Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Review Article

Renal Cancer Subtypes: Should We Be Lumping or


Splitting for Therapeutic Decision Making?
Scott M. Haake, MD; and W. Kimryn Rathmell, MD, PhD

The treatment of advanced renal cell carcinoma has posed a challenge for decades, in part because of common themes related to in-
trinsic resistance to cytotoxic chemotherapy and the obscure biology of these cancer types. Forward movement in the treatment of
the renal cell carcinomas thus can be approached in 2 ways: by splitting the tumor types along histologic and molecular features, in
the hopes of coupling highly precision-focused therapy on a subset of patients who have disease with the most potential for benefit;
or by lumping the various biologies and histologies together, to include the rarer renal cell carcinoma types with the more common
types. The former strategy satisfies the desire for customized precision in treatment delivery, whereas the latter strategy allows clini-
cians to offer a wider therapeutic menu in a set of diseases we are continuing to learn about on a physiologic and molecular level.
Cancer 2017;123:200-9. V C 2016 American Cancer Society.

KEYWORDS: clear cell, molecular subtypes, papillary, renal cell carcinoma.

INTRODUCTION
The study of biology has been propelled forward over the years by field-shifting technologic advances that bring scientific
details into greater resolution. These advances have revolutionized multiple fields, including cancer biology. Often, the
perspectives that these technologies provide result in the division of a seemingly homogeneous cohort into multiple fur-
ther, carefully defined subtypes. A prime example is lung cancer. Classically, lung cancer was subdivided into small cell
and nonsmall cell types based on histologic appearance. Subsequently, nonsmall cell lung cancer was further divided into
distinct histologic subtypes, including adenocarcinoma, squamous, and large cell, although treatment with cisplatin-based
doublets was long held as the standard of care for all of them.1 Adenocarcinoma of the lung is now further subdivided
based on the presence or absence of driver mutations. Given the development of potent inhibitors targeting the protein
products of these driver mutations, these molecularly defined subtypes of adenocarcinoma now split the disease along mu-
tation lines to guide therapeutic decisions. For example, tumors harboring epidermal growth factor receptor (EGFR)
mutations are particularly sensitive to EGFR inhibitors like erlotinib.2 Thus, the technology-driven subtyping or
“splitting” of cancers has directly impacted treatment paradigms.
The EGFR lung adenocarcinoma breakthrough is but one of several such examples in oncology in which the molecu-
lar features of a patient’s tumor directly dictates therapeutic decisions. The success of these select examples, coupled with
the momentum stemming from advanced technologies in molecular biology, has fueled enthusiasm for precision medi-
cine. Precision, also referred to as personalized, cancer medicine, as defined by the National Cancer Institute, is the use of
specific information about a patient’s tumor to aid in diagnosis, planning treatment, assessing the effects of treatment, and
prognostication. To date, if cancer care is to pursue this model of medicine, then a path can be envisioned toward further
and further “splitting” of tumors. In fact, an extreme interpretation of personalized medicine is that no 2 patients (and
thus no 2 tumors) are identical. Furthermore, we now know that individual tumors are heterogeneous and constantly
evolving.3 Thus, “splitting” to the extreme will result in the identification of genomically and phenotypically distinct sub-
regions within an individual patient’s tumor.
Identifying individual tumors in individual patients and designing individualized treatment plans based on these
data are exciting and appealing. However, practically, such pursuits make the development of novel therapeutics, provider
education, and regulation significantly more difficult. In addition, given the finite size of the human genome, the number
of ways in which a cancer cell can program itself is immense and diverse but perhaps not infinite. Thus, it is not surprising

Corresponding author: W. Kimryn Rathmell, MD, PhD, Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center,
2220 Pierce Avenue, 777 PRB, Nashville, TN 37232; Fax: (615) 343-2551; kimryn.rathmell@vanderbilt.edu

Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.

Both authors contributed equally to this work.

DOI: 10.1002/cncr.30314, Received: May 9, 2016; Revised: July 15, 2016; Accepted: July 18, 2016, Published online November 14, 2016 in Wiley Online Library
(wileyonlinelibrary.com)

200 Cancer January 15, 2017


Splitting and Lumping in RCC Treatment/Haake and Rathmell

that the pursuit of “personalized cancer medicine” has tures, and even response to treatments can be very differ-
witnessed examples of the pendulum swinging away from ent among the subtypes.
ever more subtypes or “splitting” of tumors and toward a
system, in which tumors that share a certain molecular
Histologic Subtypes of RCC
feature are “lumped” together for study purposes, includ- Clear cell RCC
ing drug development. For example, it was discovered The most common histologic subtype of RCC is the clear
that a subgroup of esophagogastric adenocarcinoma over- cell variant, representing from 70% to 75% of adult
expressed the receptor tyrosine kinase human endothelial RCCs. These tumors tend to be vascular and can have
receptor 2 (HER2). HER2 is also overexpressed in breast macroscopic evidence of hemorrhage and necrosis, as as-
cancer, in which multiple agents that target this specific tutely described by early pathologists.5 Microscopically,
protein have been tested and proven effective. Indeed, these tumors are characterized by clear cytoplasm because
when treated with targeted drugs, the metastatic HER2- of an abundant accumulation of glycogen and lipids.13
overexpressing esophagogastric adenocarcinomas demon- Certain molecular hallmarks have come to define clear
strated a response.4 Thus, in this example, HER2- cell RCC and are now thought to be critical to its patho-
overexpressing tumors can potentially be “lumped” to- genesis. For example, loss of function of the von Hippel
gether with the use of similar drugs. This system of Lindau (VHL) protein or dysregulation of its downstream
“lumping” of otherwise very dissimilar tumors, in this effectors is nearly ubiquitous in clear cell RCC. The VHL
case breast and gastric adenocarcinomas, with distinct gene is a classic 2-hit tumor suppressor, as demonstrated
sites of origin, epidemiology, and pathogenesis can be by the autosomal dominant germline hereditary cancer
viewed as the antithesis to the parsing of similar tumors risk syndrome VHL disease.14 One copy is either mutated
into further and further subdivisions. In this review, our or silenced in up to 90% of sporadic clear cell RCC
objectives are to describe these seemingly antagonistic tumors,15 whereas the second copy is typically lost
trends specifically in renal cell carcinoma (RCC) and to through chromosome 3p deletions.16 Under physiologic
explore how these trends may impact patient care in the
and normoxic conditions, the VHL protein is an impor-
near future.
tant component of an E3 ubiquitin ligase complex that
targets the hypoxia-inducible factors (HIFs) for
THE EVOLUTION OF RCC
proteasome-mediated degradation. However, biallelic loss
SUBCLASSIFICATION: A PATH OF
of VHL allows for the inappropriate stabilization of HIFs
“SPLITTING”
(irrespective of oxygen levels), which results in a proangio-
Introduction to RCC Histologic Subtypes
genic gene expression signature critical to clear cell RCC
It is now well accepted across the RCC community that
tumorigenesis.17 This “pseudohypoxic” response has
kidney tumors that look very different when examined
come to define clear cell RCC biology and distinguishes it
under magnification are distinct diseases with divergent
from other RCC histologies.
biology. This simple conclusion, which may seem self-
evident to the modern physician and investigator, actually
took decades of analysis and the molecular biology revolu- Papillary RCC
tion to become realized. For decades, most or all kidney Papillary RCC is the second most common histologic
tumors were lumped under a single title.5 Although other subtype of adult kidney cancer, representing about 15%
early pathologists described kidney tumors with papillary of cases. This variant of kidney cancer with papillary or
features,6 not until the 1970s was “papillary” RCC stud- tubulopapillary architecture was recognized early, as dis-
ied in a detailed manner with clinical and pathologic cor- cussed above.6 However, its recognition as a distinct sub-
relation.7 These and other studies marked a transition in type of kidney cancer gained traction, because it was
which investigators began to increasingly appreciate and recognized that these tumors had a distinct clinical course7
describe distinct histologic subtypes of kidney cancer. and molecular features relative to the more common clear
Thus, what were once collectively known as cell variant. Specifically, although 3p loss is nearly univer-
“hypernephromas” would eventually be “split” into at sal in clear cell RCC, early kidney cancer cytogenetic stud-
least 16 distinct subtypes through the application of vari- ies demonstrated that this chromosomal abnormality was
ous histologic and molecular criteria (Table 1).8-12 Enthu- absent in most papillary RCCs,18 thus further supporting
siasm for this system of splitting RCC into many subtypes the concept that papillary RCC was biologically and clini-
was fueled by recognition that core biology, clinical fea- cally distinct from clear cell RCC.

Cancer January 15, 2017 201


Review Article

TABLE 1. 2016 World Health Organization Classification of Kidney Tumorsa

Kidney Cancer Subtype Description

Clear cell RCC VHL mutated, most common adult kidney cancer
Multiloculated cystic clear cell renal cell neoplasm of low Low grade, little or no recurrence or metastatic potential
malignant potential
Papillary RCC Second most common adult kidney cancer, subdivided into types 1 and 2
Hereditary leiomyomatosis RCC syndrome-associated RCC Familial kidney cancer syndrome with germline mutations in fumarate
hydratase
Chromophobe RCC Lower malignant potential, associated with Bitt-Hogg-Dube syndrome
Collecting duct carcinoma Rare, poor prognosis
Renal medullary carcinoma Associated with sickle cell trait, young patients, poor prognosis
MiT family translocation RCC Young patients, poor prognosis (Choueiri 20109), rare
Succinate dehydrogenase-deficient RCC Young patients, germline SDH mutations, good prognosis (Shen 200710)
Mucinous tubular and spindle cell carcinoma Rare, good prognosis, female predominance
Tubulocystic RCC Rare, good prognosis
Acquired cystic disease-associated RCC Associated with end-stage renal disease, good prognosis
Clear cell papillary RCC May occur in end-stage renal disease and VHL disease, good prognosis
RCC, unclassified
Papillary adenoma Low grade,  1.5 cm, unencapsulated, good prognosis
Oncocytoma Benign, well differentiated, mitochondrial accumulation (Dechet 1999,11
Zerban 198712)

Abbreviations: MiT, microphthalmia transcription factor; RCC, renal cell carcinoma; SDH, succinate dehydrogenase; VHL, von Hippel Lindau.
a
Adapted from Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumors of the Urinary System and Male Genital
Organs—Part A: Renal, Penile and Testicular Tumors. Eur Urol. 2016;70:93-105.8

Later, as larger papillary RCC cohorts were stud- Thus, type 1 and 2 papillary RCCs have become fully
ied, it became evident that this disease could be further recognized not only as distinct from clear cell RCC
subdivided on the basis of histology into 2 subtypes. but also as distinct from each other.
Type 1 papillary RCC was characterized by papillary
or tubular structures covered by small, basophilic cells Other histologic subtypes of RCC
with clear cytoplasm that have small nuclei and incon- Although clear cell and papillary RCC are the 2 most
spicuous nucleoli; whereas type 2 papillary RCC was common subtypes of kidney cancer, representing 80% to
characterized by larger cells with eosinophilic cyto- 90% of cases, others have been described with increasingly
plasm, pseudostratification of cells, and prominent nu- robust molecular annotation. Chromophobe RCC repre-
cleoli.19 Moreover, familial studies reveal that germline sents approximately 5% of adult kidney cancer and micro-
mutations contribute specifically to inherited forms of scopically features distinct cell borders and voluminous
both disease types—hereditary papillary renal carcino- cytoplasm.13 Two histologic subtypes have been described
ma syndrome with germline mutations in MET proto- even for this rare tumor entity: classic and eosinophilic.
oncogene, (MET), giving rise to type 1 tumors,20,21 Classic chromophobe has a pale cytoplasm and is more
and hereditary leiomyomatosis and RCC syndrome likely to demonstrate the characteristic monosomy chro-
with germline mutations in fumarate hydratase, giving mosomal pattern often associated with chromophobe
rise to type 2 tumors. Large genomic studies have RCC, which includes loss of most or all of an entire copy
demonstrated strong molecular distinctions between of chromosome 1, 2, 6, 10, 13, 17, and often chromo-
sporadic papillary type 1 and type 2 RCC. Type 1 dis- some 21.23 In addition, the classic chromophobe subtype
ease is characterized by molecular events that result in contains more somatic mutations, including mutations in
amplification of signaling from the hepatocyte growth tumor protein 53 (TP53) and phosphatase and tensin ho-
factor receptor MET, most commonly gain-of-function molog (PTEN). In addition, the eosinophilic subtype has
somatic mutations in the tyrosine kinase domain22; dense, pink cytoplasm because of mitochondrial accumu-
whereas type 2 disease itself is more heterogeneous, lation. It is noteworthy that the eosinophilic subtype is
with loss of cyclin-dependent kinase 2a (CDKN2a) enriched with mutations in mitochondrial genes, specifi-
function, activation of the nuclear factor (erythroid-de- cally those corresponding to complex 1 of the electron
rived 2)-like 2 (NRF2) antioxidant response element transport chain.23 Therefore, like other RCC histologies,
pathway, and mutations in chromatin regulators,22 it has been demonstrated that variants of chromophobe
allowing the type 2 papillary tumors to be further sub- RCC have distinct molecular features, thus justifying their
divided into at least 3 additional subclassifications. classification as distinct cancers.

202 Cancer January 15, 2017


Splitting and Lumping in RCC Treatment/Haake and Rathmell

To extend the splitters mindset, each of these sub- tubule-derived, chromophobe RCCs rarely metastasize,
types can be further affected by the involvement of sarco- and patients do well, whereas tumors that arise from the
matoid histology. So-called sarcomatoid tumors renal medulla (namely, collecting duct and medullary car-
sometimes have such overwhelming sarcomatoid features cinomas) represent extremely aggressive disease with a
that the underlying primary tumor type is unidentifiable. poor overall prognosis. In 1 series of renal medullary carci-
Recent genetic analysis has demonstrated that a common noma, the mean survival was a mere 4 months.29 Thus,
profile of mutations can contribute to this aggressive his- precise histologic subclassification can provide a profound
tology,24 and combination chemotherapy regimens incor- contribution to the understanding of an individual
porating targeted therapies can be used for this splinter patient’s prognosis.
group.25 The clinical relevance of RCC histologic subtypes
Clear cell, papillary, and chromophobe RCCs have a informs not only prognosis but, in some instances, can
commonality, in that it is believed that they all originate guide treatment decisions. For example, in the early stud-
in the renal cortex: clear cell and papillary RCCs originate ies of immunotherapy in RCC using high-dose interleu-
in the proximal convoluted tubule, and chromophobe kin-2, researchers observed that the response rate was
RCCs originate in the distal convoluted tubule.16,22,23 In superior for patients with clear cell RCC relative to those
contrast, other kidney tumors originate outside the renal with nonclear cell RCC subtypes for reasons that remain
cortex and include collecting duct (or Bellini tumors) and poorly understood.31 Those observations also have been
medullary carcinomas. Both are thought to arise from the validated in more recent prospective analyses.32
renal medulla. Relatively less is known about these rare However, the development of a new class of medica-
kidney cancer subtypes. Collecting duct carcinomas were tions would cause a dramatic shift in the design of pro-
recognized as a distinct clinical diagnosis in the 1980s and spective RCC trials. The molecular hallmark of clear cell
are characterized by a tubulopapillary pattern and desmo- RCC (but not of other subtypes), as discussed above, is
plastic stroma26; in some cases, they are difficult to distin- biallelic loss of VHL with resultant HIF stabilization and
guish histologically from urothelial carcinomas. However, inappropriate hypoxia signaling, including profound
biologically, they may be very different from urothelial increases in angiogenesis signaling.17 This signaling
carcinomas, because the 2 cancers contain very different includes increased production of vascular epithelial
patterns of chromosomal gain and loss.27 Medullary carci- growth factor (VEGF) and subsequent signaling through
nomas most commonly arise in young patients with sickle the VEGF receptors (VEGFRs), a class of receptor tyro-
cell trait.28,29 Even less is known about their molecular bi- sine kinases. Signaling through the VEGFRs, especially
ology, although mutations in SMARCB1 (SWI/SNF-re- on endothelial cells, supports the tumor vasculature and
lated, matrix-associated, actin-dependent regulator of growth. When a novel class of small-molecule inhibitors
chromatin, subfamily B, member 1) appear to dominate was designed to target VEGFRs, the large phase 3 trials
their genetic profile,30 rendering them further distinct typically included a “clear cell component” as an inclusion
from the other histologic subtypes described. criterion for describing the histology.33,34 Those trials
established the VEGFR inhibitors as the new standard
Clinical significance of histologic subtypes of RCC first-line therapy for metastatic clear cell RCC. The exclu-
Although it is clear that several histologic subtypes exist sion of nonclear cell RCC was justified, because it was pre-
within the RCC spectrum, and detailed molecular studies dicted that these patients with VHL-intact disease would
have shed light on their divergent biology, enthusiasm for not obtain as much benefit from this class of drugs. Thus,
“splitting” this disease has been fueled by the clinical rele- for some time, it was unclear whether patients with non-
vance of the subtypes. Specifically, the histologic subtypes clear cell RCC benefited from VEGFR inhibitors.
can vary greatly in terms of prognosis. For example, ini- Attempts to design VEGFR inhibitors with an increased
tially, it seemed that papillary RCC had a favorable prog- target spectrum tailored to papillary RCC biology has had
nosis relative to clear cell RCC.7 However, with the mixed results. Specifically, foretinib has activity against
emergence of histologic papillary RCC subtypes, the story both MET and VEGFR and was tested in a phase 2 trial
became more complicated. It is now clear that patients for patients with papillary RCC.35 Although foretinib did
with MET-driven, papillary RCC type 1 tumors have a fa- have activity, the most robust responses were observed in
vorable prognosis, and most represent early stage disease; patients who had germline MET mutations (ie, most like-
whereas patients with papillary RCC type 2 tumors have a ly a small subset of those with papillary type 1 tumors).
high rate of metastasis and poor overall survival.22 Distal However, novel combinations like bevacizumab and

Cancer January 15, 2017 203


Review Article

erlotinib, which target angiogenesis and glucose transport, tion, although it has emerged as a strong prognostic
respectively, in the highly glycolytic fumarate hydratase indicator in numerous studies.40,42 Other investigators
(FH)-mutant papillary type 2 group, are being carefully have successfully merged gene expression data with
examined.36 Thus, in the antiangiogenesis era, “splitting” metabolomics profiling to parse clear cell RCC into mul-
RCC on the basis of histology has had a dramatic impact tiple molecular subtypes, including an aggressive cohort
on clinical trial design. with increased glutathione metabolism.43
Finally, building off the results from large-scale ge-
Molecular Subtypes of RCC nome sequencing projects, another schema has emerged
Given the importance of histologic subclassification in that focuses on 2 of the more commonly mutated genes
prognosis and therapy, investigators have been aggressive in clear cell RCC: protein polybromo-1 (PBRM1 [also
in identifying molecular subtypes in an attempt to build called BAP180]) and BRCA1-associated protein 1
on this success. Although some other nonclear cell RCC (BAP1).16 Both genes are involved in chromatin regula-
histologic subtypes, such as papillary and chromophobe tion, and mutations in these genes are typically mutually
RCC, can be further subdivided based on histology, clear exclusive. By using immunohistochemistry assays that
cell RCC is relatively homogenous. Because clear cell evaluate protein expression and are known to reliably
RCC represents the largest bulk of patient burden, there correlate with mutation status, large clinically annotated
has been great interest in identifying molecular subtypes tissue microarrays were used to segregate clear cell RCC
that can further inform prognosis, biology, and drug de- tumors into 1 of 4 categories: tumors that express both
velopment. One such example emerged when investi- PBRM1 and BAP1 (40%), BAP1-positive tumors
gators examined patterns of HIF staining in 160 sporadic (49%), PBRM1-positive tumors (9%), and tumors that
human clear cell RCC tumors and split the tumors into 3 are negative for both proteins (2%).44 The best prognosis
groups: VHL wild-type, tumors that were positive for was in the group that expressed both PBRM1 and BAP1
both HIF-1a and HIF-2a (H1H2), and tumors that were (and thus was expected to lack mutations in these genes),
positive only for HIF-2a (H2).37 The H1H2 tumors whereas the worst prognosis was associated with the
exhibited increased angiogenesis as well as increased pro- group that lacked expression of both proteins (and thus
tein kinase B/mammalian target of rapamycin (AKT/ were expected to be doubly mutated). Therefore, by us-
mTOR) and mitogen-activated protein kinase 1 (ERK/ ing several powerful molecular platforms, various sche-
MAPK1) signaling, whereas the H2 tumors demonstrated mas to identify prognostically significant molecular
increased c-Myc activity and enhanced proliferation, fea- subtypes of clear cell RCC have emerged, possibly indi-
tures that could impact therapy effectiveness but remain cating a future in which we can deliver personalized ther-
untested. apy based on these features.
An alternative strategy for splitting clear cell RCC Until recently, molecular subtyping of nonclear cell
was developed from gene expression data. By using RCC tumors was relatively unexplored. However, the
genome-wide messenger RNA microarray data, unsuper- Cancer Genome Atlas working group has used multiple
vised clustering identified 2 dominant clear cell RCC sub- platforms to molecularly annotate papillary RCC.22 Their
types called ccA and ccB.38 Biologically, these subtypes integrated analysis identified 4 major molecular subtypes
are distinct: ccA tumors exhibit increased angiogenesis, of papillary RCC. One subtype included predominantly
whereas ccB tumors demonstrate increased transforming papillary RCC type 1 tumors that frequently exhibit
growth factor b (TGF-b) and epithelial-to-mesenchymal MET activation, whereas the other 3 subtypes included
signaling. Perhaps most important, this classification rep- mostly papillary RCC type 2 tumors, thus reflecting the
resents clinically distinct subtypes, with ccB tumors dem- molecular heterogeneity of this subtype. Among these 3
onstrating increased tumor size, grade, and rate of molecular subtypes of papillary RCC type 2 tumors, 1
metastasis as well as decreased recurrence-free and overall was notable for its extremely poor prognosis and young
survival in multiple data sets.16,39,40 In addition, these age of onset. Termed CpG island methylator phenotype
molecular subtypes feature distinct metabolic patterns, or CIMP, these tumors were identified by their wide-
with the aggressive ccB tumors demonstrating increased spread pattern of genomic hypermethylation. They also
glucose uptake on imaging.41 However, the other clear frequently had CDKN2a silencing and included all
finding from that study was that individual tumors can tumors with FH germline mutations, a uniquely aggres-
harbor both ccA and ccB components, making this classi- sive subgroup originally identified through the association
fication strategy difficult for informing therapeutic selec- with the familial syndrome of hereditary leiomyomatosis

204 Cancer January 15, 2017


Splitting and Lumping in RCC Treatment/Haake and Rathmell

and Renal Cell Cancer (HLRCC).45 Thus, although pap- properties. For example, investigators identified 5 patients
illary type 1 RCC was relatively homogeneous, papillary with RCC who had an “exceptional” response to mTOR
type 2 RCC displayed far more molecular heterogeneity, inhibitors (median duration of response, 28 months).52
and multiple molecular subtypes were identified. One of those patients had an RCC that could not be histo-
logically subtyped, whereas the other 4 had RCCs with
THE CASE FOR LUMPING clear cell histology. Multiple specimens were obtained
Although it is relatively simpler (in the mind of an from each patient, and genomic sequencing was per-
avowed splitter) to apply definitions with greater and formed. It is noteworthy that nearly all of the samples
greater specificity, a strong rational case can be made for identified activating alterations of the mTOR pathway.
lumping strategies, particularly given the strong rein- Therefore, the use of genomic sequencing to identify a
forcement that has come from decades of trials that effec- subgroup of patients who are more likely to respond to
tively lump all of the RCCs together. This strategy mTOR inhibitors is an exciting approach to RCC thera-
simplifies enrollment in studies and provides treatment peutics. This strategy is becoming increasingly popular,
options for a greater range of patients. Although the non- and the use of basket-style trials supports even broader
clear cell histologies always make up a tiny minority of lumping of disease types according to underlying genomic
cases, clinical trials have not consistently demonstrated analysis rather than according to tissue of origin.53
inferiority for this practice.46 Moreover, despite the ex- Previously, the defining biologic features of the ma-
tensive subclassification strategies available, histological- jor subtypes of RCC were highly distinct, as detailed
ly, it is still not uncommon for tumors to be assigned a above, which warranted the strong concern against lump-
diagnosis of “unclassified” RCC or “renal epithelial tu- ing strategies. However, recent high-throughput sequenc-
mor not otherwise specified.” This is because of the wide ing efforts have revealed new themes of somatic
variation in histologic features even within subtypes and alterations that cross histologic boundaries. In addition to
a current lack of effective molecular tools to augment inactivating mutations in PTEN and related members of
pathologic assignment. Indeed, studies demonstrate the the tuberous sclerosis complex (TSC)/mTOR signaling
activity of standard frontline VEGFR-targeted therapies axis, mutations in chromatin modifiers, such as SET do-
in papillary RCC47 and even in chromophobe RCC.48 main containing 2 (SETD2) and PBRM1/BAF180, are
Even including the sarcomatoid variant classification, also prevalent across the RCC spectrum.16,22,23 Thus, as
data suggesting that VEGFR-targeted therapies can be ef- new therapeutics emerge that target cells that have specific
fective49 reinforce the lumping paradigm. In addition, somatic dependencies, the options to lump tumors
because these various histologies are treated with similar according to precision genomic strategies will only
therapies, it is possible that convergent resistance mecha- increase.
nisms could emerge that likewise could be treated For better or worse, the newest therapies in this dis-
similarly. ease rely on targets for which we have very little knowledge
The same was true for some early pivotal trials with to guide their use. The immunotherapies—formerly
another class of medications: the mTOR inhibitors. Spe- dominated by high-dose interleukin-2, which was widely
cifically, in a first-line trial comparing the mTOR inhibi- accepted to be exclusively active in clear cell RCC as well
tor temsirolimus versus interferon-a or the combination as melanoma—are now rapidly changing form with the
in patients with metastatic RCC, nearly 20% of enrollees advent of programmed death 1 (PD-1)-inhibitory, PD-1
had nonclear cell RCC histologies.50 That trial was limit- ligand (PD-L1)-inhibitory, and cytotoxic T-lymphocyte–
ed to patients who had poor-prognosis disease, as defined associated protein 4 (CTLA-4)-inhibitory antibodies.
by clinical criteria.51 Because temsirolimus proved to be These therapies hold great promise for deep, sustainable
superior to interferon-a and equivalent to the combina- responses in a surprisingly broad array of tumor types.
tion, it has emerged as an accepted therapy for patients The pivotal, positive phase 3 trial of nivolumab versus
who have poor-risk primary RCC regardless of histologic everolimus in previously treated patients that resulted in
subtype. the approval of nivolumab was limited to patients who
Although many of the studies described above have had clear cell RCC,54 although the indication was deliv-
focused primarily on defining disease biology without ered broadly to patients who had advanced, previously
much consideration to therapeutics, in fact, the biologic treated RCC. In fact, it has been demonstrated that PD-
underpinnings can make the case strongly in favor of L1 is expressed on both clear cell and nonclear cell RCC
lumping—as in lumping tumors according to biologic tumors.55,56 Despite this observation, many ongoing

Cancer January 15, 2017 205


Review Article

Figure 1. Representation of the 2 schools of thought. Top: Splitting demonstrates the narrower definitions obtained by applying
increasingly strict criteria. Possible treatment strategies for subtypes are listed. Bottom: Lumping combines groups according to
similar features or, in this example, according to therapies that have demonstrated benefit to the group. *Although programmed
death ligand 1 (PD-L1) staining has been observed in both clear cell and nonclear cell renal cell carcinoma (RCC), the activity of
checkpoint inhibitors in nonclear cell RCC has not been well studied. However, PD-L1 staining is an important example of how
biomarkers for specific classes of drugs can cross over previously defined “splitting” schemas. BAP1 indicates BRCA1-associated
protein 1; ccA/ccB, dominant clear cell RCC subtypes; CD, collecting duct; CIMP, CpG island methylator phenotype; DCT, distal
convoluted tubule; H1, hypoxia-inducible factor 1a; H2, hypoxia-inducible factor 2a; mTOR, mammalian target of rapamycin; PCT,
proximal convoluted tubule; PRBM1, protein polybromo-1; anti-VEGF, antivascular endothelial growth factor.

206 Cancer January 15, 2017


Splitting and Lumping in RCC Treatment/Haake and Rathmell

prospective trials, including those using check-point integrate such new drugs with accurate classification
inhibitors, are following the pattern from the antiangio- schemes and annotation strategies for RCCs.
genesis era and limiting inclusion to those patients whose Although a detailed understanding of RCC biologic
tumors contain a clear cell component (eg, national clini- subtypes has undoubtedly led to a deeper understanding
cal trials NCT01472081 and NCT02575222). Thus, we of the diverse biology within this disease spectrum, and
may need to wait for retrospective analyses of off-study the development of matched therapies that target the driv-
patients or novel trial designs to address the question of er biology is arguably the best approach to obtaining the
the impact of histology on patient outcomes with check- ideals of precision medicine, difficulties ascertaining this
point inhibitors. For now, however, many tumors are ideal should not paralyze drug development. For example,
finding themselves lumped together as we search for clues the checkpoint inhibitors represent an exciting, novel
to understand the arbiters of response to this new class of therapeutic approach in RCC. This method of escaping
immune therapies. immune surveillance, including expression of PD-L1, is
used by many RCC subtypes, as mentioned above. Thus,
Conclusion as more is learned about this biology across the RCC spec-
RCC is no longer recognized as a single disease but, rath- trum, new “splitting” paradigms likely will emerge that
er, as a collection of related, yet biologically distinct, can- incorporate this important biology with what has been
cers. Through the use of detailed molecular studies, we previously described. However, currently, it is practical
know that these histologically divergent tumors are also and even essential that we “lump” tumors together based
biologically divergent, with unique mutational burdens, on this biology for purposes of biomarker development
gene expression patterns, proteomic profiles, and metabo- and clinical trial design.
lomic signatures. Even among populations with histologi-
cally indistinguishable tumors, molecular profiling has FUNDING SUPPORT
allowed us to parse out biologically unique subtypes. Scott M. Haake is supported by the Conquer Cancer Foundation
These molecular subtypes are clinically relevant, as evi- Young Investigator Award program and the American Urologic As-
denced by their dissimilar recurrence and survival rates. sociation Research Scholar program. W. Kimryn Rathmell is sup-
This work has led to new knowledge and insight into the ported by a grant from the National Institutes of Health
core biology of these tumors that is unparalleled in the his- (K24CA172355).
tory of the disease (Fig. 1).
Although our understanding of the biologic sub- CONFLICT OF INTEREST DISCLOSURES
types of RCC has progressed relatively quickly, the The authors made no disclosures.
pace of clinical discovery to meet these diseases with
precision approaches has lagged. There are several REFERENCES
1. Chemotherapy in non-small cell lung cancer: a meta-analysis using
possible explanations why the identification of more updated data on individual patients from 52 randomised clinical tri-
and more molecularly distinct subtypes of RCC has als. Non-small Cell Lung Cancer Collaborative Group [serial on-
not always resulted in immediate clinical advances. line]. BMJ. 1995;311:899-909.
2. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard
First, a lack of effective therapies is an obvious hin- chemotherapy as first-line treatment for European patients with ad-
drance to improving RCC care. Regardless of how vanced EGFR mutation-positive non-small-cell lung cancer (EUR-
TAC): a multicentre, open-label, randomised phase 3 trial. Lancet
many times RCC is “split” into biologically defined Oncol. 2012;13:239-246.
subtypes, if all subtypes are resistant to available 3. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity
and branched evolution revealed by multiregion sequencing. N Engl
therapies, then little clinical progress can be made. J Med. 2012;366:883-892.
By the same token, if targeted therapies lack specific- 4. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in com-
bination with chemotherapy versus chemotherapy alone for treat-
ity for driver events, as in the case of the VEGFR- ment of HER2-positive advanced gastric or gastro-oesophageal
targeted therapies, which target the tumor vascula- junction cancer (ToGA): a phase 3, open-label, randomised con-
ture, then it is not surprising that these treatments trolled trial. Lancet. 2010;376:687-697.
5. Jellinek EO. Grawitz Tumor of Kidney. Cal State J Med. 1904;2:78-
can have broad activities in various tumor types. 79.
Thus, we have yet to develop a highly potent and 6. Kelly AOJ. On hypernephromas of the kidney. Philadelphia Med J.
1898;11:223-233.
durable, targeted therapeutic paradigm that exploits 7. Mancilla-Jimenez R, Stanley RJ, Blath RA. Papillary renal cell
the deviant biology of the renal tumor subtypes. carcinoma: a clinical, radiologic, and pathologic study of 34 cases.
Cancer. 1976;38:2469-2480.
These precise tools are on the horizon, and it will be 8. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM.
in our best interest as a community to be ready to The 2016 WHO Classification of Tumors of the Urinary System

Cancer January 15, 2017 207


Review Article

and Male Genital Organs-pt A: Renal, Penile and Testicular els of response to treatment in patients with metastatic renal cell
Tumors. Eur Urol. 2016;70:93-105. carcinoma. Clin Cancer Res. 2015;21:561-568.
9. Choueiri TK, Lim ZD, Hirsch MS, et al. Vascular endothelial growth 33. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interfer-
factor-targeted therapy for the treatment of adult metastatic Xp11.2 on alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:
translocation renal cell carcinoma. Cancer. 2010;116:5219-5225. 115-124.
10. Shen SS, Ro JY, Tamboli P, et al. Mucinous tubular and spindle cell 34. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally ad-
carcinoma of kidney is probably a variant of papillary renal cell carci- vanced or metastatic renal cell carcinoma: results of a randomized
noma with spindle cell features. Ann Diagn Pathol. 2007;11:13-21. phase III trial. J Clin Oncol. 2010;28:1061-1068.
11. Dechet CB, Bostwick DG, Blute ML, Bryant SC, Zincke H. Renal 35. Choueiri TK, Vaishampayan U, Rosenberg JE, et al. Phase II and
oncocytoma: multifocality, bilateralism, metachronous tumor devel- biomarker study of the dual MET/VEGFR2 inhibitor foretinib in
opment and coexistent renal cell carcinoma. J Urol. 1999;162:40-42. patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31:
12. Zerban H, Nogueira E, Riedasch G, Bannasch P. Renal oncocytoma: 181-186.
origin from the collecting duct. Virchows Arch B Cell Pathol Incl Mol 36. Linehan WM, Rouault TA. Molecular pathways: Fumarate
Pathol. 1987;52:375-387. hydratase-deficient kidney cancer–targeting the Warburg effect in
13. Shuch B, Amin A, Armstrong AJ, et al. Understanding pathologic cancer. Clin Cancer Res. 2013;19:3345-3352.
variants of renal cell carcinoma: distilling therapeutic opportunities 37. Gordan JD, Lal P, Dondeti VR, et al. HIF-alpha effects on c-Myc
from biologic complexity. Eur Urol. 2015;67:85-97. distinguish 2 subtypes of sporadic VHL-deficient clear cell renal car-
14. Richards FM, Phipps ME, Latif F, et al. Mapping the Von Hippel- cinoma. Cancer Cell. 2008;14:435-446.
Lindau disease tumour suppressor gene: identification of germline 38. Brannon AR, Reddy A, Seiler M, et al. Molecular stratification of
deletions by pulsed field gel electrophoresis. Hum Mol Genet. 1993; clear cell renal cell carcinoma by consensus clustering reveals dis-
2:879-882. tinct subtypes and survival patterns. Genes Cancer. 2010;1:152-
15. Nickerson ML, Jaeger E, Shi Y, et al. Improved identification of von 163.
Hippel-Lindau gene alterations in clear cell renal tumors. Clin Can- 39. Brooks SA, Brannon AR, Parker JS, et al. ClearCode34: a prognostic
cer Res. 2008;14:4726-4734. risk predictor for localized clear cell renal cell carcinoma. Eur Urol.
16. Cancer Genome Atlas Research Network. Comprehensive molecular 2014;66:77-84.
characterization of clear cell renal cell carcinoma. Nature. 2013;499: 40. Haake SM, Brooks SA, Welsh E, et al. Patients with ClearCode34-
43-49. identified molecular subtypes of clear cell renal cell carcinoma repre-
17. Shen C, Kaelin WG Jr. The VHL/HIF axis in clear cell renal carci- sent unique populations with distinct comorbidities. Urol Oncol.
noma. Semin Cancer Biol. 2013;23:18-25. 2016;34:122.e1-e.7.
18. Kovacs G, Wilkens L, Papp T, de Riese W. Differentiation between 41. Brooks SA, Khandani AH, Fielding JR, et al. Alternate metabolic
papillary and nonpapillary renal cell carcinomas by DNA analysis. programs define regional variation of relevant biological features in
J Natl Cancer Inst. 1989;81:527-530. renal cell carcinoma progression. Clin Cancer Res. 2016;22:2950-
19. Delahunt B, Eble JN. Papillary renal cell carcinoma: a clinicopatho- 2959.
logic and immunohistochemical study of 105 tumors. Mod Pathol. 42. Buttner F, Winter S, Rausch S, et al. Survival prediction of
1997;10:537-544. clear cell renal cell carcinoma based on gene expression similari-
20. Zhuang Z, Park WS, Pack S, et al. Trisomy 7-harbouring non-ran- ty to the proximal tubule of the nephron. Eur Urol. 2015;68:
dom duplication of the mutant MET allele in hereditary papillary 1016-1020.
renal carcinomas. Nat Genet. 1998;20:66-69. 43. Hakimi AA, Reznik E, Lee CH, et al. An integrated metabolic atlas
21. Lubensky IA, Schmidt L, Zhuang Z, et al. Hereditary and sporadic of clear cell renal cell carcinoma. Cancer Cell. 2016;29:104-116.
papillary renal carcinomas with c-met mutations share a distinct 44. Joseph RW, Kapur P, Serie DJ, et al. Clear cell renal cell carcinoma
morphological phenotype. Am J Pathol. 1999;155:517-526. subtypes identified by BAP1 and PBRM1 expression. J Urol. 2016;
22. Linehan WM, Spellman PT, Ricketts CJ, et al. Comprehensive mo- 195:180-187.
lecular characterization of papillary renal-cell carcinoma. N Engl J 45. Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fuma-
Med. 2016;374:135-145. rate hydratase gene cause hereditary leiomyomatosis and renal cell
23. Davis CF, Ricketts CJ, Wang M, et al. The somatic genomic land- cancer in families in North America. Am J Hum Genet. 2003;73:
scape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26: 95-106.
319-330. 46. Motzer RJ, Michaelson MD, Redman BG, et al. Activity of
24. Malouf GG, Ali SM, Wang K, et al. Genomic characterization of re- SU11248, a multitargeted inhibitor of vascular endothelial growth
nal cell carcinoma with sarcomatoid dedifferentiation pinpoints re- factor receptor and platelet-derived growth factor receptor, in
current genomic alterations. Eur Urol. 2016;70:348-357. patients with metastatic renal cell carcinoma. J Clin Oncol. 2006;24:
25. Michaelson MD, McKay RR, Werner L, et al. Phase 2 trial of suni- 16-24.
tinib and gemcitabine in patients with sarcomatoid and/or poor-risk 47. Ravaud A, Oudard S, De Fromont M, et al. First-line treatment
metastatic renal cell carcinoma. Cancer. 2015;121:3435-3443. with sunitinib for type 1 and type 2 locally advanced or metastatic
26. Fleming S, Lewi HJ. Collecting duct carcinoma of the kidney. Histo- papillary renal cell carcinoma: a phase II study (SUPAP) by the
pathology. 1986;10:1131-1141. French Genitourinary Group (GETUG). Ann Oncol. 2015;26:1123-
27. Becker F, Junker K, Parr M, et al. Collecting duct carcinomas repre- 1128.
sent a unique tumor entity based on genetic alterations [serial on- 48. Choueiri TK, Plantade A, Elson P, et al. Efficacy of sunitinib and
line]. PLoS One. 2013;8:e78137. sorafenib in metastatic papillary and chromophobe renal cell carcino-
28. Davis CJ, Jr, Mostofi FK, Sesterhenn IA. Renal medullary carcinoma. ma. J Clin Oncol. 2008;26:127-131.
The seventh sickle cell nephropathy. Am J Surg Pathol. 1995;19:1-11. 49. Tannir NM, Plimack E, Ng C, et al. A phase 2 trial of sunitinib in
29. Swartz MA, Karth J, Schneider DT, Rodriguez R, Beckwith JB, patients with advanced non-clear cell renal cell carcinoma. Eur Urol.
Perlman EJ. Renal medullary carcinoma: clinical, pathologic, immu- 2012;62:1013-1019.
nohistochemical, and genetic analysis with pathogenetic implications. 50. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon
Urology. 2002;60:1083-1089. alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;
30. Calderaro J, Moroch J, Pierron G, et al. SMARCB1/INI1 inactiva- 356:2271-2281.
tion in renal medullary carcinoma. Histopathology. 2012;61:428-435. 51. Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A,
31. Upton MP, Parker RA, Youmans A, McDermott DF, Atkins MB. Ferrara J. Survival and prognostic stratification of 670 patients
Histologic predictors of renal cell carcinoma response to interleukin- with advanced renal cell carcinoma. J Clin Oncol. 1999;17:2530-
2-based therapy. J Immunother. 2005;28:488-495. 2540.
32. McDermott DF, Cheng SC, Signoretti S, et al. The high-dose alde- 52. Voss MH, Hakimi AA, Pham CG, et al. Tumor genetic analyses of
sleukin “select” trial: a trial to prospectively validate predictive mod- patients with metastatic renal cell carcinoma and extended benefit

208 Cancer January 15, 2017


Splitting and Lumping in RCC Treatment/Haake and Rathmell

from mTOR inhibitor therapy. Clin Cancer Res. 2014;20:1955- 55. Choueiri TK, Figueroa DJ, Fay AP, et al. Correlation of PD-L1 tu-
1964. mor expression and treatment outcomes in patients with renal cell
53. Redig AJ, Janne PA. Basket trials and the evolution of clinical trial carcinoma receiving sunitinib or pazopanib: results from COM
design in an era of genomic medicine. J Clin Oncol. 2015;33:975- PARZ, a randomized controlled trial. Clin Cancer Res. 2015;21:
977. 1071-1077.
54. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus 56. Choueiri TK, Fay AP, Gray KP, et al. PD-L1 expression in
everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015; nonclear-cell renal cell carcinoma. Ann Oncol. 2014;25:2178-
373:1803-1813. 2184.

Cancer January 15, 2017 209

You might also like