You are on page 1of 14

The

new england journal

of

medicine

review article

medical progress

Renal-Cell Carcinoma
Herbert T. Cohen, M.D., and Francis J. McGovern, M.D.

n the united states, renal cancer is the 7th leading malignant condition among men and the 12th among women, accounting for 2.6 percent of all cancers.1 About 2 percent of cases of renal cancer are associated with inherited syndromes. In the United States, 36,160 new cases of renal cancer are predicted to occur in 2005, many of which are being discovered earlier because of the widespread availability of radiographic testing. Nevertheless, 12,660 deaths from the disease are predicted to occur in 2005.1 Renal-cell carcinomas arise from the renal epithelium and account for about 85 percent of renal cancers. A quarter of the patients present with advanced disease, including locally invasive or metastatic renal-cell carcinoma. Moreover, a third of the patients who undergo resection of localized disease will have a recurrence. Median survival for patients with metastatic disease is about 13 months. Thus, there is a great need for more effective surgical and medical therapies.

From the Renal and HematologyOncology Sections, Departments of Medicine and Pathology, Boston University School of Medicine (H.T.C.); and the Department of Urology, Massachusetts General Hospital and Harvard Medical School (F.J.M.) all in Boston. Address reprint requests to Dr. Cohen at the Department of Medicine, Renal Section, Boston University School of Medicine, Evans Biomedical Research Center, 650 Albany St., Rm. X-535, Boston, MA 02118, or at htcohen@bu.edu. N Engl J Med 2005;353:2477-90.
Copyright 2005 Massachusetts Medical Society.

overview
The classic presentation of renal-cell carcinoma includes the triad of flank pain, hematuria, and a palpable abdominal mass. Few patients now present in this manner. Roughly half the cases are now detected because a renal mass is incidentally identified on radiographic examination. Other common presenting features may be nonspecific, such as fatigue, weight loss, or anemia. Risk factors for renal-cell carcinoma include smoking, obesity, and hypertension,2 as well as acquired cystic kidney disease associated with end-stage renal disease. A 1.6:1.0 male predominance exists,1 and the peak incidence is in the sixth and seventh decades. Gross or microscopic hematuria is an important clinical clue to the diagnosis of renal-cell carcinoma; thus, hematuria should be evaluated promptly by a computed tomographic (CT) scan of the genitourinary tract and, in patients older than 40 years of age, by cystoscopy to rule out bladder cancer. Prognosis is closely related to the stage of disease (Fig. 1). The Heidelberg classification of renal tumors was introduced in 19976 as a means of more completely correlating the histopathological features with the identified genetic defects (Table 1).

tumor types and molecular pathogenesis


conventional or clear-cell renal-cell carcinoma

Von HippelLindau disease (number 193300 in Mendelian Inheritance in Man [MIM]) is a rare, autosomal dominant, familial cancer syndrome consisting chiefly of retinal angiomas, hemangioblastomas of the central nervous system, pheochromocytomas, and renal-cell carcinoma of the clear-cell type (Fig. 2). The von HippelLindau tumorsuppressor gene (VHL) was identified in 1993.7 In this disease, one VHL allele is inherited with a mutation. Associated focal lesions, such as renal-cell carcinoma, arise from the inactivation or silencing of the remaining normal (wild-type) VHL allele (Fig. 3). Remarkably, defects in the VHL gene also appear to be responsible for about 60 percent of

n engl j med 353;23

www.nejm.org

december 8, 2005

2477

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

Figure 1. Staging Overview and Five-Year Survival Rates for Renal Cancer. Survival data3 are based on the 1997 tumornodemetastasis (TNM) staging guidelines.4 More recent renal-cancer staging is described elsewhere.5

the cases of sporadic clear-cell renal-cell carcinoma,8 which represents a major portion of all cases of renal-cell carcinoma. VHL protein, the product of the VHL gene, functions as a tumor suppressor, inhibiting growth when reintroduced into cultures of renal-cell carcinoma.9,10 Hypoxia-inducible genes are normally inhibited by VHL protein,11 including several encoding proteins involved in angiogenesis (e.g., vascular endothelial growth factor [VEGF]), cell growth (e.g., transforming growth factor a [TGF-a]), glucose uptake (e.g., the GLUT-1 glucose transporter), and acidbase balance (e.g., carbonic anhydrase IX [CA9]). When VHL protein is lost, these proteins are overexpressed, creating a microenvironment favorable for epithelial-cell proliferation (Fig. 4A). Thus, cells deficient in VHL protein behave as if they are hypoxic, even in conditions of normoxia. VHL protein, with elongin proteins C and B, binds cul2 protein (a member of the cullin family of ubiquitin ligase proteins), indicating that some VHL protein serves as the receptor subunit of a ubiquitin ligase complex

that promotes the ubiquitination and destruction of proteins (Fig. 4B).12,13 VHL protein binds the transcriptional activators hypoxia-inducible factor 1a (HIF-1a) and 2a (HIF-2a) directly and destabilizes them.14 Furthermore, VHL protein promotes the ubiquitination and destruction of HIF-a.15-17 These VHL-regulated pathways are being studied as potential targets of therapies for clear-cell renal-cell carcinoma. HIF is the key regulator of the hypoxic response in multicellular organisms. Thus, VHL protein has a central role in oxygen sensing. For HIF-a to bind VHL protein, a proline residue must undergo hydroxylation, which is an unusual protein modification18,19 (Fig. 4B). A family of proline hydroxylases operates on HIF-a in a graded fashion, so that the extent of hydroxylation depends on oxygen tension.20,21 Hydroxylation of an asparagine residue blocks the interaction of HIF-a with the transcriptional coactivator p300.22 Thus, multiple hydroxylation steps cooperate to inhibit HIF-a activity. To correlate the genotype with the disease phe-

2478

n engl j med 353;23

www.nejm.org

december 8 , 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

Table 1. Sporadic and Hereditary Renal-Cell Carcinomas and Genetic Defects According to Histologic Appearance.* Sporadic Renal-Cell Carcinomas Histologic Appearance Incidence Gene and Frequency percent Conventional 75 VHL, 60 VHL disease FCRC Hereditary paraganglioma HPRC HLRCC BirtHoggDub syndrome BirtHoggDub syndrome VHL Chromosome 3p translocation SDHB MET FH BHD BHD Renal-Cell Carcinomas in an Inherited Syndrome Rare Syndrome Gene

Papillary Chromophobe Oncocytoma Collecting duct Unclassified

12 4 4 <1 35

MET, 13 TFE3, <1

* VHL denotes von HippelLindau, FCRC familial clear-cell renal cancer, SDHB succinate dehydrogenase B, HPRC hereditary papillary renal carcinoma, HLRCC hereditary leiomyomatosis and renal-cell cancer, and FH fumarate hydratase. Additional rare syndromes or infrequent associations are not included.

ing 5:1 male predominance. Localized papillary renal-cell carcinoma metastasizes less frequently than clear-cell renal-cell carcinoma.34 However, the survival rate for metastatic papillary renal-cell carcinoma is probably worse than that for clear-cell renal-cell carcinoma.35 The risk of both types is particularly increased among patients with endstage renal disease. Chromosome 7, which harbors the MET proto-oncogene, is duplicated in 75 percent of sporadic papillary cases. There are two subtypes of papillary renal-cell carcinoma.36 Type 1 tumors are papillary lesions covered by small cells with pale cytoplasm and small oval nuclei with indistinct nucleoli, and type 2 tumors are papillary lesions covered by large cells with abundant eosinophilic cytoplasm. Type 2 cells are typified by pseudostratification and large, spherical nuclei with distinct nucleoli. Type 2 tumors are genetically more heterogeneous, have a poorer prognosis, and may arise from type 1 tumors.37 Papillary renal-cell carcinoma occurs in several familial syndromes (MIM number 605074). Hereditary papillary renal carcinoma is an autosomal dominant disorder associated with multifocal papillary renal-cell carcinoma38 with type 1 histologic features (Fig. 3).39 The causative gene, mutations in which are responsible for hereditary papillary renal carcinoma, has been identified at chromosome 7 and encodes MET, a receptor tyrosine kinase that is normally activated by hepatocyte growth papillary renal-cell carcinoma factor 40 (Fig. 4C). In hereditary papillary renal carSporadic papillary renal-cell carcinoma has a five- cinoma, the MET receptor tyrosine kinase domain year survival rate approaching 90 percent and a strik- undergoes autoactivating amino-acidsubstitu-

notype, naturally occurring VHL mutations have been evaluated to determine their effect on HIF-a ubiquitination. An intriguing finding is that the VHL mutations that disrupt HIF-a processing are the same as those associated with the vascular manifestations of von HippelLindau disease, such as hemangioblastoma (Fig. 2).15,16,23,24 Since renalcell carcinoma develops in only a subgroup of patients with hemangioblastoma, the overexpression of HIF-a appears to be necessary for, but not sufficient to induce, renal tumorigenesis. Nevertheless, HIF-a is vitally important to the pathogenesis of this disease. VHL-induced inhibition of HIF-a is sufficient to suppress the growth of clear-cell renal-cell carcinoma in preclinical models.25,26 The cell-matrix protein fibronectin,27 chaperonin TRiC/ CCT,28 microtubules,29 and transcription factor Jade-130-32 are all molecules that interact with VHL protein in a manner that is dependent on VHL mutation, suggesting that they may also contribute to disease pathogenesis. Distinct from von HippelLindau disease, familial clear-cell renal cancer has been reported in patients with translocations of chromosome 3p at a fragile site at 3p14.33 Loss of the translocated chromosome 3p probably implicates VHL protein in the development of these tumors. Additional translocations of chromosome 3 have been associated with clear-cell renal-cell carcinoma as well.

n engl j med 353;23

www.nejm.org

december 8, 2005

2479

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

Figure 2. Schematic Representation of the Clinical Spectrum of von HippelLindau Disease and Potential Biologic Mechanisms. The three major manifestations of von HippelLindau (VHL) disease central nervous system hemangioblastoma, pheochromocytoma, and clear-cell renal-cell carcinoma are represented as Venn diagrams. Affected families may not have all three conditions, depending largely on the type of mutation in the VHL gene inherited. Circle sizes are not intended to reflect the numbers of patients in each group. Type 1 VHL disease (Panel A) is most common; it comprises clear-cell renal-cell carcinoma with central nervous system hemangioblastoma and is due to major disruptions in the VHL protein, such as those resulting from truncating mutations. Type 2 VHL disease (Panel A) includes pheochromocytoma with one, both, or neither of the other features. Type 2B disease predisposes patients to all three conditions, whereas type 2A includes hemangioblastoma and pheochromocytoma and type 2C includes only pheochromocytoma. Type 2 is most often due to more subtle VHL gene mutations, such as missense mutations that result in the substitution of a single amino acid. Besides classic von HippelLindau disease, sporadic cases of hemangioblastoma or pheochromocytoma have been shown to be due to germ-line VHL mutations. In addition, familial Chuvash polycythemia, which does not include any of the features of von HippelLindau disease, is an autosomal recessive disorder caused by homozygosity for the specific VHL missense mutation R200W. In Panel B, the spectrum of manifestations of von HippelLindau disease, along with biochemical studies, suggests the involvement of specific biochemical pathways. For example, the mutations in VHL protein that disrupt the ubiquitination and destruction of hypoxia-inducible factor a (HIF-a) are the same as those that correlate with the lesions of hemangioblastoma. Interestingly, von HippelLindau disease type 2A mutations disrupt HIF-a processing and do not cause renal cancer, suggesting that the HIF pathway plus additional, unknown VHL pathways (called X pathways) must be disrupted for clear-cell renal-cell carcinoma to develop. The probable involvement of additional pathways is supported by the multistep nature of renal tumorigenesis. Alternatively, the unknown pathway or pathways may simply reflect a higher level of expression of HIF-a in this subgroup of patients. Pheochromocytomas can arise from mutations in the gene for VHL protein that do not affect HIF-a processing, suggesting that they arise through the disruption of other, unknown VHL pathways (called Y pathways). Thus, the overexpression of HIF- a correlates closely with the development of hemangioblastoma; appears to be necessary for, but not sufficient to induce, renal tumorigenesis; and is not necessary for the development of pheochromocytoma.

tion mutations, which promote cellular transformation.41 Subsequently, chromosome 7 harboring the MET mutation is duplicated, increasing the gene dose.42,43 Only a small percentage of the cases of the sporadic papillary type have MET mutations.40 Thus, the pathogenesis of hereditary papillary renal carcinoma is usually different from that of sporadic papillary renal-cell carcinoma. Patients with the hereditary leiomyomatosis and renal-cell cancer syndrome (MIM number 605839) are at risk for cutaneous and uterine leiomyomas and solitary papillary renal-cell carcinoma with

type 2 histologic features.44 Occasionally, cases of collecting-duct or clear-cell renal-cell carcinoma occur. These cases of papillary renal-cell carcinoma metastasize early and are the most aggressive of the familial types.45 Intriguingly, FH, the gene that causes this autosomal dominant syndrome, encodes fumarate hydratase, a Krebs-cycle enzyme.46 As with the loss of a tumor-suppressor gene, the wild-type FH allele is lost in hereditary leiomyomatosis and in lesions of renal-cell carcinoma.47 Along similar lines, cases of renal-cell carcinoma with solid histologic features or cases of the clear-cell form

2480

n engl j med 353;23

www.nejm.org

december 8 , 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

Figure 3. Steps in the Development of Renal-Cell Carcinoma. In contrast to sporadic renal-cell carcinoma (Panels A and C), fewer steps are required for the development of renal-cell carcinoma in the inherited forms of the disease (Panels B and D), because all of the patients cells have a mutation that predisposes the patient to the disease. As a result, the disease associated with the familial syndromes occurs earlier and is often multifocal. Each familial renal cancer syndrome is autosomal dominant. In von HippelLindau disease, a cellular recessive mechanism is involved, since both copies of the VHL gene are inactivated (Panels A and B). VHL is a classic tumor-suppressor gene. In hereditary papillary renal carcinoma, one copy of the MET gene has an activating mutation, which is inherited (Panel D). Chromosome 7, which includes the defective MET allele, becomes duplicated, increasing the level of expression of the activated MET protein, which is a receptor tyrosine kinase for hepatocyte growth factor. Activated MET is a classic oncogene. A plus sign represents the wild-type allele; a minus sign represents a null allele. A plus sign in red type represents a mutated, activated allele; two plus signs in red type represent duplication of that allele.

n engl j med 353;23

www.nejm.org

december 8, 2005

2481

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

have been reported in patients with the hereditary paraganglioma syndrome (MIM number 115310). Certain forms of hereditary paraganglioma are associated with germ-line defects in the succinate dehydrogenase B gene.48 Succinate dehydrogenase B protein is another mitochondrial, Krebs-cycle enzyme. Thus, an intriguing connection exists among cellular ATP production, the hypoxic response, and tumorigenesis in both neuronal and kidney tissue. A number of sporadic cases of papillary renalcell carcinoma have chromosomal translocations involving the TFE3 gene at chromosome Xp11.2.49-51 Children and young adults are affected without predilection for sex, and the histologic features of such cases have been variably described as papillary renal-cell carcinoma, clear-cell renal-cell carcinoma, or a unique type of pathology. The TFE3 gene encodes a helixloophelix transcription factor related to the proto-oncogene product c-myc. Key TRE3 domains become fused with other gene products, and renal-cell carcinoma is probably due to TFE3 overexpression.
oncocytoma and chromophobe renal-cell carcinoma

Oncocytomas, which are benign, account for about 4 percent of nephrectomies performed because renal-cell carcinoma is suspected. The chromophobe variant of renal-cell carcinoma also accounts for 4 percent of all cases of renal-cell carcinoma52 and may have a benign course after surgery, provided that the tumor stage and grade are favorable.53 Oncocytoma is thought to originate from type A intercalated cells of the collecting duct, whereas chromophobe renal-cell carcinoma is thought to originate from type B intercalated cells. The BirtHoggDub syndrome (MIM number 135150) is a rare autosomal dominant disorder characterized by hair-follicle hamartomas (fibrofolliculomas) of the face and neck.54-57 About 15 percent of affected patients have multiple renal tumors, most often chromophobe or mixed chromophobeoncocytomas. Occasionally, papillary or clear-cell renal-cell carcinoma develops in patients with the BirtHoggDub syndrome. BHD, the gene implicated in the syndrome, encodes the protein folliculin,58 a suspected tumor suppressor. BHD mutations occur only rarely in sporadic renal-cell carcinoma.59,60 The BirtHoggDub renal phenotype supports the existence of a close relationship between oncocytoma and chromophobe renal-cell carcinoma.

Figure 4 (facing page). Molecular Mechanisms of the Development of Renal-Cell Carcinoma. Pathologic cooperativity between renal-cancer cells of the clear-cell type and adjacent vasculature is shown in Panel A. In clear-cell renal-cell carcinoma, hypoxiainducible factor a (HIF-a) transcription factor accumulates, resulting in the overexpression of proteins that are normally inducible with hypoxia, such as transforming growth factor a and b (TGF-a and TGF-b, respectively), vascular endothelial growth factor (VEGF), and plateletderived growth factor B chain (PDGF-B). The overexpressed VEGF, PDGF-B, and TGF-b act on neighboring vascular cells to promote tumor angiogenesis. The augmented tumor vasculature provides additional nutrients and oxygen to promote the growth of tumor cells. TGF-a acts in an autocrine manner on the tumor cells by signaling through the epidermal growth factor receptor, which promotes tumor-cell proliferation and survival. The role of von HippelLindau (VHL) protein in clear-cell renalcell carcinoma and in controlling the expression of the HIF-a transcription factors is shown in Panel B. Under normoxic conditions HIF-a is hydroxylated on two proline residues by a proline hydroxylase and on an asparagine residue by an asparagine hydroxylase. Hydroxylation (OH) by proline hydroxylase permits binding of HIF-a to VHL protein, which promotes the ubiquitination (Ub) and destruction of HIF-a by the proteasome pathway. Hydroxylation by asparagine hydroxylase blocks the interaction of HIF-a with transcriptional coactivator p300. VHL protein, with elongin proteins C and B, binds cul2 protein (a member of the cullin family of ubiquitin ligase proteins). RING-box protein Rbx1 serves as the ubiquitin transferase for the VHL skp-cullin-F-box protein (SCF) complex. In the absence of wild-type VHL protein, hydroxylated HIF-a accumulates and is able to heterodimerize with HIF-b and activate transcription at hypoxia-response elements (HREs), which are found in genes such as VEGF. In hypoxic conditions, HIF-a is not hydroxylated and so cannot bind VHL protein. Panel C shows the role of MET in papillary renal carcinoma. MET is a receptor tyrosine kinase for hepatocyte growth factor. In the absence of ligand, MET normally exists in an autoinhibited state. MET homodimerizes in the presence of ligand hepatocyte growth factor and undergoes reciprocal phosphorylation and activation. In hereditary papillary renal carcinoma and in occasional sporadic papillary renal-cell carcinoma, activating mutations of MET disinhibit the receptor, even in the absence of ligand. Furthermore, the mutated MET allele on chromosome 7 becomes duplicated, increasing the expression of the activated MET protein.

collecting-duct renal-cell carcinoma

Collecting-duct renal-cell carcinoma accounts for less than 1 percent of all cases of renal-cell carcinoma and is typically an aggressive tumor. Medullary carcinoma of the kidney, which may be a variant of the collecting-duct type, is associated with sickle

2482

n engl j med 353;23

www.nejm.org

december 8 , 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

n engl j med 353;23

www.nejm.org

december 8, 2005

2483

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

cell trait or disease. The collecting-duct form may ing. Organizations such as the VHL Family Allibe most similar to transitional-cell carcinoma of the ance (www.vhl.org) are a vital resource for patients, urothelium. families, physicians, and researchers.

management of sporadic and hereditary renal-cell carcinoma


An enhancing renal mass on a CT scan obtained after the administration of contrast material is a strong clue that renal cancer is present. A staging workup should be performed before treatment is initiated. Multiple enhancing lesions, or a family history of renal-cell carcinoma, particularly in persons younger than 50 years of age, suggests a hereditary predisposition to the disease. Von HippelLindau disease, hereditary leiomyomatosis and renal-cell cancer, and the BirtHoggDub syndrome all have extrarenal manifestations, whereas familial clearcell renal cancer and hereditary papillary renal carcinoma do not. Thus, a careful physical examination including ophthalmologic, neurologic, and dermatologic evaluation may be helpful. CT scanning or magnetic resonance imaging (MRI) of the abdomen and pelvis may reveal uterine tumors in patients with hereditary leiomyomatosis and renal-cell cancer or renal cysts or pancreatic or adrenal involvement in patients with von HippelLindau disease. Patients with hereditary renal-cell carcinoma should be closely monitored. CT before and after the administration of contrast material is the best test for detection and assessment of renal masses, with gadolinium-enhanced MRI as an alternative. Such studies can be performed at intervals ranging from every three to six months to every two to three years, depending on the size of the lesions and the type of syndrome. Larger masses require more frequent evaluation. Because small masses are usually of low grade, they can be observed until they reach 3 cm, at which time they should be removed.61,62 However, tumors caused by hereditary leiomyomatosis and renal-cell cancer should be excised immediately because of their aggressive nature. Patients with von HippelLindau disease should undergo MRI studies of the brain and spinal cord to screen for hemangioblastoma. A family pedigree should be generated, and family members at risk should be encouraged to seek medical attention. Testing is available for the VHL, MET, FH, and BHD genes. One goal of such testing is to free unaffected family members from continued cancer screen-

prognosis
Defining the prognosis of renal-cell carcinoma is important for both therapeutic decision-making and counseling patients. For metastatic renal-cell carcinoma, poor prognostic factors include a low Karnofsky performance-status score (a standard way of measuring functional impairment in patients with cancer), a high level of serum lactate dehydrogenase, a low hemoglobin level, and a high corrected level of serum calcium.63,64 The University of California, Los Angeles, Integrated Staging System was developed to evaluate the prognosis at diagnosis and in the presence of metastatic disease; it includes tumornodemetastasis (TNM) staging, the patients score on the Eastern Cooperative Oncology Group performance-status scale (another measure of functional impairment in patients with cancer), and the Fuhrman nuclear grade, which assesses histologic features of the tumor.65 This system has been used successfully in more than 4000 patients at eight international centers.66

surgical treatment
radical nephrectomy

Surgical excision is the primary treatment for renal-cell carcinoma. Radical nephrectomy, which includes removal of the kidney en bloc with Gerotas fascia, the ipsilateral adrenal gland, and regional lymph nodes, has been the standard therapy, although more limited approaches are being explored. The surgical approach is determined by the size and location of the tumor within the kidney, the TNM stage, and any special anatomical considerations. Staging and evaluation for the presence of metastases, including a careful history-taking and physical examination, should be completed before surgery. Routine laboratory studies should include measurement of the hematocrit and serum levels of creatinine, calcium, and alkaline phosphatase and a urinalysis for proteinuria. Imaging studies, such as radiographs of the chest, CT of the abdomen and pelvis, and in some cases, MRI evaluation of the renal vein and inferior vena cava, CT of the chest or head, or bone scanning may be needed. The fre-

2484

n engl j med 353;23

www.nejm.org

december 8, 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

quency of follow-up after surgery depends on the adjacent structures. Because identification of the type of renal-cell carcinoma is important, a core bistage of the tumor. opsy of the renal mass should be performed as part surgery for metastatic disease of the procedure. Ideal candidates for minimally Nephrectomy may be warranted, even in the pres- invasive percutaneous ablative therapy are patients ence of metastatic disease. The combination of in- with tumors less than 3 cm in diameter who have terferon alfa and nephrectomy is superior to inter- serious coexisting conditions and for whom stanferon alfa alone, offering a survival advantage of dard approaches would pose substantial risks. Pa3 to 10 months.67,68 Surgical excision of a solitary tients with multifocal tumors may also benefit from metastasis in patients with advanced renal-cell car- minimally invasive percutaneous procedures. Highcinoma is recommended in many cases, but this frequency focused ultrasound applied externally to approach has not yet been proved to be effective in the body is being studied as another potential minimally invasive therapy. prolonging survival.
nephron-sparing partial nephrectomy

medical treatment
Medical therapies are generally offered for locally advanced or metastatic renal-cell carcinoma (Table 2), and much of the clinical experience with this approach is in patients with the clear-cell type. Because response rates are low, the need to identify new therapeutic agents is great.74
chemotherapy

Nephron-sparing partial nephrectomy has gained acceptance for treating tumors less than 4 cm in diameter. Other indications for partial nephrectomy may include a solitary kidney, bilateral renal masses, or renal insufficiency, as well as the presence of hypertension, diabetes, or hereditary renal-cell carcinoma syndromes. Results achieved with nephronsparing surgery are similar to those with radical nephrectomy, but a disadvantage is a rate of local recurrence of 3 to 6 percent.69
laparoscopic nephrectomy

First reported in 1991,70 laparoscopic nephrectomy has accelerated the evolution toward minimally invasive surgical management of renal-cell carcinoma. The benefits of the laparoscopic approach include decreased postoperative pain, a shorter hospitalization, and a quicker recovery. The laparoscopic approach has been used for both radical nephrectomy and partial nephrectomy. The laparoscopic partial nephrectomy, however, is a technically demanding procedure with the potential for in- immunomodulatory therapies creased perioperative complications.71 The value of immunomodulatory therapy for clearcell renal-cell carcinoma is supported by reports of percutaneous ablative approaches occasional spontaneous tumor regression, infreThe most recent evolution in the surgical manage- quent complete regression of metastatic disease ment of small tumors has been percutaneous ther- with cytokine therapies, and promising early results mal ablative techniques that use radiofrequency with allogeneic stem-cell transplantation and tumor heat ablation72 or cryoablation73 to destroy tumor vaccines. The goal of immunomodulatory therapy cells. A needle probe is advanced through the skin is to boost either tumor antigenicity or host surveiland directed into the tumor under image guid- lance. Unique tumor antigens may also be inducance. Although early results of radiofrequency ab- ible in renal-cell carcinoma.79 lation and cryoablation are encouraging, larger trials with long-term follow-up are needed. The rates Interferon Alfa of complications appear to be low, but reported ad- About 14 percent of cases of metastatic clear-cell verse events include intraoperative and postopera- renal carcinoma respond to interferon alfa alone. tive hemorrhage, urinary leakage, and injury to Various doses and routes have been used.80 The me-

Rates of response to chemotherapy alone are low (roughly 4 to 6 percent).75 Drug resistance may be related to the expression of the multidrug resistance transporter in proximal-tubule cells the cells from which clear-cell and papillary renal-cell carcinoma may originate. Chemotherapy may be more efficacious for advanced nonclear-cell renal-cell carcinoma, particularly the collecting-duct type.76-78 A phase 2 trial of carboplatin and paclitaxel for the collecting-duct form of the disease is under way.

n engl j med 353;23

www.nejm.org

december 8, 2005

2485

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

Table 2. Medical Therapies for Advanced Renal Cancer.* FDA-approved regimen High-dose interleukin-2 (aldesleukin, immunomodulatory cytokine) Commonly used agents Low-dose interleukin-2 Interferon alfa (immunomodulatory cytokine) Experimental therapies Bevacizumab (humanized VEGF-neutralizing antibody) Sunitinib malate (VEGF receptor and multitargeted kinase inhibitor) Sorafenib tosylate (VEGF receptor and multitargeted kinase inhibitor) Panitumumab (human EGFR-neutralizing antibody) Gefitinib (EGFR tyrosine kinase inhibitor) Erlotinib (EGFR tyrosine kinase inhibitor) Temsirolimus (inhibitor of the mammalian target of rapamycin) Tumor vaccines Allogeneic stem-cell transplantation * FDA denotes Food and Drug Administration, VEGF vascular endothelial growth factor, and EGFR epidermal growth factor receptor. The trade name for interleukin-2 is Proleukin; for bevacizumab, Avastin; for sunitinib malate, Sutent; for sorafenib tosylate, Nexavar; for gefitinib, Iressa; for erlotinib, Tarceva.

of patients with metastatic disease. Identifying features predictive of a response to interleukin-2 would represent a further advance, and efforts are being made to identify patients with clear-cell renal carcinoma who would be likely to have a response to interleukin-2 therapy on the basis of pathological characteristics and expression of CA9.
adjuvant therapy

Given the high rate of recurrence of renal-cell carcinoma after nephrectomy, a follow-up adjuvant approach would be desirable, especially for patients with high-risk, locally advanced disease. However, conventional chemotherapy, interferon alfa, or even interleukin-283 has not proved effective as an adjuvant therapy. Approaches currently being tested include tumor vaccines and a monoclonal antibody directed against CA9.

evolving therapies
stem-cell transplantation

dian duration of response is six months and rarely exceeds two years. Because the side effects of the drug are not onerous, it appears to be a good choice to use in combination with other agents in experimental approaches.
Interleukin-2

High-dose interleukin-2 is the standard therapy for advanced renal-cell carcinoma and is the only regimen for this disease approved by the Food and Drug Administration. However, many patients with metastatic disease cannot take high-dose interleukin-2, because it causes a capillary leak syndrome or because it is not available in all treatment centers. High-dose interleukin-2 induces responses in 21 percent of patients, as compared with only 13 percent of patients who receive low-dose interleukin-2.81 The median duration of response has been reported to be 54 months overall, and for those with a complete response, the median duration of a response is yet to be reached.82 Interleukin-2 has also been used in combination with other drugs, but it is unclear whether combined therapy achieves better results than interleukin-2 alone. Thus, interleukin-2 is a highly effective therapy for a subgroup

Allogeneic stem-cell transplantation performed after the administration of a nonmarrow ablative regimen elicits a potent graft-versus-tumor effect and appears promising for treating clear-cell renalcell carcinoma.84 Protocols developed at the National Institutes of Health have used myelosuppressive pretreatment, followed by an infusion of donor CD34+ cells and T cells from an HLA-identical sibling.84 A course of immunosuppressive agents, such as cyclosporine, is used to limit graft-versushost disease and is rapidly tapered. Twenty of the first 45 patients with metastatic renal-cell carcinoma who underwent stem-cell transplantation had a response (44 percent).85 However, results in some other centers have been less promising. The responses have correlated well with the development of graft-versus-host disease and with the conversion of T-cell chimerism to full donor origin. One goal is to identify the tumor epitopes that are initiating the graft-versus-tumor response to improve treatment specificity. The two drawbacks to stemcell transplantation have been severe graft-versushost disease, which can be life-threatening, and the need for a haplotype-matched sibling donor. Prognosis is also an important guide to patient selection, since responses take several months. The next generation of strategies for stem-cell transplantation may include the use of tumor vaccines after transplantation as well as the use of cytokine therapy to boost recipient or even donor immunity.

2486

n engl j med 353;23

www.nejm.org

december 8, 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

tumor vaccines

Tumor vaccines represent a potential means of enhancing host immunity. A promising approach to the treatment of advanced clear-cell renal carcinoma uses autologous or donor dendritic cells, which initiate a primary immune response by presenting antigen in the context of costimulatory molecules. Dendritic cells can be pulsed with tumor protein,86 DNA, or RNA87; they can even be fused with tumor cells88,89 to present tumor antigens in a context favorable for therapy. Such vaccines are generally well tolerated, but they will require further optimization. Concomitant administration of cytokines may improve the response to vaccines.
target antigens

vided a key proof of principle of the efficacy of anti-angiogenic therapy and may offer additional benefit when given in combination with other drugs. Inhibitors of VEGF receptor tyrosine kinase are being developed and tested. Indeed, the multitargeted kinase inhibitors sunitinib and sorafenib have shown great promise in phase 2 and phase 3 trials, with at least stabilization of disease in as many as 70 percent of patients with cytokine-refractory disease.
TGF-aPathway Components as Molecular Targets

A goal of stem-cell or vaccine therapies is to characterize the tumor antigens involved in the immune response. One potential target is the G250 renal cancer antigen, which has been identified as CA9. The CA9 gene is a target of HIF and so is overexpressed in VHL-related clear-cell renal carcinoma, even in the earliest lesions of von HippelLindau disease.90 Thus, in cases of renal-cell carcinoma, a high proportion of CA9-positive cells may be associated with a more favorable prognosis.91 As a transmembrane protein, CA9 may also be a therapeutically useful tumor antigen.92,93 It will be important to identify additional target antigens.
therapies targeting vegf and tgf-a pathways

Originally identified as regulated by VHL, VEGF and TGF-a are now promising therapeutic targets in clear-cell renal carcinoma. The manner in which these molecules interact with the cancer epithelium and surrounding vascular endothelium leads to tumor progression (Fig. 4A). A combination of therapies based on rational targets such as these may therefore be a powerful approach to advanced renalOther Approaches cell carcinoma. Temsirolimus (CCI-779), a selective inhibitor of VEGF-Pathway Components as Molecular Targets the mammalian target of rapamycin, has shown VEGF is overexpressed throughout clear-cell renal- efficacy in a phase 2 trial of metastatic renal-cell cell carcinoma tissue and may be the most impor- carcinoma.101 Temsirolimus may inhibit HIF as tant tumor angiogenic factor. A randomized phase well. Partial responses were noted in 7 percent of 2 trial involving patients with metastatic renal-cell patients, and minor responses in 26 percent. The carcinoma investigated the efficacy of bevacizu- median survival rate was 15 months. The notable mab, a humanized VEGF-neutralizing antibody.94 activity of the drug in patients with poor prognosThis agent extended the interval before tumor pro- tic features prompted a phase 3 trial. Other opgression to 4.8 months, as compared with 2.5 tions are being pursued, including agents targetmonths for placebo. Bevacizumab therefore pro- ing HIF.

TGF-a is a potent growth factor for epithelial cells that acts through the epidermal growth factor receptor (EGFR), which is a receptor tyrosine kinase. TGF-a is overexpressed in the epithelium in clearcell renal carcinoma and is a VHL target.95 Overexpression of TGF-a is an early event in the pathogenesis of this disease.96 Furthermore, growth of renal cancer cells in culture is dependent on TGF-a.97 Thus, the TGF-a pathway is a logical choice for therapeutic intervention. Antibodies against EGFR are thought to bind EGFR and promote its down-regulation from the cell surface. A fully human monoclonal antibody against human EGFR, called panitumumab (ABX-EGF), has been evaluated in a phase 2 trial involving 88 patients with metastatic renal-cell carcinoma.98 Only one patient had a complete response, and two had partial responses a disappointing result. Small-molecule inhibitors of the EGFR tyrosine kinase are also being developed.99 The quinazolines gefitinib and erlotinib are now in phase 2 trials. In a phase 1 trial of erlotinib, just one patient with metastatic disease had a complete response.100 Erlotinib is also being tested in combination with bevacizumab, although encouraging initial results could not be confirmed in a randomized phase 2 trial.

n engl j med 353;23

www.nejm.org

december 8, 2005

2487

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

The

new england journal

of

medicine

summary and prospects for the future


The poor prognosis of advanced renal-cell carcinoma demands an aggressive search for new therapeutic agents and strategies. Leads will probably come from both a careful elucidation of the biology of each type of the disease and broader approaches, such as gene-expressionarray and proteome analyses. Much has been accomplished since the identification of the VHL gene in 1993. Already, VHL protein pathways, such as those involving VEGF and TGF-a, are being exploited therapeutically, and agents affecting these pathways might be more effective when used in combination. Identification of the MET gene was another key advance.
refer enc es
1. Jemal A, Murray T, Ward E, et al. Cancer

The mutated, activated hepatocyte growth factor receptor MET could be targeted in the papillary form of the disease.102,103 The immune responsiveness of renal-cell carcinoma provides an opportunity for the development and optimization of vaccines and other immune therapies. Preservation of as much renal function as possible and reduced rates of complications are two goals of new minimally invasive approaches to renal-cell carcinoma; other goals are to identify early markers of disease, prognosis, or responsiveness to therapy.
Supported in part by grants (RO1 CA79830 and RO1 DK67569, to Dr. Cohen) from the National Institutes of Health. We are indebted to Drs. Michael B. Atkins, William G. Kaelin, Jr., Matthew R. Smith, Walter M. Stadler, and Berton Zbar for careful review of the manuscript.

statistics, 2005. CA Cancer J Clin 2005;55: 10-30. 2. Chow WH, Gridley G, Fraumeni JF Jr, Jarvholm B. Obesity, hypertension, and the risk of kidney cancer in men. N Engl J Med 2000;343:1305-11. 3. Javidan J, Stricker HJ, Tamboli P, et al. Prognostic significance of the 1997 TNM classification of renal cell carcinoma. J Urol 1999;162:1277-81. 4. Kidney. In: Fleming ID, Cooper JS, Henson DE, et al., eds. AJCC cancer staging handbook. Philadelphia: Lippincott-Raven, 1998: 215-7. 5. Kidney. In: Greene FL, Page DL, Fleming ID, et al., eds. AJCC cancer staging manual. New York: Springer-Verlag, 2002:323-5. 6. Kovacs G, Akhtar M, Beckwith BJ, et al. The Heidelberg classification of renal cell tumours. J Pathol 1997;183:131-3. 7. Latif F, Tory K, Gnarra J, et al. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science 1993;260:1317-20. 8. Kim WY, Kaelin WG. Role of VHL gene mutation in human cancer. J Clin Oncol 2004;22:4991-5004. 9. Iliopoulos O, Kibel A, Gray S, Kaelin WG Jr. Tumour suppression by the human von Hippel-Lindau gene product. Nat Med 1995;1:822-6. 10. Chen F, Kishida T, Duh FM, et al. Suppression of growth of renal carcinoma cells by the von Hippel-Lindau tumor suppressor gene. Cancer Res 1995;55:4804-7. 11. Iliopoulos O, Levy AP, Jiang C, Kaelin WG Jr, Goldberg MA. Negative regulation of hypoxia-inducible genes by the von HippelLindau protein. Proc Natl Acad Sci U S A 1996;93:10595-9. 12. Pause A, Lee S, Worrell RA, et al. The von Hippel-Lindau tumor-suppressor gene product forms a stable complex with human CUL-2, a member of the Cdc53 family of

proteins. Proc Natl Acad Sci U S A 1997;94: 2156-61. 13. Lonergan KM, Iliopoulos O, Ohh M, et al. Regulation of hypoxia-inducible mRNAs by the von Hippel-Lindau tumor suppressor protein requires binding to complexes containing elongins B/C and Cul2. Mol Cell Biol 1998;18:732-41. 14. Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999; 399:271-5. 15. Cockman ME, Masson N, Mole DR, et al. Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. J Biol Chem 2000; 275:25733-41. 16. Ohh M, Park CW, Ivan M, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2000; 2:423-7. 17. Tanimoto K, Makino Y, Pereira T, Poellinger L. Mechanism of regulation of the hypoxia-inducible factor-1 alpha by the von Hippel-Lindau tumor suppressor protein. EMBO J 2000;19:4298-309. 18. Ivan M, Kondo K, Yang H, et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 2001;292:464-8. 19. Jaakkola P, Mole DR, Tian YM, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001;292:468-72. 20. Epstein AC, Gleadle JM, McNeill LA, et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 2001;107:43-54. 21. Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science 2001;294:1337-40.

22. Lando D, Peet DJ, Whelan DA, Gorman

JJ, Whitelaw ML. Asparagine hydroxylation of the HIF transactivation domain a hypoxic switch. Science 2002;295:858-61. 23. Clifford SC, Cockman ME, Smallwood AC, et al. 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;10:1029-38. 24. Hoffman MA, Ohh M, Yang H, Klco JM, Ivan M, Kaelin WG Jr. von Hippel-Lindau protein mutants linked to type 2C VHL disease preserve the ability to downregulate HIF. Hum Mol Genet 2001;10:1019-27. 25. Kondo K, Kim WY, Lechpammer M, Kaelin WG Jr. Inhibition of HIF2alpha is sufficient to suppress pVHL-defective tumor growth. PLoS Biol 2003;1:E83. 26. Zimmer M, Doucette D, Siddiqui N, Iliopoulos O. Inhibition of hypoxia-inducible factor is sufficient for growth suppression of VHL/ tumors. Mol Cancer Res 2004;2: 89-95. 27. Ohh M, Yauch RL, Lonergan KM, et al. The von Hippel-Lindau tumor suppressor protein is required for proper assembly of an extracellular fibronectin matrix. Mol Cell 1998;1:959-68. 28. Feldman DE, Spiess C, Howard DE, Frydman J. Tumorigenic mutations in VHL disrupt folding in vivo by interfering with chaperonin binding. Mol Cell 2003;12: 1213-24. 29. Hergovich A, Lisztwan J, Barry R, Ballschmieter P, Krek W. Regulation of microtubule stability by the von Hippel-Lindau tumour suppressor protein pVHL. Nat Cell Biol 2003;5:64-70. 30. Zhou MI, Wang H, Ross JJ, Kuzmin I, Xu C, Cohen HT. The von Hippel-Lindau tumor suppressor stabilizes novel plant homeodomain protein Jade-1. J Biol Chem 2002; 277:39887-98.

2488

n engl j med 353;23

www.nejm.org

december 8, 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

31. Zhou MI, Wang H, Foy RL, Ross JJ, Co-

hen HT. Tumor suppressor von HippelLindau (VHL) stabilization of Jade-1 protein occurs through plant homeodomains and is VHL mutation dependent. Cancer Res 2004; 64:1278-86. 32. Zhou MI, Foy RL, Chitalia VC, et al. Jade-1, a candidate renal tumor suppressor that promotes apoptosis. Proc Natl Acad Sci U S A 2005;102:11035-40. 33. Cohen AJ, Li FP, Berg S, et al. Hereditary renal-cell carcinoma associated with a chromosomal translocation. N Engl J Med 1979; 301:592-5. 34. Beck SD, Patel MI, Snyder ME, et al. Effect of papillary and chromophobe cell type on disease-free survival after nephrectomy for renal cell carcinoma. Ann Surg Oncol 2004;11:71-7. 35. Motzer RJ, Bacik J, Mariani T, Russo P, Mazumdar M, Reuter V. Treatment outcome and survival associated with metastatic renal cell carcinoma of non-clear-cell histology. J Clin Oncol 2002;20:2376-81. 36. Delahunt B, Eble JN. Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol 1997;10:537-44. 37. Gunawan B, von Heydebreck A, Fritsch T, et al. Cytogenetic and morphologic typing of 58 papillary renal cell carcinomas: evidence for a cytogenetic evolution of type 2 from type 1 tumors. Cancer Res 2003;63:6200-5. 38. Zbar B, Glenn G, Lubensky I, et al. Hereditary papillary renal cell carcinoma: clinical studies in 10 families. J Urol 1995;153: 907-12. 39. Lubensky IA, Schmidt L, Zhuang Z, et al. Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype. Am J Pathol 1999;155:517-26. 40. Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET protooncogene in papillary renal carcinomas. Nat Genet 1997;16:68-73. 41. Jeffers M, Schmidt L, Nakaigawa N, et al. Activating mutations for the met tyrosine kinase receptor in human cancer. Proc Natl Acad Sci U S A 1997;94:11445-50. 42. Zhuang Z, Park WS, Pack S, et al. Trisomy 7-harbouring non-random duplication of the mutant MET allele in hereditary papillary renal carcinomas. Nat Genet 1998; 20:66-9. 43. Fischer J, Palmedo G, von Knobloch R, et al. Duplication and overexpression of the mutant allele of the MET proto-oncogene in multiple hereditary papillary renal cell tumours. Oncogene 1998;17:733-9. 44. Launonen V, Vierimaa O, Kiuru M, et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc Natl Acad Sci U S A 2001;98:3387-92. 45. Zbar B, Klausner R, Linehan WM. Studying cancer families to identify kidney cancer genes. Annu Rev Med 2003;54:217-33. 46. Tomlinson IP, Alam NA, Rowan AJ, et

al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet 2002;30:406-10. 47. Kiuru M, Launonen V, Hietala M, et al. Familial cutaneous leiomyomatosis is a twohit condition associated with renal cell cancer of characteristic histopathology. Am J Pathol 2001;159:825-9. 48. Vanharanta S, Buchta M, McWhinney SR, et al. Early-onset renal cell carcinoma as a novel extraparaganglial component of SDHB-associated heritable paraganglioma. Am J Hum Genet 2004;74:153-9. 49. Meloni AM, Dobbs RM, Pontes JE, Sandberg AA. Translocation (X;1) in papillary renal cell carcinoma: a new cytogenetic subtype. Cancer Genet Cytogenet 1993;65:1-6. 50. Sidhar SK, Clark J, Gill S, et al. The t(X;1)(p11.2;q21.2) translocation in papillary renal cell carcinoma fuses a novel gene PRCC to the TFE3 transcription factor gene. Hum Mol Genet 1996;5:1333-8. 51. Weterman MA, Wilbrink M, Geurts van Kessel A. Fusion of the transcription factor TFE3 gene to a novel gene, PRCC, in t(X;1)(p11;q21)-positive papillary renal cell carcinomas. Proc Natl Acad Sci U S A 1996; 93:15294-8. 52. Cheville JC, Lohse CM, Zincke H, Weaver AL, Blute ML. Comparisons of outcome and prognostic features among histologic subtypes of renal cell carcinoma. Am J Surg Pathol 2003;27:612-24. 53. Peyromaure M, Misrai V, Thiounn N, et al. Chromophobe renal cell carcinoma: analysis of 61 cases. Cancer 2004;100:140610. 54. Birt AR, Hogg GR, Dube WJ. Hereditary multiple fibrofolliculomas with trichodiscomas and acrochordons. Arch Dermatol 1977; 113:1674-7. 55. Weirich G, Glenn G, Junker K, et al. Familial renal oncocytoma: clinicopathological study of 5 families. J Urol 1998;160:33540. 56. Toro JR, Glenn G, Duray P, et al. BirtHogg-Dube syndrome: a novel marker of kidney neoplasia. Arch Dermatol 1999;135: 1195-202. 57. Zbar B, Alvord WG, Glenn G, et al. Risk of renal and colonic neoplasms and spontaneous pneumothorax in the Birt-HoggDube syndrome. Cancer Epidemiol Biomarkers Prev 2002;11:393-400. 58. Nickerson ML, Warren MB, Toro JR, 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 BirtHogg-Dube syndrome. Cancer Cell 2002;2: 157-64. 59. Khoo SK, Kahnoski K, Sugimura J, et al. Inactivation of BHD in sporadic renal tumors. Cancer Res 2003;63:4583-7. 60. da Silva NF, Gentle D, Hesson LB, Morton DG, Latif F, Maher ER. Analysis of the Birt-Hogg-Dube (BHD) tumour suppressor gene in sporadic renal cell carcinoma and colorectal cancer. J Med Genet 2003;40:820-4.

61. Walther MM, Choyke PL, Glenn G, et al. Renal cancer in families with hereditary renal cancer: prospective analysis of a tumor size threshold for renal parenchymal sparing surgery. J Urol 1999;161:1475-9. 62. Herring JC, Enquist EG, Chernoff A, Linehan WM, Choyke PL, Walther MM. Parenchymal sparing surgery in patients with hereditary renal cell carcinoma: 10-year experience. J Urol 2001;165:777-81. 63. 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;17:2530-40. 64. Motzer RJ, Bacik J, Schwartz LH, et al. Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma. J Clin Oncol 2004;22:454-63. 65. Zisman A, Pantuck AJ, Dorey F, et al. Improved prognostication of renal cell carcinoma using an integrated staging system. J Clin Oncol 2001;19:1649-57. 66. Patard JJ, Kim HL, Lam JS, et al. Use of the University of California Los Angeles integrated staging system to predict survival in renal cell carcinoma: an international multicenter study. J Clin Oncol 2004;22:3316-22. 67. Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell cancer. N Engl J Med 2001;345:1655-9. 68. Mickisch GH, Garin A, van Poppel H, de Prijck L, Sylvester R. Radical nephrectomy plus interferon-alfa-based immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet 2001;358:966-70. 69. Novick AC. Nephron-sparing surgery for renal cell carcinoma. Annu Rev Med 2002; 53:393-407. 70. Clayman RV, Kavoussi LR, Soper NJ, et al. Laparoscopic nephrectomy: initial case report. J Urol 1991;146:278-82. 71. Gill IS, Matin SF, Desai MM, et al. Comparative analysis of laparoscopic versus open partial nephrectomy for renal tumors in 200 patients. J Urol 2003;170:64-8. 72. Zlotta AR, Wildschutz T, Raviv G, et al. Radiofrequency interstitial tumor ablation (RITA) is a possible new modality for treatment of renal cancer: ex vivo and in vivo experience. J Endourol 1997;11:251-8. 73. Uchida M, Imaide Y, Sugimoto K, Uehara H, Watanabe H. Percutaneous cryosurgery for renal tumours. Br J Urol 1995;75: 132-6. 74. Motzer RJ. Renal cell carcinoma: a priority malignancy for development and study of novel therapies. J Clin Oncol 2003;21: 1193-4. 75. Yagoda A, Abi-Rached B, Petrylak D. Chemotherapy for advanced renal-cell carcinoma: 1983-1993. Semin Oncol 1995;22: 42-60. 76. Gollob JA, Upton MP, DeWolf WC, Atkins MB. Long-term remission in a patient with metastatic collecting duct carcinoma

n engl j med 353;23

www.nejm.org

december 8, 2005

2489

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

medical progress

treated with taxol/carboplatin and surgery. Urology 2001;58:1058. 77. Peyromaure M, Thiounn N, Scotte F, Vieillefond A, Debre B, Oudard S. Collecting duct carcinoma of the kidney: a clinicopathological study of 9 cases. J Urol 2003;170: 1138-40. 78. Milowsky MI, Rosmarin A, Tickoo SK, Papanicolaou N, Nanus DM. Active chemotherapy for collecting duct carcinoma of the kidney: a case report and review of the literature. Cancer 2002;94:111-6. 79. Hanada K, Yewdell JW, Yang JC. Immune recognition of a human renal cancer antigen through post-translational protein splicing. Nature 2004;427:252-6. 80. Small EJ, Motzer RJ. Interferon for renal cell carcinoma. In: Belldegrun A, Ritchie AWS, Figlin RA, Oliver RTD, Vaughan ED, eds. Renal and adrenal tumors. New York: Oxford University Press, 2003:381-7. 81. Yang JC, Sherry RM, Steinberg SM, et al. Randomized study of high-dose and lowdose interleukin-2 in patients with metastatic renal cancer. J Clin Oncol 2003;21: 3127-32. 82. Fisher RI, Rosenberg SA, Fyfe G. Longterm survival update for high-dose recombinant interleukin-2 in patients with renal cell carcinoma. Cancer J Sci Am 2000;6:Suppl 1: S55-S57. 83. Clark JI, Atkins MB, Urba WJ, et al. Adjuvant high-dose bolus interleukin-2 for patients with high-risk renal cell carcinoma: a cytokine working group randomized trial. J Clin Oncol 2003;21:3133-40. 84. Childs R, Chernoff A, Contentin N, et al. Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation. N Engl J Med 2000;343:750-8. 85. Srinivasan R, Barrett J, Childs R. Allogeneic stem cell transplantation as immunotherapy for nonhematological cancers. Semin Oncol 2004;31:47-55. 86. Holtl L, Zelle-Rieser C, Gander H, et al. Immunotherapy of metastatic renal cell car-

cinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res 2002; 8:3369-76. 87. Su Z, Dannull J, Heiser A, et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res 2003;63:2127-33. 88. Marten A, Renoth S, Heinicke T, et al. Allogeneic dendritic cells fused with tumor cells: preclinical results and outcome of a clinical phase I/II trial in patients with metastatic renal cell carcinoma. Hum Gene Ther 2003;14:483-94. 89. Avigan D, Vasir B, Gong J, et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses. Clin Cancer Res 2004;10:4699-708. 90. Mandriota SJ, Turner KJ, Davies DR, et al. HIF activation identifies early lesions in VHL kidneys: evidence for site-specific tumor suppressor function in the nephron. Cancer Cell 2002;1:459-68. 91. Bui MH, Seligson D, Han KR, et al. Carbonic anhydrase IX is an independent predictor of survival in advanced renal clear cell carcinoma: implications for prognosis and therapy. Clin Cancer Res 2003;9:802-11. 92. Luiten RM, Coney LR, Fleuren GJ, Warnaar SO, Litvinov SV. Generation of chimeric bispecific G250/anti-CD3 monoclonal antibody, a tool to combat renal cell carcinoma. Br J Cancer 1996;74:735-44. 93. Steffens MG, Boerman OC, de Mulder PH, et al. Phase I radioimmunotherapy of metastatic renal cell carcinoma with 131Ilabeled chimeric monoclonal antibody G250. Clin Cancer Res 1999;5:3268s-3274s. 94. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an antivascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 2003;349:427-34. 95. Knebelmann B, Ananth S, Cohen HT, Sukhatme VP. Transforming growth factor alpha is a target for the von Hippel-Lindau

tumor suppressor. Cancer Res 1998;58:22631. 96. Everitt JI, Walker CL, Goldsworthy TW, Wolf DC. Altered expression of transforming growth factor-alpha: an early event in renal cell carcinoma development. Mol Carcinog 1997;19:213-9. 97. de Paulsen N, Brychzy A, Fournier MC, et al. Role of transforming growth factor-alpha in von HippelLindau (VHL)(/) clear cell renal carcinoma cell proliferation: a possible mechanism coupling VHL tumor suppressor inactivation and tumorigenesis. Proc Natl Acad Sci U S A 2001;98:1387-92. 98. Rowinsky EK, Schwartz GH, Gollob JA, et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol 2004;22:3003-15. 99. Arteaga CL. Overview of epidermal growth factor receptor biology and its role as a therapeutic target in human neoplasia. Semin Oncol 2002;29:Suppl:3-9. 100. Hidalgo M, Siu LL, Nemunaitis J, et al. Phase I and pharmacologic study of OSI774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J Clin Oncol 2001; 19:3267-79. 101. Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 2004;22:909-18. 102. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of nonsmall-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129-39. 103. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497-500.
Copyright 2005 Massachusetts Medical Society.

apply for jobs electronically at the nejm careercenter


Physicians registered at the NEJM CareerCenter can apply for jobs electronically using their own cover letters and CVs. You can keep track of your job-application history with a personal account that is created when you register with the CareerCenter and apply for jobs seen online at our Web site. Visit www.nejmjobs.org for more information.

2490

n engl j med 353;23

www.nejm.org

december 8, 2005

The New England Journal of Medicine Downloaded from nejm.org on March 4, 2012. For personal use only. No other uses without permission. Copyright 2005 Massachusetts Medical Society. All rights reserved.

You might also like