment activity of PDGF-B (115), but the specific molecular mechanisms by

which this happens remain to be elucidated.

As depicted in Figure 2, tumor neovascularization is a process induced by multiple

individual growth factors, cytokines, and their complex interactions. While not all of

Page 27: Novel Biomarkers in Metastatic Renal Cell Carcinoma

27

these interactions are thoroughly understood, increasing understanding of the

underlying mechanisms of neovascularization has led to the development of several

novel anti-angiogenic therapies.

Figure 2. Regulation of tumor angiogenesis (Adapted from Cao et al. Regulation of

tumor angiogenesis and metastasis by FGF and PDGF signaling pathways, Journal

of Molecular Medicine, 2008) (115).

2.2 Tumor Microenvironment

The tumor microenvironment is a complex mixture of proliferating tumor cells,

tumor stroma, blood vessels, tumor infiltrating immune cells, and a variety of

associated tissue cells. The tumor controls this unique environment through

molecular and cellular events taking place in the surrounding tissues, thus promoting

Page 28: Novel Biomarkers in Metastatic Renal Cell Carcinoma

28

tumor survival, progression, and metastasis. As a tumor progresses, the changes seen

in the tumor microenvironment resemble those seen in the process of chronic

inflammation. This ‘host reaction to the tumor’ is thought to be strongly involved in

shaping the tumor microenvironment. Although the initial goal of this response is to

wipe out the tumor, like any invader, evidence suggests that the tumor actively

downregulates these anti-tumor responses through a variety of strategies. (116, 117)

Major components of the tumor microenvironment are tumor-infiltrating

lymphocytes (TILs). TILs include B lymphocytes and T lymphocytes, which can be

further categorized into CD4+ T-cells, CD8+ cytotoxic T cells, and natural killer

(NK) cells. CD4+ T-cells differentiate into CD4+ helper and regulatory T-cells (T-

reg). While CD4+ helper T-cells coordinate immune responses through the secretion

of various cytokines, regulatory T-cells serve to limit adaptive immune responses by

cytokine- and cell-contact-dependent mechanisms. CD8+ T-cells normally function

to promote the lysis of infected, neoplastic, or otherwise targeted host cells. For T-

cells to work effectively, the antigen/antigens must be presented to T-lymphocytes

by major histocompatibility complex (MHC) molecules on the surface of an antigen-

presenting cell. NK cells, on the other hand, do not require prior antigen exposure

and they display lytic activity against cells that do not express MHC molecules. Thus,

they form an important safeguard against neoplastic cells lacking the means to

present self-antigens to T-lymphocytes. (118-120)

Other immune cells present in tumors include dendritic cells (DCs), macrophages,

and myeloid suppressor cells (MDSCs). The main function of DCs is to process and

present antigens to T-lymphocytes. Macrophages present in tumors are known as

tumor-associated macrophages (TAMs). Under physiologic conditions,

macrophages play a part in both the innate and the adaptive immune responses

through phagocytosis, antigen presentation, and the excretion of cytokines. TAMs,

however, are re-programmed to inhibit lymphocyte functions and may also possess

Page 29: Novel Biomarkers in Metastatic Renal Cell Carcinoma

29

tumor growth promoting angiogenic potential (121, 122). Similarly, MDSCs are

normal host cells of bone marrow origin with immunosuppressive and angiogenic

properties. Under pathologic conditions, systemic signals result in MDSCs

prematurely leaving the bone marrow and engaging in T-cell suppression at

extramedullary locations (123, 124). Murine models have additionally demonstrated

that MDSCs may promote tumor angiogenesis (125).

2.2.1 Tumor Escape

The tumor microenvironment forms an effective barrier against immune cell

functions. The process by which a tumor escapes immune recognition,

“immunoediting”, was first described by Dunn et al. (116). Although during the early

phases, lymphocytes responsible for immune surveillance are able to recognize and

eliminate malignant cells, tumor cells begin to undergo progressive selection

favoring the proliferation of malignant cells more likely to evade immune

recognition. The cancer cells additionally directly inhibit immune cells through

overexpression of immune-checkpoint molecules (116). Several mechanisms

leading to the dysfunction of immune cells and downregulation of the immune

response by tumors have been identified. The four main pathways by which this

happens are interference with the induction of anti-tumor responses, inadequate

effector cell function, insufficient recognition signals, and the development of

immunoresistance by the tumor (118). Given the pivotal role of immunoediting and

chronic inflammation in tumorigenesis, it is not surprising that research suggests that

the amount and presence of different immune cell populations in the tumor

microenvironment is associated with the disease course.

As described earlier, effector T-cells are responsible for the lysis of neoplastic cells.

The developing microenvironment of a growing tumor, however, prevents these

Page 30: Novel Biomarkers in Metastatic Renal Cell Carcinoma

30

immunological reactions by secreting suppressive factors expressing inhibitory

molecules and by attracting T-regs, TAMs, and MDSCs (126-128). T-regs

downregulate the proliferation and anti-tumor functions of both CD4+ and CD8+

effector T-lymphocytes (129, 130). This is achieved by the expression of various

molecules on T-regs, such as CTLA-4, preventing T-cell activation and DC functions

(131, 132). Activated T-regs also induce cell death in effector T-cells by a granzyme-

dependent pathway (133). The proliferation of T-regs is induced by interleukin-10

(IL-10), - , and prostaglandin E2, all of which

are abundant in the tumor microenvironment (134). Indeed, T-regs appear to be

present in higher numbers in more aggressive and advanced cancers (135). In RCC,

higher levels of T-regs are correlated with a poorer prognosis (136-139). Similarly,

TAMs inhibit cytotoxic T-cell responses through several mechanisms. For example,

they produce IL-10, which in turn induces the expression of programmed death

ligand 1 (PD-L1). PD-L1 is a ligand for an inhibitory cell-surface receptor,

programmed death 1 (PD-1), which negatively regulates T-cell activation (140).

TAMs additionally promote tumor-associated angiogenesis and tumor cell invasion

(141, 142). In ccRCC, higher numbers of TAMs are associated with a poor prognosis

(143-145). In pRCC, however, an opposite relationship was noted (146).

MDSCs are another immunosuppressive immune cell population that, like T-regs

and TAMs, promote tumor evasion by suppressing T-cell immunity. Among other

effects, MDSCs deplete nutrients necessary for appropriate T-cell function, block

MHC-bound peptides from being presented to T-cells, and induce the conversion of

naive T-cells to a T-reg phenotype (147-149). Research also implicates MDSCs as a

part of STAT3-VEGF-dependent angiogenesis (125). In RCC, elevated levels of

MDSCs are associated with a poor prognosis (150).

Page 31: Novel Biomarkers in Metastatic Renal Cell Carcinoma

31

Growing knowledge and understanding of immune responses and immune cells as a

part of tumorigenesis and tumor progression have sparked interest in the

development of several promising immunotherapeutic agents, such as vaccines, T-

cell modulators, and immune checkpoint inhibitors.

2.2.2 Tumor Heterogeneity

Previous work on solid cancers has demonstrated that extensive genetic

heterogeneity exists among individual tumors (151-153). This intratumor

heterogeneity may contribute to treatment failure and drug resistance (154-156), and

can manifest as spatial heterogeneity, with an uneven distribution of genetically

diverse tumor subpopulations, and temporal heterogeneity, with dynamic variations

in the genetic diversity of an individual tumor over time.

Gerlinger et al. investigated gene-expression signatures in RCC from spatially

separated tumor samples obtained from primary renal carcinomas and associated

metastatic sites. Up to 69% of all somatic mutations were not detectable across every

tumor region. Additionally, gene-expression signatures of both a good and poor

prognosis were detected in different regions of the same tumor. (157) As

personalized medicine approaches traditionally rely on single tumor biopsy samples,

intratumor heterogeneity may have significant consequences regarding

antineoplastic treatment strategies. It is thought that tumor heterogeneity underlies

resistance to therapy, thus complicating the selection of globally effective therapies

(158). These observations emphasize the importance of multidimensional

therapeutic approaches, as well as the need to develop ways to dynamically monitor

patients during treatment.

Page 32: Novel Biomarkers in Metastatic Renal Cell Carcinoma

32

3 Diagnosis and Management of Renal Cell Carcinoma

Since renal cell carcinoma often develops without any early warning signs, a rather

high proportion of patients present with metastases at diagnosis. The most common

clinical presentations of the disease are hematuria, abdominal pain, and a palpable

mass in the flank or abdomen. This “classic triad”, however, is only present in less

than 10% of patients (159). Research has shown that in 25–30% of patients, the

disease has metastasized at the time of diagnosis (160, 161)

While surgical resection remains the golden standard of care for localized RCCs,

relapse occurs in almost a third of the patients (162).

3.1 Diagnosis

The widespread application of computed tomography (CT) and ultrasonography for

other indications has led to the increased detection of RCCs. More than 50% of RCCs

are detected incidentally (163). RCC is sometimes referred to as the “internist’s

cancer”, since in addition to the “classic triad” of symptoms, the disease is sometimes

accompanied by hypercalcemia, unexplained fever, erythrocytosis, and Stauffer’s

syndrome (signs of cholestasis unrelated to tumor infiltration of the liver or intrinsic

liver disease, which typically resolves after kidney tumor resection). Laboratory

examinations of serum creatinine, hemoglobin, leukocyte and platelet counts, lactate

dehydrogenase (LDH), C-reactive protein (CRP), and albumin-corrected calcium are

recommended if suspicion of RCC arises. (164)

Page 33: Novel Biomarkers in Metastatic Renal Cell Carcinoma

33

3.1.1 Imaging

RCC is usually suspected based on ultrasonography findings, prompting further

imaging with computed tomography. According to the ESMO 2016 guidelines,

contrast-enhanced chest, abdominal, and pelvic CT are mandatory for accurate

staging. The use of bone scanning and CT or magnetic resonance imaging (MRI) of

the brain is recommended only when suggestive clinical symptoms or laboratory

findings are present and not as a part of routine clinical practice. (164)

In most cases, a single examination using CT provides sufficient information for

accurate staging and surgical planning with no need for additional imaging (165).

MRI is mainly an alternative in patients requiring further imaging and in cases of

allergies, pregnancy, or surveillance. MRI is also an excellent alternative in patients

with renal insufficiency. Positron emission tomography/computed tomography

(PET/CT) scanning is not a standard investigation in the diagnosis and staging of

RCC and its use is not recommended. (164)

3.1.2 Staging and Pathology Assessment

For staging, the tumor-node-metastasis (TNM) staging system should be used. The

TNM staging system is presented in Table 1.

A core needle biopsy is recommended before treatment with ablative therapies, as

well as in patients with an unresectable solid renal mass and metastatic disease before

starting systemic therapy. It is additionally practical in patients with significant

comorbidities where the risk of biopsy is outweighed by the risk of surgery. (166,

167). The role of core needle biopsy is less clear regarding solitary renal masses

<4 cm in diameter, as the probability of malignancy within a small renal mass has

been found to be inversely related to tumor size (168). In a study by Halverson et al.

Page 34: Novel Biomarkers in Metastatic Renal Cell Carcinoma

34

in which patients underwent both percutaneous renal mass biopsy and subsequent

partial or radical nephrectomy, there was complete concordance between the

histology rendered from core needle biopsy and that rendered by surgery (169).

Given that core needle biopsy has a high sensitivity and specificity in confirming the

histopathological nature of a tumor, it probably has a role when deciding on the

active surveillance and operative management of a small renal mass (167). A

specimen provided by nephrectomy, however, provides the final histopathological

diagnosis, classification, and grading. Regarding the assessment of pathology, the

ESMO 2016 guidelines recommend reporting the tumor histological subtype, the

International Society of Urological Pathology (ISUP) nucleolar grading, sarcomatoid

and/or rhabdoid differentiation, the presence of necrosis and presence of microscopic

vascular invasion, in addition to TNM staging in routine clinical practice. (164)

Page 35: Novel Biomarkers in Metastatic Renal Cell Carcinoma

35

Table 1. TNM classification System

Primary Tumor (T)

TX Primary tumor cannot be assessedT0 No evidence of primary tumorT1 limited to the kidney

T1aT1b

T2 Tumor >7.0 cm in greatest dimension, limited to the kidney

T2aT2b

T3 Tumor extends into major veins or perinephric tissues but not into the ipsilateral adrenal gland and not beyond Gerota’s fascia

T3a

T3b

T3c

T4 Tumor invades beyond Gerota’s fascia (including contiguous extension into the ipsilateral adrenal gland)

Tumor

Tumor >10 cm, limited to the kidney

Tumor grossly extends into the renal vein or its segmental (muscle containing) branches, or tumor invades perirenal and/or renal sinus fat (peripelvic) but not beyond Gerota’s fascia

Tumor grossly extends into the vena cava below the diaphragm

Tumor grossly extends into the vena cava above the diaphragm or invades the wall of the vena cava

Regional Lymph Nodes (N)

NXN0 N1 N2

Regional lymph nodes cannot be assessed No regional lymph node metastasisMetastasis in regional lymph node(s)More than one lymph node involved

Distant Metastases (M)

cM0 cM1 pM1

Clinically no distant metastasisClinically distant metastasisPathologically proven distant metastasis, e.g. needle biopsy

Anatomic stage/prognostic groups

Stage IStage IIStage III

Stage IV

T1T2T1-2 T3T4Any

N0N0N1AnyAnyAny

M0M0M0M0M0M1

Adapted from the AJCC Cancer Staging Handbook, 7th edition (2010)

Page 36: Novel Biomarkers in Metastatic Renal Cell Carcinoma

36

3.1.3 Small Renal Masses

An increasingly common and problematic clinical entity encountered by clinicians

is small renal masses (SRMs) less than 4 cm in size. Based on epidemiological

studies, SRMs account for nearly one-half of all newly diagnosed renal masses (170).

Therapeutic options regarding SRMs include extirpative surgery (radical or partial

nephrectomy), ablative therapies, and active surveillance (AS). Kutikov et al.

demonstrated that 20–30% of solitary renal masses presumed to be renal cell

carcinoma on preoperative imaging had benign pathologic findings on resection

(171). Furthermore, in those lesions that are RCC, the majority of tumors are of low

grade and unlikely to develop metastases (172, 173).

There are no definitive clinical guidelines for the management of SRMs. General

recommendations for patient selection favoring AS include increased age, decreased

life expectancy, suitability for surgery, and a decreased risk of metastatic disease.

Several studies have shown that the risk of metastatic progression while on AS is

<2%, making it a viable alternative to surgery (174-178). The main trigger for

operative intervention, on the other hand, is believed to be the tumor growth rate

(GR). Evidence regarding tumor GR is somewhat controversial, as some studies have

demonstrated a high GR among AS patients developing metastases (174, 179), while

a number of studies have demonstrated a low or zero GR for tumors of malignant

pathology (176). Although the malignant potential of an SRM is likely to be defined

by several important factors, progression to metastatic disease is exceptionally low

in tumors that demonstrate a low or zero GR and remain <3 cm in diameter (177,

180).

In a multi-institutional prospective clinical trial investigating AS for the management

of SMRs, the authors demonstrated non-inferiority of AS versus primary

intervention. At five years, cancer-specific survival was 99% and 100% for primary

intervention and AS, respectively, suggesting that AS should become part of the

Page 37: Novel Biomarkers in Metastatic Renal Cell Carcinoma

37

standard discussion for the management of SRMs (180). An informed decision by

the patient and physician, including a discussion of the small but real risk of cancer

progression, loss of a window of opportunity for nephron-sparing surgery, and the

lack of curative treatments for mRCC should, however, precede any treatment

decisions.

3.2 Prognostication

For localized RCC, several multivariable models such as the University of California

Los Angeles Integrated Staging System (UISS) and SSIGN score have been

developed to predict cancer-specific survival after nephrectomy. In these models,

prognostication is mainly based on clinical and histological parameters of the tumor,

as presented in Table 2. Although these models can predict the natural history of

RCC, they are not designed to account for the effect of targeted therapies in patients

with mRCC. (181, 182)

Table 2. The UISS and SSIGN prognostic models assessing postoperative cancer-

specific mortality

Model Sample Size Target population

Predictors C-index

UISS 661 RCC of all stages

- AJCC- Fuhrman grade - ECOG-PS

82–86%

SSIGN 1801 Localized clear cell RCC

- TNM (1997)- Tumor size - Nuclear grade - Tumor necrosis

81–82%

AJCC = American Joint Committee on Cancer; ECOG-PS = Eastern Cooperative Oncology Group

Performance Status

Page 38: Novel Biomarkers in Metastatic Renal Cell Carcinoma

38

Prognostication of metastatic RCC relies heavily on traditional markers of risk. In

addition to pathological and histological staging and grading, several clinical and

biological prognostic markers have been identified.

While it seems that age, gender, and race are not associated with the disease course

or outcome, the association between the performance status and prognosis is well

established. Two different scales are used to evaluate the patient’s performance

status, namely the Eastern Cooperative Oncology Group (ECOG) scale and

Karnofsky performance status (KPS) score; both have demonstrated good reliability

and validity.

Other validated prognostic factors for mRCC include serum hemoglobin lower than

the lower limit of normal, platelets greater than the upper limit of normal, serum

LDH greater than the upper limit of normal, corrected serum calcium greater than

the upper limit of normal, neutrophils greater than the upper limit of normal, and a

time from diagnosis to treatment of <1 year.

The two most widely used prognostic models combining these markers into risk

classifications are the Memorial Sloan Kettering Cancer Center (MSKCC) risk

classification (183) and the International Metastatic Renal Cell Carcinoma Database

Consortium (IMDC) risk classification (184). These models apply exclusively to

patients with mRCC.

In addition to the more traditional markers of risk, gene signatures are known to

detect risk groups in RCC (185, 186). However, in current clinical practice, no

specific molecular markers can be recommended for routine use, as is discussed later.

Page 39: Novel Biomarkers in Metastatic Renal Cell Carcinoma

39

3.3 Treatment

Surgical resection of the tumor is the golden standard of care in localized RCC.

While radical nephrectomy remains the most frequently used alternative, several

novel minimally invasive techniques have emerged, mostly for the treatment of T1

tumors. Current evidence suggests that localized RCCs are best managed by partial

nephrectomy (or nephron-sparing surgery) when feasible. The role of techniques

such as radio-frequency ablation and cryoablation remains to be further elucidated.

Disease recurrence occurs in 20–30% of patients after partial or radical nephrectomy.

(187). The role of adjuvant therapies in localized RCC is discussed separately in

conjunction with systemic therapies.

The treatment of mRCC mainly relies on systemic therapy, with surgery having a

less well-defined role than in localized disease.

3.3.1 Cytoreductive Nephrectomy

A combined analysis by Flanigan et al. investigating the role of cytoreductive

nephrectomy in the era of immunotherapy demonstrated that nephrectomy plus

interferon was associated with longer median overall survival than interferon alone

(13.6 vs. 7.8 months), representing a 31% decrease in the risk of death (P = 0.002)

(188). Since then, cytoreductive nephrectomy has been a part of routine clinical

practice in patients with a good performance status and large primary tumors with

limited volumes of metastatic disease. It is not recommended, however, in patients

with a poor performance status (164).

A recent phase III multicenter trial, CARMENA, investigated the role of

nephrectomy among 450 mRCC patients with an intermediate or poor MSKCC risk

classification. In this study, patients were randomized to undergo nephrectomy and

then receive sunitinib or receive sunitinib alone. The median OS was 18.4 months in

Page 40: Novel Biomarkers in Metastatic Renal Cell Carcinoma

40

the sunitinib-alone group and 13.9 months in the nephrectomy–sunitinib group,

demonstrating the non-inferiority of sunitinib alone versus nephrectomy followed by

sunitinib. (189) However, as Motzer and Russo pointed out in their editorial, the

interpretation of the results from the CARMENA trial is complicated by several

factors, including slow enrollment, a lack of information regarding the selection

factors for performing nephrectomy, and the high percentage of poor-risk patients

(43%). As such, these data should emphasize the importance of patient selection

rather than lead to the abandonment of nephrectomy. (190)

3.3.2 Metastasectomy and Other Local Therapeutic Options for Metastases

Common metastatic sites in RCC include the lung, bone, liver, and brain, but

metastases can be found at any anatomical site (191, 192). In addition to traditional

surgery, options for local therapy of metastases include whole brain radiotherapy,

conventional radiotherapy, stereotactic radiosurgery, stereotactic body

radiotherapy, cyberknife radiotherapy, and hypofractionated radiotherapy.

The benefits of metastasectomy as well as other local therapeutic options have been

under fervent discussion, with no general guidelines existing as to whether a patient

should be referred for local treatment of metastases. Dabestani et al. systematically

reviewed the potential benefits of these treatments, suggesting a survival benefit

with complete metastasectomy vs. either incomplete or no metastasectomy (193).

Complete metastasectomy was additionally associated with improved symptom

control. The role of other local therapies is less clear, with consensus mainly

existing on the palliative benefits in selected patients.

Page 41: Novel Biomarkers in Metastatic Renal Cell Carcinoma

41

The ESMO 2016 guidelines do not recommend metastasectomy or other local

therapies in routine practice, but rather after multidisciplinary review for selected

patients (164).

3.3.3 Systemic Therapy

The low response rates of mRCC to classical cytotoxic agents such as

fluoropyrimidines or vinblastine, as well as hormonal treatment and radiotherapy,

has inevitably led to the development of alternative therapies. As ccRCC is

considered an immunogenic tumor, immunotherapy comprised the first wave of

systemic therapies. As our knowledge of the underlying mechanisms and the

pathogenesis of RCC has evolved, an array of new therapies has emerged from anti-

VEGF and tyrosine kinase inhibition to modern approaches of dual inhibition and

immune checkpoint inhibition.

3.3.3.1 Immunotherapy

Before the advent of targeted therapies, systemic therapy of mRCC comprised

interferon- -2 (IL-2).

IL-2 is a cytokine that enhances the proliferation and function of T-cell lymphocytes.

Although high-dose IL-2 therapy only produces modest response rates of 15–16% in

the treatment of mRCC, the responses that do occur seem durable (194).

Additionally, research has shown that approximately 5–7% of patients achieve

complete remission (195, 196). High-dose IL-2 treatment, however, is associated

with severe adverse events, such as capillary leak syndrome, resembling the clinical

manifestations of septic shock and producing major morbidity in patients receiving

Page 42: Novel Biomarkers in Metastatic Renal Cell Carcinoma

42

IL-2 (197). The significant toxicity profile has limited the use of high-dose IL-2 in

the treatment of mRCC.

, and antiproliferative

activities. the treatment of

mRCC patients (198, 199). Negrier et al. demonstrated that the

and low-dose IL-2 resulted in prolonged progression-free survival and improved

response rates as compared to either agent alone (200). No benefit was seen

regarding overall survival, however.

Current guidelines suggest high-dose IL-

bevacizumab as alternatives for the standard options among patients with a good or

intermediate prognosis (164). In the era of targeted therapies, however, the role and

use of these agents has become limited.

3.3.3.2 Tyrosine Kinase Inhibitors (TKIs)

The introduction of molecularly targeted therapies revolutionized the treatment of

advanced RCC. Inhibition of the VEGF pathway has emerged as the primary

therapeutic intervention for most patients with advanced disease. TKIs are multi-

targeted kinase inhibitors that inhibit signaling in a variety of key receptors such as

VEGFR-1, -2, and -3, PDGFR- - -RET, macrophage colony-stimulating

factor 1 (CSF-1R), FMS-like tyrosine kinase 3 receptor (FLT3), and c-KIT (201).

Among the first TKIs approved for the treatment of mRCC were sunitinib and

sorafenib. Sorafenib was approved after showing prolonged PFS and increased ORR

when compared to placebo among mRCC patients resistant to standard therapy

(202). It is currently recommended as one of several options in the second line

setting. Sunitinib is the current standard of care for treatment-naïve mRCC patients

Page 43: Novel Biomarkers in Metastatic Renal Cell Carcinoma

43

after having demonstrated significantly longer median PFS and a higher objective

(201).

Pazopanib, a second-generation TKI, has since been approved after demonstrating

non-inferiority when compared to sunitinib, providing yet another option for first-

line therapy (203).

Other current second-line options include axitinib, a highly potent and selective

inhibitor of the various kinase domains of VEGFRs, distinguishing it from previous

multi-targeted TKIs. In a phase III randomized clinical trial (AXIS), axitinib

demonstrated an improved PFS when compared to sorafenib in the second-line

treatment setting (204). Axitinib has also demonstrated clinical activity and safety in

the first-line setting, but no significant PFS benefit was noted in a phase III

randomized comparison between axitinib and sorafenib (205).

3.3.3.3 mTOR Inhibitors

The mammalian target of rapamycin (mTOR) is a downstream effector of the PI3-

K/Akt pathway that exerts its biological functions as two distinct complexes,

mTORC1 and mTORC2, as depicted earlier (206, 207). Everolimus and

temsirolimus are allosteric inhibitors of mTOR that have shown clinical activity in

mRCC. A phase III trial enrolling 626 mRCC patients with poor prognostic features

comparing temsirolimus, IFN , and the combination of both showed that

temsirolimus alone resulted in longer OS and PFS in this patient population (208).

Based on these results, temsirolimus is still recommended as a first-line therapy for

poor-risk mRCC patients (164). However, the use of temsirolimus in first-line

treatment for patients with a poor risk classification has been called into question by

Page 44: Novel Biomarkers in Metastatic Renal Cell Carcinoma

44

the results from the RECORD-3 trial, in which sunitinib showed superiority over

everolimus, even among patients with a poor prognosis (209).

Everolimus is the standard of care for mRCC patients whose disease progresses after

initial anti-VEGFR therapy. It was approved based on the results from Renal Cell

Cancer Treatment With Oral RAD001 Given Daily (RECORD-1), a phase III trial,

in which everolimus exhibited superior efficacy to placebo (210).

Although mTOR inhibitors initially showed promising clinical activity in mRCC,

their effects have proven to be far from durable and only a subset of patients

experience significant clinical benefit from these agents.

The cellular action mechanisms of TKIs and mTOR inhibitors are depicted in Figure

3.

Page 45: Novel Biomarkers in Metastatic Renal Cell Carcinoma

45

Figure 3. Cellular action mechanisms of TKIs and mTOR inhibitors (Adapted from

Rini et al. Resistance to targeted therapy in renal-cell carcinoma, Lancet 2009)

(211).

3.3.3.4 Novel Approaches

Most patients treated with either VEGF- or mTOR-targeted therapy ultimately

develop resistance to these drugs. Whether, and more importantly how, this

phenomenon is embedded in the intricate network of complex regulation and

feedback loops these different pathways possess remains to be elucidated. With first-

line sunitinib or pazopanib, the median PFS ranges from 8 to 11 months for all

Page 46: Novel Biomarkers in Metastatic Renal Cell Carcinoma

46

patients (201, 203). Additionally, only a few treatments have shown a survival

benefit in previously treated mRCC patients.

The upregulation of prometastatic MET and AXL as a result of VHL protein

dysfunction in ccRCC has been implicated as a potential mediator of resistance to

anti-VEGFR therapy (212). Cabozantinib is an inhibitor of tyrosine kinases,

including MET, VEGFR, and AXL (213). In the fairly recent phase III METEOR

trial comparing cabozantinib and everolimus among patients who progressed after

initial VEGFR tyrosine-kinase treatment, cabozantinib became the first agent to

show a survival benefit in all efficacy endpoints (PFS, OS, and ORR) in previously

treated mRCC patients (214). Given the dim estimations of a PFS of only 5.6 months

for first-line VEGFR-targeted therapy among patients with an intermediate–poor risk

classification (215), cabozantinib has also been investigated in the first-line setting

in the CABOSUN trial. In this setting, cabozantinib significantly increased median

PFS (8.2 vs. 5.6 months) and was additionally associated with an increased reduction

in the rate of progression when compared to sunitinib (216). As these results

demonstrate a possible role for this dual inhibitor in the first-line setting,

cabozantinib is currently recommended as an alternative after prior to anti-VEGFR

therapy (164).

As Dunn et al. described, one of the mechanisms by which tumors evade host

immune reactions is by directly inhibiting immune cells through overexpression of

immune checkpoint molecules (116). Recently, immune checkpoint blockade has

become a new avenue of immunotherapy in several tumor types.

PD-L1 is a ligand for an inhibitory cell-surface receptor, programmed death 1 (PD-

1), which negatively regulates T-cell activation (140). In RCC, studies have shown

that PD-L1 expression is associated with a poor prognosis (217-219). Nivolumab is

a PD-1 immune checkpoint inhibitor that selectively blocks the interaction between

PD-1 and PD-1L. Nivolumab has shown consistent efficacy in the treatment of

Page 47: Novel Biomarkers in Metastatic Renal Cell Carcinoma

47

advanced melanoma, both alone and in combination with a CTLA-4 inhibitor,

ipilimumab (220). In addition to melanoma, nivolumab has been approved for the

treatment of squamous and nonsquamous non-small-cell lung cancer and Hodgkin’s

disease (221, 222). In advanced RCC, nivolumab has been granted approval based

on the results from a phase III study comparing nivolumab with everolimus in

previously treated mRCC patients. In this setting, nivolumab was associated with

improved OS and ORR, as well as fewer grade 3 or 4 adverse events (223).

According to the ESMO 2016 guidelines, nivolumab is currently recommended in

second- and third-line settings, depending on the patient’s risk classification (164).

Several ongoing studies are evaluating nivolumab as well as other anti-PD-1/PD-L1

agents in combination with VEGF-targeted therapies.

3.3.3.5 Optimal Sequencing

With several new agents available for the treatment of advanced RCC, sequencing

of these therapies is of great relevance. The treatment strategies according to the

ESMO 2016 guidelines portrayed in Figure 4 are on the verge of a major change.

When choosing the first-line treatment, the options have traditionally been relatively

limited. Current data support the use of VEGF tyrosine kinase therapies, sunitinib

and pazopanib, in this setting. In some institutions, high-dose interleukin-2 therapy

is used for young and healthy patients with a good performance status, as it is the

only therapy associated with durable long-term responses (194-196). Recent data

from the CheckMate-214 and CABOSUN studies are, however, likely to change the

first-line treatment paradigm of mRCC. Results from the phase II CABOSUN trial

showed a median PFS of 8.6 months compared with 5.3 months for previously

untreated mRCC patients taking cabozantinib or sunitinib, respectively (P = 0.0008)

Page 48: Novel Biomarkers in Metastatic Renal Cell Carcinoma

48

(224). Similarly, results from the CheckMate-214 study demonstrated significantly

improved OS with combination immunotherapy (nivolumab+ipililumab) in

comparison to standard of care sunitinib among 1082 treatment-naïve mRCC

patients (225).

Optimal sequencing of drug classes beyond first-line treatment remains unknown.

Options include TKIs, cabozantinib, nivolumab, and the combination of lenvatinib,

a receptor tyrosine kinase inhibitor, and everolimus. In the AXIS trial, which led to

the approval of axitinib, patients with first-line immunotherapy demonstrated a

median PFS of 12 months on axitinib as compared to 6.5 months on sorafenib,

supporting its use among patients with prior immunotherapy (226). As approval for

the combination of lenvatinib and everolimus was based on a phase II study with

only 100 patients (227) as compared to the large randomized phase III trials

investigating cabozantinib and nivolumab (214, 228), the level of evidence favors

the use of cabozantinib and nivolumab among post-TKI patients. Ongoing trials

investigating these agents alone and/or in combinations in first- and later-line

settings may change current clinical guidelines regarding the optimal sequencing of

varied therapies.

Page 49: Novel Biomarkers in Metastatic Renal Cell Carcinoma

49

Figu

re 4

. ESM

O G

uide

lines

201

6 (A

dapt

ed fr

om E

SMO

Clin

ical

Pra

ctic

e G

uide

lines

for d

iagn

osis

, tre

atm

ent a

nd fo

llow

-

up. E

scud

ier e

t al.

Ann.

Onc

ol. 2

016)

(164

).

1 Pa

tient

s cat

egor

ized

into

goo

d, in

term

edia

te, a

nd p

oor r

isk g

roup

s bas

ed o

n IM

DC

risk

cla

ssifi

catio

n.

Page 50: Novel Biomarkers in Metastatic Renal Cell Carcinoma

50

3.3.3.6 Adjuvant Therapy in RCC

The only curative treatment for patients with stage I–III RCC is surgery. However,

the 5-year relapse rates after surgical treatment in patients with stage II–III disease

are 30% to 40% (162). With an increasing variety of novel agents now available for

systemic therapy, the search for effective adjuvant therapy, i.e. systemic therapy

following surgery, in underway. The first positive phase III adjuvant therapy trial (S-

TRAC) investigated the use of sunitinib in an adjuvant setting. With disease-free

survival (DFS) as the primary endpoint, the study demonstrated a median DFS

duration of 6.8 years versus 5.6 years in the placebo arm. However, the largest trial

to date, the ASSURE trial, in which nearly 2000 patients with completely resected

RCC were randomly assigned to sunitinib, sorafenib, or placebo, showed no

significant differences between treatment arms in DFS or OS (229). Important

differences between S-TRAC and ASSURE are that whereas S-TRAC recruited

patients with pT3-4 disease, ASSURE also enrolled patients with PT1b and pT2

disease. Additionally, S-TRAC only included patients with ccRCC histology, and

the final dropout rates due to treatment toxicity were significantly different in the

two studies. A recent phase III trial, PROTECT, evaluated the efficacy and safety of

pazopanib versus placebo among 1538 post-nephrectomy patients with localized or

locally advanced RCC. Results from the primary analyses of DFS showed no benefit

of pazopanib over placebo (230). However, a relationship between dose exposure

and an improved clinical outcome was noted, suggesting that patients achieving

higher levels of pazopanib exposure derived more clinical benefit from the treatment

(231).

As promising as the results from S-TRAC are, there are several questions

surrounding these findings. It has been hypothesized that adjuvant sunitinib may

simply delay the time to recurrence with no effect on the cure rate. In addition,

Page 51: Novel Biomarkers in Metastatic Renal Cell Carcinoma

51

concern over possible resistance to therapies for metastatic RCC after adjuvant

sunitinib has been raised. (232)

Specific reasons as to why sunitinib improved DFS in S-TRAC and not in ASSURE

remain unanswered. Similarly, it has been hypothesized that the lack of significant

benefit in the PROTECT trial could be due to the lack of efficacy with 600 mg

pazopanib. In addition to the three completed adjuvant trials, there are several

ongoing phase III trials (depicted in Table 3), which may shed light on these

questions. A closer examination of whether the differences between trials reflect

differences in patient selection, dosing, and/or study design is of critical importance.

Several trials investigating immunotherapy agents in an adjuvant setting are

additionally recruiting patients. These trials are targeting high-risk populations, and

they include PROSPER investigating nivolumab, IMMotion investigating

atezolizumab, KEYNOTE investigating pembrolizumab, and CheckMate

investigating the combination of nivolumab and ipilimumab. As these agents have

shown impressive clinical activity in patients with mRCC, the results are eagerly

anticipated.

Page 52: Novel Biomarkers in Metastatic Renal Cell Carcinoma

52

Table 3. Completed and ongoing adjuvant trials

Trial Randomization TreatmentDetails

N Inclusion Criteria(Stage/Grade)

Inclusion Criteria (Histology)

Results

ASSURE Sorafenib

Sunitinib

400 mg daily, 54 wks

37.5 mg daily (4 wks on/2 wks off), 54 wks

1943 pT1b N0 M0 (grade 3-4)pT2-pT4 N0 M0pT (any) N1 M0

Any histology

DFS:HR 0.97 (CI 0.80-1.17) vs. placebo

DFS:HR 1.02 (CI 0.85-1.23) vs. placebo

S-TRAC Sunitinib 50 mg daily (4 wks on/2 wks off), 9 cycles

615 pT3 N0 M0 (grades 2-4)pT4 N0 M0pT (any) N1 M0

ccRCC only

DFS:HR 0.76 (CI 0.59-0.98; P = 0.03)

PROTECT Pazopanib 600 mg daily, 1 year

1500 pT2 N0 M0 (grades 3-4)pT3-4 N0 M0pT (any) N1 M0

ccRCC only

DFS:HR 0.86 (CI 0.70-1.06)

ATLAS Axitinib 5 mg twice per day, 3 years

724 pT2 N0 M0pT (any) N1 M0

ccRCC only

Ongoing

SORCE Sorafenib 400 mg daily, 1 year or 400 mg daily, 3 years

1420 pT1a N0 M0 (grade 4)pT1b N0 M0 (grades 3-4) pT2-4 N0 M0pT1b-4 N1 M0

Any histology

Ongoing

EVEREST Everolimus 10 mg daily

1545 pT1b N0 M0 (grades 3-4)pT2-4 N0 M0pT (any) N1 M0

Any histology

Ongoing

DFS = disease-free survival

Page 53: Novel Biomarkers in Metastatic Renal Cell Carcinoma

53

4 Biomarkers in mRCC

The term ‘biomarker’ refers to a broad subcategory of medical signs, which can be

measured accurately and reproducibly. They stand in contrast to medical symptoms

perceived by patients themselves and are by definition objective, quantifiable

characteristics of biological processes and are commonly used in basic and clinical

research. Examples of biomarkers include everything from blood pressure to more

complex tests of blood and other tissues. (233)

The ever-growing understanding of the biology and the molecular mechanisms

underlying the pathogenesis of RCC have offered novel means to predict tumor

behavior. Indeed, since the discovery of the inactivation of the VHL gene in the

majority of ccRCCs, several new and promising tissue- and blood-based biomarkers

have been identified. The complexity of the molecular pathways and the

heterogeneous nature and wide diversity of RCCs at the molecular level have,

however, made the incorporation of these biomarkers into clinical practice

challenging. Presently, the prognostication of RCC relies heavily on clinical factors

and, even more importantly, despite molecularly-targeted therapies comprising the

foundation of treatment strategies in mRCC, treatment decisions are solely based on

clinical factors. Current biomarkers mainly provide information regarding the

outcome independent of treatment, and no validated predictive biomarkers are

available. The search for predictive biomarkers identifying patients for more

personalized treatment of advanced RCC is rigorous and ongoing.

Page 54: Novel Biomarkers in Metastatic Renal Cell Carcinoma

54

4.1 Prognostic and Predictive Biomarkers

Prognostic biomarkers are biomarkers used to identify the likelihood of a clinical

event such as disease progression or recurrence, whereas predictive biomarkers

identify individuals more likely to experience a favorable or unfavorable effect from

exposure to a medical product or agent than similar individuals without the

biomarker. As discussed earlier, the current prediction of a patient’s clinical outcome

mainly relies on clinical and pathological variables in both localized and metastatic

RCC; these variables, however, poorly reflect the individual tumor biology seen in

RCC. Biomarkers improving prognostication are urgently needed to allow for more

accurate prognostic models.

Most of the identified biomarkers in RCC are directly associated with the VHL

defect. The VHL protein (pVHL) regulates hypoxia inducible factor- -

which is a transcription factor inducing the transcription of several hypoxia-regulated

genes (234). Under normal oxygen tension, pVHL binds to HIF-

subsequently destroyed by the cell through ubiquitinization. With genetic alterations

rendering the VHL gene inactive, however, the loss of pVHL leads to dysregulation

of this cascade, resulting in overexpression of various angiogenic proteins. (235)

Enhanced understanding of this molecular pathway played a key role in identifying

new approaches in drug development for mRCC and has since also led to the

discovery of several potential biomarkers. Several molecular markers have indeed

been investigated, but so far none have proven reliable in predicting the treatment

outcome. Their use is therefore not recommended in routine clinical practice.

However, the success of targeted therapies relies on appropriate patient selection to

identify patients likely to respond to treatment and to avoid unnecessary toxicity.

Page 55: Novel Biomarkers in Metastatic Renal Cell Carcinoma

55

4.1.1 Clinical-related Biomarkers

Motzer et al. were the first to investigate pretreatment clinical features and survival.

In a study among 670 mRCC patients, a low Karnofsky performance status, high

serum lactate dehydrogenase, low hemoglobin, high corrected calcium, and a time

from diagnosis to treatment of <1 year were identified to associate with shorter

survival. Based on the presence or absence of these risk factors, a risk classification,

the Memorial Sloan Kettering Cancer Center (MSKCC) prognostic model, was

constructed, categorizing patients into favorable, intermediate, and poor risk groups

for which the median survival times were separated by 6 months or more. In the

favorable risk group with zero risk factors, the median time to death was 20 months.

In the intermediate and poor risk groups, median survival was 10 and 4 months,

respectively. (236)

Later, in 2009, Heng et al. validated the use of components of the MSKCC prognostic

model in a landmark retrospective study among 645 mRCC patients treated with

VEGF pathway inhibitors. In addition to the previously described risk factors, Heng

et al. demonstrated that high pretreatment platelet and neutrophil counts were

associated with shorter survival. Based on these findings, a prognostic model, the

International Metastatic Renal Cell Carcinoma Database Consortium (IMDC), was

constructed, demonstrating a median OS of 27 and 8.8 months in intermediate and

poor-risk patients, respectively. Among patients with a favorable risk profile, the

median OS was not reached, and the 2-year OS was 75%. (237) These results were

later externally validated among 1028 mRCC patients treated with sunitinib,

sorafenib, pazopanib, bevacizumab, or axitinib (184).

The MSKCC and IMDC prognostic models are still used for the prognostication of

mRCC patients, as well as the stratification of patients for clinical trials. As these

models have mainly been used in the era of cytokines and anti-VEGF therapies, their

Page 56: Novel Biomarkers in Metastatic Renal Cell Carcinoma

56

role with modern immunotherapy agents remains to be elucidated. The MSKCC and

IMDC prognostic models are depicted in Table 4.

The further search for new predictive and prognostic markers has additionally

provided preliminary evidence of hyponatremia as a prognostic factor for mRCC

patients. Jeppesen et al. were the first to demonstrate that baseline sodium levels

below the normal range were associated with shorter survival among mRCC patients

treated with IL-2 and IFN- (238). Since then, several studies have shown a similar

association between hyponatremia and a poor outcome among mRCC patients

treated with tyrosine kinase inhibitors (239-241). Sodium values are not, however,

routinely used in clinical practice to improve the prognostication of mRCC patients.

Page 57: Novel Biomarkers in Metastatic Renal Cell Carcinoma

57

Table 4. Comparison between the MSKCC and the IMDC Prognostic Risk

Criteria for RCC

MSKCC Criteria IMDC CriteriaRisk factorsKarnofsky performance status < 80%

x x

Time from diagnosis to treatment < 1 year

x x

xHemoglobin level below LLN

x x

Corrected serum calcium level above ULN

x x

Platelet count above ULN xNeutrophil count above ULN

x

Entry Population CriteriaPatients with metastatic RCC

Treated with interferon alfa as initial systemic therapy

Treated with first-line TKI therapy

Distribution of Risk Groups0 criteria (favorable) 168 pts (25%) 133 pts (21%)1–2 criteria (intermediate) 355 pts (53%) 301 pts (47%)

ia (poor) 147 pts (22%) 152 pts (32%)Median OS, by Risk Group0 criteria (favorable) 20.0 mo Not reached1–2 criteria (intermediate) 10.0 mo 27.0 mo

4.0 mo 8.8 mo LDH = lactate dehydrogenase; LLN = lower limit of normal; ULN = upper limit of normal; pts =

patients; mo = months

Page 58: Novel Biomarkers in Metastatic Renal Cell Carcinoma

58

4.1.2 Vascular Endothelial-derived Growth Factor

The VEGF family comprises five different mammalian ligands: VEGF-A, VEGF-B,

VEGF-C, VEGF-D, and placenta growth factor (PLGF). These growth factors

function as regulators of angiogenesis and lymphangiogenesis, and they bind to three

different but structurally related receptor tyrosine kinases: VEGFR-1, VEGFR-2,

and VEGFR-3 (242).

In RCC, where angiogenesis is an essential event of tumorigenesis, upregulation of

VEGF due to the loss of pVHL is well documented (243, 244). VEGF-A, which is

the most widely studied member of the family, has been shown to promote

tumorigenesis through a variety of ways. It acts on tumor endothelial cells to increase

their proliferation, migration, and permeability and it inhibits vessel maturation

(245). An immunomodulatory role has also been suggested (246). Since many tumor

cells express VEGFRs, VEGF-A may also possess more direct effects in supporting

tumor growth and invasion (247).

Previous research has demonstrated that a low baseline concentration of VEGF-A is

prognostic for overall survival in mRCC in univariate analyses (248, 249). High

serum VEGF-A levels have additionally been associated with the tumor stage and

grade and a poor prognosis (250). Rini et al. reported that low VEGF-C and soluble

VEGFR-3 concentrations were associated with longer PFS and higher response rates

in bevacizumab-refractory patients treated with sunitinib (251). These results are

supported by a phase III multicenter trial demonstrating an association with low

baseline levels of VEGF-C and prolonged PFS among a total of 750 mRCC patients

(248). Additionally, soluble VEGFR-3 concentrations correlated with the outcome

among patients treated with sunitinib, but not among patients on the

Page 59: Novel Biomarkers in Metastatic Renal Cell Carcinoma

59

arm, suggesting a possible role for soluble VEGFR-3 in predicting sunitinib

treatment efficacy (248).

Evidence regarding soluble VEGFR-2 as a biomarker remains controversial.

Gruenwald et al. noticed decreased soluble VEGFR-2 concentrations during

sunitinib treatment, but this kinetic modulation was insufficient to predict the tumor

response (252). On the other hand, Terakawa et al. showed that increased VEGFR-2

expression in a specimen obtained from radical nephrectomy correlated significantly

with longer PFS (253).

4.1.3 Carbonic Anhydrase IX

Carbonic anhydrase IX (CAIX) is a HIF-1a-regulated, transmembrane protein that

regulates intracellular pH in response to hypoxia. In ccRCC, CAIX is overexpressed

due to inactivation of the VHL gene product, resulting in overexpression of HIF-1a,

a transcription factor for CAIX (25).

CAIX is present in more than 80% of primary and metastatic RCCs, whereas it is

detected in only 9% of normal kidneys (254). Several studies have reported increased

CAIX expression as an independent predictor of longer disease-specific survival in

mRCC (255-257), but recent studies have been unable to confirm this finding (186,

258). Considering that 94–100% of ccRCCs stain positively for CAIX (254, 257), it

may be helpful in establishing a diagnosis, but its value as a biomarker improving

prognostication or predicting the treatment response is unclear.

Page 60: Novel Biomarkers in Metastatic Renal Cell Carcinoma

60

4.1.4 Cytokine and Angiogenic Factors

Several cytokines as well as other proteins of the angiogenic cascade have been

evaluated as biomarkers for the response in RCC. A recent retrospective analysis of

phase II and phase III trials evaluated 17 different cytokine and angiogenic factors

(CAFs) among patients with mRCC treated with pazopanib, identifying seven

promising biomarkers: interleukin 6 (IL-6), interleukin 8 (IL-8), VEGF, osteopontin,

E-selectin, hepatocyte growth factor (HGF), and tissue inhibitor of

metalloproteinases (TIMP)-1. In the study, low concentrations of these factors

correlated with increased tumor shrinkage, PFS, or both. IL-6, IL-8, and osteopontin

were additionally shown to be stronger prognostic markers than any single clinical

classification when stratified by the ECOG performance status, MSKCC risk group,

or HENG risk group. Based on the results, a CAF signature was built demonstrating

a significantly shorter PFS and OS for pazopanib-treated patients in the high CAF

group (259).

Another study investigating CAFs in sorafenib-treated mRCC patients identified six

baseline markers that significantly correlated with PFS. Higher concentrations of

osteopontin, CAIX, VEGF, collagen IV (ColIV), and soluble VEGFR-2 were

associated with shorter PFS, while the opposite relationship was seen for tumor

necrosis factor-related apoptosis-inducing ligand (TRAIL). In a dichotomized CAF

signature for these six biomarkers, ‘signature-negative’ patients demonstrated

improved PFS with a hazard ratio of 0.2 (P = 0.00002). Furthermore, a significant

interaction was noted between the CAF signature and treatment arm (sorafenib alone

or sorafenib + , suggesting a possible predictive role for this biomarker panel

(260).

Page 61: Novel Biomarkers in Metastatic Renal Cell Carcinoma

61

While several studies have yielded promising results for CAFs as prognostic, and

even predictive, biomarkers in mRCC, some issues have limited their clinical

usefulness. First, CAFs have mainly been investigated among mRCC patients treated

with TKIs. Little or no data exist regarding CAFs in mRCC patients treated with

other therapies such as mTOR inhibitors. Given the significant number of therapies

available for mRCC, prospective studies comparing CAFs between different

treatment arms are needed. Second, methodological problems regarding

measurement and cut-off values for these biomarkers exist, limiting their more

widespread application in clinical settings.

4.1.5 Single Nucleotide Polymorphisms

Single nucleotide polymorphisms (SNPs) are variations occurring in a single

nucleotide at a specific position in the genome. They are common mutations that can

alter the function of genes in any chromosome. Several studies have investigated the

possible association of various SNPs and the outcome in mRCC. The main focus has

been on identifying prognostic and predictive SNPs for VEGF-targeted therapy.

Two studies investigating SNPs involved in the pharmacokinetic and

pharmacodynamic pathways of sunitinib suggested that polymorphisms in VEGFR1,

VEGFR3, CYP3A5, NR1/3, and ABCB1 might be able to define a subset of patients

with a decreased sunitinib response and tolerance (261, 262). Additionally, three

separate studies investigated two SNPs of VEGFR1 in sunitinib-treated patients,

demonstrating a favorable association between these mutations and the outcome

(263-265). However, a recent meta-analysis by Liu et al. was unable to verify this

association, thus questioning their clinical use as biomarkers of the sunitinib

response (266).

Page 62: Novel Biomarkers in Metastatic Renal Cell Carcinoma

62

Escudier et al. investigated 15 different SNPs among patients in the phase III AXIS

trial, a study comparing axitinib and sorafenib in a second-line setting. Although the

authors were able to identify a polymorphism in VEGFR2 predicting improved OS

and PFS for sorafenib-treated patients, the authors concluded that

sensitivity/specificity limitations preclude its use for selecting patients for sorafenib

treatment (267).

For pazopanib-treated patients, three polymorphisms in IL-8 and HIF-1a and five

polymorphisms in HIF-1a, NR1/2, and VEGF-A were shown to be significantly

associated with PFS and the response rate in a study by Xu et al. (268). The same

authors pursued these suggestive associations among patients from the phase III trial

COMPARZ comparing pazopanib and sunitinib, and from an observational study of

sunitinib-treated patients. In a combined analysis, IL-8 polymorphisms were

associated with shorter OS in both pazopanib- and sunitinib-treated mRCC patients,

raising the possibility that IL-8 polymorphisms may be associated with the outcome,

irrespective of the treatment (269).

4.1.6 Immune Markers

Several clinical and laboratory factors have been investigated and identified as being

prognostic and predictive for mRCC patients treated -2. These

include the performance status, clear cell histology, MSKCC risk classification, C-

reactive protein (CRP), neutrophil levels, and the number of metastatic sites. For

modern immunotherapy agents, the search for predictive and prognostic biomarkers

is ongoing and some preliminary results have already shown promise.

Page 63: Novel Biomarkers in Metastatic Renal Cell Carcinoma

63

PD-1 is an immune checkpoint molecule expressed in activated T and B cells. It

binds to two ligands, PD-L1 and PD-L2. The interaction of PD-1 and PD-L1 leads

to immune suppression through negative regulation of activated T cell effector

functions. In RCC, PD-L1 is overexpressed in 30% of tumors and correlates with a

more advanced tumor stage, higher Fuhrman grade, sarcomatoid differentiation, and

poor survival (270, 271). With the emerging role of anti-PD-1/PD-L1 agents in the

treatment of mRCC, PD-L1 expression is currently being investigated as a potential

predictive biomarker for patients treated with these agents. A phase I trial using a

5% cut-off value for PD-L1 expression among previously treated mRCC patients

reported higher response rates for nivolumab among PD-L1-positive patients as

compared to PD-L1-negative patients (22% vs. 8%) (219). Research has revealed

similar associations among patients with metastatic melanoma and non-small-cell

lung cancer (272). In the recent phase III CheckMate 214 study comparing the

combination of nivolumab and ipilimumab with sunitinib, the ORR significantly

favored the combination over sunitinib in intermediate/poor-risk patients with

baseline PD-L1 expression of (58% vs. 25%; P = 0.0002) (225). Additionally,

in a phase I study of atezolizumab, an anti-PD-L1 agent, exploratory subanalyses

demonstrated that upregulation of PD-L1 in on-treatment biopsies as compared to

baseline expression was associated with an improved response rate, suggesting a

possible role as an on-treatment predictive biomarker (273).

Several issues, however, limit the use of PD-L1 expression as a predictive biomarker.

First and foremost, it fails to identify all responders and not all PD-L1-positive

tumors respond to treatment. Furthermore, there are currently no standardized cut-

off points to define the positivity of a sample.

Another promising immune-related biomarker is tumor T-cell infiltration. In a phase

I study investigating nivolumab, baseline tumor CD3+ and CD8+ T-cell infiltrates

correlated with a decrease in the tumor burden (272). A prospectively designed

Page 64: Novel Biomarkers in Metastatic Renal Cell Carcinoma

64

exploratory analysis from the S-TRAC trial also identified tumor CD8+ T-cell

density as predicting longer DFS for sunitinib but not placebo (274).

Other biomarkers under investigation include PD-L2 expression, the presence of

CD20+ B-cells, gene expression profiling, and the T-cell receptor repertoire, but

none of them have so far been validated.

4.1.7 Mechanism-based Adverse Events

Current understanding depicts treatment-related toxicity events as on-target effects

resulting from the inhibition of a given pathway. It therefore stands to reason that

these on-target effects are associated with treatment efficacy and could serve as

surrogate markers of the pharmacodynamic effect. Research has indeed shown that

a class-specific adverse event of VEGFR inhibitors, hypertension (HTN), associates

with an improved outcome. Following the preliminary results of Bono et al.,

demonstrating a favorable association of bevacizumab-induced HTN and the

outcome (275), a phase III trial investigating the combination of bevacizumab and

interferon- vs. interferon- later confirmed these findings. In the study,

patients on combination therapy who developed grade 2 HTN had significantly

longer PFS (13.2 vs. 8.0 months; P = 0.001) and OS (41.6 vs. 16.2 months;

P = 0.001) as compared to patients who did not develop HTN (276). A retrospective

analysis among 544 mRCC patients by Rini et al. revealed a similar association of

sunitinib-induced HTN and the outcome (277). Supporting evidence exists for

axitinib (278), sorafenib (226), and tivozanib (279).

Page 65: Novel Biomarkers in Metastatic Renal Cell Carcinoma

65

In addition to hypertension, several other class-specific adverse events have been

examined as potential biomarkers of anti-VEGFR treatment efficacy. Donskov et al.

examined hand-foot syndrome, fatigue, neutropenia, and thrombocytopenia among

sunitinib-treated mRCC patients. In the analyses, neutropenia was significantly

associated with longer PFS and OS, and hand-foot syndrome with longer OS (280).

Similarly, Rautiola et al. demonstrated that in addition to HTN, neutropenia and

thrombocytopenia were predictors of improved treatment efficacy. A biomarker

profile categorizing patients into favorable, intermediate, and poor risk profiles

according to the presence of these AEs was constructed, demonstrating a clear

difference in OS to the benefit of patients with all three AEs vs. patients with none

of the AEs (OS; not reached vs. 5.3 months; P < 0.001) (281). Evidence regarding

treatment-induced hypothyroidism is less clear. While many smaller trials have

suggested a possible association of hypothyroidism and the treatment outcome, a

large meta-analysis comprising several retrospective and prospective trials found no

significant association of acquired hypothyroidism and the outcome for mRCC

patients treated with sunitinib (282).

For mTOR inhibitors, previous research has shown that non-infectious pneumonitis,

a class effect of mTOR inhibitors, may associate with an improved outcome. In the

largest study to date, Atkinson et al. demonstrated that pneumonitis independently

predicted improved OS (HR 0.32; P < 0.001) for mRCC patients treated with

everolimus. Other class-specific AEs, including serum cholesterol, triglyceride, and

glucose levels, have also been investigated. In a phase III trial among intermediate

and poor-risk temsirolimus-treated mRCC patients, increases in on-treatment serum

cholesterol levels above baseline values were associated with longer PFS (HR 0.81,

P < 0.0001) and OS (HR 0.77, P < 0.0001). However, no such association was noted

for hypertriglyceridemia or hyperglycemia (283).

Page 66: Novel Biomarkers in Metastatic Renal Cell Carcinoma

66

4.2 Incorporation of Biomarkers into Clinical Practice and Future Directions

Despite the widespread use of targeted therapies in mRCC, reliable predictive

biomarkers are still lacking, and the accepted biomarkers are mainly prognostic and

clinical-related. Even though mechanism-based toxicities, such as hypertension,

seem to serve as surrogate markers of treatment efficacy, they do not help in patient

selection. As discussed earlier, several promising results exist regarding modern

immunotherapy agents. However, most of the published evidence remains at an

immature stage with inconsistencies for many of the investigated biomarkers. In

addition to problems arising from the lack of cut-off points and standardization,

tumor heterogeneity and the use of archival tissue samples remain unresolved issues.

Promising avenues that have recently emerged are the sampling of circulating tumor

DNA (ctDNA) and microRNA (miRNA) profiling. ctDNA is obtained from

peripheral blood and is therefore non-invasive and allows for dynamic evaluation of

tumor genomics and the response to treatment. A few studies have already shown

that ctDNA may be a useful tool in monitoring the tumor burden and response to

treatment (284, 285). Similarly, miRNA from blood plasma is easily obtained and

recent studies have demonstrated that specific miRNA signatures may predict the

disease outcome and recurrence in ccRCC (286-288).

With the renaissance of immunotherapy agents in the treatment of mRCC, the

integration and optimization of these therapies into new treatment algorithms is of

vital importance. While decades of research have demonstrated the immunological

and molecular heterogeneity of RCC, novel technologies such as real-time PCR,

microarrays, and next-generation sequencing are bringing us closer to identifying

robust and reliable prognostic and predictive biomarkers, allowing for a molecularly

stratified biomarker-guided treatment approach.

Page 67: Novel Biomarkers in Metastatic Renal Cell Carcinoma

67

AIMS OF THE STUDY

The aim of the study was to discover markers predicting treatment efficacy for

targeted therapies used in the treatment of mRCC and to gain knowledge of those

deeper factors through which we could enhance treatment efficacy.

Specific aims of the study were:

1) To identify clinical prognostic and predictive biomarkers for mRCC patients

treated with targeted therapy;

2) To identify molecular prognostic and predictive biomarkers for mRCC

patients treated with targeted therapy;

3) To investigate the mechanism-based treatment-related adverse events of

targeted therapies and their correlation with clinical efficacy.

Page 68: Novel Biomarkers in Metastatic Renal Cell Carcinoma

68

PATIENTS AND METHODS

1. Patients and Treatment

The patient cohorts are described in detail in the original publications I–IV. The first

study included 303 consecutive mRCC patients treated with a first-line anti-VEGF

agent, either sunitinib or pazopanib, at the Comprehensive Cancer Center, Helsinki

University Hospital, between October 18, 2006 and December 31, 2014. None of the

patients had received prior TKI therapy. Eighteen patients (5.9%) had received prior

interferon alfa. Both sunitinib and pazopanib were administered according to

standard care until disease progression or unacceptable toxicity. Altogether, 23%

(N = 59) of the patients started sunitinib with intermittent dosing (4 weeks on-

treatment, 2 weeks off-treatment) and 77% (N = 208) with continuous dosing.

Pazopanib was administered continuously.

The second study comprised mRCC patients treated with first-line sunitinib at the

Comprehensive Cancer Center, Helsinki University Hospital, between October 18,

2006 and May 31, 2012. We were able to identify 137 patients for whom sufficient

clinical data and sufficient histologic specimens were available.

Studies III and IV were performed in collaboration with Aarhus University Hospital,

Denmark. We identified a total of 85 patients who were treated with everolimus after

prior anti-VEGFR therapy failure at the Comprehensive Cancer Center, Helsinki

University Hospital, between October 18, 2006 and December 31, 2014. A similar

cohort comprising 148 patients treated between January 25, 2010 and June 6, 2016

at Aarhus University Hospital, Denmark, was collected. In both cohorts, everolimus

was administered according to standard care until disease progression or

unacceptable toxicity.

Page 69: Novel Biomarkers in Metastatic Renal Cell Carcinoma

69

1.1 Assessment of the Tumor Response and Adverse Events

In all studies, the response to treatment was assessed by computed tomography at 8-

to 12-week intervals. Treatment efficacy was reported according to the Response

Evaluation Criteria in Solid Tumors (RECIST) versions 1.0 and 1.1. Adverse events

were captured every 4–6 weeks and were graded according to the Common

Terminology Criteria for Adverse Events (CTCAE) versions 3.0 and 4.0.

1.2 Assessment of Pneumonitis

To correctly assess whether a patient developed pneumonitis during everolimus

treatment, medical files, including CT scan reports, were retrospectively reviewed

for pneumonitis. The radiographic studies were subjected to a blinded review by an

experienced radiologist for findings indicative of pneumonitis. A more detailed

description is provided in the original publication (III). Figure 5 depicts typical

radiographic findings indicative of pneumonitis.

Figure 5. CT scans at the onset of pneumonitis showing peripheral, bilateral ground

glass opacities.

Page 70: Novel Biomarkers in Metastatic Renal Cell Carcinoma

70

1.3 Immunohistochemistry

In the second study, the expression of c-Met was analyzed from formalin-fixed

paraffin-embedded samples by using an anti-c-Met rabbit monoclonal ready-to-use

antibody by Roche, and BenchMark XT immunostainer by Ventana Medical

Systems. The staining was carried out according to the manufacturer’s protocol. c-

Met staining was scored by two independent evaluators with good concordance

(kappa value 0.7) according to intensity. A 4-tier system was used with 0 as no

staining, 1+ weak, 2+ strong, and 3+ as very strong staining intensity. The c-Met

immunohistochemistry score was also determined by assessing the proportion of

stained cells as the intensity of the staining. Negative (0): fewer than 50% of tumor

cells with membrane and/or cytoplasmic

tumor cells with weak or higher membrane and/or cytoplasmic staining but <50% of

tumor cells with a

cells with moderate or strong membrane and/or cytoplasmic staining but <50% of

tumor cells with a

with a strong membrane and/or cytoplasmic staining intensity. Figure 6 displays

examples of each category.

Figure 6. Examples of negative, moderate, strong, and very strong staining (A–D)

of anti-c-Met antibody with 400x magnification. (Reprinted from Peltola KJ et al.

Correlation of c-Met Expression and Outcome in Patients With Renal Cell

Carcinoma Treated With Sunitinib. Clin Genitourin Cancer. 2017 Aug;15(4):487-

494 with the permission of Elsevier)

Page 71: Novel Biomarkers in Metastatic Renal Cell Carcinoma

71

2. Statistical Analysis

All the statistical analyses were performed using IBM SPSS Statistics for Windows

(versions 22.0–24.0, Armonk, NY, USA, IBM Corp.). Tests used to assess the

association of various clinicopathological factors were the Mann-Whitney U-test for

continuous data and the chi-squared test for categorical data. In all original

publications, OS was defined as the time from treatment initiation to death from any

cause, and (ii) PFS as the time from treatment initiation to the first event (tumor

progression or death from any cause). The Kaplan-Meyer method was used to

estimate the median survival times with 95% confidence intervals for both OS and

PFS, censoring the patients who were alive or had no disease progression at the last

follow-up visit.

Uni- and multivariate Cox proportional hazard models were used to assess the

association of OS and PFS with various clinicopathological factors. Interactions

between the factors of interest were tested with inclusion of the interaction terms in

the models. The proportional hazards assumptions were assessed graphically,

obtaining plots of (log(-logS(t)) versus time and Schoenfeld residuals versus time.

Uni- and multivariate logistic regression models were used to investigate the effects

of various clinicopathological factors and categorical end-point data. The results are

expressed as odds ratios (ORs) with 95% CI.

To control for time bias when investigating potentially time-biased parameters, time-

dependent Cox regression models and landmark survival analyses were applied.

According to the landmark method, PFS and OS were defined as the time from the

landmark to progression or death from any cause.

All statistical tests were two-sided and P-values of <0.05 were considered as

statistically significant.

Page 72: Novel Biomarkers in Metastatic Renal Cell Carcinoma

72

3. Ethical Considerations

The Hospital District of Helsinki and Uusimaa (HUS) accepted the research plan

§36/28.4.2014 and the Ethics Committee of the Department of Surgery of HUS

accepted the research plan §190/23.10.2013. The National Authority for Welfare and

Health licensed the use of pathological archive material.

Page 73: Novel Biomarkers in Metastatic Renal Cell Carcinoma

73

RESULTS AND DISCUSSION

Ever since the introduction of targeted therapies in the treatment of metastatic RCC

in the mid-2000s, the search for reliable biomarkers has been of great interest. Even

though there are presently no such biomarkers that could identify patients for specific

treatments, the ever-growing knowledge of the intricate molecular and immunogenic

mechanisms of tumor biology in RCC has brought us closer to the moment of finding

clinically applicable predictive biomarkers. This is even more important now that the

first-line options to treat advanced RCC are on the verge of a major change.

1. The use of angiotensin system inhibitors (ASIs) in the treatment

of TKI-induced hypertension (I)

Hypertension (HTN) is a class effect of VEGF receptor tyrosine kinases and it has

been reported as an adverse event of all agents in this class (289-292). This class

effect also seems to function as a surrogate marker of treatment efficacy in patients

with mRCC (277, 278, 281, 293). As antihypertensive medication, angiotensin

system inhibitors have been of interest in the treatment of TKI-induced HTN, as

xenograft models have demonstrated that ASIs may possess antiangiogenic potential

(294, 295)

We investigated treatment-induced hypertension and the use of ASIs among 303

consecutive mRCC patients treated with a first-line anti-VEGFR agent. Of these 303

patients, 197 (65%) patients had baseline hypertension and 126 (64%) of these

Page 74: Novel Biomarkers in Metastatic Renal Cell Carcinoma

74

patients had ASI as the baseline antihypertensive medication, either as monotherapy

or in combination with other antihypertensive medication. Treatment-induced HTN,

defined as a recurrent, persistent, or symptomatic rise in diastolic BP of >20 mmHg

or a systolic BP rise to >150 mmHg, if previously within the normal range (CTCAE

, developed among 110 patients, of whom the majority were ASI

users (82.7%).

We found no difference in the outcome between patients with and without baseline

HTN (OS: 20.3 months (95%CI 16.4–24.2 months) vs. 20.1 months (95%CI 15.5–

24.7 months); PFS: 8.2 months (95%CI 6.6–9.7 months) vs. 8.2 months (95%CI 5.4–

11.0 months); P = 0.54 and P = 0.72, respectively). Similarly, we found no

significant differences in outcomes between patients with and without baseline ASI

use. On the other hand, patients who developed HTN during treatment had

significantly longer OS (unadjusted HR 0.41, 95%CI 0.30–0.55, P < 0.001) and PFS

(HR 0.43, 95%CI 0.33–0.56, P < 0.001). The same was true for patients receiving

ASI as a new drug or dose escalation (N = 91) when compared to the remaining

patients (OS: 37.7 months (95%CI 26.0–49.5) vs. 16.5 months (95%CI 12.9–20.2),

HR 0.38, 95%CI 0.26–0.54, P < 0.001; PFS: 16.7 months (95%CI 6.1–27.2) vs. 6.6

months (95%CI 5.8–7.5), HR 0.41, 95% CI 0.30–0.57, P < 0.001).

Due to the strong multicollinearity of HTN and ASI use, they could not be fitted in

the same multivariate analyses. We therefore conducted subgroup analyses among

patients with TKI-induced HTN. In these analyses, ASI users (N = 91) had longer

OS (37.5 vs. 18.1 months; P = 0.001) and PFS (17.1 vs. 7.2 months; P = 0.004) than

ASI nonusers. These results were supported by multivariate analyses conducted in

the same subset of patients, demonstrating superior OS (adjusted HR, 0.35; 95%CI

Page 75: Novel Biomarkers in Metastatic Renal Cell Carcinoma

75

0.18–0.66; P = 0.001) and PFS (adjusted HR, 0.42; 95%CI 0.23–0.77; P = 0.005)

for ASI users when adjusted for the baseline HENG risk classification.

Regarding TKI-induced HTN, our results are well in line with previously published

findings. In our patient cohort, treatment-induced hypertension was detected in

36.3% of patients. The previously reported incidence of TKI-induced all-grade HTN

has varied between 17% and 49.6% (296). Several studies have depicted TKI-

induced HTN as a predictive biomarker of treatment efficacy (278, 280, 281, 293,

297, 298). We found a similar association in our data, with TKI-induced HTN having

a strong association with prolonged OS and PFS, even when corrected for the

possible time bias from longer treatment. Due to disproportionate group sizes,

patients treated with sunitinib (88%) and pazopanib (22%) were not separately

analyzed.

Several retrospective studies have investigated the use of ASIs in cancer patients. In

a meta-analysis comparing ASIs in a variety of cancer patients, their use was

associated with a significant improvement in DFS and OS. Benefits were noted

among patients with urinary tract cancer, colorectal cancer, pancreatic cancer, and

prostate cancer (299). In RCC, a few studies have reported a significant association

between ASI use and an improved clinical outcome among mRCC patients receiving

VEGF-targeted therapy. All of the previous studies have investigated ASI use at

baseline or within 30 days of therapy initiation (300-302). The largest study to date,

investigating this association among 4736 patients, demonstrated a significant

improvement in OS (HR 0.84, P = 0.0105) for ASI users vs. users of other

antihypertensive medication. Additionally, this association was more prominent

among patients on VEGF-targeted therapy (302). Contradicting evidence also exists,

however, as Sorich et al. recently noted no difference in the outcome for baseline

ASI users vs. nonusers among 1545 mRCC patients treated with sunitinib and

Page 76: Novel Biomarkers in Metastatic Renal Cell Carcinoma

76

pazopanib. In this study, however, for patients treated with sunitinib there was also

a trend towards improved survival and baseline ASI use. (303) There are no obvious

differences between these studies explaining the contrasting results, but classifying

baseline ASI use within the first 30 days of VEGF-targeted therapy may contribute

to the disparity, as TKI-induced HTN, a validated mechanism-based adverse event,

often manifests within the first weeks of therapy. Furthermore, the concomitant use

of other antihypertensive medication was in most studies unaccounted for.

To the best of our knowledge, our study is the first to investigate ASI use in the

treatment of TKI-induced HTN. Within this subset of patients, we demonstrated a

significant association between the use of ASIs and an improved outcome. There are

currently no guidelines for cancer patients suffering from HTN as a result VEGF-

targeted therapies. The pathogenesis of TKI-induced hypertension is also not

completely understood. Key hypotheses include the inhibition of endothelial nitric

oxide (NO) synthase, increased vascular stiffness, inhibition of the renin-angiotensin

system, and a decrease in the density of microvessels leading to an increase in

systemic vascular resistance (304-307). Regarding the role of the renin-angiotensin

system in cancer development, preclinical data have shown that angiotensin II may

promote cancer development, as it is involved in the regulation of apoptotic

mechanisms and angiogenesis through the up-regulation of VEGF expression and

neovascularization (308, 309). It additionally facilitates cellular growth and

proliferation through several growth factors (310). In RCC, angiotensin II has been

demonstrated to correlate with tumor aggressiveness and decreased survival (311).

In murine models, ASIs have been shown to prevent the development of metastatic

lung nodules, as well as potentiate the antiangiogenic effects of sunitinib (294, 295).

Page 77: Novel Biomarkers in Metastatic Renal Cell Carcinoma

77

McKay et al. hypothesized that the improved outcome noted among ASI users could

represent a synergistic interaction of TKIs and ASIs (302). The mechanisms by

which this happens are yet to be elucidated, but it has been suggested that ASIs may

reduce sarcopenia (312). Earlier research has shown that sarcopenia combined with

a body mass index of <25 kg m 2 predicts sunitinib-induced early dose-limiting

toxicities and sorafenib-induced dose-limiting toxicities in mRCC patients (313,

314). The use of ASIs could thus improve the therapeutic index of VEGF-targeted

therapies, resulting in improved efficacy.

Our results support the hypothesis of possible synergistic interactions between TKIs

and ASIs. Based on our study, conclusions on whether the possible underlying

interactions are the result of combinatory effects on angiogenesis, tumor cell

proliferation, reduced blood flow to the tumor, or improvement in the therapeutic

index cannot be drawn. In the absence of published guidelines for the treatment of

HTN in this setting, ASIs do, however, present an interesting option as

antihypertensive medication, as they might offer a survival benefit as well as feasible

management of TKI-induced HTN in patients with mRCC. We do recognize that for

these results to be properly validated, further studies, preferably in a randomized

prospective setting, are warranted.

2. c-Met expression is associated with the outcome among mRCC

patients treated with sunitinib (II)

c-Met, a receptor tyrosine kinase, is involved in cell growth/differentiation,

neovascularization, and tissue repair in normal tissues. Interfering and directly

Page 78: Novel Biomarkers in Metastatic Renal Cell Carcinoma

78

activating mutations leading to dysregulation of c-Met and its ligand, hepatocyte

growth factor (HGF), have been implicated in tumor development, invasion, and

angiogenesis in several malignancies (83-85). In RCC, both clear cell and papillary

subtypes have demonstrated excessive activation of the c-Met/HGF pathway (315,

316). Furthermore, overexpression of c-Met is associated with poor pathologic

features and prognosis in RCC (317). In our study, we sought to analyze the possible

association of c-Met expression and the outcome among mRCC patients treated with

first-line sunitinib.

c-Met expression was analyzed from 137 formalin-fixed paraffin-embedded tumor

samples collected at diagnosis. The samples consisted of resected primary RCC

tumors (N = 134) and core needle biopsies (N = 3) originally retrieved for diagnostic

purposes. Clinical data were collected, including patient characteristics, treatments,

adverse events, hospitalizations, and outcomes. c-Met expression was divided into

two groups, with low expression consisting of expression levels 0 to 1 and high

expression consisting of levels 2 and 3.

Among the 137 patients, 78 (56.9%) patients had low c-Met expression and 59

(43.1%) high c-Met expression. Patients with low c-Met were significantly older

than patients with high c-Met (P = 0.007). Additionally, patients with low c-Met

were significantly more likely to belong to the favorable HENG criteria risk group

(P < 0.001) and have a clear cell histology (P = 0.024).

In survival analyses, patients with low c-Met expression had significantly longer PFS

(14.3 months vs. 6.5 months; P <0.001) and OS (32.1 months vs. 20.1; P = 0.049)

than patients with high c-Met expression. In multivariate analyses, low c-MET

Page 79: Novel Biomarkers in Metastatic Renal Cell Carcinoma

79

expression remained significantly associated with improved PFS when adjusted for

the HENG risk classification (adjusted HR 0.61; P = 0.016). For OS, we found no

statistically significant association (adjusted HR 0.75; P = 0.34). In a logistic

regression model investigating c-Met expression and the response to treatment,

patients with high c-Met expression were more than four times more likely to have

PD as their best response to sunitinib therapy (unadjusted OR 4.19; P = 0.004).

Based on previous hypotheses regarding an association between c-Met and the

presence of bone metastases, we conducted further subgroup analyses among

patients with (N = 31) and without (N = 106) bone metastases present at the onset of

treatment. Even though the level of c-Met expression and presence of bone

metastases were not significantly associated with each other (P = 0.47), in survival

analyses, low c-Met expression was associated with improved OS (unadjusted HR

0.63; P = 0.034) and PFS (unadjusted HR 0.47; P < 0.001) in patients without bone

metastases, but not among patients with bone metastases.

To the best of our knowledge, this is the first study to establish an association

between c-Met expression and the outcome among patients treated with first-line

anti-VEGFR therapy. Our results are similar to previously published data in the sense

that higher c-Met expression was associated with a poor prognosis and response rate.

We hypothesize that c-Met expression could thus serve as a biomarker defining

patients who are more likely to benefit from alternative therapy.

Regarding the reasons why c-Met expression was associated with PFS but not OS in

multivariate analyses, we hypothesize that this difference may be associated with the

availability of active therapies beyond first-line sunitinib. Additionally, the number

Page 80: Novel Biomarkers in Metastatic Renal Cell Carcinoma

80

of registered events was higher when using PFS as the endpoint (129 events for PFS

vs. 113 events for OS), possibly leading to a lack of statistical power in OS analysis.

In the oncology literature, there is a general trend for the HR for OS to be closer to

1.00 than the HR for PFS. Due to the longer median duration of OS, a greater number

of events is required to reach statistical significance. (318)

Preclinical evidence suggests that inhibition of VEGF activity leads to the induction

of compensatory mechanisms upregulating growth factors and cytokines. The

developing resistance is thought to be largely indirect, with angiogenic tumors

adapting to the presence of angiogenesis inhibitors. While the specific therapeutic

target remains inhibited, several adaptive mechanisms, including the activation of

alternative pro-angiogenic pathways, recruitment of bone marrow-derived

proangiogenic-cells, and increased pericyte coverage of the tumor vasculature, lead

to tumor evasion and ultimately resistance to antiangiogenic therapy (319). In a

xenograft study investigating sunitinib efficacy, the investigators demonstrated

increased levels of HGF in the stroma of sunitinib-resistant tumors, suggesting that

HGF-stimulated c-Met signaling may enable tumor cells to bypass the desired effects

of anti-VEGFR therapy (320). Regulation of c-Met activity during anti-VEGF

therapy is illustrated in Figure 7. Given the possible role of the c-Met/HGF pathway

in the development of resistance to anti-VEGFR therapy, our results stand to reason,

as patients with high c-Met expression achieved less benefit from sunitinib treatment.

Previous research in breast cancer has depicted c-Met as a mediator of the

interactions between tumor cells and the bone microenvironment, suggesting that it

is involved in the bone metastatic process (321). D’Amico et al. demonstrated that

c-Met expression on RCC stem cells drives renal cancer progression to bone, since

RCC stem cells expressing c-Met directly formed bone lesions. Furthermore, in the

same study, treatment with a selective c-Met inhibitor hindered the development of

Page 81: Novel Biomarkers in Metastatic Renal Cell Carcinoma

81

bone metastases, indicating that HGF/c-Met signaling is relevant in the metastatic

process induced by RCC stem cells (322). These findings are supported by clinical

data from the METEOR phase III trial, in which patients with bone metastases

treated with the dual inhibitor cabozantinib had a 46% risk reduction for OS and 67%

risk reduction for PFS when compared to patients treated with everolimus.

Additionally, the objective response rate in bone scans was 17% vs 0%, for

cabozantinib and everolimus, respectively (323). In our analyses, we found that in

the subgroup of patients without bone metastases, low c-Met expression predicted

an improved outcome. The implications of this finding are presently unclear, but it

could be hypothesized that low c-Met expression among patients without bone

metastases represents a subset of patients in whom resistance to antiangiogenic

therapy has not yet developed or is less likely to develop.

In the METEOR phase III trial comparing cabozantinib and everolimus among

mRCC patients after previous VEGFR tyrosine-kinase treatment, the authors

additionally investigated MET expression levels and the treatment outcome. While

the study did demonstrate relevant clinical activity for cabozantinib vs. everolimus,

it did not provide evidence for a significant relationship between the treatment

modality, c-Met expression, and the outcome. However, in as many as 41.5% of

patients, c-Met expression could not be evaluated. Additionally, archival tumor

tissue rather than fresh biopsies was used in the analyses (324). A recent phase II

trial investigating crizotinib, a small-molecule TKI inhibiting MET, anaplastic

lymphoma kinase (ALK), and ROS proto-oncogene 1 receptor tyrosine kinase

(ROS1), demonstrated that crizotinib induced long-lasting disease control in

metastatic papillary renal cell carcinoma type 1 with MET mutation and long-term

stable disease in a case with MET amplification (325). These results are of special

interest now that the CABOSUN trial has demonstrated that cabozantinib may have

Page 82: Novel Biomarkers in Metastatic Renal Cell Carcinoma

82

a role as first-line therapy and has indeed already been approved by the Food and

Drug Administration (FDA) to be used as such in the United States (326). Our

results, along with the growing evidence implicating the c-Met/HGF pathway as one

of the key regulators of tumorigenesis and angiogenic escape, warrant further studies

investigating c-Met as biomarker among patients treated with dual c-Met/VEGFR

inhibitors.

Page 83: Novel Biomarkers in Metastatic Renal Cell Carcinoma

83 Fi

gure

7. R

egul

atio

n of

c-M

et a

ctiv

ity d

urin

g an

ti-V

EGF

ther

apy.

PTP

1B=

prot

ein

tyro

sine

pho

spha

tase

1B

.(Ad

apte

d fro

m M

cCar

ty

J., G

liobl

asto

ma

Resis

tanc

e to

Ant

i-VEG

F Th

erap

y: H

as th

e C

halle

nge

been

MET

? C

linic

al C

ance

r Res

earc

h 20

13) (

327)

.

Page 84: Novel Biomarkers in Metastatic Renal Cell Carcinoma

84

3. Everolimus-induced pneumonitis predicts the treatment outcome

among mRCC patients treated with everolimus (III)

Several studies have depicted the treatment-related adverse events associated with

targeted therapies as possible predictive markers of treatment efficacy. Treatment-

induced hypertension was one of the first biomarkers shown to be favorably

associated with the efficacy of sunitinib treatment (277). Since then, multiple

different adverse events have been associated with the outcome. In addition to

hypertension, Donskov et al. demonstrated that on-treatment neutropenia was

significantly associated with longer OS and PFS and hand-foot syndrome with OS

(280). Similar results were recorded by Rautiola et al., who demonstrated that the

development of hypertension, neutropenia, and thrombocytopenia was significantly

associated with OS and PFS (281). For mTOR inhibitors, such on-treatment

biomarkers predicting treatment efficacy do not presently exist. We sought to

investigate the possible association between everolimus-induced pneumonitis and

the treatment outcome.

Our study population comprised two patient cohorts: cohort A (N = 85) and a

validation cohort B (N = 148), which were analyzed separately. In cohort A, 29

(34.1%) patients had CT-verified pneumonitis during everolimus treatment. Eight

cases were grade 1 (27.6%), 18 cases grade 2 (62.1%), and three cases were grade 3

(10.3%). No grade 4 (life-threatening) pneumonitis was recorded. In cohort B, 29

(19.6%) patients had CT-verified pneumonitis. Among patients with pneumonitis,

the median time to onset was 2.4 months (range 0.4–7.5 months) in cohort A and 2.8

months (range 0.1–14.1 months) in cohort B.

Page 85: Novel Biomarkers in Metastatic Renal Cell Carcinoma

85

In survival analyses, everolimus-induced pneumonitis was significantly associated

with OS (cohort A: 24.7 vs. 8.5 months; P < 0.001; cohort B: 12.9 vs. 6.0 months;

P = 0.02) and PFS (cohort A: 5.5 vs. 3.2 months; P = 0.002; cohort B: 6.0 vs. 2.8

months; P = 0.02) in both cohorts. Additionally, pneumonitis was associated with

an improved clinical benefit rate (CBR): 57.1% vs. 24.1% and 79.3% vs. 24.1% for

cohorts A and B, respectively. In multivariate analyses, pneumonitis remained

significantly associated with both OS (cohort A: HR, 0.22; 95%CI 0.12–0.44;

P < 0.001; cohort B: HR 0.58; 95%CI 0.36–0.94; P = 0.03) and PFS (cohort A: HR

0.37; 95%CI 0.21–0.66; P = 0.001; cohort B: HR 0.61; 95%CI 0.39–0.95; P = 0.03)

when adjusted for age, the number of prior treatment lines, and the MSKCC

classification for previously treated patients.

Pneumonitis, like all treatment-related adverse events, is dependent on the time spent

on the study treatment. To account for this possible time bias resulting from longer

treatment, we performed a multivariate analysis with pneumonitis as a time-

dependent covariate. To provide sufficient statistical power, the two patients cohorts

were combined for this analysis after it was verified that the association between

pneumonitis and OS was not significantly different in the two cohorts. In a Cox

regression model adjusted for age, gender, cohort, the number of previous treatment

lines, and the MSKCC risk classification for previously treated patients, CT-verified

pneumonitis was independently associated with longer OS (HR, 0.67; 95%CI 0.46–

0.97; P = 0.03). A landmark analysis demonstrated similar results with a significant

association between pneumonitis and improved OS (17.4 vs. 7.8 months; P = 0.01).

Page 86: Novel Biomarkers in Metastatic Renal Cell Carcinoma

86

In cohort B, data had also been collected on patients who had clinical symptoms of

pneumonitis/pneumonia but no radiological evidence of either. We therefore divided

patients in cohort B into three groups: patients with pneumonitis verified by CT

(N = 29), patients with clinical symptoms of pneumonitis/pneumonia (N = 25), and

patients with no clinical or radiological signs of pneumonitis/pneumonia (N = 94).

In a subgroup analysis comparing OS, PFS, and the CBR between these groups, there

was no statistically significant difference in any of the endpoints between patients

with CT-verified pneumonitis and patients with clinical symptoms of

pneumonitis/pneumonia.

In this study, we demonstrated that everolimus-induced pneumonitis significantly

associated with all efficacy endpoints. Furthermore, we validated these findings in

an independent patient cohort. Previous work on this subject has revealed that

mTOR-inhibitor-induced pneumonitis associates with OS among mRCC patients

treated with everolimus/temsirolimus (328). Research has additionally shown that

the mean tumor shrinkage and SD according to the RECIST were significantly higher

in patients with pneumonitis (329). However, neither study controlled for bias

resulting from longer treatment, an important aspect when investigating adverse

events caused by a given treatment. We accounted for the possible time bias by

performing a landmark analysis, as well as multivariate analyses with pneumonitis

as a time-dependent covariable. Both methods demonstrated a significant association

between pneumonitis and longer OS, suggesting that our results are unlikely to be

impacted by time bias.

In our study, the incidence of CT-verified pneumonitis was 34.1% and 19.6% in

cohorts A and B, respectively. In the previous literature, the incidence of pneumonitis

has varied widely, with reported figures between 13.5% and 48.7% among mRCC

Page 87: Novel Biomarkers in Metastatic Renal Cell Carcinoma

87

patients treated with mTOR inhibitors (210, 329-332). In a large meta-analysis

comprising 2233 patients with breast cancer, neuroendocrine tumors, or mRCC, the

reported pulmonary toxicity was only 10.4% (333). Prior work has, however,

demonstrated that mTOR-inhibitor-related adverse events are more common among

patients with compromised renal function (334, 335), a phenomenon more prevalent

among patients with mRCC. Another possible explanation for the discrepancy in the

reported incidence of mTOR-inhibitor-induced pneumonitis may be due to its non-

specific clinical and radiological nature, leading to inaccurate reporting of this

adverse event. Clinically, pneumonitis manifests heterogeneously and symptoms

may include fever, fatigue, coughing, and dyspnea, nonspecific signs and symptoms

that do not facilitate diagnosis. Moreover, the traditional chest X-ray may not show

any abnormalities (335). As the diagnosis of pneumonitis is difficult and is often

made by excluding other causes such as infections, it is of relevance that clinicians

recognize pneumonitis as a possible, and a rather common, adverse event of mTOR-

inhibitor treatment.

The precise pathogenesis of mTOR-inhibitor-induced pneumonitis remains unclear.

Hypothesized mechanisms include both direct toxic effects and immunological

toxicity, and the combination of both. In a study by Morelon et al. investigating

sirolimus-associated interstitial pneumonitis, patients displayed signs of

lymphocytic alveolitis in bronco-alveolar lavage fluid analyses (336). The authors

suggested that this might represent an autoimmune response to exposed cryptic

antigens, leading to lymphocytic alveolitis and interstitial pneumonitis. The rapid

response of pneumonitis to treatment withdrawal, however, is in support of more

direct toxic effects (337, 338). A dose-dependent effect has also been suggested.

Weiner et al. investigated the relationship between plasma concentration levels of

sirolimus and pneumonitis. While the risk of pneumonitis was indeed higher among

Page 88: Novel Biomarkers in Metastatic Renal Cell Carcinoma

88

patients with increased levels of sirolimus, pneumonitis also occurred at relatively

low concentration levels (335). With regard to our finding of pneumonitis being

associated with a favorable outcome, no clear evidence exists on whether this

represents an on-target dose-dependent effect or whether there are more intricate

immunological pathways at work.

The management of mTOR-inhibitor-induced pneumonitis comprises treatment

withdrawal or dose reduction and corticosteroid administration, as shown by the

RECORD-1 trial, in which among the 18 patients with grade 2 pneumonitis,

complete resolution was obtained in 11 patients and 1 patient improved to grade 1 as

a result of corticosteroid therapy (10/18), dose adjustment (12/18), and/or

discontinuation (3/18) (332). Similarly, in an overview among organ transplant

patients receiving mTOR inhibitors, treatment withdrawal resulted in complete

resolution in 94.3% of pneumonitis cases (339). Even though most cases of

pneumonitis are relatively unaggressive and respond well to the therapeutic measures

described above, life-threatening lung toxicity may occasionally occur (337, 340).

Current guidelines suggest that everolimus treatment can be continued, or

temporarily interrupted, among patients with mild symptoms (grades 1 to 2). In more

severe cases of pneumonitis (grade 3), reintroduction of everolimus may also be

attempted (341, 342). While we did not investigate how patients with pneumonitis

were managed after the diagnosis of this adverse event, our results do prompt careful

consideration regarding permanent treatment withdrawal, as these patients seem to

draw clear benefit from everolimus treatment.

Our results add to the growing field of on-treatment biomarker research in mRCC.

They demonstrate pneumonitis to be a strong biomarker of everolimus efficacy. As

pneumonitis occurs relatively early after treatment initiation, with a median onset

Page 89: Novel Biomarkers in Metastatic Renal Cell Carcinoma

89

after 2.4 and 2.8 months in cohorts A and B, respectively, it may also be clinically

applicable in reassuring the patient and the treating physician of clinical benefit

during therapy.

4. Hyponatremia associates with a poor outcome among mRCC

patients treated with everolimus (IV)

Hyponatremia is commonly encountered among cancer patients. Most frequently, it

occurs with small cell lung cancer (SCLC) as a result of the syndrome of

inappropriate secretion of antidiuretic hormone (SIADH) (343). It is also relatively

common among other tumor types, although the distribution of causes is different.

In mRCC, the reported incidence of hyponatremia varies between 14–25% (238-

241).

Previous research has shown that hyponatremia is a predictor of a poor outcome in

several different medical conditions, such as liver cirrhosis (344), congestive heart

failure (345), and infectious diseases such as pneumonia (346), childhood meningitis

(347), and necrotizing soft-tissue infection (348). Furthermore, several studies have

reported a similar association between hyponatremia and a poor outcome in cancer

patients (349-351).

Vasudev et al. were the first to show that hyponatremia is associated with shorter OS

and DFS among pre-nephrectomy patients in localized RCC (352). Since then,

evidence has accumulated on the possible prognostic role of hyponatremia in mRCC.

Indeed, hyponatremia has been shown to associate with a poor outcome among

mRCC patients treated with cytokines (238) and TKIs (239-241). Evidence

Page 90: Novel Biomarkers in Metastatic Renal Cell Carcinoma

90

regarding the prognostic value of hyponatremias among everolimus-treated mRCC

patients is lacking, however.

The mechanisms underlying hyponatremia are not entirely understood. Of the

several mechanisms that have been suggested, an intriguing hypothesis is the

possible association of chronic inflammation and hyponatremia through elevated

interleukin-6 levels (240).

Our aim was to evaluate baseline and on-treatment sodium as prognostic biomarkers

in mRCC patients treated with everolimus. We additionally evaluated the association

between baseline sodium and baseline neutrophils or thrombocytes, markers that are

often elevated in chronic inflammation.

Our study population comprised 233 mRCC patients treated with everolimus at

Helsinki University Central Hospital, Finland (N = 85) and Aarhus University

Hospital, Denmark (N = 148). Among these 233 patients, 65 (27.9%) had sodium

below the lower limit of normal (LLN) at baseline. Within 12 weeks of treatment

initiation, 41 (18.4%) patients had sodium <LLN. Of the 65 patients with baseline

Baseline

sodium <LLN was significantly associated with elevated baseline neutrophils above

the upper limit of normal (ULN) (P = 0.002), and a similar tendency was noted for

baseline sodium <LLN and thrombocytes >ULN (P = 0.08). Additionally, baseline

sodium correlated inversely with baseline neutrophils (Spearman’s r = -0.23;

P = 0.001) and baseline thrombocytes (Spearman’s r = -0.25; P < 0.001) when

evaluated as continuous variables.

In univariate survival analyses, baseline sodium <LLN was significantly associated

with shorter OS (6.1 months vs. 10.3 months; P < 0.001) and PFS (2.8 months vs.

3.5 months; P = 0.04). Patients with on-treatment hyponatremia had significantly

Page 91: Novel Biomarkers in Metastatic Renal Cell Carcinoma

91

shorter OS (5.4 months vs. 9.9 months; P < 0.001) and PFS (2.8 months vs. 4.0

months; P < 0.001), as well as a significantly worse CBR (17.5% vs. 50.0%;

P < 0.001). In multivariate analyses adjusted for the IMDC risk classification and

factors not included in the IMDC classification that were significantly associated

with the outcome in univariate analysis (at P < 0.1), baseline sodium <LLN

remained significantly associated with OS (adjusted HR 1.46; P = 0.02) and on-

treatment sodium <LLN with OS (adjusted HR 1.80; P = 0.002) and PFS (adjusted

HR 1.71; P = 0.004).

We additionally conducted subgroup analyses among patients with and without

baseline sodium <LLN and on-treatment sodium <LLN, demonstrating that shifts

from baseline sodium values <LLN to , and vice versa, were associated with

the outcome. Results from the subgroup analyses are depicted in Table 5.

Since baseline neutrophil and sodium values were inversely correlated, we

additionally analyzed the association between baseline hyponatremia and the

outcome among patients with (N = 40) and without baseline neutrophilia (N = 193).

Among patients with normal baseline neutrophil values, baseline sodium <LLN was

associated with shorter OS (6.9 vs. 11.4 months; unadjusted HR 1.96; P < 0.001).

Among patients with baseline neutrophils >ULN, baseline sodium <LLN was not

predictive of shorter OS, but survival was equally modest in both groups (5.2 vs. 4.2

months; unadjusted HR 0.83; P = 0.60). Similarly, among patients with normal

baseline thrombocytes (N = 189), baseline sodium <LLN was associated with

shorter OS (5.9 vs. 11.0 months; unadjusted HR 2.29; P < 0.001), whereas in patients

with baseline thrombocytes >ULN (N = 44), baseline hyponatremia did not

significantly affect OS (7.3 vs 5.8 months; unadjusted HR 1.04; P = 0.91).

Page 92: Novel Biomarkers in Metastatic Renal Cell Carcinoma

92

Table 5. Shifts in baseline/on-treatment sodium values and OS

A. Baseline sodium - a reference group

Baseline

Sodium

Sodium

during

treatment

N Median OS HR P

147 11.1 Ref.

<LLN 11 5.6 2.00 0.037

<LLN 35 8.3 1.68 0.012

<LLN <LLN 30 5.4 2.6 <0.001

B. Two different reference groups demonstrating the dynamic association of sodium

values and OS

Baseline

Sodium

Sodium

during

treatment

N Median OS HR P

147 11.1 Ref.

<LLN 11 5.6 2.00 0.037

<LLN 35 8.3 Ref.

<LLN <LLN 30 5.4 1.60 0.08

Page 93: Novel Biomarkers in Metastatic Renal Cell Carcinoma

93

Our results demonstrate that both baseline and on-treatment hyponatremia are

associated with shorter OS and on-treatment hyponatremia additionally with shorter

PFS and a worse CBR. To the best of our knowledge, this is the largest study to

confirm the prognostic role of hyponatremia among everolimus-treated patients. Our

subgroup analyses additionally suggest that sodium is probably a dynamic

biomarker, as shifts from baseline , and vice

versa, were associated with OS among this patient cohort. This dynamic nature of

the prognostic value of hyponatremia has not previously been investigated.

As the negative prognostic impact of hyponatremia has been demonstrated in mRCC

patients treated with cytokines (238) and TKIs (239-241), possible future

applications of these results in clinical practice may include the incorporation of

sodium in prognostic models. The assessment of hyponatremia among mRCC

patients treated with novel immunotherapy agents is therefore warranted.

Additionally, the normalization of sodium values during treatment may reassure the

treating physician of clinical benefit and thus encourage the continuation of

everolimus therapy.

The underlying mechanisms of hyponatremia are unclear. In addition to SIADH,

other possibilities include renal dysfunction, poor adrenal gland function, existing

comorbidities, concomitant medications (such as diuretics and steroids), cancer

therapy, and/or its adverse effects, such as diarrhea and vomiting (353-357). One

intriguing hypothesis is the association of chronic inflammation and hyponatremia.

Chronic inflammation has a well-established role in tumorigenesis (358).

Experimental and clinical studies suggest that chronic inflammation may lead to the

overproduction of interleukin-6 (IL-6), which induces neutrophilia and the secretion

of ADH, resulting in hyponatremia (359). In our analyses, there was a significant

association between baseline sodium and baseline neutrophils/thrombocytes.

Furthermore, our subgroup analyses demonstrated that baseline hyponatremia was

Page 94: Novel Biomarkers in Metastatic Renal Cell Carcinoma

94

associated with worse OS and PFS among patients with baseline

neutrophils/th

neutrophils/thrombocytes >ULN, where OS and PFS were modest. We find these

results in support of the notion that hyponatremia may be associated with chronic

inflammation in mRCC. More research is needed in this area, however.

Another interesting question is whether the treatment of hyponatremia associates

with the treatment outcome. Several recommendations and treatment algorithms for

hyponatremia exist, but none specifically address patients with cancer (360). In a

retrospective analysis among 57 hyponatremic patients with cancer of various

origins, the median OS for the 32 patients among whom sodium levels returned to

normal was significantly longer (13.6 vs. 5.1 months; P < 0.001) (361). It is,

however, difficult to establish whether the improvement in OS reflects the treatment

of hyponatremia, the continuation of anticancer therapy, an improvement in the

patients’ clinical condition, or bias resulting from the longer follow-up. An opposite

view depicts the negative prognostic impact of hyponatremia to result from the

underlying pathology rather than hyponatremia itself (362). Considering our results,

further studies examining hyponatremia among cancer patients are warranted.

In conclusion, we demonstrated that sodium independently associated with the

outcome among mRCC patients treated with everolimus. As it is a readily available,

inexpensive, and reproducible laboratory parameter, it has great potential as a

clinically applicable prognostic biomarker. Additionally, monitoring on-treatment

sodium values may in the future aid in clinical decision making. The association of

baseline neutrophilia/thrombocytosis and hyponatremia is an intriguing finding,

supporting previous evidence of chronic inflammation as a driver of disease

progression and metastasis and warranting further investigations among mRCC

patients and cancer patients in general.

Page 95: Novel Biomarkers in Metastatic Renal Cell Carcinoma

95

Limitations of the Study Materials and Methods

In study I, the methods used to assess treatment-induced hypertension as well as the

use of antihypertensive medication were based on data obtained from patient case

records. This may have led to underestimation of the incidence of hypertension if it

was not properly documented. Similarly, this may have led to overestimation of the

true use of antihypertensive medication, as patient case records rather than

prescription records were used. Furthermore, data regarding why some patients were

treated with ASIs and some not were lacking, possibly representing confounding

factors, e.g. renal dysfunction.

In study II, archival tumor samples rather than fresh tumor biopsies were used. This

may have affected our results in a few ways. Firstly, from a technical standpoint, the

detected c-Met expression may vary based on the age of the sample. Secondly, c-

Met expression seen in an older sample may not represent the true c-Met expression

of the tumor at the onset of systemic therapy due to tumor biology and behavior.

However, there was no significant difference in the median age of tumor samples for

low and high c-Met expression (5.1 vs. 5.0 years; P = 0.62). In further analyses, we

did note a statistically significant association between the frequency of low and high

c-Met expression and the age of the tissue samples, with older samples having a

higher percentage of low c-Met expression as compared to the less aged samples

(65.2% vs. 48.5%, P = 0.049). As this might call into question the fidelity of the c-

Met assays for the older samples, we hypothesize that the difference might rather be

explained by the more aggressive tumor behavior in patients with high c-Met

expression. As described earlier, all patients in this study were identified from

hospital case records based on the initiation of sunitinib treatment. Patients with

older tissue samples therefore had a longer time between the collection of the sample

(nephrectomy) and initiation of sunitinib therapy, most likely due to less aggressive

Page 96: Novel Biomarkers in Metastatic Renal Cell Carcinoma

96

tumor behavior. Although this might explain the association between the tissue

sample age and c-Met expression, no definitive conclusions on the fidelity of the c-

Met assays can be drawn, and we recognize this as a potential limitation of this study.

Additionally, the number of patients with bone metastases was relatively low

(N = 31) and this could be the reason why c-Met expression did not reach statistical

significance among this patient subgroup.

In study III, a potential limitation in addition to its retrospective nature arises from

the assessment of pneumonitis. Due to overlapping symptoms and similar radiologic

findings, pneumonitis can be difficult to differentiate from other diseases of the lung

parenchyma such as pneumonia, and we recognize this as a potential limitation. The

study did, however, comprise two independent patient cohorts and the radiographs

were subjected to a blinded radiologic review. Additionally, when comparing the

difference in the outcome between patients with CT-

patients with clinical symptoms of pneumonitis/pneumonia.

In study IV, we assessed on-treatment hyponatremia based on the highest sodium

value within 12 weeks of treatment initiation. This may have led to underestimation

of the true incidence of hyponatremia, as a patient would be categorized as

normonatremic with only one on-treatment normonatremic sodium value. This

stricter assessment of hyponatremia may therefore limit the clinical applicability of

the results concerning on-treatment hyponatremia.

Page 97: Novel Biomarkers in Metastatic Renal Cell Carcinoma

97

Conclusions

As the past decade has seen marked advances in the treatment of metastatic RCC in

the form of targeted agents, including sorafenib, sunitinib, bevacizumab, pazopanib,

axitinib, everolimus, and temsirolimus, a number of additional targets aside from

VEGF and mTOR have been identified. Recent phase III trials have demonstrated

that cabozantinib and the combination of nivolumab and ipilimumab have relevant

clinical activity in first-line therapy when compared with sunitinib.

Recommendations regarding first-line therapy are likely to be updated in the near

future based on the results from CABOSUN and Checkmate-214. (224, 225)

As the number of treatment options for mRCC has increased, it has become

increasingly important to identify predictive biomarkers that would allow the

clinician to identify patients who are more likely to benefit from the differing

treatments. At present, there are no biomarkers in clinical use. A few have shown

promise in this regard, including the tumor PD-L1 expression level for nivolumab-

treated patients.

In our study, we demonstrated that high levels of c-Met expression associate with a

worse outcome among mRCC patients treated with sunitinib. We also found that

high c-Met expression is associated with a poor outcome among patients without

bone metastases at baseline, suggesting that the prognostic role may vary based on

the location of the metastases. These results are of special interest given the results

of the CABOSUN trial and should be further investigated among patients treated

with cabozantinib. In the future, the evaluation of tumor c-Met expression may be

incorporated into clinical practice to improve the prognostication of mRCC. The role

of c-Met expression as a possible predictive biomarker, however, remains to be

determined.

Page 98: Novel Biomarkers in Metastatic Renal Cell Carcinoma

98

We additionally demonstrated that the use of angiotensin system inhibitors may have

beneficial effects in conjunction with sunitinib and pazopanib in the treatment of

TKI-induced hypertension. In the future, these results may guide the choice of

antihypertensive medication for patients being treated with angiogenesis inhibitors.

However, for these results to be properly validated, the use of ASIs in the treatment

of TKI-induced HTN needs to be investigated in a randomized prospective setting.

Finally, we identified on-treatment biomarkers for everolimus-treated patients. To

the best of our knowledge, we showed for the first time that treatment-related

pneumonitis is significantly associated with longer overall survival and progression-

free survival, as well as a higher clinical benefit rate, in two independent patient

cohorts. These results verified pneumonitis as a strong marker of everolimus

efficacy. Furthermore, our results demonstrated that both baseline and on-treatment

hyponatremia independently associate with shorter overall survival and that on-

treatment shifts in sodium values from baseline , and vice versa, are

associated with the outcome. These findings add to the growing field of on-treatment

biomarker research in metastatic renal cell carcinoma.

The routine use and incorporation of biomarkers into clinical practice, whether to aid

in treatment selection or to reassure clinicians of the clinical benefit during treatment,

is slowly developing. Future directions in mRCC biomarker research should include

further investigation of promising leads such as tumor PD-L1 expression and c-Met

expression.

Page 99: Novel Biomarkers in Metastatic Renal Cell Carcinoma

99

Acknowledgements

This study was carried out in the Laboratory of Molecular Oncology, University of

Helsinki and in the Department of Oncology, Helsinki University Hospital. It was

financed by grants from the Oncologists Association, the Finnish-Norwegian

Medicine Foundation and the Ida Montin Foundation.

I express my sincere gratitude to my supervisor Adjunct Professor and Chief Medical

Officer Petri Bono for his active guidance and advice regarding research, academic

writing and medicine in general. Petri, You always found the time to answer my late

calls and emails despite Your busy schedule. Your enthusiasm, passion and

determination, as well as Your ever-positive attitude, made this journey not only

possible, but also a pleasant one.

I am also grateful to Associate Professor Frede Donskov for great collaboration and

especially his invaluable guidance in our two last studies. Frede, I sincerely enjoyed

our conversations via email – Your ideas and rigorous approach to both statistical

analyses and the questions behind them truly drove me forward.

I thank the members of my thesis committee, Docent Pipsa Saharinen, Docent Juha

Klefström and Medical Doctor Harry Nisen for constructive comments and for

encouraging me to think “outside the box”.

I want to thank the reviewers of this thesis, Docent Petter Boström and Docent Peeter

Karihtala, for their efficient work around a tight schedule and their observant

feedback and critique helping me to improve my thesis.

Dr. Roy Siddall is warmly acknowledged for the fast and high-quality editing of the

language.

Page 100: Novel Biomarkers in Metastatic Renal Cell Carcinoma

100

I want to thank all the collaborators: Katriina Peltola for all the help in writing and

editing, Tuija Poussa for her assistance with statistical analyses, Marjut Laukka for

her valuable help with the interpretation of radiographs and Juhana Rautiola for not

only his input and expertise, but also for his friendship. I would also like to thank all

other coauthors: Heikki Joensuu, Erkki Hänninen and Ari Ristimäki.

A special thanks goes to my friend and colleague Jaakko Allonen, with whom I

shared a work space for a better part of a year. We had great conversations, not only

about research and work, but life in general. I also want to thank other friends and

colleagues who have helped and supported me along the way at work, in personal

life or both: Antti Kerppola, Tuomas Pyrhönen, Tuomas Kaprio, Timo Laihinen,

Nico Aksentjeff, Olavi Parkkonen and Lauri Jouhi.

My parents and my brothers deserve my deepest thanks for love and support. And

Johanna – thank you for acting as my “practice audience” for so many times. Your

patience with me extends far beyond one can imagine. Your love, as well as Your

practical and sensible attitude towards life, reminded me to keep my feet on the

ground.

Kauniainen

September 2018

Patrick Penttilä

Page 101: Novel Biomarkers in Metastatic Renal Cell Carcinoma

101

References

1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA: a cancer journal for clinicians. 2017;67(1):7-30. 2. Znaor A, Lortet-Tieulent J, Laversanne M, Jemal A, Bray F. International variations and trends in renal cell carcinoma incidence and mortality. Eur Urol. 2015;67(3):519-30. 3. Levi F, Ferlay J, Galeone C, Lucchini F, Negri E, Boyle P, et al. The changing pattern of kidney cancer incidence and mortality in Europe. BJU international. 2008;101(8):949-58. 4. Chow WH, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nature reviews Urology. 2010;7(5):245-57. 5. Sanchez-Martin FM, Millan-Rodriguez F, Urdaneta-Pignalosa G, Rubio-Briones J, Villavicencio-Mavrich H. Small renal masses: incidental diagnosis, clinical symptoms, and prognostic factors. Advances in urology. 2008:310694. 6. Kane CJ, Mallin K, Ritchey J, Cooperberg MR, Carroll PR. Renal cell cancer stage migration: analysis of the National Cancer Data Base. Cancer. 2008;113(1):78-83. 7. Hunt JD, van der Hel OL, McMillan GP, Boffetta P, Brennan P. Renal cell carcinoma in relation to cigarette smoking: meta-analysis of 24 studies. International journal of cancer. 2005;114(1):101-8. 8. Sunela KL, Kataja MJ, Kellokumpu-Lehtinen PL. Influence of body mass index and smoking on the long-term survival of patients with renal cell cancer. Clinical genitourinary cancer. 2013;11(4):458-64. 9. Pischon T, Lahmann PH, Boeing H, Tjonneland A, Halkjaer J, Overvad K, et al. Body size and risk of renal cell carcinoma in the European Prospective Investigation into Cancer and Nutrition (EPIC). International journal of cancer. 2006;118(3):728-38. 10. Calle EE, Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer. 2004;4(8):579-91. 11. Adams KF, Leitzmann MF, Albanes D, Kipnis V, Moore SC, Schatzkin A, et al. Body size and renal cell cancer incidence in a large US cohort study. American journal of epidemiology. 2008;168(3):268-77. 12. Haggstrom C, Rapp K, Stocks T, Manjer J, Bjorge T, Ulmer H, et al. Metabolic factors associated with risk of renal cell carcinoma. PloS one. 2013;8(2):e57475. 13. Chow WH, Gridley G, Fraumeni JF, Jr., Jarvholm B. Obesity, hypertension, and the risk of kidney cancer in men. The New England journal of medicine. 2000;343(18):1305-11. 14. Vatten LJ, Trichopoulos D, Holmen J, Nilsen TI. Blood pressure and renal cancer risk: the HUNT Study in Norway. British journal of cancer. 2007;97(1):112-4.

Page 102: Novel Biomarkers in Metastatic Renal Cell Carcinoma

102

15. Stewart JH, Vajdic CM, van Leeuwen MT, Amin J, Webster AC, Chapman JR, et al. The pattern of excess cancer in dialysis and transplantation. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2009;24(10):3225-31. 16. Vajdic CM, McDonald SP, McCredie MR, van Leeuwen MT, Stewart JH, Law M, et al. Cancer incidence before and after kidney transplantation. Jama. 2006;296(23):2823-31. 17. Bonsib SM. Renal cystic diseases and renal neoplasms: a mini-review. Clinical journal of the American Society of Nephrology : CJASN. 2009;4(12):1998-2007. 18. Rakowski SK, Winterkorn EB, Paul E, Steele DJ, Halpern EF, Thiele EA. Renal manifestations of tuberous sclerosis complex: Incidence, prognosis, and predictive factors. Kidney international. 2006;70(10):1777-82. 19. Gudbjartsson T, Jonasdottir TJ, Thoroddsen A, Einarsson GV, Jonsdottir GM, Kristjansson K, et al. A population-based familial aggregation analysis indicates genetic contribution in a majority of renal cell carcinomas. International journal of cancer. 2002;100(4):476-9. 20. Hemminki K, Li X. Familial risks of cancer as a guide to gene identification and mode of inheritance. International journal of cancer. 2004;110(2):291-4. 21. Gandaglia G, Ravi P, Abdollah F, Abd-El-Barr A-E-RM, Becker A, Popa I, et al. Contemporary incidence and mortality rates of kidney cancer in the United States. Canadian Urological Association Journal. 2014;8(7-8):247-52. 22. Renshaw AA. Subclassification of renal cell neoplasms: an update for the practising pathologist. Histopathology. 2002;41(4):283-300. 23. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol. 2016;70(1):93-105. 24. Decastro GJ, McKiernan JM. Epidemiology, clinical staging, and presentation of renal cell carcinoma. The Urologic clinics of North America. 2008;35(4):581-92; vi. 25. Gnarra JR. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7(1):85. 26. Foster K, Prowse A, van den Berg A, Fleming S, Hulsbeek MM, Crossey PA, et al. Somatic mutations of the von Hippel-Lindau disease tumour suppressor gene in non-familial clear cell renal carcinoma. Human molecular genetics. 1994;3(12):2169-73. 27. Lopez JI, Erramuzpe A, Guarch R, Cortes JM, Pulido R, Llarena R, et al. CD34 immunostaining enhances a distinct pattern of intratumor angiogenesis with prognostic implications in clear cell renal cell carcinoma. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica. 2017;125(2):128-33. 28. Delahunt B, Eble JN. Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tumors. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 1997;10(6):537-44. 29. Muglia VF, Prando A. Renal cell carcinoma: histological classification and correlation with imaging findings. Radiologia Brasileira. 2015;48(3):166-74.

Page 103: Novel Biomarkers in Metastatic Renal Cell Carcinoma

103

30. Gurel S, Narra V, Elsayes KM, Siegel CL, Chen ZE, Brown JJ. Subtypes of renal cell carcinoma: MRI and pathological features. Diagnostic and interventional radiology (Ankara, Turkey). 2013;19(4):304-11. 31. Eble JN. Mucinous tubular and spindle cell carcinoma and post-neuroblastoma carcinoma: newly recognised entities in the renal cell carcinoma family. Pathology. 2003;35(6):499-504. 32. Nagy A, Wilhelm M, Sukosd F, Ljungberg B, Kovacs G. Somatic mitochondrial DNA mutations in human chromophobe renal cell carcinomas. Genes, chromosomes & cancer. 2002;35(3):256-60. 33. Brunelli M, Eble JN, Zhang S, Martignoni G, Delahunt B, Cheng L. Eosinophilic and classic chromophobe renal cell carcinomas have similar frequent losses of multiple chromosomes from among chromosomes 1, 2, 6, 10, and 17, and this pattern of genetic abnormality is not present in renal oncocytoma. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2005;18(2):161-9. 34. Speicher MR, Schoell B, du Manoir S, Schrock E, Ried T, Cremer T, et al. Specific loss of chromosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe renal cell carcinomas revealed by comparative genomic hybridization. The American journal of pathology. 1994;145(2):356-64. 35. Khoo SK, Kahnoski K, Sugimura J, Petillo D, Chen J, Shockley K, et al. Inactivation of BHD in sporadic renal tumors. Cancer research. 2003;63(15):4583-7. 36. Farrow GM, Harrison EG, Jr., Utz DC. Sarcomas and sarcomatoid and mixed malignant tumors of the kidney in adults. 3. Cancer. 1968;22(3):556-63. 37. Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia. The American journal of surgical pathology. 2013;37(10):1469-89. 38. de Peralta-Venturina M, Moch H, Amin M, Tamboli P, Hailemariam S, Mihatsch M, et al. Sarcomatoid differentiation in renal cell carcinoma: a study of 101 cases. The American journal of surgical pathology. 2001;25(3):275-84. 39. Cheville JC, Lohse CM, Zincke H, Weaver AL, Leibovich BC, Frank I, et al. Sarcomatoid renal cell carcinoma: an examination of underlying histologic subtype and an analysis of associations with patient outcome. The American journal of surgical pathology. 2004;28(4):435-41. 40. Haas NB, Nathanson KL. Hereditary Renal Cancer Syndromes. Advances in chronic kidney disease. 2014;21(1):10.1053/j.ackd.2013.10.001. 41. Zbar B, Glenn G, Merino M, Middelton L, Peterson J, Toro J, et al. Familial renal carcinoma: clinical evaluation, clinical subtypes and risk of renal carcinoma development. The Journal of urology. 2007;177(2):461-5; discussion 5. 42. Adeniran AJ, Shuch B, Humphrey PA. Hereditary Renal Cell Carcinoma Syndromes: Clinical, Pathologic, and Genetic Features. The American journal of surgical pathology. 2015;39(12):e1-e18. 43. Richards FM. Molecular pathology of von HippelLindau disease and the VHL tumour suppressor gene. Expert reviews in molecular medicine. 2001;2001:1-27.

Page 104: Novel Biomarkers in Metastatic Renal Cell Carcinoma

104

44. Kaelin WG, Jr. The von Hippel-Lindau tumor suppressor protein and clear cell renal carcinoma. Clin Cancer Res. 2007;13(2 Pt 2):680s-4s. 45. Maher ER, Yates JR, Harries R, Benjamin C, Harris R, Moore AT, et al. Clinical features and natural history of von Hippel-Lindau disease. The Quarterly journal of medicine. 1990;77(283):1151-63. 46. Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet. 1997;16(1):68-73. 47. Lubensky IA, Schmidt L, Zhuang Z, Weirich G, Pack S, Zambrano N, et al. Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype. The American journal of pathology. 1999;155(2):517-26. 48. Schmidt L, Junker K, Nakaigawa N, Kinjerski T, Weirich G, Miller M, et al. Novel mutations of the MET proto-oncogene in papillary renal carcinomas. Oncogene. 1999;18(14):2343-50. 49. Schmidt LS, Nickerson ML, Angeloni D, Glenn GM, Walther MM, Albert PS, et al. Early onset hereditary papillary renal carcinoma: germline missense mutations in the tyrosine kinase domain of the met proto-oncogene. The Journal of urology. 2004;172(4 Pt 1):1256-61. 50. Zbar B, Glenn G, Lubensky I, Choyke P, Walther MM, Magnusson G, et al. Hereditary papillary renal cell carcinoma: clinical studies in 10 families. The Journal of urology. 1995;153(3 Pt 2):907-12. 51. Zbar B, Tory K, Merino M, Schmidt L, Glenn G, Choyke P, et al. Hereditary papillary renal cell carcinoma. The Journal of urology. 1994;151(3):561-6. 52. Nickerson ML, Warren MB, Toro JR, Matrosova V, Glenn G, Turner ML, et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dube syndrome. Cancer cell. 2002;2(2):157-64. 53. Pavlovich CP, Walther MM, Eyler RA, Hewitt SM, Zbar B, Linehan WM, et al. Renal tumors in the Birt-Hogg-Dube syndrome. The American journal of surgical pathology. 2002;26(12):1542-52. 54. Linehan WM, Rouault TA. Molecular pathways: Fumarate hydratase-deficient kidney cancer--targeting the Warburg effect in cancer. Clin Cancer Res. 2013;19(13):3345-52. 55. Tomlinson IP, Alam NA, Rowan AJ, Barclay E, Jaeger EE, Kelsell D, et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet. 2002;30(4):406-10. 56. Linehan WM. Identification of the genes for kidney cancer. Cancer biology & therapy. 2006;5(6):696-9. 57. Linehan WM, Gnarra JR, Lerman MI, Latif F, Zbar B. Genetic basis of renal cell cancer. Important advances in oncology. 1993:47-70. 58. Linehan WM, Ricketts CJ. The metabolic basis of kidney cancer. Seminars in cancer biology. 2013;23(1):46-55.

Page 105: Novel Biomarkers in Metastatic Renal Cell Carcinoma

105

59. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499. 60. Maxwell PH. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271. 61. Guo G, Gui Y, Gao S, Tang A, Hu X, Huang Y, et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nat Genet. 2011;44(1):17-9. 62. Ziello JE, Jovin IS, Huang Y. Hypoxia-Inducible Factor (HIF)-1 Regulatory Pathway and its Potential for Therapeutic Intervention in Malignancy and Ischemia. The Yale Journal of Biology and Medicine. 2007;80(2):51-60. 63. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science (New York, NY). 2001;292(5516):468-72. 64. Krieg M, Haas R, Brauch H, Acker T, Flamme I, Plate KH. Up-regulation of hypoxia-inducible factors HIF-1alpha and HIF-2alpha under normoxic conditions in renal carcinoma cells by von Hippel-Lindau tumor suppressor gene loss of function. Oncogene. 2000;19(48):5435-43. 65. Klatte T, Pantuck AJ. Molecular biology of renal cortical tumors. The Urologic clinics of North America. 2008;35(4):573-80; vi. 66. Fu L, Wang G, Shevchuk MM, Nanus DM, Gudas LJ. Generation of a mouse model of Von Hippel-Lindau kidney disease leading to renal cancers by expression of a constitutively active mutant of HIF1alpha. Cancer research. 2011;71(21):6848-56. 67. Lessi F, Mazzanti CM, Tomei S, Di Cristofano C, Minervini A, Menicagli M, et al. VHL and HIF-1alpha: gene variations and prognosis in early-stage clear cell renal cell carcinoma. Medical oncology (Northwood, London, England). 2014;31(3):840. 68. 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. 69. Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013;45(8):860-7. 70. Xu K, Ding Q, Fang Z, Zheng J, Gao P, Lu Y, et al. Silencing of HIF-1alpha suppresses tumorigenicity of renal cell carcinoma through induction of apoptosis. Cancer gene therapy. 2010;17(3):212-22. 71. Fu L, Wang G, Shevchuk MM, Nanus DM, Gudas LJ. Activation of HIF2alpha in kidney proximal tubule cells causes abnormal glycogen deposition but not tumorigenesis. Cancer research. 2013;73(9):2916-25. 72. Gordan JD, Simon MC. Hypoxia-inducible factors: central regulators of the tumor phenotype. Current opinion in genetics & development. 2007;17(1):71-7. 73. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes & development. 2004;18(16):1926-45. 74. Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6(9):729-34.

Page 106: Novel Biomarkers in Metastatic Renal Cell Carcinoma

106

75. Pópulo H, Lopes JM, Soares P. The mTOR Signalling Pathway in Human Cancer. International Journal of Molecular Sciences. 2012;13(2):1886-918. 76. Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441(7092):424-30. 77. Porta C, Figlin RA. Phosphatidylinositol-3-kinase/Akt signaling pathway and kidney cancer, and the therapeutic potential of phosphatidylinositol-3-kinase/Akt inhibitors. The Journal of urology. 2009;182(6):2569-77. 78. Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG, Jr. TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer cell. 2003;4(2):147-58. 79. Sourbier C, Lindner V, Lang H, Agouni A, Schordan E, Danilin S, et al. The phosphoinositide 3-kinase/Akt pathway: a new target in human renal cell carcinoma therapy. Cancer research. 2006;66(10):5130-42. 80. He L, Fan C, Gillis A, Feng X, Sanatani M, Hotte S, et al. Co-existence of high levels of the PTEN protein with enhanced Akt activation in renal cell carcinoma. Biochimica et biophysica acta. 2007;1772(10):1134-42. 81. Wysocki PJ. mTOR in renal cell cancer: modulator of tumor biology and therapeutic target. Expert review of molecular diagnostics. 2009;9(3):231-41. 82. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nature reviews Molecular cell biology. 2011;12(1):21-35. 83. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nature reviews Molecular cell biology. 2003;4(12):915-25. 84. Christensen JG, Burrows J, Salgia R. c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Letters. 2005;225(1):1-26. 85. Corso S, Comoglio PM, Giordano S. Cancer therapy: can the challenge be MET? Trends in molecular medicine. 2005;11(6):284-92. 86. Organ SL, Tsao M-S. An overview of the c-MET signaling pathway. Therapeutic Advances in Medical Oncology. 2011;3(1 Suppl):S7-S19. 87. Paumelle R, Tulasne D, Kherrouche Z, Plaza S, Leroy C, Reveneau S, et al. Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene. 2002;21(15):2309-19. 88. Xiao GH, Jeffers M, Bellacosa A, Mitsuuchi Y, Vande Woude GF, Testa JR. Anti-apoptotic signaling by hepatocyte growth factor/Met via the phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase pathways. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(1):247-52. 89. Graveel C, Su Y, Koeman J, Wang LM, Tessarollo L, Fiscella M, et al. Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(49):17198-203. 90. Olivero M, Valente G, Bardelli A, Longati P, Ferrero N, Cracco C, et al. Novel mutation in the ATP-binding site of the MET oncogene tyrosine kinase in a HPRCC family. International journal of cancer. 1999;82(5):640-3.

Page 107: Novel Biomarkers in Metastatic Renal Cell Carcinoma

107

91. Bussolino F, Di Renzo MF, Ziche M, Bocchietto E, Olivero M, Naldini L, et al. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. The Journal of cell biology. 1992;119(3):629-41. 92. Grant DS, Kleinman HK, Goldberg ID, Bhargava MM, Nickoloff BJ, Kinsella JL, et al. Scatter factor induces blood vessel formation in vivo. Proceedings of the National Academy of Sciences of the United States of America. 1993;90(5):1937-41. 93. Zhang YW, Su Y, Volpert OV, Vande Woude GF. Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin 1 regulation. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(22):12718-23. 94. Hara S, Nakashiro K, Klosek SK, Ishikawa T, Shintani S, Hamakawa H. Hypoxia enhances c-Met/HGF receptor expression and signaling by activating HIF-1alpha in human salivary gland cancer cells. Oral oncology. 2006;42(6):593-8. 95. Kitajima Y, Ide T, Ohtsuka T, Miyazaki K. Induction of hepatocyte growth factor activator gene expression under hypoxia activates the hepatocyte growth factor/c-Met system via hypoxia inducible factor-1 in pancreatic cancer. Cancer science. 2008;99(7):1341-7. 96. Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer cell. 2003;3(4):347-61. 97. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57-70. 98. Hickey MM, Simon MC. Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Current topics in developmental biology. 2006;76:217-57. 99. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, Abramsson A, et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. The Journal of cell biology. 2003;161(6):1163-77. 100. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407(6801):242-8. 101. Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A, et al. Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes & development. 1994;8(16):1897-909. 102. Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fujiwara Y, Gendron-Maguire M, et al. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature. 1995;376(6535):70-4. 103. Eklund L, Saharinen P. Angiopoietin signaling in the vasculature. Experimental cell research. 2013;319(9):1271-80. 104. Chae JK, Kim I, Lim ST, Chung MJ, Kim WH, Kim HG, et al. Coadministration of angiopoietin-1 and vascular endothelial growth factor enhances collateral vascularization. Arteriosclerosis, thrombosis, and vascular biology. 2000;20(12):2573-8.

Page 108: Novel Biomarkers in Metastatic Renal Cell Carcinoma

108

105. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science (New York, NY). 1999;284(5422):1994-8. 106. Goede V, Coutelle O, Neuneier J, Reinacher-Schick A, Schnell R, Koslowsky TC, et al. Identification of serum angiopoietin-2 as a biomarker for clinical outcome of colorectal cancer patients treated with bevacizumab-containing therapy. British journal of cancer. 2010;103(9):1407-14. 107. Helfrich I, Edler L, Sucker A, Thomas M, Christian S, Schadendorf D, et al. Angiopoietin-2 levels are associated with disease progression in metastatic malignant melanoma. Clin Cancer Res. 2009;15(4):1384-92. 108. Sfiligoi C, de Luca A, Cascone I, Sorbello V, Fuso L, Ponzone R, et al. Angiopoietin-2 expression in breast cancer correlates with lymph node invasion and short survival. International journal of cancer. 2003;103(4):466-74. 109. Soutter AD, Nguyen M, Watanabe H, Folkman J. Basic fibroblast growth factor secreted by an animal tumor is detectable in urine. Cancer research. 1993;53(21):5297-9. 110. Malek AM, Connors S, Robertson RL, Folkman J, Scott RM. Elevation of cerebrospinal fluid levels of basic fibroblast growth factor in moyamoya and central nervous system disorders. Pediatric neurosurgery. 1997;27(4):182-9. 111. Nguyen M, Watanabe H, Budson AE, Richie JP, Hayes DF, Folkman J. Elevated levels of an angiogenic peptide, basic fibroblast growth factor, in the urine of patients with a wide spectrum of cancers. J Natl Cancer Inst. 1994;86(5):356-61. 112. Cao R, Brakenhielm E, Pawliuk R, Wariaro D, Post MJ, Wahlberg E, et al. Angiogenic synergism, vascular stability and improvement of hind-limb ischemia by a combination of PDGF-BB and FGF-2. Nature medicine. 2003;9(5):604-13. 113. Lu H, Xu X, Zhang M, Cao R, Brakenhielm E, Li C, et al. Combinatorial protein therapy of angiogenic and arteriogenic factors remarkably improves collaterogenesis and cardiac function in pigs. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(29):12140-5. 114. Nissen LJ, Cao R, Hedlund EM, Wang Z, Zhao X, Wetterskog D, et al. Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. The Journal of clinical investigation. 2007;117(10):2766-77. 115. Cao Y, Cao R, Hedlund E-M. R Regulation of tumor angiogenesis and metastasis by FGF and PDGF signaling pathways. Journal of Molecular Medicine. 2008;86(7):785-9. 116. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22. 117. Postow MA, Harding J, Wolchok JD. Targeting immune checkpoints: releasing the restraints on anti-tumor immunity for patients with melanoma. Cancer journal (Sudbury, Mass). 2012;18(2):153-9. 118. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27(45):5904-12.

Page 109: Novel Biomarkers in Metastatic Renal Cell Carcinoma

109

119. Whiteside TL. The role of immune cells in the tumor microenvironment. Cancer treatment and research. 2006;130:103-24. 120. Simonson WTN, Allison KH. Tumour-infiltrating lymphocytes in cancer: implications for the diagnostic pathologist. Diagnostic Histopathology. 2011;17(2):80-90. 121. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends in immunology. 2002;23(11):549-55. 122. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Frontiers in bioscience : a journal and virtual library. 2008;13:453-61. 123. Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, Mandruzzato S, et al. Myeloid-derived suppressor cell heterogeneity and subset definition. Current opinion in immunology. 2010;22(2):238-44. 124. Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nature reviews Immunology. 2004;4(12):941-52. 125. Kujawski M, Kortylewski M, Lee H, Herrmann A, Kay H, Yu H. Stat3 mediates myeloid cell-dependent tumor angiogenesis in mice. The Journal of clinical investigation. 2008;118(10):3367-77. 126. Franciszkiewicz K, Boissonnas A, Boutet M, Combadiere C, Mami-Chouaib F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer research. 2012;72(24):6325-32. 127. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annual review of immunology. 2007;25:267-96. 128. Lindenberg JJ, Fehres CM, van Cruijsen H, Oosterhoff D, de Gruijl TD. Cross-talk between tumor and myeloid cells: how to tip the balance in favor of antitumor immunity. Immunotherapy. 2011;3(1):77-96. 129. Bacchetta R, Gambineri E, Roncarolo MG. Role of regulatory T cells and FOXP3 in human diseases. The Journal of allergy and clinical immunology. 2007;120(2):227-35; quiz 36-7. 130. Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunological reviews. 2006;212:28-50. 131. Jago CB, Yates J, Camara NO, Lechler RI, Lombardi G. Differential expression of CTLA-4 among T cell subsets. Clinical and experimental immunology. 2004;136(3):463-71. 132. Misra N, Bayry J, Lacroix-Desmazes S, Kazatchkine MD, Kaveri SV. Cutting edge: human CD4+CD25+ T cells restrain the maturation and antigen-presenting function of dendritic cells. Journal of immunology (Baltimore, Md : 1950). 2004;172(8):4676-80. 133. Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. Journal of immunology (Baltimore, Md : 1950). 2005;174(4):1783-6.

Page 110: Novel Biomarkers in Metastatic Renal Cell Carcinoma

110

134. Bergmann C, Strauss L, Zeidler R, Lang S, Whiteside TL. Expansion of human T regulatory type 1 cells in the microenvironment of cyclooxygenase 2 overexpressing head and neck squamous cell carcinoma. Cancer research. 2007;67(18):8865-73. 135. Bohling SD, Allison KH. Immunosuppressive regulatory T cells are associated with aggressive breast cancer phenotypes: a potential therapeutic target. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2008;21(12):1527-32. 136. Cesana GC, DeRaffele G, Cohen S, Moroziewicz D, Mitcham J, Stoutenburg J, et al. Characterization of CD4+CD25+ regulatory T cells in patients treated with high-dose interleukin-2 for metastatic melanoma or renal cell carcinoma. J Clin Oncol. 2006;24(7):1169-77. 137. Liotta F, Gacci M, Frosali F, Querci V, Vittori G, Lapini A, et al. Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU international. 2011;107(9):1500-6. 138. Polimeno M, Napolitano M, Costantini S, Portella L, Esposito A, Capone F, et al. Regulatory T cells, interleukin (IL)-6, IL-8, vascular endothelial growth factor (VEGF), CXCL10, CXCL11, epidermal growth factor (EGF) and hepatocyte growth factor (HGF) as surrogate markers of host immunity in patients with renal cell carcinoma. BJU international. 2013;112(5):686-96. 139. Siddiqui SA, Frigola X, Bonne-Annee S, Mercader M, Kuntz SM, Krambeck AE, et al. Tumor-infiltrating Foxp3-CD4+CD25+ T cells predict poor survival in renal cell carcinoma. Clin Cancer Res. 2007;13(7):2075-81. 140. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. The Journal of experimental medicine. 2009;206(6):1327-37. 141. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006;124(2):263-6. 142. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4(1):71-8. 143. Dannenmann SR, Thielicke J, Stockli M, Matter C, von Boehmer L, Cecconi V, et al. Tumor-associated macrophages subvert T-cell function and correlate with reduced survival in clear cell renal cell carcinoma. Oncoimmunology. 2013;2(3):e23562. 144. Komohara Y, Hasita H, Ohnishi K, Fujiwara Y, Suzu S, Eto M, et al. Macrophage infiltration and its prognostic relevance in clear cell renal cell carcinoma. Cancer science. 2011;102(7):1424-31. 145. Xu L, Zhu Y, Chen L, An H, Zhang W, Wang G, et al. Prognostic value of diametrically polarized tumor-associated macrophages in renal cell carcinoma. Annals of surgical oncology. 2014;21(9):3142-50. 146. Hutterer GC, Pichler M, Chromecki TF, Strini KA, Klatte T, Pummer K, et al. Tumour-associated macrophages might represent a favourable prognostic indicator in patients with papillary renal cell carcinoma. Histopathology. 2013;63(3):309-15.

Page 111: Novel Biomarkers in Metastatic Renal Cell Carcinoma

111

147. Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer research. 2006;66(2):1123-31. 148. Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nature medicine. 2007;13(7):828-35. 149. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer research. 2010;70(1):68-77. 150. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nature medicine. 2012;18(8):1254-61. 151. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, et al. An integrated genomic analysis of human glioblastoma multiforme. Science (New York, NY). 2008;321(5897):1807-12. 152. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469(7331):539-42. 153. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609-15. 154. Lee AJ, Endesfelder D, Rowan AJ, Walther A, Birkbak NJ, Futreal PA, et al. Chromosomal instability confers intrinsic multidrug resistance. Cancer research. 2011;71(5):1858-70. 155. Campbell PJ, Yachida S, Mudie LJ, Stephens PJ, Pleasance ED, Stebbings LA, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467(7319):1109-13. 156. Gerlinger M, Swanton C. How Darwinian models inform therapeutic failure initiated by clonal heterogeneity in cancer medicine. British journal of cancer. 2010;103(8):1139-43. 157. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. The New England journal of medicine. 2012;366(10):883-92. 158. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nature reviews Clinical oncology. 2018;15(2):81-94. 159. Ritchie AW, Chisholm GD. The natural history of renal carcinoma. Seminars in oncology. 1983;10(4):390-400. 160. Gupta K, Miller JD, Li JZ, Russell MW, Charbonneau C. Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): a literature review. Cancer Treat Rev. 2008;34(3):193-205. 161. Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. The New England journal of medicine. 1996;335(12):865-75.

Page 112: Novel Biomarkers in Metastatic Renal Cell Carcinoma

112

162. Ljungberg B, Campbell SC, Choi HY, Jacqmin D, Lee JE, Weikert S, et al. The epidemiology of renal cell carcinoma. Eur Urol. 2011;60(4):615-21. 163. Volpe A, Panzarella T, Rendon RA, Haider MA, Kondylis FI, Jewett MA. The natural history of incidentally detected small renal masses. Cancer. 2004;100(4):738-45. 164. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27(suppl 5):v58-v68. 165. Guzzo TJ, Pierorazio PM, Schaeffer EM, Fishman EK, Allaf ME. The accuracy of multidetector computerized tomography for evaluating tumor thrombus in patients with renal cell carcinoma. The Journal of urology. 2009;181(2):486-90; discussion 91. 166. Silverman SG, Gan YU, Mortele KJ, Tuncali K, Cibas ES. Renal masses in the adult patient: the role of percutaneous biopsy. Radiology. 2006;240(1):6-22. 167. Caoili EM, Davenport MS. Role of Percutaneous Needle Biopsy for Renal Masses. Seminars in Interventional Radiology. 2014;31(1):20-6. 168. Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL, Zincke H. Solid renal tumors: an analysis of pathological features related to tumor size. The Journal of urology. 2003;170(6 Pt 1):2217-20. 169. Halverson SJ, Kunju LP, Bhalla R, Gadzinski AJ, Alderman M, Miller DC, et al. Accuracy of determining small renal mass management with risk stratified biopsies: confirmation by final pathology. The Journal of urology. 2013;189(2):441-6. 170. Nguyen MM, Gill IS, Ellison LM. The evolving presentation of renal carcinoma in the United States: trends from the Surveillance, Epidemiology, and End Results program. The Journal of urology. 2006;176(6 Pt 1):2397-400; discussion 400. 171. Kutikov A, Fossett LK, Ramchandani P, Tomaszewski JE, Siegelman ES, Banner MP, et al. Incidence of benign pathologic findings at partial nephrectomy for solitary renal mass presumed to be renal cell carcinoma on preoperative imaging. Urology. 2006;68(4):737-40. 172. Rothman J, Egleston B, Wong YN, Iffrig K, Lebovitch S, Uzzo RG. Histopathological characteristics of localized renal cell carcinoma correlate with tumor size: a SEER analysis. The Journal of urology. 2009;181(1):29-33; discussion -4. 173. Thompson RH, Hill JR, Babayev Y, Cronin A, Kaag M, Kundu S, et al. Metastatic renal cell carcinoma risk according to tumor size. The Journal of urology. 2009;182(1):41-5. 174. Chawla SN, Crispen PL, Hanlon AL, Greenberg RE, Chen DY, Uzzo RG. The natural history of observed enhancing renal masses: meta-analysis and review of the world literature. The Journal of urology. 2006;175(2):425-31. 175. Kouba E, Smith A, McRackan D, Wallen EM, Pruthi RS. Watchful waiting for solid renal masses: insight into the natural history and results of delayed intervention. The Journal of urology. 2007;177(2):466-70; discussion 70. 176. Kunkle DA, Egleston BL, Uzzo RG. Excise, ablate or observe: the small renal mass dilemma--a meta-analysis and review. The Journal of urology. 2008;179(4):1227-33; discussion 33-4.

Page 113: Novel Biomarkers in Metastatic Renal Cell Carcinoma

113

177. Pierorazio PM, Hyams ES, Mullins JK, Allaf ME. Active Surveillance for Small Renal Masses. Reviews in Urology. 2012;14(1-2):13-9. 178. Smaldone MC, Kutikov A, Egleston BL, Canter DJ, Viterbo R, Chen DY, et al. Small renal masses progressing to metastases under active surveillance: a systematic review and pooled analysis. Cancer. 2012;118(4):997-1006. 179. Mason RJ, Abdolell M, Trottier G, Pringle C, Lawen JG, Bell DG, et al. Growth kinetics of renal masses: analysis of a prospective cohort of patients undergoing active surveillance. Eur Urol. 2011;59(5):863-7. 180. Pierorazio PM, Johnson MH, Ball MW, Gorin MA, Trock BJ, Chang P, et al. Five-year analysis of a multi-institutional prospective clinical trial of delayed intervention and surveillance for small renal masses: the DISSRM registry. Eur Urol. 2015;68(3):408-15. 181. Zisman A, Pantuck AJ, Dorey F, Said JW, Shvarts O, Quintana D, et al. Improved prognostication of renal cell carcinoma using an integrated staging system. J Clin Oncol. 2001;19(6):1649-57. 182. Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL, Zincke H. An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score. The Journal of urology. 2002;168(6):2395-400. 183. Motzer RJ, Escudier B, Bukowski R, Rini BI, Hutson TE, Barrios CH, et al. Prognostic factors for survival in 1059 patients treated with sunitinib for metastatic renal cell carcinoma. British journal of cancer. 2013;108(12):2470-7. 184. Heng DY, Xie W, Regan MM, Harshman LC, Bjarnason GA, Vaishampayan UN, et al. External validation and comparison with other models of the International Metastatic Renal-Cell Carcinoma Database Consortium prognostic model: a population-based study. Lancet Oncol. 2013;14(2):141-8. 185. Jones J, Otu H, Spentzos D, Kolia S, Inan M, Beecken WD, et al. Gene signatures of progression and metastasis in renal cell cancer. Clin Cancer Res. 2005;11(16):5730-9. 186. Rini B, Goddard A, Knezevic D, Maddala T, Zhou M, Aydin H, et al. A 16-gene assay to predict recurrence after surgery in localised renal cell carcinoma: development and validation studies. Lancet Oncol. 2015;16(6):676-85. 187. MacLennan S, Imamura M, Lapitan MC, Omar MI, Lam TB, Hilvano-Cabungcal AM, et al. Systematic review of oncological outcomes following surgical management of localised renal cancer. Eur Urol. 2012;61(5):972-93. 188. Flanigan RC, Mickisch G, Sylvester R, Tangen C, Van Poppel H, Crawford ED. Cytoreductive nephrectomy in patients with metastatic renal cancer: a combined analysis. The Journal of urology. 2004;171(3):1071-6. 189. Méjean A, Ravaud A, Thezenas S, Colas S, Beauval J-B, Bensalah K, et al. Sunitinib Alone or after Nephrectomy in Metastatic Renal-Cell Carcinoma. New England Journal of Medicine.0(0):null. 190. Motzer RJ, Russo P. Cytoreductive Nephrectomy - Patient Selection Is Key. The New England journal of medicine. 2018.

Page 114: Novel Biomarkers in Metastatic Renal Cell Carcinoma

114

191. Bianchi M, Sun M, Jeldres C, Shariat SF, Trinh QD, Briganti A, et al. Distribution of metastatic sites in renal cell carcinoma: a population-based analysis. Ann Oncol. 2012;23(4):973-80. 192. Sountoulides P, Metaxa L, Cindolo L. Atypical presentations and rare metastatic sites of renal cell carcinoma: a review of case reports. Journal of medical case reports. 2011;5:429. 193. Dabestani S, Marconi L, Hofmann F, Stewart F, Lam TB, Canfield SE, et al. Local treatments for metastases of renal cell carcinoma: a systematic review. Lancet Oncol. 2014;15(12):e549-61. 194. Bukowski RM. Natural history and therapy of metastatic renal cell carcinoma: the role of interleukin-2. Cancer. 1997;80(7):1198-220. 195. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol. 1995;13(3):688-96. 196. Fisher RI, Rosenberg SA, Fyfe G. Long-term survival update for high-dose recombinant interleukin-2 in patients with renal cell carcinoma. The cancer journal from Scientific American. 2000;6 Suppl 1:S55-7. 197. Siegel JP, Puri RK. Interleukin-2 toxicity. J Clin Oncol. 1991;9(4):694-704. 198. Minasian LM, Motzer RJ, Gluck L, Mazumdar M, Vlamis V, Krown SE. Interferon alfa-2a in advanced renal cell carcinoma: treatment results and survival in 159 patients with long-term follow-up. J Clin Oncol. 1993;11(7):1368-75. 199. Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. The Cochrane database of systematic reviews. 2005(1):Cd001425. 200. Negrier S, Escudier B, Lasset C, Douillard JY, Savary J, Chevreau C, et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal-cell carcinoma. Groupe Francais d'Immunotherapie. The New England journal of medicine. 1998;338(18):1272-8. 201. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. The New England journal of medicine. 2007;356. 202. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M. Sorafenib in advanced clear-cell renal-cell carcinoma. The New England journal of medicine. 2007;356. 203. Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. The New England journal of medicine. 2013;369(8):722-31. 204. Motzer RJ, Escudier B, Tomczak P, Hutson TE, Michaelson MD, Negrier S, et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol. 2013;14(6):552-62.

Page 115: Novel Biomarkers in Metastatic Renal Cell Carcinoma

115

205. Hutson TE, Lesovoy V, Al-Shukri S, Stus VP, Lipatov ON, Bair AH, et al. Axitinib versus sorafenib as first-line therapy in patients with metastatic renal-cell carcinoma: a randomised open-label phase 3 trial. Lancet Oncol. 2013;14(13):1287-94. 206. Banumathy G, Cairns P. Signaling pathways in renal cell carcinoma. Cancer biology & therapy. 2010;10(7):658-64. 207. Hanna SC, Heathcote SA, Kim WY. mTOR pathway in renal cell carcinoma. Expert review of anticancer therapy. 2008;8(2):283-92. 208. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. The New England journal of medicine. 2007;356. 209. Knox JJ, Barrios CH, Kim TM, Cosgriff T, Srimuninnimit V, Pittman K, et al. Final overall survival analysis for the phase II RECORD-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic RCC. Ann Oncol. 2017;28(6):1339-45. 210. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet (London, England). 2008;372(9637):449-56. 211. Rini BI, Atkins MB. Resistance to targeted therapy in renal-cell carcinoma. The Lancet Oncology. 2009;10(10):992-1000. 212. Zhou L, Liu XD, Sun M, Zhang X, German P, Bai S, et al. Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene. 2016;35(21):2687-97. 213. Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther. 2011;10(12):2298-308. 214. Choueiri TK. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17(7):917. 215. Ko JJ, Choueiri TK, Rini BI, Lee JL, Kroeger N, Srinivas S, et al. First-, second-, third-line therapy for mRCC: benchmarks for trial design from the IMDC. British journal of cancer. 2014;110(8):1917-22. 216. Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, et al. Cabozantinib Versus Sunitinib As Initial Targeted Therapy for Patients With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The Alliance A031203 CABOSUN Trial. Journal of Clinical Oncology. 2016;35(6):591-7. 217. Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res. 2007;13. 218. Thompson RH, Kuntz SM, Leibovich BC, Dong H, Lohse CM, Webster WS, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer research. 2006;66(7):3381-5.

Page 116: Novel Biomarkers in Metastatic Renal Cell Carcinoma

116

219. Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H, et al. Immunomodulatory Activity of Nivolumab in Metastatic Renal Cell Carcinoma. Clin Cancer Res. 2016;22(22):5461-71. 220. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. The New England journal of medicine. 2017;377(14):1345-56. 221. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. The New England journal of medicine. 2015;373(2):123-35. 222. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. The New England journal of medicine. 2015;372(4):311-9. 223. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. The New England journal of medicine. 2015;373(19):1803-13. 224. Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, et al. Cabozantinib Versus Sunitinib As Initial Targeted Therapy for Patients With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The Alliance A031203 CABOSUN Trial. J Clin Oncol. 2017;35(6):591-7. 225. Escudier B TN, McDermott DF, et al. CheckMate 214: Efficacy and safety of nivolumab plus ipilimumab vs sunitinib for treatment-naive advance or metastatic renal cell carcinoma, including IMDC risk and PD-L1 expression subgroups. Presented at 2017 ESMO Congress; Madrid, Spain; September 8–12, 2017 Abstract LBA5. 2017. 226. Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet (London, England). 2011;378(9807):1931-9. 227. Leonetti A, Leonardi F, Bersanelli M, Buti S. Clinical use of lenvatinib in combination with everolimus for the treatment of advanced renal cell carcinoma. Therapeutics and clinical risk management. 2017;13:799-806. 228. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. New England Journal of Medicine. 2015;373(19):1803-13. 229. Haas NB, Manola J, Uzzo RG, Flaherty KT, Wood CG, Kane C, et al. Adjuvant sunitinib or sorafenib for high-risk, non-metastatic renal-cell carcinoma (ECOG-ACRIN E2805): a double-blind, placebo-controlled, randomised, phase 3 trial. Lancet (London, England). 2016;387(10032):2008-16. 230. Motzer RJ, Haas NB, Donskov F, Gross-Goupil M, Varlamov S, Kopyltsov E, et al. Randomized Phase III Trial of Adjuvant Pazopanib Versus Placebo After Nephrectomy in Patients With Localized or Locally Advanced Renal Cell Carcinoma. J Clin Oncol. 2017;35(35):3916-23. 231. Sternberg C, Donskov F, Haas NB, Doehn C, Russo P, Elmeliegy MA, et al. Pazopanib Exposure Relationship with Clinical Efficacy and Safety in the Adjuvant Treatment of Advanced Renal Cell Carcinoma. Clin Cancer Res. 2018.

Page 117: Novel Biomarkers in Metastatic Renal Cell Carcinoma

117

232. Patel DN, Figlin RA, Kim HL. Adjuvant treatment for renal cell carcinoma: do we finally have a major breakthrough? Clinical advances in hematology & oncology : H&O. 2016;14(11):907-14. 233. Strimbu K, Tavel JA. What are Biomarkers? Current opinion in HIV and AIDS. 2010;5(6):463-6. 234. Masoud GN, Li W. HIF-1alpha pathway: role, regulation and intervention for cancer therapy. Acta pharmaceutica Sinica B. 2015;5(5):378-89. 235. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nature cell biology. 2000;2(7):423-7. 236. Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A, Ferrara J. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol. 1999;17(8):2530-40. 237. Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, 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. J Clin Oncol. 2009;27(34):5794-9. 238. Jeppesen AN, Jensen HK, Donskov F, Marcussen N, von der Maase H. Hyponatremia as a prognostic and predictive factor in metastatic renal cell carcinoma. British journal of cancer. 2010;102(5):867-72. 239. Furukawa J, Miyake H, Kusuda Y, Fujisawa M. Hyponatremia as a powerful prognostic predictor for Japanese patients with clear cell renal cell carcinoma treated with a tyrosine kinase inhibitor. International journal of clinical oncology. 2015;20(2):351-7. 240. Kawashima A, Tsujimura A, Takayama H, Arai Y, Nin M, Tanigawa G, et al. Impact of hyponatremia on survival of patients with metastatic renal cell carcinoma treated with molecular targeted therapy. International journal of urology : official journal of the Japanese Urological Association. 2012;19(12):1050-7. 241. Schutz FA, Xie W, Donskov F, Sircar M, McDermott DF, Rini BI, et al. The impact of low serum sodium on treatment outcome of targeted therapy in metastatic renal cell carcinoma: results from the International Metastatic Renal Cell Cancer Database Consortium. Eur Urol. 2014;65(4):723-30. 242. Tugues S, Koch S, Gualandi L, Li X, Claesson-Welsh L. Vascular endothelial growth factors and receptors: anti-angiogenic therapy in the treatment of cancer. Molecular aspects of medicine. 2011;32(2):88-111. 243. Skubitz KM, Skubitz AP. Differential gene expression in renal-cell cancer. The Journal of laboratory and clinical medicine. 2002;140(1):52-64. 244. Takahashi M, Rhodes DR, Furge KA, Kanayama H, Kagawa S, Haab BB, et al. Gene expression profiling of clear cell renal cell carcinoma: gene identification and prognostic classification. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(17):9754-9.

Page 118: Novel Biomarkers in Metastatic Renal Cell Carcinoma

118

245. Greenberg JI, Shields DJ, Barillas SG, Acevedo LM, Murphy E, Huang J, et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature. 2008;456(7223):809-13. 246. Ohm JE, Gabrilovich DI, Sempowski GD, Kisseleva E, Parman KS, Nadaf S, et al. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood. 2003;101(12):4878-86. 247. Lee TH, Seng S, Sekine M, Hinton C, Fu Y, Avraham HK, et al. Vascular endothelial growth factor mediates intracrine survival in human breast carcinoma cells through internally expressed VEGFR1/FLT1. PLoS medicine. 2007;4(6):e186. 248. Harmon CS, DePrimo SE, Figlin RA, Hudes GR, Hutson TE, Michaelson MD, et al. Circulating proteins as potential biomarkers of sunitinib and interferon-alpha efficacy in treatment-naive patients with metastatic renal cell carcinoma. Cancer chemotherapy and pharmacology. 2014;73(1):151-61. 249. 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. Clin Cancer Res. 2010;16(19):4853-63. 250. Bukowski RM. Prognostic factors for survival in metastatic renal cell carcinoma: update 2008. Cancer. 2009;115(10 Suppl):2273-81. 251. Rini BI, Michaelson MD, Rosenberg JE, Bukowski RM, Sosman JA, Stadler WM, et al. Antitumor activity and biomarker analysis of sunitinib in patients with bevacizumab-refractory metastatic renal cell carcinoma. J Clin Oncol. 2008;26(22):3743-8. 252. Gruenwald V, Beutel G, Schuch-Jantsch S, Reuter C, Ivanyi P, Ganser A, et al. Circulating endothelial cells are an early predictor in renal cell carcinoma for tumor response to sunitinib. BMC Cancer. 2010;10:695. 253. Terakawa T, Miyake H, Kusuda Y, Fujisawa M. Expression level of vascular endothelial growth factor receptor-2 in radical nephrectomy specimens as a prognostic predictor in patients with metastatic renal cell carcinoma treated with sunitinib. Urol Oncol. 2013;31(4):493-8. 254. Uemura H, Nakagawa Y, Yoshida K, Saga S, Yoshikawa K, Hirao Y, et al. MN/CA IX/G250 as a potential target for immunotherapy of renal cell carcinomas. British journal of cancer. 1999;81(4):741-6. 255. Dornbusch J, Zacharis A, Meinhardt M, Erdmann K, Wolff I, Froehner M, et al. Analyses of potential predictive markers and survival data for a response to sunitinib in patients with metastatic renal cell carcinoma. PloS one. 2013;8(9):e76386. 256. Kim HL, Seligson D, Liu X, Janzen N, Bui MH, Yu H, et al. Using tumor markers to predict the survival of patients with metastatic renal cell carcinoma. The Journal of urology. 2005;173(5):1496-501. 257. Bui MH, Seligson D, Han KR, Pantuck AJ, Dorey FJ, Huang Y, 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(2):802-11. 258. Choueiri TK, Cheng S, Qu AQ, Pastorek J, Atkins MB, Signoretti S. Carbonic anhydrase IX as a potential biomarker of efficacy in metastatic clear-cell renal cell

Page 119: Novel Biomarkers in Metastatic Renal Cell Carcinoma

119

carcinoma patients receiving sorafenib or placebo: analysis from the treatment approaches in renal cancer global evaluation trial (TARGET). Urol Oncol. 2013;31(8):1788-93. 259. Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, 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(8):827-37. 260. Zurita AJ, Jonasch E, Wang X, Khajavi M, Yan S, Du DZ, 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(1):46-52. 261. van der Veldt AA, Eechoute K, Gelderblom H, Gietema J, Guchelaar HJ, van Erp NP, et al. Genetic polymorphisms associated with a prolonged progression-free survival in patients with metastatic renal cell cancer treated with sunitinib. Clin Cancer Res. 2011;17(3):620-9. 262. Garcia-Donas J, Esteban E, Leandro-Garcia LJ, Castellano DE, del Alba AG, Climent MA, 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(12):1143-50. 263. Beuselinck B, Jean-Baptiste J, Schoffski P, Couchy G, Meiller C, Rolland F, et al. Validation of VEGFR1 rs9582036 as predictive biomarker in metastatic clear-cell renal cell carcinoma patients treated with sunitinib. BJU international. 2016;118(6):890-901. 264. Beuselinck B, Karadimou A, Lambrechts D, Claes B, Wolter P, Couchy G, et al. VEGFR1 single nucleotide polymorphisms associated with outcome in patients with metastatic renal cell carcinoma treated with sunitinib - a multicentric retrospective analysis. Acta oncologica. 2014;53(1):103-12. 265. Dornbusch J, Walter M, Gottschalk A, Obaje A, Junker K, Ohlmann CH, et al. Evaluation of polymorphisms in angiogenesis-related genes as predictive and prognostic markers for sunitinib-treated metastatic renal cell carcinoma patients. Journal of cancer research and clinical oncology. 2016;142(6):1171-82. 266. Liu X, Swen JJ, Boven E, Castellano D, Gelderblom H, Mathijssen RH, et al. Meta-analysis on the association of VEGFR1 genetic variants with sunitinib outcome in metastatic renal cell carcinoma patients. Oncotarget. 2017;8(1):1204-12. 267. Escudier B, Rini BI, Motzer RJ, Tarazi J, Kim S, Huang X, et al. Genotype Correlations With Blood Pressure and Efficacy From a Randomized Phase III Trial of Second-Line Axitinib Versus Sorafenib in Metastatic Renal Cell Carcinoma. Clinical genitourinary cancer. 2015;13(4):328-37.e3. 268. Xu CF, Bing NX, Ball HA, Rajagopalan D, Sternberg CN, Hutson TE, et al. Pazopanib efficacy in renal cell carcinoma: evidence for predictive genetic markers in angiogenesis-related and exposure-related genes. J Clin Oncol. 2011;29(18):2557-64. 269. Xu CF, Johnson T, Garcia-Donas J, Choueiri TK, Sternberg CN, Davis ID, et al. IL8 polymorphisms and overall survival in pazopanib- or sunitinib-treated patients with renal cell carcinoma. British journal of cancer. 2015;112(7):1190-8.

Page 120: Novel Biomarkers in Metastatic Renal Cell Carcinoma

120

270. Jilaveanu LB, Shuch B, Zito CR, Parisi F, Barr M, Kluger Y, et al. PD-L1 Expression in Clear Cell Renal Cell Carcinoma: An Analysis of Nephrectomy and Sites of Metastases. Journal of Cancer. 2014;5(3):166-72. 271. Kang MJ, Kim KM, Bae JS, Park HS, Lee H, Chung MJ, et al. Tumor-infiltrating PD1-Positive Lymphocytes and FoxP3-Positive Regulatory T Cells Predict Distant Metastatic Relapse and Survival of Clear Cell Renal Cell Carcinoma. Translational oncology. 2013;6(3):282-9. 272. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF. Safety, Activity, and Immune Correlates of Anti–PD-1 Antibody in Cancer. N Engl J Med. 2012;366. 273. Powderly JD KH, Hodi FS, et al. . Biomarkers and associations with the clinical activity of PD-L1 blockade in a MPDL3280A study. J Clin Oncol. 2013;Suppl; abstr 3001. 274. George DJ, Martini JF, Staehler M, Motzer RJ, Magheli A, Escudier B, et al. Immune Biomarkers Predictive for Disease-Free Survival with Adjuvant Sunitinib in High-Risk Locoregional Renal Cell Carcinoma: From Randomized Phase III S-TRAC Study. Clin Cancer Res. 2018. 275. Bono P, Elfving H, Utriainen T, Osterlund P, Saarto T, Alanko T, et al. Hypertension and clinical benefit of bevacizumab in the treatment of advanced renal cell carcinoma. Ann Oncol. 2009;20(2):393-4. 276. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Archer L, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. J Clin Oncol. 2010;28(13):2137-43. 277. Rini BI, Cohen DP, Lu DR, Chen I, Hariharan S, Gore ME. Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst. 2011;103. 278. Rini BI, Schiller JH, Fruehauf JP, Cohen EE, Tarazi JC, Rosbrook B, et al. Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res. 2011;17(11):3841-9. 279. Nosov DA, Esteves B, Lipatov ON, Lyulko AA, Anischenko AA, Chacko RT, et al. Antitumor activity and safety of tivozanib (AV-951) in a phase II randomized discontinuation trial in patients with renal cell carcinoma. J Clin Oncol. 2012;30(14):1678-85. 280. Donskov F, Michaelson MD, Puzanov I, Davis MP, Bjarnason GA, Motzer RJ, et al. Sunitinib-associated hypertension and neutropenia as efficacy biomarkers in metastatic renal cell carcinoma patients. British journal of cancer. 2015;113(11):1571-80. 281. Rautiola J, Donskov F, Peltola K, Joensuu H, Bono P. Sunitinib-induced hypertension, neutropaenia and thrombocytopaenia as predictors of good prognosis in patients with metastatic renal cell carcinoma. BJU international. 2016;117(1):110-7. 282. Nearchou A, Valachis A, Lind P, Akre O, Sandstrom P. Acquired Hypothyroidism as a Predictive Marker of Outcome in Patients With Metastatic Renal Cell

Page 121: Novel Biomarkers in Metastatic Renal Cell Carcinoma

121

Carcinoma Treated With Tyrosine Kinase Inhibitors: A Literature-Based Meta-Analysis. Clinical genitourinary cancer. 2015;13(4):280-6. 283. Lee CK, Marschner IC, Simes RJ, Voysey M, Egleston B, Hudes G, et al. Increase in cholesterol predicts survival advantage in renal cell carcinoma patients treated with temsirolimus. Clin Cancer Res. 2012;18(11):3188-96. 284. Pal SK, Sonpavde G, Agarwal N, Vogelzang NJ, Srinivas S, Haas NB, et al. Evolution of Circulating Tumor DNA Profile from First-line to Subsequent Therapy in Metastatic Renal Cell Carcinoma. European Urology.72(4):557-64. 285. Ball MW, Gorin MA, Guner G, Pierorazio PM, Netto G, Paller CJ, et al. Circulating Tumor DNA as a Marker of Therapeutic Response in Patients With Renal Cell Carcinoma: A Pilot Study. Clinical genitourinary cancer. 2016;14(5):e515-e20. 286. Shu X, Hildebrandt MA, Gu J, Tannir NM, Matin SF, Karam JA, et al. MicroRNA profiling in clear cell renal cell carcinoma tissues potentially links tumorigenesis and recurrence with obesity. British journal of cancer. 2017;116(1):77-84. 287. Petillo D, Kort EJ, Anema J, Furge KA, Yang XJ, Teh BT. MicroRNA profiling of human kidney cancer subtypes. International journal of oncology. 2009;35(1):109-14. 288. Slaby O, Jancovicova J, Lakomy R, Svoboda M, Poprach A, Fabian P, et al. Expression of miRNA-106b in conventional renal cell carcinoma is a potential marker for prediction of early metastasis after nephrectomy. Journal of experimental & clinical cancer research : CR. 2010;29:90. 289. Li Y, Li S, Zhu Y, Liang X, Meng H, Chen J, et al. Incidence and risk of sorafenib-induced hypertension: a systematic review and meta-analysis. Journal of clinical hypertension. 2014;16(3):177-85. 290. Qi WX, He AN, Shen Z, Yao Y. Incidence and risk of hypertension with a novel multi-targeted kinase inhibitor axitinib in cancer patients: a systematic review and meta-analysis. British journal of clinical pharmacology. 2013;76(3):348-57. 291. Qi WX, Lin F, Sun YJ, Tang LN, He AN, Yao Y, et al. Incidence and risk of hypertension with pazopanib in patients with cancer: a meta-analysis. Cancer chemotherapy and pharmacology. 2013;71(2):431-9. 292. Launay-Vacher V, Deray G. Hypertension and proteinuria: a class-effect of antiangiogenic therapies. Anticancer Drugs. 2009;20(1):81-2. 293. Bono P, Rautiola J, Utriainen T, Joensuu H. Hypertension as predictor of sunitinib treatment outcome in metastatic renal cell carcinoma. Acta oncologica. 2011;50(4):569-73. 294. Miyajima A, Kosaka T, Asano T, Asano T, Seta K, Kawai T, et al. Angiotensin II type I antagonist prevents pulmonary metastasis of murine renal cancer by inhibiting tumor angiogenesis. Cancer research. 2002;62(15):4176-9. 295. Verhoest G, Dolley-Hitze T, Jouan F, Belaud-Rotureau MA, Oger E, Lavenu A, et al. Sunitinib combined with angiotensin-2 type-1 receptor antagonists induces more necrosis: a murine xenograft model of renal cell carcinoma. BioMed research international. 2014;2014:901371.

Page 122: Novel Biomarkers in Metastatic Renal Cell Carcinoma

122

296. Semeniuk-Wojtas A, Lubas A, Stec R, Szczylik C, Niemczyk S. Influence of Tyrosine Kinase Inhibitors on Hypertension and Nephrotoxicity in Metastatic Renal Cell Cancer Patients. Int J Mol Sci. 2016;17(12). 297. Rini BI, Garrett M, Poland B, Dutcher JP, Rixe O, Wilding G. Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol. 2013;53. 298. Rixe O, Billemont B, Izzedine H. Hypertension as a predictive factor of Sunitinib activity. Ann Oncol. 2007;18(6):1117. 299. Song T, Choi CH, Kim MK, Kim ML, Yun BS, Seong SJ. The effect of angiotensin system inhibitors (angiotensin-converting enzyme inhibitors or angiotensin receptor blockers) on cancer recurrence and survival: a meta-analysis. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation (ECP). 2017;26(1):78-85. 300. Keizman D, Huang P, Eisenberger MA, Pili R, Kim JJ, Antonarakis ES, et al. Angiotensin system inhibitors and outcome of sunitinib treatment in patients with metastatic renal cell carcinoma: a retrospective examination. European journal of cancer (Oxford, England : 1990). 2011;47(13):1955-61. 301. Izzedine H, Derosa L, Le Teuff G, Albiges L, Escudier B. Hypertension and angiotensin system inhibitors: impact on outcome in sunitinib-treated patients for metastatic renal cell carcinoma. Ann Oncol. 2015;26(6):1128-33. 302. McKay RR, Rodriguez GE, Lin X, Kaymakcalan MD, Hamnvik OP, Sabbisetti VS, et al. Angiotensin system inhibitors and survival outcomes in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2015;21(11):2471-9. 303. Sorich MJ, Kichenadasse G, Rowland A, Woodman RJ, Mangoni AA. Angiotensin system inhibitors and survival in patients with metastatic renal cell carcinoma treated with VEGF-targeted therapy: A pooled secondary analysis of clinical trials. International journal of cancer. 2016;138(9):2293-9. 304. Wei W, Jin H, Chen ZW, Zioncheck TF, Yim AP, He GW. Vascular endothelial growth factor-induced nitric oxide- and PGI2-dependent relaxation in human internal mammary arteries: a comparative study with KDR and Flt-1 selective mutants. Journal of cardiovascular pharmacology. 2004;44(5):615-21. 305. Yang R, Thomas GR, Bunting S, Ko A, Ferrara N, Keyt B, et al. Effects of vascular endothelial growth factor on hemodynamics and cardiac performance. Journal of cardiovascular pharmacology. 1996;27(6):838-44. 306. Kappers MH, van Esch JH, Sluiter W, Sleijfer S, Danser AH, van den Meiracker AH. Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 levels. Hypertension (Dallas, Tex : 1979). 2010;56(4):675-81. 307. Horowitz JR, Rivard A, van der Zee R, Hariawala M, Sheriff DD, Esakof DD, et al. Vascular endothelial growth factor/vascular permeability factor produces nitric oxide-dependent hypotension. Evidence for a maintenance role in quiescent adult endothelium. Arteriosclerosis, thrombosis, and vascular biology. 1997;17(11):2793-800.

Page 123: Novel Biomarkers in Metastatic Renal Cell Carcinoma

123

308. Tamarat R, Silvestre JS, Durie M, Levy BI. Angiotensin II angiogenic effect in vivo involves vascular endothelial growth factor- and inflammation-related pathways. Laboratory investigation; a journal of technical methods and pathology. 2002;82(6):747-56. 309. Fernandez LA, Twickler J, Mead A. Neovascularization produced by angiotensin II. The Journal of laboratory and clinical medicine. 1985;105(2):141-5. 310. Daemen MJ, Lombardi DM, Bosman FT, Schwartz SM. Angiotensin II induces smooth muscle cell proliferation in the normal and injured rat arterial wall. Circulation research. 1991;68(2):450-6. 311. Dolley-Hitze T, Jouan F, Martin B, Mottier S, Edeline J, Moranne O, et al. Angiotensin-2 receptors (AT1-R and AT2-R), new prognostic factors for renal clear-cell carcinoma? British journal of cancer. 2010;103(11):1698-705. 312. Huillard O, Xylinas E, Peyromaure M, Alexandre J, Goldwasser F. Angiotensin System Inhibitors in Renal Cell Carcinoma--Letter. Clin Cancer Res. 2016;22(2):524. 313. Huillard O, Mir O, Peyromaure M, Tlemsani C, Giroux J, Boudou-Rouquette P, et al. Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients. British journal of cancer. 2013;108(5):1034-41. 314. Antoun S, Baracos VE, Birdsell L, Escudier B, Sawyer MB. Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma. Ann Oncol. 2010;21(8):1594-8. 315. Fay AP, Signoretti S, Choueiri TK. MET as a Target in Papillary Renal Cell Carcinoma. Clinical Cancer Research. 2014;20(13):3361-3. 316. Miyata Y, Kanetake H, Kanda S. Presence of phosphorylated hepatocyte growth factor receptor/c-Met is associated with tumor progression and survival in patients with conventional renal cell carcinoma. Clin Cancer Res. 2006;12(16):4876-81. 317. Gibney GT. c-Met is a prognostic marker and potential therapeutic target in clear cell renal cell carcinoma. Ann Oncol. 2013;24(2):343. 318. Saad ED, Buyse M. Statistical controversies in clinical research: end points other than overall survival are vital for regulatory approval of anticancer agents. Annals of Oncology. 2016;27(3):373-8. 319. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8(8):592-603. 320. Shojaei F, Lee JH, Simmons BH, Wong A, Esparza CO, Plumlee PA, et al. HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Cancer research. 2010;70(24):10090-100. 321. Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/Met and beta-catenin/Wnt pathways. European journal of cancer (Oxford, England : 1990). 2010;46(9):1679-91. 322. D'Amico L, Belisario D, Migliardi G, Grange C, Bussolati B, D'Amelio P, et al. C-met inhibition blocks bone metastasis development induced by renal cancer stem cells. Oncotarget. 2016.

Page 124: Novel Biomarkers in Metastatic Renal Cell Carcinoma

124

323. Escudier B, Powles T, Motzer RJ, Olencki T, Aren Frontera O, Oudard S, et al. Cabozantinib, a New Standard of Care for Patients With Advanced Renal Cell Carcinoma and Bone Metastases? Subgroup Analysis of the METEOR Trial. J Clin Oncol. 2018:Jco2017747352. 324. Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17(7):917-27. 325. Schoffski P, Wozniak A, Escudier B, Rutkowski P, Anthoney A, Bauer S, et al. Crizotinib achieves long-lasting disease control in advanced papillary renal-cell carcinoma type 1 patients with MET mutations or amplification. EORTC 90101 CREATE trial. European journal of cancer (Oxford, England : 1990). 2017;87:147-63. 326. Choueiri TK, Halabi S, Sanford B, Hahn O, Michaelson MD, Walsh M, et al. CABOzantinib versus SUNitinib (CABOSUN) as initial targeted therapy for patients with metastatic renal cell carcinoma (mRCC) of poor and intermediate risk groups: Results from ALLIANCE A031203 trial. Annals of Oncology. 2016;27(suppl 6). 327. McCarty JH. Glioblastoma resistance to anti-VEGF therapy: has the challenge been MET? Clin Cancer Res. 2013;19(7):1631-3. 328. Atkinson BJ, Cauley DH, Ng C, Millikan RE, Xiao L, Corn P, et al. Mammalian target of rapamycin (mTOR) inhibitor-associated non-infectious pneumonitis in patients with renal cell cancer: predictors, management, and outcomes. BJU international. 2014;113(3):376-82. 329. Dabydeen DA, Jagannathan JP, Ramaiya N, Krajewski K, Schutz FA, Cho DC. Pneumonitis associated with mTOR inhibitors therapy in patients with metastatic renal cell carcinoma: incidence, radiographic findings and correlation with clinical outcome. European journal of cancer (Oxford, England : 1990). 2012;48. 330. Albiges L, Chamming's F, Duclos B, Stern M, Motzer RJ, Ravaud A, et al. Incidence and management of mTOR inhibitor-associated pneumonitis in patients with metastatic renal cell carcinoma. Ann Oncol. 2012;23(8):1943-53. 331. Amato RJ, Jac J, Giessinger S, Saxena S, Willis JP. A phase 2 study with a daily regimen of the oral mTOR inhibitor RAD001 (everolimus) in patients with metastatic clear cell renal cell cancer. Cancer. 2009;115(11):2438-46. 332. White DA, Camus P, Endo M, Escudier B, Calvo E, Akaza H, et al. Noninfectious pneumonitis after everolimus therapy for advanced renal cell carcinoma. American journal of respiratory and critical care medicine. 2010;182(3):396-403. 333. Iacovelli R, Palazzo A, Mezi S, Morano F, Naso G, Cortesi E. Incidence and risk of pulmonary toxicity in patients treated with mTOR inhibitors for malignancy. A meta-analysis of published trials. Acta oncologica. 2012;51(7):873-9. 334. Sola E, Lopez V, Gutierrez C, Cabello M, Burgos D, Molina MG, et al. Late conversion to mammalian target of rapamycin inhibitor/proliferation signal inhibitors in kidney transplant patients: clinical experience in the last 5 years. Transplantation proceedings. 2010;42(8):2859-60. 335. Weiner SM, Sellin L, Vonend O, Schenker P, Buchner NJ, Flecken M, et al. Pneumonitis associated with sirolimus: clinical characteristics, risk factors and outcome--a

Page 125: Novel Biomarkers in Metastatic Renal Cell Carcinoma

125

single-centre experience and review of the literature. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2007;22(12):3631-7. 336. Morelon E, Stern M, Israel-Biet D, Correas JM, Danel C, Mamzer-Bruneel MF, et al. Characteristics of sirolimus-associated interstitial pneumonitis in renal transplant patients. Transplantation. 2001;72(5):787-90. 337. Pham PT, Pham PC, Danovitch GM, Ross DJ, Gritsch HA, Kendrick EA, et al. Sirolimus-associated pulmonary toxicity. Transplantation. 2004;77(8):1215-20. 338. Singh U, Gupta A, Jasuja S. Sirolimus-induced pneumonitis. Indian Journal of Nephrology. 2009;19(2):80-1. 339. Schrader J, Sterneck M, Klose H, Lohse AW, Nashan B, Fischer L. Everolimus-induced pneumonitis: report of the first case in a liver transplant recipient and review of treatment options. Transplant international : official journal of the European Society for Organ Transplantation. 2010;23(1):110-3. 340. Kahan BD, Knight R, Schoenberg L, Pobielski J, Kerman RH, Mahalati K, et al. Ten years of sirolimus therapy for human renal transplantation: the University of Texas at Houston experience. Transplantation proceedings. 2003;35(3 Suppl):25s-34s. 341. Porta C, Osanto S, Ravaud A, Climent MA, Vaishampayan U, White DA, et al. Management of adverse events associated with the use of everolimus in patients with advanced renal cell carcinoma. European journal of cancer (Oxford, England : 1990). 2011;47(9):1287-98. 342. Willemsen AE, Grutters JC, Gerritsen WR, van Erp NP, van Herpen CM, Tol J. mTOR inhibitor-induced interstitial lung disease in cancer patients: Comprehensive review and a practical management algorithm. International journal of cancer. 2016;138(10):2312-21. 343. Berghmans T, Paesmans M, Body JJ. A prospective study on hyponatraemia in medical cancer patients: epidemiology, aetiology and differential diagnosis. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer. 2000;8(3):192-7. 344. Luca A, Angermayr B, Bertolini G, Koenig F, Vizzini G, Ploner M, et al. An integrated MELD model including serum sodium and age improves the prediction of early mortality in patients with cirrhosis. Liver Transplantation. 2007;13(8):1174-80. 345. Rossi J, Bayram M, Udelson JE, Lloyd-Jones D, Adams KF, Oconnor CM, et al. Improvement in hyponatremia during hospitalization for worsening heart failure is associated with improved outcomes: insights from the Acute and Chronic Therapeutic Impact of a Vasopressin Antagonist in Chronic Heart Failure (ACTIV in CHF) trial. Acute cardiac care. 2007;9(2):82-6. 346. Nair V, Niederman MS, Masani N, Fishbane S. Hyponatremia in Community-Acquired Pneumonia. American Journal of Nephrology. 2007;27(2):184-90. 347. Chao YN, Chiu NC, Huang FY. Clinical features and prognostic factors in childhood pneumococcal meningitis. Journal of microbiology, immunology, and infection = Wei mian yu gan ran za zhi. 2008;41(1):48-53.

Page 126: Novel Biomarkers in Metastatic Renal Cell Carcinoma

126

348. Yaghoubian A, de Virgilio C, Dauphine C, Lewis RJ, Lin M. Use of admission serum lactate and sodium levels to predict mortality in necrotizing soft-tissue infections. Archives of surgery (Chicago, Ill : 1960). 2007;142(9):840-6; discussion 4-6. 349. Choi JS, Bae EH, Ma SK, Kweon SS, Kim SW. Prognostic impact of hyponatraemia in patients with colorectal cancer. Colorectal disease : the official journal of the Association of Coloproctology of Great Britain and Ireland. 2015;17(5):409-16. 350. Kim WR, Biggins SW, Kremers WK, Wiesner RH, Kamath PS, Benson JT, et al. Hyponatremia and mortality among patients on the liver-transplant waiting list. The New England journal of medicine. 2008;359(10):1018-26. 351. Huo TI, Lin HC, Hsia CY, Huang YH, Wu JC, Chiang JH, et al. The MELD-Na is an independent short- and long-term prognostic predictor for hepatocellular carcinoma: A prospective survey. Digestive and Liver Disease. 2008;40(11):882-9. 352. Vasudev NS, Brown JE, Brown SR, Rafiq R, Morgan R, Patel PM, et al. Prognostic factors in renal cell carcinoma: association of preoperative sodium concentration with survival. Clin Cancer Res. 2008;14(6):1775-81. 353. Goel A, Kalra OP. Hyponatremia in older individuals. The Journal of the Association of Physicians of India. 2010;58:663-4. 354. Hamdi T, Latta S, Jallad B, Kheir F, Alhosaini MN, Patel A. Cisplatin-induced renal salt wasting syndrome. Southern medical journal. 2010;103(8):793-9. 355. Oren RM. Hyponatremia in congestive heart failure. The American journal of cardiology. 2005;95(9a):2b-7b. 356. Sahay M, Sahay R. Hyponatremia: A practical approach. Indian Journal of Endocrinology and Metabolism. 2014;18(6):760-71. 357. Spital A. Diuretic-induced hyponatremia. Am J Nephrol. 1999;19(4):447-52. 358. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74. 359. Mastorakos G, Weber JS, Magiakou MA, Gunn H, Chrousos GP. Hypothalamic-pituitary-adrenal axis activation and stimulation of systemic vasopressin secretion by recombinant interleukin-6 in humans: potential implications for the syndrome of inappropriate vasopressin secretion. The Journal of clinical endocrinology and metabolism. 1994;79(4):934-9. 360. Hoorn EJ, Zietse R. Diagnosis and Treatment of Hyponatremia: Compilation of the Guidelines. Journal of the American Society of Nephrology : JASN. 2017;28(5):1340-9. 361. Balachandran K, Okines A, Gunapala R, Morganstein D, Popat S. Resolution of severe hyponatraemia is associated with improved survival in patients with cancer. BMC Cancer. 2015;15:163. 362. Chawla A, Sterns RH, Nigwekar SU, Cappuccio JD. Mortality and serum sodium: do patients die from or with hyponatremia? Clinical journal of the American Society of Nephrology : CJASN. 2011;6(5):960-5.