Kidney Metastatic Bone Disease


Primary tumor (T)

TX: Primary tumor cannot be assessed

T0: No evidence of primary tumor

T1: Tumor ≤7 cm in greatest dimension, limited to the kidney

T1a: Tumor ≤4 cm in greatest dimension, limited to the kidney

T1b: Tumor >4 cm but ≤7 cm in greatest dimension, limited to the kidney

T2: Tumor >7 cm in greatest dimension, limited to the kidney

T2a: Tumor >7 cm but ≤10 cm in greatest dimension, limited to the kidney

T2b: Tumor >10 cm, limited to the kidney

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

T3a: Tumors spreads into renal vein or its muscles or perirenal and/or renal sinus fat, but not beyond Gerota’s fascia

T3b: Tumor extends into vena cava below the diaphragm

T3c: Tumor extends into the vena cava above the diaphragm or invades the wall of vena cava

T4: Tumor invades beyond Gerota’s fascia and extends into the contiguous adrenal gland

Regional lymph nodes (N)

NX: Regional lymph nodes cannot be assessed

N0: No regional lymph node metastasis

N1: Metastasis to regional lymph nodes

Distant metastasis (M)

M0: No distant metastasis

M1: Distant metastasis

Stage grouping

Stage I

T1

N0

M0

Stage II

T2

N0

M0

Stage III

T1–T2

N1

M0

T3

N0–N1

M0
 
Stage IV

T4

Any N

M0

Any T

Any N

M1
 

Used with permission of the American Joint Committee on Cancer (AJCC), Chicago, IL. The original and primary source for this information is the AJCC Cancer Staging Manual, Seventh Edition (2010) published by Springer Science+Business Media





Biology of RCC, Targeted Therapy, and Immunotherapy


Renal cell carcinoma tumors are significantly different with respect to their cell type origin, type of genetic mutation, and in turn, responsiveness to different modes of therapy and clinical course. Four hereditary forms of RCC have been identified with specific genetic components: Birt-Hogg-Dube (BHD1 aka Folliculin gene), familial leiomyomatosis and RCC (fumarate hydratase gene), hereditary papillary RCC (c-MET proto-oncogene), and von Hippel–Lindau (VHL gene). The role of VHL gene in RCC has been studied extensively and has resulted in the identification of several new targets for molecular therapies.

Von Hippel–Lindau disease was first described in the medical literature in 1894 in a report describing two siblings with abnormal, bilateral vascular retinal growths [13]. Eugene von Hippel described similar blood vessel tumors in members of one family in 1904 [14]. Arvid Lindau, a Swedish pathologist, described the presence of vascular tumors within the CNS associated with retinal tumors [15]. Since then, additional tumors have been described in the setting of VHL disease, namely clear cell renal cell carcinomas, pheochromocytomas, and pancreatic tumors of the islet cells. Approximately 50 % of patients with VHL disease will develop RCC, commonly after the third decade of life. Elegant genetic mapping studies performed on DNA of von Hippel–Lindau disease patients led to localization of the VHL gene to the short arm of chromosome 3 in 1988 by Seizinger et al. [16]. Analyses of DNA from RCC tumors of patients without VHL disease showed that 33–66 % of sporadic RCC tumors, predominantly clear cell RCC, contain the VHL mutation [17, 18]. The VHL tumor suppressor, VHL protein (pVHL) , has been identified as a regulator of hypoxia-inducible genes based on observation that cells lacking pVHL have abnormally high amounts of hypoxia-inducible mRNA in the presence of normal oxygen levels [19]. It is an indirect regulation by a protein complex containing pVHL that marks hypoxia inducible factor (HIF) with ubiquitin for destruction by proteasomes. Absent or nonfunctional pVHL then leads to over-accumulation of HIF which, in turn, greatly increases transcription of HIF target genes including genes coding for various growth factors [20]. Additionally, HIF has been implemented in facilitating metastatic process through upregulation of the transcription factor TWIST, a master regulator of gastrulation and mesoderm-specification implicated in metastasis of hepatocellular carcinomas [21] as well as downregulation of intercellular adhesion molecules (integrins, E-cadherin) and upregulation of matrix metalloproteinases (MMP2, MMP9) [22]. Understanding of these pathways was crucial for the development of targeted therapy.

As of 2014, there are seven FDA-approved drugs for use in mRCC utilizing four different mechanisms of action. Bevacizumab (Avastin) is a IgG1 monoclonal antibody able to recognize and bind circulating extracellular vascular endothelial growth factor (VEGF) molecules and thus preventing them from binding to the VEGF receptor on endothelial cells and pericytes. Activation of VEGF receptors initiates a signaling cascade leading to angiogenesis necessary to support tumor cells growth. Axitinib (Inlyta) and Pazopanib (Votrient) are both kinase inhibitors effective against tyrosine kinases associated with VEGF receptors. Sunitinib (Sutent) and Sorafenib (Nexavar) are also kinase inhibitors, but unlike Axitinib and Pazopanib, they have activity against intracellular kinase Raf-1 in addition to activity against tyrosine kinases associated with VEGF and platelet derived growth factor (PDGF) receptors [23]. Temsirolimus (Torisel) and Everolimus (Afinitor) are inhibitors of the mammalian target of rapamycin (mTOR) , a kinase involved in regulation of cell proliferation, survival, and transcription of HIF [24]. In general, targeted therapies are well tolerated with relatively mild side-effects: rashes, hypertension, hand/foot syndrome, and diarrhea [25]. Summary of the seven currently available and FDA approved agents for targeted therapy in mRCC and their performance in initial trials can be found in Table 7.2. Figure 7.1 shows the molecular targets of targeted therapy.


Table 7.2
Comparison of targeted therapy agents’ performances in clinical trials














































































































































































Agent

Type

Line of therapy

Route

FDA approved

Study

Results

Results 2

Notes

Sunitinib Sutent

TKI (VEGFR and PDGFR)

1st

PO

January 2006

Sunitinib vs. IFN

mPFS (mo) 11 vs. 5 [58]

OS (mo) 26.4 vs. 21.8 [58]
 
         
Sunitinib vs. Pazopanib

Pazopanib noninferior HR 1.05 (95 % CI 0.9–1.22) [59]
 
QOL and safety better with Pazopanib
         
Sunitinib vs. Pazopanib

70 % Patients preferred Pazopanib [60]
 
Double blind crossover study

Sorafenib Nexavar

TKI (VEGFR and PDGFR)

2nd

PO

December 2005

Sorafenib vs. placebo

mPFS (mo) 17.8(17.8) vs. 15.2(14.3) P = 0.146(0.029) [61]
 
() After censoring post-cross over placebo survival data
         
Sorafinib vs. Axinitib

See under Axinitib
   

Pazopanib Votrient

TKI (VEGFR)

1st

PO

October 2009

Pazopanib vs. placebo

mPFS (mo) 9.2 vs. 4.2 [62] (HR 0.46, P < 0.001)
   
         
Pazopanib vs. Sunitinib

See under Sunitinib
   

Axitinib Inlyta

TKI (VEGFR)

2nd

PO

January 2012

Axinitib vs. Sorafenib

mPFS (mo) 6.7 vs. 4.7 [63]
   
         
Axinitib vs. Sorafenib

mPFS (mo) 8.3 vs. 5.7 (HR 0.656, 95 % CI 0.55–0.78) [64]

OS (mo) 20.1 vs. 19.2 (HR 0.969, 95 % CI 0.8–1.174) [64]
 
         
Axinitib vs. Sorafenib

mPFS (mo) 13.7 vs. 6.6 [65]
 
ECOG score 0
           
mPFS (mo) 6.5 vs. 6.4 [65]
 
ECOG score 1

Temsirolimus Torisel

mTOR inhibitor

1st

IV

May 2007

Temsirolimus vs. IFNα

mPFS (mo) 5.5 vs. 3.1 (HR 0.66, 95 % CI 0.53–0.81) [66]

OS (mo) 10.9 vs. 7.3 (HR 0.73, 95 % CI 0.58–0.92) [66]
 

Everolimus Afinitor

mTOR inhibitor

2nd

PO

March 2009

Everolimus vs. placebo

mPFS (mo) 4.0 vs. 1.9 [67]
 
FDA approved for use after failure of Sunitinib or Sorafenib

Bevacizumab Avastin

Monoclonal antibody to VEGF

1st

IV

July 2009

Bevacizumab + IFN vs. IFN

mPFS (mo) 10.2 vs. 5.4 [68]

OS (mo) 22.9 vs. 20.6 [68]

FDA approved only for use in combination with IFNα-2a
         
Bevacizumab + IFN vs. IFN

mPFS (mo) 8.5 vs. 5.1 [34]

OS (mo) 18.3 vs. 17.4 [34]
 


A303926_1_En_7_Fig1_HTML.gif


Fig. 7.1
Interaction between RCC tumor cell (brown) and endothelial cell (red). Temsirolimus and Everolimus (1) are inhibitors of the mammalian target of Rapamycin (mTOR) which is a part of the signaling cascade from growth receptor (black) leading to increased cell growth, motility, survival, and upregulation of hypoxia-inducible factors (HIF). Von Hippel–Lindau tumor suppressor (pVHL) degrades HIF. High levels of HIF increase secretion of platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and tumor growth factor (TGF). Sunitinib and Sorafenib (2) are receptor kinase inhibitors with activity against both vascular endothelial growth factor receptors (VEGFR, green) and platelet derived growth factor receptors (PDGFR, red). Axitinib and Pazopanib (3) are tyrosine kinase inhibitors with specific activity against VEGFR. Signaling from PDGFR, VEGFR, and epidermal growth factor receptors (EGFR, blue) increases growth and proliferation of endothelial cells as well as pericytes leading to increased tumor neovascularization. Bevacizumab (4) is a monoclonal antibody against VEGF-A and prevents it from binding to the VEGFR

Immunotherapy with cytokines interferon-α and interleukin-2 has been utilized in the treatment of advanced RCC since their clinical trials in early 1980s. The exact mechanism of either of these agents is not fully understood. While IFN-α has some antiproliferative and direct antitumor properties [26], IL-2 has a wide-ranging stimulatory effect on the immune system including both T and B cells, monocytes, macrophages, and natural killer cells leading to tumor cell cytotoxicity [27].

Initial clinical trials of IL-2 showed response rates of over 30 %, but subsequent studies had lower response rates between 15 and 23 % [28, 29]. Most importantly, 7–9 % of patients treated with HD IL-2 had a durable complete response. Median duration of completed responses was not reached at the time of analysis, but have been estimated to be >80 months with 20 % patients surviving for 10 years following their treatment [28]. The efficacy of Il-2 appears to be dose-related as suggested by the results of a three arm trial with high IV dose, low IV dose, and SQ dose of IL-2 with response rates of 15, 10, and 8 %, respectively [30]. Unfortunately, IL-2 in high doses is very poorly tolerated and needs to be administered in an inpatient setting, preventing its wide spread use in all patients with mRCC despite its ability to induce a durable complete response. Such a response has not been seen with any targeted agent yet developed.

Until the advent of targeted therapy, IFN-α had been the agent of choice in the initial treatment of mRCC. Its response rates are generally lower (10–15 %) than those of IL-2 and durable complete responses are quite rare at less than 2 % [31, 32]. Unlike IL-2, IFN-α is relatively well tolerated and easily administered in the outpatient setting. Even though it has been shown to be inferior in terms of survival to the new mTOR and tyrosine kinase inhibitors in several comparative trials, it is still used in combination with VEGF monoclonal antibodies (Bevacizumab) [33, 34].


Prognostic Factors of Metastatic Renal Cell Carcinoma’s Clinical Behavior


Renal cell carcinoma remains the deadliest of all genitourinary cancers. It is a complex disease with highly variable natural history and biological behavior. Approximately 30–40 % of newly diagnosed patients with RCC have evidence of metastatic disease. Additionally, 20–40 % of patients who initially presented with localized disease will develop metastases, frequently within 2 years. The majority of metastatic cases (up to 90 %) develop in the setting of clear cell RCC [35].

Prior to the advent of immunotherapy in early 1990s, the prognosis of patients with metastatic renal cell carcinoma (mRCC) was abysmal with a 10-year survival being virtually nonexistent. Introduction of high dose interleukin-2 therapy (FDA approved for treatment of advanced RCC in 1992) created a breakthrough in the management of advanced RCC. The overall response rates were between 21 and 23 % with durable complete responses seen in only 5–7 % of patients. Historically, the role of surgery in the form of either a cytoreductive nephrectomy or metastasectomy in this setting was purely palliative for cases of persistent hematuria, intractable pain, paraneoplastic manifestations, or constitutional symptoms. With the advent of immunotherapy, debulking of the primary tumor with cytoreductive nephrectomy has been shown to offer a survival benefit in a selected patient population and is now considered the standard of care [36]. However, high dose IL-2 therapy has a long list of specific toxicities related to hyperstimulation of the immune system ranging from relatively mild flu-like symptoms to life-threatening cardiovascular toxicities. These are similar to those seen in sepsis and septic shock [37]. There is a predominate vascular leak syndrome characterized by a widespread capillary leakage leading to a drop in systemic vascular resistance and intravascular volume. This can lead to a decrease in end-organ perfusion, renal insufficiency with oligouria and pulmonary edema [38]. Admission to an intensive care unit is common practice when administering IL-2.

The toxicities of high dose IL-2 treatment created a need for a prognostic model that would identify patients able to withstand the treatment and benefit from it based on clinical features of their disease. One such model was developed and published in 1999 from the Memorial Sloan-Kettering Cancer Center based on data obtained from 24 clinical trials totaling 670 patients with mRCC treated between 1975 and 1996. Multivariate analysis of numerous patient characteristics identified five pretreatment clinical features of mRCC associated with shorter survival: Karnofsky performance status <80 %, high serum lactate dehydrogenase (>1.5 times the upper limit of normal), low hemoglobin (below the lower limit of normal), elevated corrected plasma calcium levels (>10 mg/dl), and absence of prior nephrectomy. The mean overall survival was found to correlate strongly with the number of adverse prognostic factors [39]. The MSKCC model was later found to be predictive of survival in a dataset of 353 patients from Cleveland Clinic [40] and remains widely used in clinical practice today, helping to guide clinical decisions in the treatment of mRCC patients.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 4, 2017 | Posted by in ORTHOPEDIC | Comments Off on Kidney Metastatic Bone Disease

Full access? Get Clinical Tree

Get Clinical Tree app for offline access