Renal (Kidney) Cancer: Connections in Metabolism at Krebs cycle and Histone Modulation
Curator: Demet Sag, PhD, CRA, GCP
Through Histone Modulation
Renal cell carcinoma accounts for only 3% of total human malignancies but it is still the most common type of urological cancer with a high prevalence in elderly men (>60 years of age).
Most kidney cancers are renal cell carcinomas (RCC). RCC lacks early warning signs and 70 % of patients with RCC develop metastases. Among them, 50 % of patients having skeletal metastases developed a dismal survival of less than 10 % at 5 years.
There are three main histopathological entities:
- Clear cell RCC (ccRCC), dominant in histology (65%)
- Papillary (15-20%) and
- Chromophobe RCC (5%).
There are very rare forms of RCC shown in collecting duct, mucinous tubular, spindle cell, renal medullary, and MiTF-TFE translocation carcinomas.
Subtypes of clear cell and papillary RCC, and a new subtype, clear cell papillary http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f6.jpg
Different subtypes of clear cell RCC can be defined by HIF patterns as well as by transcriptomic expression as defined by ccA and ccB subtypes. Papillary RCC also demonstrates distinct histological subtypes. A recently described variant denoted as clear cell papillary RCC is VHL wildtype (VHL WT), while other clear cell tumors are characterized by VHL mutation, loss, or inactivation (VHL MT).
KEY POINTS
- Renal cell cancer is a disease in which malignant (cancer) cells form in tubules of the kidney.
- Smoking and misuse of certain pain medicines can affect the risk of renal cell cancer.
- Signs of renal cell cancer include
- Blood in your urine, which may appear pink, red or cola colored
- A lump in the abdomen.
- Back pain just below the ribs that doesn’t go away
- Weight loss
- Fatigue
- Intermittent fever
Factors that can increase the risk of kidney cancer include:
- Older age.
- High blood pressure (hypertension).
- Treatment for kidney failure.(long-term dialysis to treat chronic kidney failure)
- Certain inherited syndromes.
- von Hippel-Lindau disease
Tests that examine the abdomen and kidneys are used to detect (find) and diagnose renal cell cancer.
The following tests and procedures may be used:
There are 3 treatment approaches for Renal Cancer:
Stages of Renal Cancer:
Stage I |
Tumour of a diameter of 7 cm (approx. 23⁄4 inches) or smaller, and limited to the kidney. No lymph node involvement or metastases to distant organs. |
Stage II |
Tumour larger than 7.0 cm but still limited to the kidney. No lymph node involvement or metastases to distant organs. |
Stage III
any of the following |
Tumor of any size with involvement of a nearby lymph node but no metastases to distant organs. Tumour of this stage may be with or without spread to fatty tissue around the kidney, with or without spread into the large veins leading from the kidney to the heart. |
Tumour with spread to fatty tissue around the kidney and/or spread into the large veins leading from the kidney to the heart, but without spread to any lymph nodes or other organs. |
Stage IV
any of the following |
Tumour that has spread directly through the fatty tissue and the fascia ligament-like tissue that surrounds the kidney. |
Involvement of more than one lymph node near the kidney |
Involvement of any lymph node not near the kidney |
Distant metastases, such as in the lungs, bone, or brain. |
Grade Level |
Nuclear Characteristics |
Grade I |
Nuclei appear round and uniform, 10 μm; nucleoli are inconspicuous or absent. |
Grade II |
Nuclei have an irregular appearance with signs of lobe formation, 15 μm; nucleoli are evident. |
Grade III |
Nuclei appear very irregular, 20 μm; nucleoli are large and prominent. |
Grade IV |
Nuclei appear bizarre and multilobated, 20 μm or more; nucleoli are prominent |
GENETICS:
90% or more of kidney cancers are believed to be of epithelial cell origin, and are referred to as renal cell carcinoma (RCC), which are further subdivided based on histology into clear-cell RCC (75%), papillary RCC (15%),
chromophobe tumor (5%), and oncocytoma (5%).
Nephrectomy continues to be the cornerstone of treatment for localized renal cell carcinoma (RCC). Research is still underway to developed targeted agents against the vascular endothelial growth factor (VEGF) molecule and related pathways as well as inhibitors of the mammalian target of rapamycin (mTOR),
clear cell RCC (ccRCC) doesn’t respond well to radiation chemotherapy due to high radiation resistancy. The hallmark genetic features of solid tumors such as KRAS or TP53 mutations are also absent. However, there is a well-designed association presented between ccRCC and mutations in the VHL gene
Hereditary RCC, accounts for around 4% of cases, has been a relatively dominant area of RCC genetics.
Causative genes have been identified in several familial cancer syndromes that predispose to RCC including
- VHLmutations in von Hippel-Lindau disease that predispose to ccRCC and VHL is somatically mutated in up to 80% of ccRCC
- METmutations in familial papillary renal cancer,
- dominantly activating kinase domainMET mutation reported in 4–10% of sporadic papillary RCC[2].
- FH (fumarate hydratase) mutations in hereditary leiomyomatosis and renal cell cancer that predispose to papillary RCC
- FLCN(folliculin) mutations in Birt-Hogg-Dubé syndrome that predispose to primarily chromophobe RCC.
In addition, there are germline mutations:
- in theTSC1/2 genes predispose to tuberous sclerosis complex where approximately 3% of cases develop ccRCC,
- in the SDHB(succinate dehydrogenase type B) in patients with paraganglioma syndrome shows elevated risk to develop multiple types of RCC.
GWAS in almost 6000 RCC cases demonstrated that loci on 2p21 and 11q13.3 play a role in RCC. Although EPAS1 gene encoding a transcription factor operative in hypoxia-regulated responses in 2p21 , 11q13.3 has no known coding genes.
There has been, however, comparatively less progress in the elaboration of the somatic genetics of sporadic RCC.
Absent mutations in sporadic RCC:
- somaticFH mutations
- somatic mutations ofTSC12 and SDHB
Present mutations in sporadic ccRCC (chromophobe RCC) are
- TSC1mutations occur in 5% of ccRCCs and
- somatic mutations inFLCN rare
- may predict for extraordinary sensitivity to mTORC1 inhibitors clinically.
The COSMIC database reports somatic point mutations in TP53 in 10% of cases, KRAS/HRAS/NRAS combined ≤1%, CDKN2A 10%, PTEN 3%, RB1 3%, STK11/LKB1 ≤1%, PIK3Ca ≤1%, EGFR1% and BRAF ≤1% in all histological samples. Further information can be found at (http://www.sanger.ac.uk/ genetics/CGP/cosmic/) for the RCC somatic genetics.
HIF- and hypoxia-mediated epigenetic regulation work together due to histone modification because HIF activate several chromatin demethylases, including JMJD1A (KDM3A), JMJD2B (KDM4B), JMJD2C (KDM4C) and JARID1B (KDM5B), all of which are directly targeted by HIF.
Overview of Histone 3 modifications implicated in RCC genetics http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f1.jpg
A number of histone modifying genes are mutated in renal cell carcinoma. These include the H3K36 trimethylase SETD2, the H3K27 demethylase UTX/KDM6A, the H3K4 demethylase JARID1C/KDM5C and the SWI/SNF complex compenent PBRM1, shown in this cartoon to represent their relative activities on Histone H3.
Hyper-methylation is observed on RASSF1 highly (50% f RCC) yet less on VHL and CDKN2A, yet there is a methylation and silencing observed on TIMP3 and secreted frizzled-related protein 2.
RCC is ONE OF THE “CILIOPATHIES” among Polycystic Kidney Disease (PKD), Tuberous Sclerosis Complex (TSC) and VHL Syndrome. The main display of cysts is dysfunctional primary cilia.
Mol Cancer Res. Author manuscript; available in PMC 2013 Jan 1.
Mol Cancer Res. 2012 Jul; 10(7): 859–880. Published online 2012 May 25. doi: 10.1158/1541-7786.MCR-12-0117
pVHL mutants are categorized as Class A, B and C depending on the affected step in pVHL protein quality control http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f2.jpg
VHL proteostasis involves the chaperone mediated translocation of nascent VHL peptide from the ribosome to the TRiC/CCT chaperonin, where folding occurs in an ATP dependent process. The VBC complex is formed while VHL is bound to TRiC, and the mature complex is then released. Three different classes of mutation exist: Class A mutations prevent binding of VHL to TRiC, and abrogate folding into a mature complex. Class B mutations prevent association of Elongins C and B to VHL. Class C mutations inhibit interaction between VHL and HIF1 a.
- familial papillary renal cell carcinoma.
# 144700. RENAL CELL CARCINOMA, NONPAPILLARY; RCC |
ICD+, Links |
|
NONPAPILLARY RENAL CARCINOMA 1 LOCUS, INCLUDED
Cytogenetic locations: 3p25.3 , 3p25.3 , 3q21.1 , 8q24.13 , 12q24.31 , 17p11.2 , 17q12
Matching terms: renal, familial, papillary, carcinoma, cell |
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4358399/bin/467fig3.jpg
Model for the control of the fate of nephron progenitor cells. Eya1 lies genetically upstream of Six2. Six2 labels the nephron progenitor cells, which can either maintain a progenitor state and self-renew or differentiate via the Wnt4-mediated MET. Wnt4 expression is under the direct control of Wt1. β-Catenin is involved in both progenitor cell fates through activation of different transcriptional programs. Active nuclear phosphorylated Yap/Taz shifts the progenitor balance toward the self-renewal fate. Eya1 and Six2 interact directly with Mycn, leading to dephosphorylation of Mycn pT58, stabilization of the protein, increased proliferation, and potentially a shift of the nephron progenitor toward self-renewal. Genes activated in Wilms’ tumors are depicted in green, and inactivated genes are in blue. Deregulation of Yap/Taz in Wilms’ tumors results in phosphorylated Yap not being retained in the cytoplasm as it should, but it translocates to the nucleus and thus shifts the progenitor cell balance toward self-renewal. This model is likely a simplification, as it presumes that all Wilms’ tumors, regardless of causative mutation, are caused by the same mechanism.
Epigenetic aberrations associated with Wilms’ tumor
Chinese Case Study: PMCID: PMC4471788
They u8ndertook this study based on association of low circulating adiponectin concentrations with a higher risk of several cancers, including renal cell carcinoma. Thus they demonstrated that by case–control study that ADIPOQ rs182052 is significantly associated with ccRCC risk.
They investigated the frequency of three single nucleotide polymorphisms (SNPs), rs182052G>A, rs266729C>G, rs3774262G>A, in the adiponectin gene (ADIPOQ). 1004 registered patients with clear cell renal cell carcinoma (ccRCC) compared with 1108 healthy subjects (n = 1108).
The first table presents the characteristics of 1004 patients with clear cell renal cell carcinoma and 1108 cancer-free controls from a Chinese Han population. The Second and third table shows the SNP results.
Table 1: The characteristics of the examined population.
Variable |
Cases, n (%) |
Controls, n (%) |
P-value† |
1004 (100) |
1108 (100) |
Age, years |
≤44 |
195 (19.4) |
230 (20.8) |
0.559 |
45–64 |
580 (57.8) |
644 (58.1) |
≥65 |
229 (22.8) |
234 (21.1) |
Sex |
Male |
711 (70.8) |
815 (73.6) |
0.160 |
Female |
293 (29.2) |
293 (26.4) |
BMI, kg/m2 |
<25 |
480 (47.8) |
589 (53.2) |
0.014 |
≥25 |
524 (52.2) |
519 (46.8) |
Smoking status |
Never |
455 (45.3) |
529 (47.7) |
0.265 |
Ever/current |
549 (54.7) |
579 (52.3) |
Hypertension |
No |
639 (63.6) |
780 (70.4) |
0.001 |
Yes |
365 (36.4) |
328 (29.6) |
Fuhrman grade |
I |
40 (4.0) |
|
|
II |
380 (37.8) |
|
|
III |
347 (34.6) |
|
|
IV |
175 (17.4) |
|
|
Missing |
62 (6.2) |
|
|
Stage at diagnosis |
I |
738 (73.5) |
|
|
II |
71 (7.1) |
|
|
III |
19 (1.9) |
|
|
IV |
176 (17.5) |
|
|
†Pearson’s χ2-test.
Table 2:
Association between ADIPOQ single nucleotide polymorphisms (SNP) and clear cell renal cell carcinoma risk
SNP |
HWE |
Cases, n(%) |
Controls, n(%) |
Crude OR (95% CI) |
P-value |
Adjusted OR (95% CI) |
P-value† |
rs182052 |
GG |
0.636 |
249 (24.8) |
315 (28.4) |
1.00 |
|
1.00 |
|
AG |
|
485 (48.3) |
544 (49.1) |
1.13 (0.92–1.39) |
0.253 |
1.11 (0.90–1.37) |
0.331 |
AA |
|
270 (26.9) |
249 (22.5) |
1.37 (1.08–1.75) |
0.010 |
1.36 (1.07–1.74) |
0.013 |
AG/AA versusGG |
|
|
|
1.20 (0.99–1.46) |
0.060 |
1.19 (0.98–1.45) |
0.086 |
AA versusGG/AG |
|
|
|
1.28 (1.04–1.57) |
0.019 |
1.27 (1.04–1.56) |
0.019 |
rs266729 |
CC |
0.143 |
502 (50.0) |
572 (51.6) |
1.00 |
|
1.00 |
|
CG |
|
398 (39.6) |
434 (39.2) |
1.05 (0.88–1.25) |
0.635 |
1.05 (0.87–1.26) |
0.633 |
GG |
|
104 (10.4) |
102 (9.2) |
1.16 (0.86–1.57) |
0.324 |
1.17 (0.86–1.58) |
0.307 |
CG/GG versusCC |
|
|
|
1.07 (0.91–1.29) |
0.456 |
1.07 (0.90–1.27) |
0.445 |
GG versus CC/CG |
|
|
|
1.19 (0.83–1.59) |
0.377 |
1.15 (0.86–1.54) |
0.353 |
rs3774262 |
GG |
0.106 |
482 (48.0) |
523 (47.2) |
1.00 |
|
1.00 |
|
AG |
|
420 (41.8) |
459 (41.4) |
0.99 (0.83–1.20) |
0.938 |
0.99 (0.82–1.19) |
0.905 |
AA |
|
102 (10.2) |
126 (11.4) |
0.88 (0.66–1.17) |
0.381 |
0.90 (0.67–1.20) |
0.463 |
AG/AA versusGG |
|
|
|
0.98 (0.80–1.16) |
0.711 |
0.97 (0.82–1.15) |
0.722 |
AA versusGG/AG |
|
|
|
0.88 (0.67–1.18) |
0.372 |
0.90 (0.68–1.19) |
0.465 |
Bold values indicate significance.
†Adjusted for age, sex, BMI, smoking status, and hypertension. CI, confidence interval; OR, odds ratio; HWE, Hardy–Weinberg equilibrium.
Table 3:
Association between ADIPOQ single nucleotide polymorphisms (SNP) and clear cell renal cell carcinoma risk
SNP |
HWE |
Cases, n(%) |
Controls, n(%) |
Crude OR (95% CI) |
P-value |
Adjusted OR (95% CI) |
P-value† |
rs182052 |
GG |
0.636 |
249 (24.8) |
315 (28.4) |
1.00 |
|
1.00 |
|
AG |
|
485 (48.3) |
544 (49.1) |
1.13 (0.92–1.39) |
0.253 |
1.11 (0.90–1.37) |
0.331 |
AA |
|
270 (26.9) |
249 (22.5) |
1.37 (1.08–1.75) |
0.010 |
1.36 (1.07–1.74) |
0.013 |
AG/AA versusGG |
|
|
|
1.20 (0.99–1.46) |
0.060 |
1.19 (0.98–1.45) |
0.086 |
AA versusGG/AG |
|
|
|
1.28 (1.04–1.57) |
0.019 |
1.27 (1.04–1.56) |
0.019 |
rs266729 |
CC |
0.143 |
502 (50.0) |
572 (51.6) |
1.00 |
|
1.00 |
|
CG |
|
398 (39.6) |
434 (39.2) |
1.05 (0.88–1.25) |
0.635 |
1.05 (0.87–1.26) |
0.633 |
GG |
|
104 (10.4) |
102 (9.2) |
1.16 (0.86–1.57) |
0.324 |
1.17 (0.86–1.58) |
0.307 |
CG/GG versusCC |
|
|
|
1.07 (0.91–1.29) |
0.456 |
1.07 (0.90–1.27) |
0.445 |
GG versus CC/CG |
|
|
|
1.19 (0.83–1.59) |
0.377 |
1.15 (0.86–1.54) |
0.353 |
rs3774262 |
GG |
0.106 |
482 (48.0) |
523 (47.2) |
1.00 |
|
1.00 |
|
AG |
|
420 (41.8) |
459 (41.4) |
0.99 (0.83–1.20) |
0.938 |
0.99 (0.82–1.19) |
0.905 |
AA |
|
102 (10.2) |
126 (11.4) |
0.88 (0.66–1.17) |
0.381 |
0.90 (0.67–1.20) |
0.463 |
AG/AA versusGG |
|
|
|
0.98 (0.80–1.16) |
0.711 |
0.97 (0.82–1.15) |
0.722 |
AA versusGG/AG |
|
|
|
0.88 (0.67–1.18) |
0.372 |
0.90 (0.68–1.19) |
0.465 |
Bold values indicate significance.
†Adjusted for age, sex, BMI, smoking status, and hypertension. CI, confidence interval; OR, odds ratio; HWE, Hardy–Weinberg equilibrium.
Molecular Genetics Level for Physiology (Function):
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4503866/bin/10585_2015_9731_Fig6_HTML.jpg
a The protein–protein interaction for the identified 8 proteins in STRING (10 necessary proteins/genes were added into the network so as to find the potential strong connection among them. The red dotted lines circled three main pathways. b The ingenuity pathway analysis (IPA) for all these 18 genes showing that oxidative phosphorylation, mitochondria dysfunction and granzyme A are the significantly activated pathways (fold change over 1.5, P < 0.05). c The possible mechanism related mitochondria functions: unspecific condition like inflammation, carcinogens, radiation (ionizing or ultraviolet), intermittent hypoxia, viral infections which is carcinogenesis in our study that damages a cell’s oxidative phosphorylation. Any of these conditions can damage the structure and function of mitochondria thus activating a respiratory chain changes (Complex I, II, III, IV) and also cytochrome c release. When the mitochondrial dysfunction persists, it produces genome instability (mtDNA mutation), and further lead to malignant transformation (metastasis) via increased ROS and apoptotic resistance. (Color figure online)
RENAL CELL CARCINOMA AND METABOLISM goes hand to hand in genes encoding enzymes of the Krebs cycle suppress tumor formation in kidney cells. This includes Succinate dehydrogenase (SDH), Fumarate hydratase (FH). As a result of accumulation of succinate or fumarate causes the inhibition of a family of 2-oxoglutarate-dependent dioxygeneases.
The FH and SDH genes function as two-hit tumor suppressor genes.
SDH has a complex of 4 different polypeptides (SDHA-D) function in electron transfer, catalyzes the conversion of succinate to fumarate. Furthermore, heterozygous germline mutations in SDHsubunits predispose to pheochromocytoma/paraganglioma. FH function to convert fumarate to malate. When its mutations presented as heterozygous germline, it predisposes hereditary leiomyomatosis and renal cell cancer (HLRCC). Among them about 20–50% of HLRCC families are typically papillary-type 2 (pRCC-2) and overwhelmingly aggressive.RCC is increasingly being recognized as a metabolic disease, and key lesions in nutrient sensing and processing have been detected.
Regulation of Prolyl Hydroxylases and Keap1 by Krebs cycle http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f4.jpg
Regulation of Prolyl Hydroxylases by Tricarboxylic Acid (TCA) Cycle Intermediates. Prolyl hydroxylases use TCA cycle intermediates to help catalyze the oxygen, iron and ascorbate dependent- addition of a hydroxyl side chain to a Pro402 and Pro564 of HIF alpha subunits, leading to VHL binding and degradation. Defects in either fumarate hydratase or succinate dehydrogenase will drive up levels of fumarate and succinate, which competitively bind prolyl hydroxylases, and prevent HIF prolyl hydroxylation. This results in higher intracellular HIF levels.
Regulation of mTORC1 http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f5.jpg
HIF regulation and mTOR pathway connections. Hypoxia blocks HIF expression in a TSC1/2 and REDD dependent pathway [155]. HIF1α appears to be both TORC1 and TORC2 dependent, whereas HIF2α is only TORC2 dependent [275]. Signaling via TORC2 appears to upregulate HIF2α in an AKT dependent manner [69].
TREATMENT:
Based on the types of renal cancers the treatment method may vary but the general scheme is:
Drugs Approved for Kidney (Renal Cell) Cancer
Food and Drug Administration (FDA) approved drugs for kidney (renal cell) cancer. Some of the drug names link to NCI’s Cancer Drug Information summaries.
T cell regulation in RCC http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3399969/bin/nihms380694f7.jpg
Immune regulation of renal tumor cells. A: When an antigen presenting cell (APC) engages a T-cell via a cognate T-cell receptor (TCR) and CD28, T-cell cell activation occurs. B: Early and late T-cell inhibitory signals are mediated via CTLA-4 and PD-1 receptors, and this occurs via engagement of the APC via B7 and PD-L1, respectively. C: Inhibitory antibodies against CTLA-4 and PD-1 can overcome T-cell downregulation and once again allow cytokine production.
Phase III Trials of Targeted Therapy in Metastatic Renal Cell Carcinoma
Trial |
Number
of
patients |
Clinical setting |
RR (%) |
PFS (months) |
OS (months) |
VEGF-Targeted Therapy |
*AVOREN
Bevacizumab +
IFNa
vs.IFNa[270] |
649 |
First-line |
31 vs. 12 |
10.2 vs. 5.5
(p<0.001) |
23.3 vs. 21.3
(p=0.129) |
*CALBG 90206
Bevacizumab +
IFNa
vs.IFNa[271] |
732 |
First-line |
25.5 vs. 13 |
8.4 vs. 4.9
(p<0.001) |
18.3 vs. 17.4
(p=0.069) |
Sunitinib vs.
IFNa[248] |
750 |
First-line |
47 vs. 12 |
11 vs. 5
(p=0.0001) |
26.4 vs. 21.8
(p=0.051) |
*TARGET
Sorafenib vs.
Placebo[272] |
903 |
Second-line
(post-cytokine) |
10 vs. 2 |
5.5 vs. 2.8
(p<0.01) |
17.8vs.15.2
(p=0.88) |
Pazopanib vs.
placebo[273] |
435 |
First line/second line
(post-cytokine) |
30 vs. 3 |
9.2 vs. 4.2
(p<0.0001) |
22.9 vs. 20.5
(p=0.224) |
*AXIS
Axitinib vs.
sorafenib [269] |
723 |
Second line
(post-sunitinib, cytokine,
bevacizumab or
temsirolimus) |
19 vs. 9
(p=0.0001) |
6.7 vs. 4.7
(p<0.0001) |
Not reported |
mTOR-Targeted Therapy |
*ARCC
Temsirolimus
vs. Tem + IFNa
vs. IFNa[249] |
624 |
First line, ≥ 3 poor risk
featuresa |
9 vs. 5 |
3.8 vs. 1.9 for
IFNa
monotherapy
(p=0.0001) |
10.9 vs. 7.3 for
IFNa(p=0.008) |
*RECORD-1
Everolimus vs.
placebo [274] |
410 |
Second line
(post sunitinib and/or
sorafenib) |
2 vs. 0 |
4.9 vs. 1.9
(p<0.0001) |
14.8 vs. 14.5 |
RCC renal cell carcinoma, RR response rate, OS overall survival, PFS progression free survival, VEGFvascular endothelial growth factor, IFNa interferon alpha, mTOR mammalian target of rapamycin. AVORENAVastin fOr RENal cell cancer, CALBG Cancer and Leukemia Group B. TARGET Treatment Approaches in Renal Cancer Global Evaluation Trial. AXIS Axitinib in Second Line. ARCC Advanced Renal-Cell Carcinoma. RECORD-1 REnal Cell cancer treatment withOral RAD001 given Daily.
aIncluding serum lactate dehydrogenase level of more than 1.5 times the upper limit of the normal range, a hemoglobin level below the lower limit of the normal range; a corrected serum calcium level of more than 10 mg per deciliter (2.5 mmol per liter), a time from initial diagnosis of renal-cell carcinoma to randomization of less than 1 year, a Karnofsky performance score of 60 or 70, or metastases in multiple organs.
Table: RCC-Associated Antigens (RCCAA) Recognized by T Cells.
Antigen |
Antigen
Category |
Frequency of
Expression
Among RCC
Tumors (%) |
CD8+ T cell
recognition:
Patients with
HLA Class I
Allele(s) |
CD4+ T cell
recognition:
Patients with
HLA Class II
Allele(s) |
References found in Open Access J Urol. Author manuscript; available in PMC 2013 Jul 8. |
Survivina |
ML |
100 |
Multiple |
Multiple |
114 |
OFA-iLR |
OF |
100 |
A2 |
NR |
115, 116 |
IGFBP3a, b |
ML |
97 |
NR |
Multiple |
117, 118 |
EphA2a |
ML |
> 90 |
A2 |
DR4 |
17, 44, 119 |
RU2AS |
Antisense
transcript |
> 90 |
B7 |
NR |
120 |
G250
(CA-IX) a, b |
RCC |
90 |
A2, A24 |
Multiple |
47, 51 |
EGFRa, b |
ML |
85 |
A2 |
NR |
121, 122 |
HIFPH3a |
ML |
85 |
A24 |
NR |
123 |
c-Meta |
ML |
> 80 |
A2 |
NR |
124 |
WT-1a |
ML |
80 |
A2, A24 |
NR |
125–128 |
MUC1a, b |
ML |
76 |
A2 |
DR3 |
46, 129, 130 |
5T4 |
ML |
75 |
A2, Cw7 |
DR4 |
54, 131–133 |
iCE |
aORF |
75 |
B7 |
NR |
134 |
MMP7a |
ML |
75 |
A3 |
Multiple |
117, 135, 136 |
Cyclin D1a |
ML |
75 |
A2 |
Multiple |
117, 137, 138 |
HAGE b |
CT |
75 |
A2 |
DR4 |
139 |
hTERT a, b |
ML |
> 70 |
Mutliple |
Multiple |
140–142 |
FGF-5 |
Protein splice variant |
> 60 |
A3 |
NR |
143 |
mutVHLa, b |
ML |
> 60 |
NR |
NR |
144 |
MAGE-A3 b |
CT |
60 |
Multiple |
Multiple |
145 |
SART-3 |
ML |
57 |
Mulitple |
NR |
146–149 |
SART-2 |
ML |
56 |
A24 |
NR |
150 |
PRAME b |
CT |
40 |
Multiple |
NR |
151–154 |
p53a, b |
Mutant/WT
ML |
32 |
Mutliple |
Multiple |
155, 156 |
MAGE-A9b |
CT |
>30 |
A2 |
NR |
157 |
MAGE-A6b |
CT |
30 |
Mutliple |
DR4 |
18, 158 |
MAGE-D4b |
CT |
30 |
A25 |
NR |
159 |
Her2/neua |
ML |
10–30 |
Multiple |
Multiple |
45, 160–164 |
SART-1a |
ML |
25 |
Multiple |
NR |
165–167 |
RAGE-1 |
CT (ORF2/5) |
21 |
Mutliple |
Multiple |
151, 157, 168, 169 |
TRP-1/ gp75 |
ML |
11 |
A31 |
DR4 |
151, 170–172 |
A summary is provided for RCCAA that have been defined at the molecular level. RCCAA are characterized with regard to their antigen category, their prevalence of (over)expression among total RCC specimens evaluated, whether RCCAA expression is modulated by hypoxia or tumor DNA methylation status, and which HLA class I and class II alleles have been reported to serve as presenting molecules for T cell recognition of peptides derived from a given RCCAA.
Abbreviations: CT = Cancer-Testis Antigens; ML = Multi-lineage Antigens; NR = Not Reported; OF = Oncofetal Antigen; aORF = altered open reading frame; ORF = open reading frame; RCC = Renal cell carcinoma; WT = Wild-Type;
aHypoxia-Induced;
bHypomethylation-Induced.
Expected Impact on Teff versus Suppressor Cells |
Co-Therapeutic Agent |
Teff
priming |
Teff
function |
Teff
survival |
Teff
(TME) |
Treg/
MDSC |
References found in Open Access J Urol. Author manuscript; available in PMC 2013 Jul 8. |
Cytokines |
|
|
|
|
|
|
IL-2 |
↑ |
↑ |
+/− |
↑ |
↑ (Treg) |
173–175 |
IL-7 |
↑ |
↑ |
↑ |
↑ |
↑ (Treg) |
176–178 |
IL-12 |
↑ |
↑ |
↑ |
↑ |
– (Treg), ↓ (MDSC) |
179–181 |
IL-15 |
↑ |
↑ |
↑ |
↑ |
↑ (Treg)* |
182, 183 |
IL-18 |
↑ |
↑ |
↑ |
↑ |
↓ (Treg) |
184–186 |
IL-21 |
↑ |
↑ |
↑ |
? |
+/− (Treg) |
187–190 |
IFN-α |
↑ |
↑ |
↑ |
↑ |
+/− (Treg) |
175, 191–194 |
IFN-γ |
↑ |
↑ |
-? |
? ↑ |
↑ (Treg); ↑ ?(MDSC) |
195–197 |
GM-CSF |
↑ |
↑ |
? |
↑ |
↑ (Treg); ↑(MDSC) |
198–202 |
Coinhibitory Antagonist |
|
|
|
|
|
|
CTLA-4 |
↑ |
↑ |
? |
↑ |
↓ (Treg) |
203, 204 |
PD1/PD1L |
↑ |
↑ |
↑ |
↑ |
↓ (Treg) |
205–207 |
Costimulatory Agonist |
|
|
|
|
|
|
CD40/CD40L |
↑ |
↑ |
↑ |
↑ |
↑ (Treg); ↑(MDSC) |
208–211 |
GITR/GITRL |
↑ |
↑ |
↑ |
↑ |
↓ (Treg); ↓ (MDSC) |
212, 213 |
OX40/OX86 |
↑ |
↑ |
↑ |
↑ |
↑↓ (Treg); ↓ (MDSC) |
214–219 |
4-1BB/4-1BBL |
↑ |
↑ |
↑ |
↑ |
↑ (Treg) |
220–224 |
TLR Agonists |
|
|
|
|
|
|
Imiquimod (TLR7) |
↑ |
↑ |
↑ |
↑ |
? |
225–227 |
Resiquimod (TLR8) |
↑ |
↑ |
↑ |
? |
? |
228, 229 |
CpG (TLR9) |
↑ |
↑ |
↑ |
↑ |
↓ (Treg) |
230–232 |
Anti-Angiogenic |
|
|
|
|
|
|
VEGF-Trap |
– |
– |
? |
? |
– |
233 |
Sunitinib |
↑ |
↑ |
? |
↑ |
↓ (Treg/MDSC) |
98, 100, 234 |
Sorafenib |
↓ |
↓ |
↓ |
? |
↓ (MDSC) |
235 |
Bevacizumab |
↑ |
↑ |
? |
? |
↓ (MDSC) |
236, 237 |
Gefitinib (IRESSA) |
? |
? |
? |
? |
? |
238, 239 |
Cetuximab |
? |
↑ |
? |
? |
? |
240 |
mTOR Inhibitors |
|
|
|
|
|
|
Temsirolimus/Everolimus |
↓ |
↓ |
↓ |
? |
↓ (Treg) |
241 |
Treg/MDSC Inhibitors |
|
|
|
|
|
|
Iplimumab (CTLA-4) |
↑ |
↑ |
? |
↑ |
↓ (Treg) |
242, 243 |
ONTAK (CD25) |
+/− |
+/− |
? |
? |
↓ (Treg) |
244 |
Anti-TGFβ/TGFβR |
↑ |
↑ |
↑ |
↑ |
↓ (Treg) |
245–247 |
Anti-IL10/IL10R |
↑ |
↑ |
↑ |
+/− |
↓ (Treg) |
248, 249 |
Anti-IL35/IL35R |
↑? |
↑? |
↑? |
↑? |
↓ (Treg) |
250 |
1-methyl trytophan |
↑ |
↑ |
? |
? |
↓ (MDSC) |
251 |
ATRA |
↑ |
↑ |
? |
? |
↑ (Treg), ↓ (MDSC) |
90–93 |
Agents that are currently or soon-to-be in clinical trials are summarized with regard to their anticipated impact(s) on Type-1 anti-tumor T cell (Te) activation, function, survival and recruitment into the TME. Additional anticipated effects of drugs on suppressor cells (Treg and MDSC) are also summarized. Key: ↑, agent is expected to increase parameter; ↓, agent is expected to inhibit parameter; +/−, minimal increase or decrease is expected in parameter as a consequence of treatment with agent; ?, unknown effect of agent on parameter.
Abbreviations: ATRA, all-trans retinoic acid; CTLA-4, cytotoxic T Lymphocyte antigen 4; GITR(L), glucocorticoid-induced TNF receptor (ligand); GM-CSF, granulocyte-macrophage colony stimulating factor; IFN, interferon; IL, interleukin; MDSC, myeloid-derived suppressor cell; PD1/PD1L, programmed cell death 1 (ligand); TGF-β(R), tumor necrosis factor-β(receptor); TLR, Toll-like receptor; TME, tumor microenvironment; Treg, regulatory T cell; VEGF, vascular endothelial growth factor.
Alternative and Complementary Therapies for Cancer:
- Art therapy
- Dance or movement therapy
- Exercise
- Meditation
- Music therapy
- Relaxation exercises
Mol Cancer Res. 2012 Jul; 10(7): 859–880. Published online 2012 May 25. doi: 10.1158/1541-7786.MCR-12-0117 PMCID: PMC3399969 NIHMSID: NIHMS380694
State-of-the-science: An update on renal cell carcinoma
Eric Jonasch,1 Andrew Futreal,1 Ian Davis,2 Sean Bailey,2 William Y. Kim,2 James Brugarolas,3 Amato Giaccia,4 Ghada Kurban,5 Armin Pause,6 Judith Frydman,4 Amado Zurita,1 Brian I. Rini,7 Pam Sharma,8Michael Atkins,9 Cheryl Walker,8,* and W. Kimryn Rathmell2,*
Go to:
REFERENCES
Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas.[Nat Genet. 1997]
Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer.[Nat Genet. 2002]
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-Dubé syndrome.[Cancer Cell. 2002]
Tuberous sclerosis-associated renal cell carcinoma. Clinical, pathological, and genetic features.[Am J Pathol. 1996]
Tumor risks and genotype-phenotype-proteotype analysis in 358 patients with germline mutations in SDHB and SDHD.[Hum Mutat. 2010]
Genome-wide association study of renal cell carcinoma identifies two susceptibility loci on 2p21 and 11q13.3.[Nat Genet. 2011]
Mutations of the VHL tumour suppressor gene in renal carcinoma.[Nat Genet. 1994]
Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas.[Nat Genet. 1997]
Few FH mutations in sporadic counterparts of tumor types observed in hereditary leiomyomatosis and renal cell cancer families.[Cancer Res. 2002]
Interplay between pVHL and mTORC1 pathways in clear-cell renal cell carcinoma.[Mol Cancer Res. 2011]
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing.[N Engl J Med. 2012]
Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma.[Nature. 2011]
Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes.[Nature. 2010]
Histone methyltransferase gene SETD2 is a novel tumor suppressor gene in clear cell renal cell carcinoma.[Cancer Res. 2010]
The von Hippel-Lindau tumor suppressor protein regulates gene expression and tumor growth through histone demethylase JARID1C.[Oncogene. 2012]
HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing.[Science. 2001]
Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation.[Science. 2001]
Hypoxia induces trimethylated H3 lysine 4 by inhibition of JARID1A demethylase.[Cancer Res. 2010]
Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma.[Proc Natl Acad Sci U S A. 1994]
Inactivation of the von Hippel-Lindau (VHL) tumour suppressor gene and allelic losses at chromosome arm 3p in primary renal cell carcinoma: evidence for a VHL-independent pathway in clear cell renal tumourigenesis.[Genes Chromosomes Cancer. 1998]
DNA methylation and histone modifications cause silencing of Wnt antagonist gene in human renal cell carcinoma cell lines.[Int J Cancer. 2008]
Global levels of histone modifications predict prognosis in different cancers.[Am J Pathol. 2009]
A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma.[Cancer Invest. 2011]
Review Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway.[Mol Cell. 2008]
Review Targeting HIF-1 for cancer therapy.[Nat Rev Cancer. 2003]
Review Hypoxia-inducible factors: central regulators of the tumor phenotype.[Curr Opin Genet Dev. 2007]
Review Role of VHL gene mutation in human cancer.[J Clin Oncol. 2004]
Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes.[Nature. 2010]
Genetic and functional studies implicate HIF1α as a 14q kidney cancer suppressor gene.[Cancer Discov. 2011]
HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma.[Cancer Cell. 2008]
Software and database for the analysis of mutations in the VHL gene.[Nucleic Acids Res. 1998]
Genetic analysis of von Hippel-Lindau disease.[Hum Mutat. 2010]
The von Hippel-Lindau tumor suppressor protein is required for proper assembly of an extracellular fibronectin matrix.[Mol Cell. 1998]
pVHL modification by NEDD8 is required for fibronectin matrix assembly and suppression of tumor development.[Mol Cell Biol. 2004]
Contrasting effects on HIF-1alpha regulation by disease-causing pVHL mutations correlate with patterns of tumourigenesis in von Hippel-Lindau disease.[Hum Mol Genet. 2001]
Characterization of a von Hippel Lindau pathway involved in extracellular matrix remodeling, cell invasion, and angiogenesis.[Cancer Res. 2006]
The von Hippel-Lindau tumor suppressor gene inhibits hepatocyte growth factor/scatter factor-induced invasion and branching morphogenesis in renal carcinoma cells.[Mol Cell Biol. 1999]
A role for mitochondrial enzymes in inherited neoplasia and beyond.[Nat Rev Cancer. 2003]
Mitochondrial tumour suppressors: a genetic and biochemical update.[Nat Rev Cancer. 2005]
Mitochondrial tumour suppressors: a genetic and biochemical update.[Nat Rev Cancer. 2005]
Identification of the von Hippel-Lindau disease tumor suppressor gene.[Science. 1993]
Vascular tumors in livers with targeted inactivation of the von Hippel-Lindau tumor suppressor.[Proc Natl Acad Sci U S A. 2001]
mTOR: from growth signal integration to cancer, diabetes and ageing.[Nat Rev Mol Cell Biol. 2011]
Genomic expression and single-nucleotide polymorphism profiling discriminates chromophobe renal cell carcinoma and oncocytoma.[BMC Cancer. 2010]
Classification of renal neoplasms based on molecular signatures.[J Urol. 2006]
Genetic subtyping of renal cell carcinoma by comparative genomic hybridization.[Recent Results Cancer Res. 2003]
Papillary renal cell carcinoma. Prognostic value of morphological subtypes in a clinicopathologic study of 43 cases.[Virchows Arch. 2003]
Prognostic impact of carbonic anhydrase IX expression in human renal cell carcinoma.[BJU Int. 2007]
Survivin expression in renal cell carcinoma.[Cancer Invest. 2008]
High expression levels of survivin protein independently predict a poor outcome for patients who undergo surgery for clear cell renal cell carcinoma.[Cancer. 2006]
Mcm2, Geminin, and KI67 define proliferative state and are prognostic markers in renal cell carcinoma.[Clin Cancer Res. 2005]
Prognostic impacts of cytogenetic findings in clear cell renal cell carcinoma: gain of 5q31-qter predicts a distinct clinical phenotype with favorable prognosis.[Cancer Res. 2001]
Chromosome 14q loss defines a molecular subtype of clear-cell renal cell carcinoma associated with poor prognosis.[Mod Pathol. 2011]
Loss of chromosome 9p is an independent prognostic factor in patients with clear cell renal cell carcinoma.[Mod Pathol. 2008]
Chromosome 9p deletions identify an aggressive phenotype of clear cell renal cell carcinoma.[Cancer. 2010]
Gene expression profiling predicts survival in conventional renal cell carcinoma.[PLoS Med. 2006]
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]
Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma.[Cancer Res. 2010]
Therapeutic vaccination against metastatic renal cell carcinoma by autologous dendritic cells: preclinical results and outcome of a first clinical phase I/II trial.[Cancer Immunol Immunother. 2002]
Immunotherapy for metastatic renal cell carcinoma.[BJU Int. 2007]
Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy.[J Clin Oncol. 1995]
Phase II study of vinorelbine in patients with androgen-independent prostate cancer.[Ann Oncol. 2001]
CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones.[Nature. 1992]
CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation.[J Exp Med. 1995]
Mechanisms of T-cell inhibition: implications for cancer immunotherapy.[Expert Rev Vaccines. 2010]
Enhancement of antitumor immunity by CTLA-4 blockade.[Science. 1996]
Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation.[J Exp Med. 1999]
Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients.[Proc Natl Acad Sci U S A. 2003]
Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial.[Clin Cancer Res. 2010]
Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis.[J Immunother. 2007]
PD-1 and its ligands in tolerance and immunity.[Annu Rev Immunol. 2008]
New strategies in kidney cancer: therapeutic advances through understanding the molecular basis of response and resistance.[Clin Cancer Res. 2010]
Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial.[Lancet. 2008]
Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial.[Lancet. 2011]
Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial.[Lancet. 2008]
American Cancer Society. Cancer Facts & Figures 2014. 2014. http://www.cancer.org/research/cancerfactsstatistics/cancerfactsfigures2014/. Accessed Oct. 1, 2014.
Amin A, Plimack ER, Infante JR, Ernstoff MS, Rini BI, Mcdermott DF, Knox JJ, Pal SK, Voss MH, Sharma P, Kollmannsberger CK, Heng DYC, Sprattin JL, Shen Y, Kurland JF, Gagnier P, Hammers HJ. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with sunitinib or pazopanib in patients (pts) with metastatic renal cell carcinoma (mRCC). J Clin Oncol 32:5s (suppl; abstr 5010), 2014.
Atkins MB, Kudchadkar RR, Sznol M, Mcdermott DF, Lotem M, Schacther J, Wolchok JD, Urba WJ, Kuzel T, Schuchter LM, Slingluff CL, Ernstoff MS, Fay JW, Friedlander PA, Gajewski T, Zarour H, Rotem-Yehudar R, Sosman JA. Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma. J Clin Oncol 32:5s (suppl; abstr 9001), 2014.
Beck KE, Blansfield JA, Tran KQ, Feldman AL, Hughes MS, Royal RE, Kammula US, Topalian SL, Sherry RM, Kleiner D, Quezado M, Lowy I, Yellin M, Rosenberg SA, Yang JC. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J Clin Oncol 24(15):2283-2289, 2006.
Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A, Nagler A. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res 14(10):3044-3051, 2008.
Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, Mcmiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll DM, Lowy I, Topalian SL. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28(19):3167-3175, 2010.
Camacho LH, Antonia S, Sosman J, Kirkwood JM, Gajewski TF, Redman B, Pavlov D, Bulanhagui C, Bozon VA, Gomez-Navarro J, Ribas A. Phase I/II trial of tremelimumab in patients with metastatic melanoma. J Clin Oncol 27(7):1075-1081, 2009.
Cho DC, Sosman JA, Sznol M, Gordon MS, Hollebecque A, Mcdermott DF, Delord JP, Rhee IP, Mokatrin A, Kowantez M, Funke RP, Fine GD, Powles T. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC). J Clin Oncol 31 (suppl; abstr 4505), 2013.
Choueiri TK, Fishman MN, Escudier B, Kim JJ, Kluger H, Stadler WM, Perez-Gracia JL, Mcneel DG, Curti BD, Harrison MR, Plimack ER, Appleman LJ, Fong L, Drake CG, Cohen LJ, Srivastava S, Jure-Kunkel M, Hong Q, Kurland JF, Sznol M. Immunomodulatory activity of nivolumab in previously treated and untreated metastatic renal cell carcinoma (mRCC): Biomarker-based results from a randomized clinical trial. J Clin Oncol 32:5s (suppl; abstr 5012), 2014.
Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev (1):CD001425, 2005.
Downey SG, Klapper JA, Smith FO, Yang JC, Sherry RM, Royal RE, Kammula US, Hughes MS, Allen TE, Levy CL. Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin Cancer Res 13(22):6681-6688, 2007.
Drake CG, Mcdermott DF, Sznol M. Survival, safety, and response duration results of nivolumab (Anti-PD-1; BMS-936558; ONO-4538) in a phase I trial in patients with previously treatedmetastatic renal cell carcinoma (mRCC): long-term patient followup. J Clin Oncol 31 (suppl; abstr 4514), 2013.
Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 3(11):991-998, 2002.
Elfiky AA, Sonpavde G. Novel molecular targets for the therapy of renal cell carcinoma. Discov Med13(73):461-471, 2012.
FDA. Pembrolizumab. http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm412861.htm. 2014. Accessed Oct. 16, 2014.
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 13(3):688-696, 1995.
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 34(3):193-205, 2008.
Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369(2):134-144, 2013.
Hammers H, Plimack ER, Infante JR, Ernstoff MS, Rini BI, Mcdermott B, Razak AR, Pal SK, Voss MH, Sharma P, Kollmannsberger C, Heng DY, Spratlin J, Shen Y, Kurland J, Gagnier P, Amin A. Phase I study of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma. ASCO Annual Meeting. J Clin Oncol 32:5s (suppl; abstr 4504), 2014.
Hodi FS, O’day SJ, Mcdermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC. Improved survival with ipilimumab in patients with metastatic melanoma.N Engl J Med 363(8):711-723, 2010.
Infante JR, Powderly JD, Burris HA, Kittaneh M, Grice JH, Smothers JF, Brett S, Fleming ME, May R, Marshall S, Devenport M, Pillemer S, Pardoll DM, Chen L, Langermann S, Lorusso P. Clinical and pharmacodynamic (PD) results of a phase I trial with AMP-224 (B7-DC Fc) that binds to the PD-1 receptor. J Clin Oncol 31 (suppl; abstr 3044), 2013.
Intlekofer AM, Thompson CB. At the bench: preclinical rationale for CTLA-4 and PD-1 blockade ascancer immunotherapy. J Leukoc Biol 94(1):25-39, 2013.
Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26:677-704, 2008.
Kirkwood JM, Lorigan P, Hersey P, Hauschild A, Robert C, Mcdermott D, Marshall MA, Gomez-Navarro J, Liang JQ, Bulanhagui CA. Phase II trial of tremelimumab (CP-675,206) in patients with advanced refractory or relapsed melanoma. Clin Cancer Res 16(3):1042-1048, 2010.
Lieu C, Bendell J, Powderly JD, Pishvaian MJ, Hochster H, Eckhardt SG, Funke RP, Rossi C, Waterkamp D, Hurwitz H. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or chemotherapy (chemo) in patients (pts) with locally advanced or metastatic solid tumors. Ann Oncol 25 (suppl 4; abstr 1049o), 2014.
McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, Kirkwood JM, Gordon MS, Sosman JA, Ernstoff MS, Tretter CP, Urba WJ, Smith JW, Margolin KA, Mier JW, Gollob JA, Dutcher JP, Atkins MB. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol 23(1):133-141, 2005.
McDermott DF, Sznol M, Sosman JA, Soria JC, Gordon MS, Hamid O, Delord JP, Fasso M, Wang Y, Bruey J, Fine GD, Powles T. Immune correlates and long term follow up of a phase Ia study of MPDL3280A, an engineered PD-L1 antibody, in patients with metastatic renal cell carcinoma (mRCC). Ann Oncol 25 (Suppl 4; abstr 809o), 2014.
Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 7(2):95-106, 2007.
Millward M, Underhill C, Lobb S, Mcburnie J, Meech SJ, Gomez-Navarro J, Marshall MA, Huang B, Mather CB. Phase I study of tremelimumab (CP-675 206) plus PF-3512676 (CPG 7909) in patients with melanoma or advanced solid tumours. Br J Cancer 108(10):1998-2004, 2013.
Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, Nathan P, Staehler M, De Souza P, Merchan JR, Boleti E, Fife K, Jin J, Jones R, Uemura H, De Giorgi U, Harmenberg U, Wang J, Sternberg CN, Deen K, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 369(8):722-731, 2013.
Motzer RJ, Jonasch E, Agarwal N. NCCN Clinical Practice Guidelines in Oncology: Kidney Cancer. Version 1. 2015. National Comprehensive Cancer Network.http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Accessed Oct. 1, 2014.
Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, Vaishampayan UN, Drabkin HA, George S, Logan TF, Margolin KA, Plimack ER, Lambert AM, Waxman IM, Hammers HJ. Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol, epub ahead of print, Dec. 1, 2014.
National Cancer Institute. SEER Stat Fact Sheets: Kidney and renal pelvis cancer. Surveillance,Epidemiology, and End Results Program. 2014. http://seer.cancer.gov/statfacts/html/kidrp.html. Accessed Oct. 1, 2014.
Negrier S, Escudier B, Lasset C, Douillard JY, Savary J, Chevreau C, Ravaud A, Mercatello A, Peny J, Mousseau M, Philip T, Tursz T. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal-cell carcinoma. Groupe Francais d’Immunotherapie. N Engl J Med 338(18):1272-1278, 1998.
O’Day SJ, Hamid O, Urba WJ. Targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4). Cancer110(12):2614-2627, 2007.
Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med 65:185-202, 2014.
Pages C, Gornet JM, Monsel G, Allez M, Bertheau P, Bagot M, Lebbe C, Viguier M. Ipilimumab-induced acute severe colitis treated by infliximab. Melanoma Res 23(3):227-230, 2013.
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer12(4):252-264, 2012.
Park J-J, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, Augustine MM, Yao S, Tsushima F, Narazaki H, Anand S. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 116(8):1291-1298, 2010.
Patnaik A, Kang SP, Tolcher AW, Rasco DW, Papadopoulos KP, Beeram M, Drengler R, Chen C, Smith L, Perez C, Gergich K, Lehnert M. Phase I study of MK-3475 (anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. J Clin Oncol 30 (suppl; abstr 2512), 2012.
Ribas A, Hodi FS, Kefford R, Hamid O, Daud A, Wolchok JD, Hwu WJ, Gangadhar TC, Patnaik A, Joshua AM, Hersey P, Weber JS, Dronca R, Zarour H, Gergich K, Li XN, Iannone R, Kang SP, Ebbinghaus SW, Robert C. Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL). J Clin Oncol 32:5s (suppl; abstr LBA9000), 2014.
Ribas A, Kefford R, Marshall MA, Punt CJ, Haanen JB, Marmol M, Garbe C, Gogas H, Schachter J, Linette G, Lorigan P, Kendra KL, Maio M, Trefzer U, Smylie M, Mcarthur GA, Dreno B, Nathan PD, Mackiewicz J, Kirkwood JM, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol 31(5):616-622, 2013.
Rini BI, Stein M, Shannon P, Eddy S, Tyler A, Stephenson JJ Jr, Catlett L, Huang B, Healey D, Gordon M. Phase 1 dose-escalation trial of tremelimumab plus sunitinib in patients with metastatic renal cell carcinoma. Cancer 117(4):758-767, 2011.
Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, Weber JS, Joshua AM, Hwu WJ, Gangadhar TC, Patnaik A, Dronca R, Zarour H, Joseph RW, Boasberg P, Chmielowski B, Mateus C, Postow MA, Gergich K, Elassaiss-Schaap J, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet 384(9948):1109-1117, 2014.
Robert C, Thomas L, Bondarenko I, O’day S, M DJ, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, Davidson N, Richards J, Maio M, Hauschild A, Miller WH Jr, Gascon P, Lotem M, Harmankaya K, Ibrahim R, Francis S, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364(26):2517-2526, 2011.
Sheridan C. Cautious optimism surrounds early clinical data for PD-1 blocker. Nat Biotechnol30(8):729-730, 2012.
Tarhini AA, Cherian J, Moschos SJ, Tawbi HA, Shuai Y, Gooding WE, Sander C, Kirkwood JM. Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage IV melanoma. J Clin Oncol 30(3):322-328, 2012.
Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 24(2):207-212, 2012a.
Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, Mcdermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26):2443-2454, 2012b.
Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, Mak TW. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science270(5238):985-988, 1995.
Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol 30(21):2691-2697, 2012.
Weber JS, Minor DR, D’Angelo S, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH, Grob J, Lao C, Linette G, Grossmann K, Hassel J, Lorigan P, Maio M, Sznol M, Lambert A, Yang A, Larkin J. A phase 3 randomized, open-label study of nivolumab (anti-PD-1; BMS-936558; ONO-4538) versus investigator’s choice chemotherapy (ICC) in patients with advanced melanoma after prior anti-CTLA-4 therapy. ESMO Annual Meetings. Abstract #LBA3_PR. 2014.
Westin JR, Chu F, Zhang M, Fayad LE, Kwak LW, Fowler N, Romaguera J, Hagemeister F, Fanale M, Samaniego F, Feng L, Baladandayuthapani V, Wang Z, Ma W, Gao Y, Wallace M, Vence LM, Radvanyi L, Muzzafar T, Rotem-Yehudar R, et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol 15(1):69-77, 2014.
Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, et al. Nivolumab plus ipilimumab in advanced melanoma.N Engl J Med 369(2):122-133, 2013.
Yang JC, Hughes M, Kammula U, Royal R, Sherry RM, Topalian SL, Suri KB, Levy C, Allen T, Mavroukakis S, Lowy I, White DE, Rosenberg SA. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother30(8):825-830, 2007.
Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol8(6):467-477, 2008.
[Discovery Medicine; ISSN: 1539-6509; Discov Med 18(101):341-350, December 2014.Copyright © Discovery Medicine. All rights reserved.]
Related Articles
Like this:
Like Loading...
Read Full Post »