ArticlePDF AvailableLiterature Review

Metastatic Renal Cell Carcinoma Management: From Molecular Mechanism to Clinical Practice

Frontiers
Frontiers in Oncology
Authors:
  • Azienda ospedaliera SAndrea

Abstract and Figures

The therapeutic sc"enario of metastatic renal cell cancer (mRCC) has noticeably increased, ranging from the most studied molecular target therapies to those most recently introduced, up to immune checkpoint inhibitors (ICIs). The most recent clinical trials with an ICI-based combination of molecular targeted agents and ICI show how, by restoring an efficient immune response against cancer cells and by establishing an immunological memory, it is possible to obtain not only a better radiological response but also a longer progression-free and overall survival. However, the role of tyrosine kinase inhibitors (TKIs) remains of fundamental importance, especially in patients who, for clinical characteristics, tumor burden and comorbidity, could have greater benefit from the use of TKIs in monotherapy rather than in combination with other therapies. However, to use these novel options in the best possible way, knowledge is required not only of the data from the large clinical trials but also of the biological mechanisms, molecular pathways, immunological mechanisms, and methodological issues related to both new response criteria and endpoints. In this complex scenario, we review the latest results of the latest clinical trials and provide guidance for overcoming the barriers to decision-making to offer a practical approach to the management of mRCC in daily clinical practice. Moreover, based on recent literature, we discuss the most innovative combination strategies that would allow us to achieve the best clinical therapeutic results.
This content is subject to copyright.
Metastatic Renal Cell Carcinoma
Management: From Molecular
Mechanism to Clinical Practice
Michela Roberto
1,2
*, Andrea Botticelli
1,3
, Martina Panebianco
1,4
, Anna Maria Aschelter
4
,
Alain Gelibter
3
, Chiara Ciccarese
5
, Mauro Minelli
6
, Marianna Nuti
7
, Daniele Santini
8
,
Andrea Laghi
2
, Silverio Tomao
9
and Paolo Marchetti
1,3
1
Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy,
2
Department of Medical-Surgical
Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy,
3
Medical Oncology Unit, Policlinico Umberto
I, Sapienza University of Rome, Rome, Italy,
4
Medical Oncology Unit, Azienda Ospedaliero Universitaria SantAndrea,
Rome, Italy,
5
Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy,
6
Department of Medical Oncology, Azienda Ospedaliera San Giovanni Addolorata, Rome, Italy,
7
Department of Experimental
Medicine, University of Rome Sapienza Rome, Rome, Italy,
8
Department of Medical Oncology, University Campus Bio-
Medico, Rome, Italy,
9
Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I,
Sapienza University of Rome, Rome, Italy
The therapeutic sc"enario of metastatic renal cell cancer (mRCC) has noticeably
increased, ranging from the most studied molecular target therapies to those most
recently introduced, up to immune checkpoint inhibitors (ICIs). The most recent clinical
trials with an ICI-based combination of molecular targeted agents and ICI show how, by
restoring an efcient immune response against cancer cells and by establishing an
immunological memory, it is possible to obtain not only a better radiological response
but also a longer progression-free and overall survival. However, the role of tyrosine kinase
inhibitors (TKIs) remains of fundamental importance, especially in patients who, for clinical
characteristics, tumor burden and comorbidity, could have greater benet from the use of
TKIs in monotherapy rather than in combination with other therapies. However, to use
these novel options in the best possible way, knowledge is required not only of the data
from the large clinical trials but also of the biological mechanisms, molecular pathways,
immunological mechanisms, and methodological issues related to both new response
criteria and endpoints. In this complex scenario, we review the latest results of the latest
clinical trials and provide guidance for overcoming the barriers to decision-making to offer
a practical approach to the management of mRCC in daily clinical practice. Moreover,
based on recent literature, we discuss the most innovative combination strategies that
would allow us to achieve the best clinical therapeutic results.
Keywords: renal cancer carcinoma, targeted therapy, tyrosine kinase inhibitor (TKI), immune checkpoints inhibitor,
new biomarkers
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576391
Edited by:
Eugenio Zoni,
University of Bern, Switzerland
Reviewed by:
Eric Jonasch,
University of Texas MD Anderson
Cancer Center, United States
Michael Staehler,
Ludwig Maximilian University of
Munich, Germany
*Correspondence:
Michela Roberto
michela.roberto@uniroma1.it
Specialty section:
This article was submitted to
Genitourinary Oncology,
a section of the journal
Frontiers in Oncology
Received: 23 January 2021
Accepted: 29 March 2021
Published: 22 April 2021
Citation:
Roberto M, Botticelli A, Panebianco M,
Aschelter AM, Gelibter A, Ciccarese C,
Minelli M, Nuti M, Santini D, Laghi A,
Tomao S and Marchetti P (2021)
Metastatic Renal Cell Carcinoma
Management: From Molecular
Mechanism to Clinical Practice.
Front. Oncol. 11:657639.
doi: 10.3389/fonc.2021.657639
REVIEW
published: 22 April 2021
doi: 10.3389/fonc.2021.657639
INTRODUCTION
Renal cancer is the 10th most common cancer in Italy, with
approximately 13,400 new cases per year (1), 7080% have clear
cell histology, while papillary, medullary, chromophobe, and
other forms classied as non-clear cell histology are rare.
Approximately 25% of patients present with the advanced-stage
disease since their diagnosis, and among those undergoing
nephrectomy, about one-third experience a distant recurrence
during the rest of their lives and are initiated to systemic treatment.
Despite the signicant therapeutic improvements, the 5-year
survival rate of patients with metastatic renal cell cancer (mRCC)
remains poor, especially in patients with unfavorable prognostic
factors (2). The two validated prognostic models for the
classication of patients with mRCC within clinical trials are
the Memorial Sloan Kettering Cancer Centre (MSKCC) model
(3) and the International mRCC Database Consortium (IMDC)
that date back to 2005 and 2009, respectively (4). Although more
than 10 years have elapsed, and in the meantime, drug molecules
with new mechanisms of action have been developed, clinical
trials still stratify patients into those with favorable (with 0 poor
prognostic factors), intermediate (with 12poorprognostic
factors), or poor risk in the presence of at least three of the
following prognostic factors: less than 1 year from diagnosis to
treatment time, a Karnofsky PS score of <80 at the start of
treatment, anemia, neutrophil or platelet count greater than the
normal upper limit, or hypercalcemia (corrected Ca >10 mg/dl or
>2.5 mmol/L).
The therapeutic scenario of mRCC has undergone incredible
enrichment in recent years, ranging from the most studied
tyrosine kinase inhibitor (TKI)-targeted therapies (anti-
vascular endothelial growth factor (VEGF) and anti-mTOR) to
those most recently introduced (anti-MET, anti-RET, and anti-
FGFR) up to immunotherapy (IO) (anti-PD-1, anti-PD-L1, and
anti-CTLA-4). Literature data on new therapeutic indications
with cabozantinib in both the rst and second lines (5,6),
nivolumab after anti-VEGF TKI progression (7), nivolumab
combined with ipilimumab in naive patients with poor
prognostic factors, and pembrolizumab combined with axitinib
in all prognostic subgroups (8), have modied the prognosis of
patients with mRCC. Especially, patients classied as
intermediaterisk pass from a historical median survival of
approximately 20 months to 3 years in the front line, which is
almost equal to that of patients with favorable prognosis. On the
one hand, we have seen a considerable improvement in the
therapeutic algorithm of mRCC [clinical guidelines reported
different therapeutic options only in patients with intermediate
prognosis (1)]. On the other hand, the rate of development in the
identication of new prognostic and predictive factors has not
been the same throughout. Therefore, MSKCC/IMDC remains
the standard prognostic classication criteria. However, in light
of the complex mechanisms of action of the new TKI molecules,
such as cabozantinib or combinations of TKIs and IO, or even
more combinations of different immune checkpoint inhibitors
(ICIs), are we sure that these oldcriteria are sufcient?
In the era of precision medicine, in which knowledge of the
molecular and genomic aspects of renal cancer has become ever
wider, how can we think that criteria based on obvious clinical
considerations (poor performance status and a short
progression-free interval) and hematochemical parameters are
sufcient to determine a therapeutic choice? To make the best
use of new drugs and associations and to propose new
therapeutic sequences, better knowledge is required not only of
the data derived from the large clinical trials but also of the basic
biology, the complexity of involved molecular pathways, the
immunology of tumours, and methodological problems related
to both new response criteria and new endpoints. In this complex
scenario, this review aims to provide a practical approach to the
management of advanced renal cancer, framing the new results
in daily clinical practice and providing points for reections to
overcome decision-making barriers based on physician
therapeutic choice.
THE HETEROGENEITY OF RENAL TUMOR
RCC includes a heterogeneous group of tumors that are
characterized by different clinical and genomic factors and are
increasingly well dened in both syndromic and sporadic
settings (9). These tumor types originate from different cells;
for example, clear cell and papillary carcinomas arise from the
proximal or parietal kidney cells, whereas chromophobe
carcinomas arise from the intercalated cells (10)andare
characterized by different genomic drivers that lead to
tumorigenesis. In more than 90% of clear cell RCC cases,
large-scale genomic sequencing has identied chromothripsis
of chromosome 3p, typically with a concurrent gain of 5q (>67%)
and loss of 15q (45%) (9). In particular, the loss of 3p results in
the inactivation of Von HippelLindau disease tumor suppressor
protein (VHL). Mutations in genes encoding other components
of the VHL complex [such as TCEB1 (also known as ELOC)]
also lead to VHL inactivation (1113). pVHL is part of a
multiprotein complex with ubiquitin ligase activity. Within this
complex, pVHL is the subunit that recognizes protein substrates,
stimulating their ubiquitination and proteasome-dependent
degradation. The main target of this complex is the
transcription factor hypoxia-inducible factor 1 (HIF-1a),
which plays a key role in the cellular response to hypoxic
conditions. It stimulates the transcription of genes involved in
promoting angiogenesis and invasive growth. In renal cancer
cells, this complex does not function; therefore, HIF-1a
accumulates in cells and activates a cascade of other genes that
encode factors that induce hypoxia, including VEGF or those
involved in alternative pathways to VEGF, such as broblast
growth factor receptor (FGFR), platelet-derived growth factor
receptors (PDGFRs), AXL, and c-MET, all of which are involved
in angiogenesis, tumor growth, and survival (14).
Zinc-nger and homeobox protein 2 (ZHX2) is a VHL target.
VHL loss-of-function mutations usually result in an increased
abundance and nuclear localization of ZHX2. Loss of ZHX2
inhibits signaling through the transcription factor NF-kB, and
ZHX2 binds to many NF-kB target genes, revealing that ZHX2 is
a potential therapeutic target for RCC (15).
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576392
VHL inactivation alone is insufcient for RCC tumorigenesis,
and several gene mutations contribute to tumor heterogeneity
that characterizes RCC. Intratumoral heterogeneity, dened as
the presence of genetically different clones in different
subpopulations of the same tumor, is a typical renal tumor
condition (16). Accordingly, phylogenetic studies show how
the tumorigenesis in the RCC follows an evolutionary model,
tree-like: in the trunk lies the main mutation (e.g. the VHL gene
in the clear cell tumor) that paves the way for tumorigenesis, and
from the trunk, different subclonal mutations branch out, which
contribute to tumor growth and progression. Data from the
TRACERx renal study have identied secondary mutations and
chromosomal changes involved in tumor evolution (17).
Excluding hereditary forms, which cover only 4% of cases, for
sporadic forms, The Cancer Genome Atlas (TCGA) has identied 19
genes involved in addition to VHL, including BAP1, PBRM1, SETD2,
KDM5C, KDM6A, mTOR, PTEN, PIK3CA,andp53 (18). The
constitutive activation of the mTOR cascade plays an equally
important role in renal tumorigenesis through the loss of p53
expression or mutation of genes such as PI3K and PTEN. Therefore,
TKI therapies directed against one or more of these factors will always
be a therapeutic weapon of fundamental importance, as these are
precisely targeted against the genetic mechanisms based on
tumorigenesis and proliferation of renal cancer cells (Figure 1).
In addition to proper genetic damage, we must consider the
variations induced by the environment (epigenetics), alterations
in receptor expression, and all the complexity that revolves
around the tumor microenvironment.
Systemic inammation is frequently observed in advanced
RCC (19). Nevertheless, the functional correlation between
inammation and RCC metastasis remains unclear. Recent
data have demonstrated that cancer cells can secrete cytokines
and chemokines through a process known as cancer-cell-
intrinsic inammation, altering the immune landscape (2022).
Cancer-cell-intrinsic inammation contributes to cancer
metastasis and the initial progression of cancers. The driver
gene mutations responsible for the inammation in different
tumors are TP53 and KRAS mutations (2326). These mutations
lead to increased cytokine release, which recruits myeloid cells in
the primary tumor microenvironment or (pre-) metastatic sites.
It has been demonstrated that epigenetic remodeling
determines the massive expression of inammation-related
genes in RCC. Synchronous inhibition of the bromodomain
and extra-terminal motif suppressed C-X-C-type chemokines
in clear cell RCC cells and decreased neutrophil-dependent lung
metastasis, suggesting a potential therapeutic strategy (27).
The cells of the immune system (T cells, B cells, and natural
killer cells), which represent the targets of known ICIs, such as
anti-CTLA4, anti-PD-1, or anti-PD-L1, are found within the
tumor microenvironment. In addition to playing a key role in the
carcinogenesis process, some parameters such as the expression
of PD-L1 have been associated with a worse prognosis (28)as
well as a higher degree of tumor aggressiveness (29). Thus, the
use of ICIs that block PD-1/PD-L1 binding or amplify the overall
immune response ndsinthisbiologicalrationaleitshigh
activity in patients with mRCC (Figure 1).
It remains evident that the intratumoral heterogeneity
problem is responsible for the difculty in identifying a single
driver mutation and for overcoming mechanisms of clonal
selection during targeted treatment (30). To make things
FIGURE 1 | Representation of the main pathways involved in the mechanisms of tumorigenesis and proliferation of renal cancer cells and their targeted agents.
PD1, programmed cell death-1 receptor; PD-L1, programmed death-ligand 1; CTLA4, cytotoxic T-lymphocyte-associated protein 4; CD80, cluster of differentiation
80; CD86, cluster of differentiation 86; MHC, major histocompatibility complex; PI3K, phosphatidylinositol-3-kinase; AKT, serine/threonine kinase 1; mTOR,
mechanistic target of rapamycin; FGF, broblast growth factor; PDGF, platelet-derived growth factor; VEGF, vascular endothelial growth factor; cMET, mesenchymal
epithelial transition factor; AXL, AXL receptor tyrosine kinase; FGFR, broblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; VEGFR,
vascular endothelial growth factor receptor.
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576393
worse, a microenvironment response exists: tumors treated with
anti-angiogenic agents present an inammatory inltrate consisting
mainly of regulatory T cells (CD4
+
FOXP3
+
) and express high levels
of PD-L1, thus demonstrating the conditions associated with a worse
prognosis (31). These ndings suggest that the immunosuppressive
phenotype found in metastatic sites, for example, is the result of close
communication between the occurrence of anti-angiogenic
treatment-resistant subclones and the enrichment of inammatory
inltration with Treg cells to evade the anti-tumor immune response.
Given the above-mentioned data, the rationale for combining TKIs
with ICIs has become increasingly clear.
THE LATEST APPROVED THERAPEUTIC
STRATEGIES IN MRCC
Cabozantinib
Cabozantinib is a multi-targeting TKI directed against the
receptors of factors involved in tumor growth, angiogenesis,
pathological bone remodeling, chemoresistance, and metastatic
progression of cancer, such as VEGF, MET, GAS6(AXL), RET,
ROS1, TYRO3, MER, KIT (stem cell factor), TRKB, Fms-like
tyrosine kinase-3 (FLT3), and TIE-2 (32). Based on its broad
mechanism of action, it is believed to overcome resistance to
anti-VEGF agents, such as sunitinib and pazopanib; thus, it was
rst tested as a second-line therapy in patients previously treated
with anti-VEGF therapy (5)andsubsequentlyasrst-line
therapy in patients with intermediatepoor-risk prognosis (6).
In the phase III METEOR trial, 658 patients with mRCC, who
had previously been treated with at least one VEGF tyrosine
kinase receptor inhibitor (VEGFR-TKI), were randomized 1:1 to
receive cabozantinib (n = 330) or everolimus (n = 328), including
those who may have previously been treated with other therapies,
including cytokines and antibodies directed against VEGF, the
PD-1 receptor, or other ligands. Additionally, patients with
treated brain metastases were included. The primary endpoint
of the study was progression-free survival (PFS). Secondary
endpoints were objective response rate (ORR) and overall
survival (OS). Most patients were males (75%), with a median
age of 62 years. Seventy-one percent of patients had previously
been treated with only one VEGFR-TKI. In 41% of patients,
sunitinib was the single VEGFR-TKI previously received.
According to the MSCKK criteria for the prognostic risk
category, in 46% of patients, the prognosis was favorable; in
42%, it was intermediate (one risk factor); and in 13%, it was
poor (two or three risk factors). In 54% of patients, three or more
organs, including the lungs (63%), lymph nodes (62%), liver
(29%), and bones (22%), had metastatic disease. The median
duration of treatment was 7.6 months (range 0.320.5) for
patients who received cabozantinib and 4.4 months (range 0.2
18.9) for patients who received everolimus. A statistically
signicant improvement has been demonstrated in PFS for
cabozantinib compared to everolimus (7.4 months compared
to 3.9 months, hazard ratio [HR] = 0.51 [0.410.62], p = 0.0001).
In a subsequent interim analysis, a statistically signicant
improvement was also demonstrated in terms of OS [320
events, median value of 21.4 months compared to 16.5
months; HR = 0.66 (0.53, 0.83), p = 0.0003]. Comparable OS
results were observed with a follow-up analysis (descriptive) at
430 events. Exploratory analyses of PFS and OS in the intent-to-
treat population also showed consistent results in favor of
cabozantinib compared to everolimus in different subgroups
dened by age (<65 years compared to 65 years), sex, risk
group, ECOG status (0 compared to 1), time from diagnosis to
randomisation (<1 year compared to 1 year), tumor expression
of MET (high compared to low compared to unknown), bone
metastasis, visceral metastasis, number of VEGFR-TKIs
previously received (one vs two), and duration of rst
treatment with VEGFR-TKI (6 months vs >6 months). Dose
reductions were more frequent with cabozantinib than with
everolimus, but no statistically signicant difference in terms of
discontinuation of severe adverse events was reported (5,33).
The safety and efcacy of the rst-line cabozantinib were
evaluated in the CABOSUN study, a randomized, open-label,
controlled vs sunitinib phase II study, which enrolled 157 mRCC
patients, classied as intermediate or poor risk according to
IMDC criteria. The patients (n = 157) were randomized 1:1 to
receive cabozantinib (n = 79) or sunitinib with a schedule of 4
weeks on/2 weeks off (n = 78). The patients were stratied
according to the IMDC risk category (81% intermediate and
19% poor) and the presence or absence of bone metastases.
Approximately 75% of patients underwent nephrectomy before
the start of treatment. The primary endpoint was the PFS, and
the secondary endpoints were ORR and OS. Most patients were
males (78%) with a median age of 62 years. Most patients (87%)
had an ECOG performance status of 0 or 1; 13% had an ECOG
performance status of 2. Thirty-six percent of the patients had
bone metastases. The study has reached the primary endpoint of
statistically signicant improvement of the PFS for cabozantinib
compared to sunitinib [8.6 months regarding 5.3 months; HR =
0.48 (0.320.73), p = 0.0005]. Patients showed a favorable effect
with cabozantinib compared to sunitinib irrespective of MET
status (positive or negative); however, cabozantinib demonstrated
greater activity in patients with positive MET status than that in
patients with negative MET status [HR = 0.32 (0.16 and 0.63) vs
0.67 (0.37 and 1.23)]. In addition, compared to the treatment with
sunitinib, treatment with cabozantinib has been associated with a
trend of longer OS (30.3 months compared to 21.0 months; HR
0.74 [0.471.14]) (6).
In the two aforementioned studies, the most frequently
reported serious adverse events with cabozantinib were
hypocalcemia, hypokalemia, thrombocytopenia, hypertension,
palm-plantar erythrodysesthesia syndrome, proteinuria, and
gastrointestinal events (abdominal pain, inammation of the
mucous membranes, constipation, diarrhea, and vomiting) and
were generally found during the rst 8 weeks of treatment. In the
METEOR study, dosing reductions and dosing interruptions of
59.8 and 70%, respectively, occurred in relation to an adverse
event caused by cabozantinib. In CABOSUN, where patients
were naïve to treatment, the percentages of reduction and
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576394
treatment interruption were quite similar (46 and 73% of
patients, respectively). Therefore, it does not seem to be a
condition of drug toxicity. However, hypertension has been
observed more frequently in the population of naïve patients
(67%) than in patients included in the METEOR trial who had
been previously treated with anti-VEGF targets (37%).
Nivolumab: Monotherapy and ICI
Combination Therapy
Nivolumab was the rst anti-PD-1 ICI approved for the
treatment of mRCC, rst as monotherapy in patients
previously exposed to a VEGFR-TKI and then in combination
with ipilimumab as the rst-line treatment in patients with
intermediate- and poor-risk prognosis. According to data from
the Phase III Checkmate 025, patients who progressed during or
after 12 previous anti-angiogenic regimens were eligible for
treatment with nivolumab monotherapy (34). This study
included patients regardless of tumor PD-L1 status and with a
70% Karnofsky performance status (KPS). Patients with a history
of brain metastasis or concomitant brain metastasis, previously
treated with an mTOR inhibitor, affected with an autoimmune
disease in the active phase, or with medical conditions requiring
systemic immunosuppression were excluded from the study. A
total of 821 patients were randomized to receive nivolumab (n =
410) or everolimus (n = 411). The study reached the primary
endpoint of efcacy (median OS equal to 25 months with
nivolumab compared to 19.6 months with everolimus, HR =
0.73 [0.70.93], p = 0.0018). Secondary endpoints included ORR
and PFS, as evaluated by the investigator. In this study,
nivolumab was shown to be better than everolimus in pre-
treated patients in terms of ORR (25 vs 5%, p < 0.001, HR for
OS = 0.73; 95% condence interval (CI) = 0.570.93). However,
no signicant advantages in terms of PFS have been reported.
Nivolumab in combination with ipilimumab proved to be
superior to sunitinib as the rst-line therapy in the Phase III
study Checkmate 214 (8). The study included patients with
mRCC, with clear cell components that were not previously
treated. The primary efcacy population included patients at
intermediate/poor-risk according to the IMDC criteria. A total of
1,096 patients were enrolled, of which 847 at intermediate/poor-
risk were randomized to nivolumab in combination with
ipilimumab (n = 425) for four cycles followed by nivolumab
monotherapy or sunitinib (n = 422). The primary endpoints
were the OS, ORR, and PFS. Patients with mRCC with
intermediate/poor prognosis according to IMDC reported a
statistically signicant benet in terms of both OS and ORR
(HR for OS = 0.63, 95% CI = 0.440.89; ORR 42 vs 27%, p <
0.001), regardless of the expression level of PD-L1, although in
the PD-L1 >1% group, the advantage was even more signicant
(HR = 0.52; 95% CI = 0.340.78). The PFS was not signicantly
different between the two groups (HR = 0.82; 95% CI = 0.64
1.05). In addition, in the 249 patients at favorable risk,
nivolumab plus ipilimumab was detrimental in terms of OS
compared to sunitinib (HR = 1.13 [0.641.99] p = 0.6710). In
terms of tolerability, the combination of ipilimumab and
nivolumab was burdened with a higher toxicity than sunitinib
(22 vs 12% of patients, respectively, discontinued treatment for
toxicity) (8) and compared to IO with a single agent, resulting in
a more severe immune-related toxicity percentage (35).
However, a more recent report on the Checkmate 214 study
demonstrated that patient-reported outcomes were more
favorable with nivolumab plus ipilimumab than sunitinib in
patients at intermediate or poor risk, leading to fewer symptoms
and better health-related quality of life (36). Moreover, to better
characterize the association between outcomes and IMDC risk in
CheckMate 214, a post-hoc analysis (n = 1051) of efcacy by the
number of IMDC risk factors was completed. ORR with
nivolumab plus ipilimumab was consistent across zero to six
IMDC risk factors, whereas with sunitinib, it decreased with an
increasing number of risk factors. The benets of nivolumab plus
ipilimumab over sunitinib in terms of ORR (4044% vs 1638%),
OS (HR = 0.500.72), and PFS (HR = 0.440.86) were
consistently observed in subgroups with one, two, three, or
four to six IMDC risk factors. These results demonstrate the
benetofrst-line nivolumab plus ipilimumab over sunitinib
across all intermediate- and poor-risk groups, regardless of the
number of IMDC risk factors (37).
Thanks to the data reported, the combination of nivolumab
and ipilimumab was approved by ESMO guidelines in
intermediate- and poor-risk prognostic subgroups of mRCC.
Moreover, a post-hoc analysis of nivolumab plus ipilimumab
or sunitinib in IMDC intermediate/poor-risk patients with
previously untreated mRCC with sarcomatoid features showed
an ORR of 56.7% (CI = 43.269.4, p < 0.001) in the combination
arm against 19.2% (9.632.5) of standard treatment and a rate of
complete response (CR) of 18.3% in the experimental group,
whereas no CR was observed in the sunitinib arm (38).
Elderly patients with pre-treated mRCC may benet from
therapy with nivolumab or nivolumab plus ipilimumab as a rst-
line option (7,39), and salvage-line cabozantinib may offer the
best survival outcomes, although evidence suggests that the
majority of rst-line treatments have worse efcacy in older
patients than in younger patients (40,41).
Despite the undeniable benets of ICIs in the treatment of
mRCC, some aspects must be considered: i) only a subset of
patients achieves objective responses, ii) some patients have a
delayed response, and iii) a signicant number of patients do not
benet even clinically. In detail, although the so-called comboIO is
particularly active as the upfront treatment in patients with
intermediate/poor prognosis, it cannot be a universal choice for
all patients, but only for those patients tfor a more intensive
combined treatment. Moreover, the ipilimumabnivolumab
combination was less effective than sunitinib in patients over 75
years of age, who represent most of those we met in clinical practice.
Therefore, IO is an important strategy both as rst- and second-line
treatment in patients with mRCC, but TKI agents remain the
central focus of mRCC treatment in all therapeutic lines. Several
hypotheses have been formulated regarding the lack of efcacy of
ICIs in all patients, and among these, tumor heterogeneity and the
dynamism of the tumor microenvironment typical of renal cancer
cells seem to be the main conditions (29,42).
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576395
The Combination of VEGF-Targeting
Agents With ICIs
The upfront combination of VEGF-targeting agents with ICIs is
emerging as a therapeutic alternative that could overcome the
limitations of IO alone as well as target both the cascade of
angiogenesis and the tumor microenvironment (Figure 1). Anti-
VEGFR inhibitors, in addition to their intrinsic anti-angiogenic
effect, showed immunomodulatory effects: unlocking the
inhibitory brake of VEGF, promoting inltration and
activation of effector cells, and inhibiting immunosuppressive
cells (43). Although the initial studies of sunitinib or pazopanib
associated with nivolumab had negative results for the high rates
of liver and gastrointestinal toxicity (44), new combinations are
proving to be active and well tolerated (4547).
In the IMmotion151 study, the anti-PD-L1 atezolizumab
combined with the anti-VEGF bevacizumab performed better
than sunitinib monotherapy in patients with PD-L1-positive
tumors (HR = 0.74 [95% CI = 0.570.96]; p = 0.02]; however,
in the intention-to-treat (ITT) population, the median OS was
33.6 months in the combination arm vs 34.9 months in the
sunitinib arm, and the results (HR = 0.93) had not yet crossed the
signicance boundary (45). A pre-specied subgroup analysis of
IMmotion151 demonstrated a signicant benet in terms of PFS
in patients with mRCC with sarcomatoid features in the
bevacizumab plus atezolizumab treatment arm when compared
with the sunitinib treatment arm (48).
Other promising combinations always used as rst-line
treatment are axitinib plus avelumab and axitinib plus
pembrolizumab, tested in the Javelin Renal 101 (46) and the
Keynote-426 (47) trials, respectively.
The Javelin Renal 101 randomized 442 and 444 patients to
the avelumab plus axitinib and sunitinib arms, respectively, and
showed that the combination treatment was higher than
sunitinib monotherapy in terms of PFS and ORR, regardless
of the PD-L1 status and prognostic risk category (46). The last
update of the study conrmed previous results; in particular, in
the overall population, the median PFS was 13.3 (95% CI =
11.1e15.3) vs 8.0 months (95% CI = 6.7e9.8), HR = 0.69 [95%
CI = 0.5740.825]; p < 0.0001); moreover, the combination
prolonged PFS2 compared with sunitinib. However, OS data
(primary endpoint of both studies) are still immature (49).
The Keynote 426 study is a phase 3 trial that randomly
assigned 861 patients with previously untreated advanced RCC
to receive pembrolizumab plus axitinib or sunitinib. The primary
endpoints were the OS and PFS in the ITT population. The key
secondary endpoint was ORR. After a median follow-up of 12.8
months, this study observed a signicant benet in terms of PFS
(15.1 vs 11.1%, HR = 0.69; 95% CI = 0.570.84; p = 0.001) and
ORR (59.3 vs 35.7%, p = 0.001) in favor of the combined
treatment arm, disregarding the status of PD-L1 and the
prognostic risk category (47).
The results of the extended follow-up of the randomized
phase III study KEYNOTE-426 (median follow-up 30.6
months) conrmed the benet for the experimental arm,
which was proven statistically signicant in terms of median
OS [not reached with pembrolizumab and axitinib vs 35.7
months (95% CI = 33.3not reached) with sunitinib; HR =
0.68 (95% CI = 0.550.85), p = 0.0003], median PFS [15.4
months with pembrolizumab and axitinib (12.718.9) vs 11.1
months for sunitinib (95% CI = 9.112.5); HR = 0.71 (95% CI =
0.600.84), p = 0.0001], and ORR (60% in the combo arm vs 40%
in the sunitinib group). Although the trial was not designed to
observe differences between risk categories, it should be noted
that the benet in terms of OS was particularly evident in the
population at intermediate and unfavorable risk
[pembrolizumab plus axitinib vs sunitinib: HR = 0.63 (95%
CI = 0.500.81)], while it was not signicant in the favorable
risk group [HR = 1.06; (95% CI = 0.601.86)]. Moreover, in
terms of toxicity, no signicant news emerged with the continued
follow-up of patients in the study. The most frequent treatment-
related grade 3 or higher adverse events (10% of patients in both
groups) were hypertension [95 (22%) of 429 patients in the
pembrolizumab group plus axitinib vs 84 (20%) of 425 patients
in the sunitinib group], increased alanine aminotransferase levels
[54 (13%) vs 11 (3%)], and diarrhea [46 (11%) vs 23 (5%)] (50).
The fact that the advantage in OS for the combination,
already known from the rst analysis, is maintained over time,
although half of the patients randomized to only sunitinib had
then received progression IO (vs. 8% in the experimental arm),
suggests the synergistic activity of the combination of
pembrolizumab plus axitinib, which may therefore not be
reproducible by their use in sequence. With regard to drug
synergy, the role of a single agent in the overall result may also
be different: while axitinib is more responsible for shrinkage,
pembrolizumab could then be more decisive in maintaining the
volumetric reduction effect over time (51).
Furthermore, although all the front-line combination trials
enrolled patients with clear cell RCC, exploratory post-hoc
analyses from these studies demonstrated that patients with
sarcomatoid differentiation, which has historically been
associated with worse prognosis, derive marked benets from
ICI-based therapy. Based on these data, the Food and Drug
Administration (FDA) and EMA in 2019 approved the axitinib
pembrolizumab combination as the rst-line treatment for
patients with clear cell mRCC in the all-risk category.
CheckMate-9ER is a randomized controlled trial comparing
the combination of nivolumab and cabozantinib vs sunitinib as a
rst-line treatment for mRCC with a clear cell component and
any IMDC risk group. In the rst analysis of the study, the
superiority of the combination arm over standard treatment was
shown to meet all three efcacy endpoints, with a 40% reduced
risk of death [HR = 0.60 (98.89% CI = 0.400.89); p = 0.0010;
median OS not reached in both arms]. In patients treated with
the combination cabozantinib and nivolumab, the median PFS,
the primary endpoint of the study, is doubled compared to
patients who received only sunitinib: 16.6 months compared to
8.3 months [HR = 0.51 (95% CI = 0.410.64), p = 0.0001]. In
addition, cabozantinib in combination with nivolumab showed a
higher ORR (56 vs 27%) and 8% of patients compared to 5% who
achieved a complete response. Moreover, the combination
treatment was associated with a longer response duration
compared to sunitinib, with a median duration of 20.2 months
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576396
compared to 11.5 months. In addition, patients treated with the
combination showed a lower rate of discontinuation of treatment
than those treated with sunitinib (44.4 vs 71.3%) and a
signicantly lower rate of discontinuation for disease
progression than sunitinib (27.8 vs 48.1%). All these key
efcacy results are consistent in pre-specied subgroups and all
risk categories according to the IMDC and PD-L1 expression
(52,53). Based on this study, the ESMO guidelines proposed the
combination of nivolumab and cabozantinib as a valid rst-line
therapy in all prognostic subgroups (52).
Unlike the KEYNOTE 426 trial, no signicant results were
obtained in terms of OS when the experimental arm was compared
with the standard treatment in the other phase III studies. In fact, in
the Javelin Renal 101 study, OS data were immature in the 2019
publication, while in the IMmotion-151 trial, OS was not met.
Among the new drug combinations tested in mRCC, there is
also the one examined in the phase Ib/II TiNivo study, which
evaluated the efcacy and safety of combination therapy with
nivolumab plus tivozanib, a highly potent and selective VEGFR-
TKI approved by the European Medicines Agency (EMA) for
rst-line treatment of patients with mRCC (54), and showed a
generally tolerable prole and promising anti-tumor efcacy (55).
HOW THERAPEUTIC ALGORITHM HAS
CHANGED IN MRCC TREATMENT WITH
THE APPROVAL OF COMBO?
For a decade, it has been wondered what the best sequence
treatment between TKImTORiTKI vs TKITKImTORi is.
However, the next future question will be much more complex
since there are no comparative studies, clear prognostic factors,
or predictive markers, thus making a weighted choice between
the various options available in the rst- and second-line
very difcult.
The new treatment strategies range from molecular targeted
agents such as cabozantinib, able to overcome some anti-
angiogenic mechanisms of resistance, through ICIs, such as
nivolumab, as a single agent, up to the combinations of ICIs
(nivolumab + ipilimumab), or between ICIs with VEGF-
targeting agents (atezolizumab + bevacizumab, pembrolizumab +
axitinib, avelumab + axitinib, cabozantinib + nivolumab, and
others under investigation).
The paradigm of rst-line treatment in advanced RCC, rmly
occupied for more than 10 years by monotherapy with anti-
angiogenic TKIs, such as sunitinib or pazopanib, has changed,
and combinations of ICIs, either with each other or with TKIs,
have shown efcacy compared to monotherapy with TKIs. In
light of the results of recent combinations, except for
comorbidity and clinical contraindications, in the rst-line, the
therapeutic proposal is to administer all prognostic classes the
combination TKI/IO (axitinib plus pembrolizumab/nivolumab
plus cabozantinib) or IO/IO (nivolumab plus ipilimumab), and
considering the combination IO/IO for patients with
sarcomatoid components, whereas all other cases remain valid
for TKI monotherapy, in particular, cabozantinib in the
intermediate- and high-risk subgroups untforcombo
treatment, and pazopanib or sunitinib in the good risk unt
for combo (56).
Whether the objective is to achieve a complete response (CR)
as well as a long survival (or possible cure), ipilimumab plus
nivolumab or nivolumab plus cabozantinib would be the
treatment of choice. In fact, the CR rates in CheckMate 214,
CheckMate9ER, Keynote426, and Javeline Renal 101 were 9, 8,
5.8, and 3.8%, respectively. In contrast, we should also consider
that a higher rate of progressive disease (PD) was observed as the
best response to treatment in the CheckMate 214 trial, while the
lowest was observed in CheckMate9ER. The toxicity prole is a
further discriminant in the choice of combination treatment. In
fact, for the IOIO combination, the major toxicities are limited
to the induction phase with ipilimumab, while for the combination
of an ICI and a VEGFR-TKI, safety issues tend to persist over time
due to the prolonged administration of both agents.
As the eld stands now, the immuno-target combination
could represent a particularly valid opportunity, especially in
patients with a coldphenotype, whose tumor is characterized by
poor immune inltration and are considered less likely to
respond to ICI-based treatment alone.
Given the lack of head-to-head comparative studies, both
experience and common sense must guide the choice of a
physician according to the following considerations: i) patient
characteristics, comorbidities, drug interactions with concurrent
therapy, occupation, preferences of patient, and side effects that
can affect the quality of life; ii) neoplasia features, its histology, if
it has a representative sarcomatoid components, the genetic
structure, the burden of cancer disease, and the location of the
metastases and their related symptoms; iii) balancing the risk and
benet of treatment itself: for safety, we should consider that the
trade-off between efcacy and safety that a rst-line patient is
willing to accept is usually unbalanced in favor of efcacy; iv)
biological aggressiveness of the tumor: in the case of an
aggressive disease, the combo IO/TKI seems a very reasonable
choice to control disease growth while waiting for the tail effect of
IO; otherwise, one could head for the long-term benet of the
IO/IO combo, as well as for complete response, trying to spare
the additional toxicity derived from the continuous use of the
VEGFR-TKI.
Moreover, a recent meta-analysis network on the choice of
the rst-line showed that cabozantinib is the best molecular
targeted agent for the advantage in terms of PFS in patients at
intermediate/poor risk compared to sunitinib, with a 91% chance
of giving the best benet in PFS (57). Therefore, the choice will
be conditioned by our primary endpoints, even if they do not
always coincide with those of large clinical trials.
Taking into account that the IMDC prognostic model was
developed at the time of a rst-line anti-VEGF-based therapy
(58) and that neither validated prognostic models in rst-line
with ICIs or with the immuno-target combo nor data on the
second-lines are available, the therapeutic algorithm of mRCC
could be revised in the following way (Figure 2): i) for the rst-
line, to assess whether the patient is considered tfor a
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576397
combination strategy; ii) for subsequent lines, taking into
account what has been done previously (in immuno-naive
patients, the choice could fall on nivolumab or another TKI
such as cabozantinib, while in TKI-naive patients, the choice
could fall on an anti-VEGF TKI such as sunitinib, pazopanib, or
cabozantinib). Nevertheless, data on pazopanib or cabozantinib
as second-line treatment after ICI-based treatment are not
available. However, cabozantinib demonstrated impressive PFS
and OS when administered post-IO in patients with mRCC,
according to ndings from the METEOR sub-analysis
33
and
recent retrospective real-world studies (59,60). After disease
progression to rst-line TKI-based monotherapy, the factors that
could guide the choice towards a second-line of treatment in
favor of another TKI are low or intermediate risk, long duration
of rst-line treatment with VEGFR-TKI, good tolerability to
previous treatment lines, low tumor burden, slow progression,
revascularization of pre-existing lesions, and high probability of
receiving further treatment lines. In favor of IO-based second-
line treatment, we have considered the following factors: high
risk, short duration of rst-line treatment with VEGFR-TKI,
poor tolerability to TKI, dose reductions and interruptions, high
tumor burden, rapid progression, progression not guided by
angiogenesis, and low probability of receiving further therapy.
Currently, there are no data comparing the available strategies
that combine two IO agents or TKI plus IO, but increasing
evidence suggests that some biomarkers and genetic features
could guide optimal treatment options for patients.
WHAT TO EXPECT FROM
DIAGNOSTIC IMAGING?
The complex therapeutic scenario described above makes
imaging evaluation extremely challenging, both at the time of
diagnosis and in assessing the response to treatment. At the time
of diagnosis, owing to high intratumoral heterogeneity and
heterogeneity between the gene expression proles of primary
cancer and its metastases, tumor genomic characterization is
necessary. Considering the technical difculties and morbidity in
performing multiple renal biopsies (61), a solution may be
represented by radiogenomics. Radiogenomics, a result of
advances in both computational hardware and machine-
learning algorithms, is an emerging eld in which quantitative
information is extracted from radiological images (radiomics)
and is correlated with tumor genomic proling (62). Although
FIGURE 2 | Proposed therapeutic algorithm for the treatment of mRCC in and beyond the rst-line setting. The choice of treatment is based on (1) i) patient
characteristics: comorbidities, potential drug interactions with the concomitant therapy, occupation, patient preferences and the side effects that can affect the quality
of life; ii) and tumor characteristics: histology, if it has a representative sarcomatoid component, the genetic structure, the tumor burden, site of metastases, and
related symptoms. (2) MSKCC/IMDC prognostic classication; *if not previously carried out, **if the patient does not have autoimmune disease in the active phase,
solid organ transplant, or interstitial pneumopathy or if the patient requires high doses of corticosteroids. TKI, tyrosine kinase inhibitor Pazopanib and cabozantinib
are still not indicated as second-line treatment after immunotherapy; however, real-world analysis of patients treated with cabozantinib after anti-PD-1 treatment
reported promising results.
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576398
studies are still preliminary (63,64), it is expected that
quantitative imaging data might become a useful biomarker for
assessing tumor prognosis, treatment selection, and prediction of
treatment response.
With the advent of anti-VEGF and TKIs and then ICIs, the
evaluation of response to therapy made it necessary to introduce
new objective response criteria [i.e. modied Choi (65), SACT
(66), iRECIST (67)], since conventional RECIST (66) is not
adequate for categorizing patient response. However, there are
still open issues regarding the assessment of pseudo-progression
and dissociated response (68,69), both of which are strongly
associated with the clinical benetofICIsandhyper-
progression. Further challenges will await radiology with the
advent of combined treatments. The solution will probably be
found in the integrated analysis of imaging data (from different
sources, including CT, MRI, and PET, combining morphological
and functional studies, targeting tumor perfusion and
cellularity), tumor mutational burden, and biological markers.
Once collected, this large amount of data will be processed by
high-speed processors driven by articial intelligence.
POSSIBLE FUTURE PREDICTIVE AND
PROGNOSTIC BIOMARKERS
PD-L1 Expression
Several studies have demonstrated the negative prognostic role of
the expression of PD-L1 in the setting of mRCC (7072). The
expression of PD-L1 on tumor cells was associated with a higher
tumor stage and a worse response to TKI therapy in two post-hoc
analyses of the COMPARZ study and the METEOR and
CABOSUN trials (28,7274). In addition, a meta-analysis
including more than 1,300 patients showed that higher PD-L1
expression correlated with an approximately doubling risk of
death (75). In contrast, the predictive role of PD-L1 in response
to IO is still controversial, and the results obtained in the
exploratory analyses of clinical trials investigating ICIs are
inconclusive (7,8,4547). In the CheckMate 025 trial, PD-L1
expression was associated with poor survival independent of the
treatment received, but not with response to nivolumab (7). The
CheckMate 214 trial showed a higher PFS in the ipilimumab plus
nivolumab arm than in the sunitinib arm for IMDC intermediate
poor-risk patients, with PD-L1 expression in 1% or greater of cells
(median PFS 22.8 vs 5.9 months), but this advantage was not
observed when PD-L1 was less than 1% (median PFS, 11 vs 10.4
months). Conversely, a better ORR and OS for IO over an anti-
vascular agent was reported regardless of tumor PD-L1 expression
level (8). In the IMmotion 151 trial, the magnitude of benet
derived from the combination therapy with atezolizumab plus
bevacizumab increased in patients with PD-L1 expression by more
than 1% of tumor-inltrating lymphocytes compared with the ITT
population (45). In the JAVELIN Renal 101 and KEYNOTE-426
trials, the combination therapy showed a benet over sunitinib
irrespective of PD-L1 expression (49,50).
The above-mentioned results suggest that the expression of
PD-L1 in mRCC cannot completely predict the responsiveness of
tumors to ICIs. Its role remains controversial and warrants
further investigation. Moreover, the assessment method and
tumor heterogeneity are the major limitations of the evaluation
of PD-L1 (76). The technique used for the IHC analysis has an
elevated variability among the different methods available, and
the scoring systems are not concordant for the target cells
evaluated, whether tumor cells, immune cells inltrating the
neoplastic stroma, or a combination of both; additionally, there is
no validated positivity cut-off (77,78). Furthermore, PD-1 and
PD-L2 evaluation should be considered, and their role should be
claried (79). Finally, the expression of PD-L1 is dynamic,
changing depending on the history of the disease and the
treatments received. In addition, intratumoral variability and a
different expression in the primitive tumor and metastases,
which would explain the high response rates obtained despite
the negativity of PD-L1 in the primitive lesion, should be
considered when the expression of this biomarker is
examined (80).
Tumor Mutational Burden
TMB is dened as the total number of mutations per coding area
of the tumor genome, measured as mutations per megabase
(mutations/Mb) (81). In tumors with high TMB, there is an
increased production of surface neoantigens that stimulate the
anti-tumor immune system response, which could explain the
potential association between TMB and response to ICIs (82). In
the setting of mRCC, TMB is variable, ranging from a very low
level in chromophobe type to a higher value in clear cell and
papillary tumors and is not concordant with the clinically
dened prognostic groups according to IMDC and MSKCC
(83). Regarding its prognostic role, the data in the literature
are discordant, since some studies observed a correlation
between improved survival and increased TMB, while others
demonstrated a negative prognostic role (84,85). Concerning the
predictive signicance of TMB, no association between TMB and
survival, PD-L1 expression on tumor cells, or clinical benet was
observed (86).
Microenvironment
RCC is characterized by a heterogeneous population of tumor-
inltrating immune cells; however, conicting data have been
obtained to date. Inltration of effector T cells, such as CD8
+
lymphocytes, and M1 macrophages may be associated with a
better prognosis, whereas inltration of regulatory T cells, such
as Tregs and M2 macrophages, have a poorer outcome (8790).
In contrast, high intra- and peri-tumoral CD8
+
cell density was
also correlated with poor prognosis (91). It was demonstrated
that PD-L1 expression on tumor cells could lead to higher CD8
+
T cell inltration, distinguishing two groups of tumors with
CD8
+
inltrate, and the group with low expression of immune
checkpoints and localization of mature dendritic cells was
associated with a good prognosis (92).
Concerning the predictive role of the microenvironment, a
comprehensive analysis of patients enrolled in four clinical trials
on nivolumab demonstrated a poorer and greater response in
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 6576399
correlation to the overexpression of genes involved in metabolic
functions (e.g. UGT1A) and the increased expression of immune
markers (e.g. BACH2 and CCL3), respectively (93). Moreover, an
exploratory analysis of the IMmotion150 trial reported that a T-
effector immune gene in association with the expression of PD-
L1 and the inltration of T CD8
+
cells correlates with a higher
ORR and prolonged PFS in the atezolizumab arm (45). In
particular, it was observed that VEGF blockade could promote
the inltration of T cells into the tumor microenvironment, thus
potentiating the mechanism of action of ICIs (94).
Circulating Tumor Markers
Circulating tumor DNA (ctDNA) and circulating tumor cells
(CTCs) are peripherally detectable tumor-derived materials. These
markers could detect primary and metastatic sites non-invasively
and evaluate the response to therapy (9597). Variable frequencies
of genomic alterations were detected in the front-line and second-
line treatment settings, showing an increased incidence of genomic
alterations, particularly those affecting TP53 and MTOR, after rst-
line treatment with VEGFR-TKI therapy (96). These differences
could reect treatment-selective pressures and the effect offront-line
therapy on ctDNA load, but might also simply depend on the
technical limitations of ctDNA assessment in this disease (98).
Other circulating protein and lipid markers have
demonstrated predictive and prognostic value in advanced
disease. Based on 52 circulating markers, a cohort of 69
patients treated with rst-line sorafenib was grouped by either
an angiogenic or an inammatory signature, with correlations to
PFS (HR = 0.2 vs 2.25; p = 0.0002) (99). Additional markers in
serum have been investigated, such as soluble VEGF, circulating
microRNAs, carbonic anhydrase 9, and inammatory markers,
such as IL-6 and IL-8, but most of these studies were conducted
in the era of targeted therapies (99103), and new dedicated
investigations are required to address the dramatic changes in
treatment paradigms brought about by the advent of ICIs.
Genomic and Transcriptomic
Environments
There are three possible treatment strategies in the therapeutic
landscape of mRCC: angiogenic inhibitors at one end, IO at the
other, and combinations of the two classes in the middle. The
challenge, however, is identifying the subset of patients who
could benet from one therapeutic class alone to avoid the
unnecessary toxicity of combination approaches.
Using RNA-based analyses, four distinct molecular subgroups
associated with different responses and survival were dened:
Cluster 3 had the best prognosis with high angiogenic gene
expression and was associated with a better outcome under anti-
angiogenic therapy (PBRM1 mutation was frequently associated)
(104); Cluster 4, with upregulation of the immune pathway, had a
worse prognosis, with a frequent sarcomatoid differentiation and
expression of PD-L1 (105); and Clusters 1 and2 were intermediate
clusters with a lower expression of angiogenic and immune genes.
These results may have the potential to inform treatment
personalization in patients with mRCC (106).
The phase 2 IMmotion150 trial investigated the efcacy, as
measured by PFS, of atezolizumab with or without bevacizumab
against sunitinib in patients with untreated mRCC and
correlated differential gene expression signatures (angiogenesis,
T-effector, and myeloid) with therapeutic response. Highly
angiogenic tumors, which coincided with tumors exhibiting
PBRM1 mutations, seemed to benet more from sunitinib, but
not from atezolizumab either alone or in combination with
bevacizumab. The combination treatment improved clinical
benets compared with sunitinib in T-effector high tumours.
Tumors with T-effector high and lower myeloid inammation-
associated gene expression beneted from atezolizumab
monotherapy. Instead, in T-effector high tumors, a
concomitant high myeloid inammation predicted a worse
response to IO alone. Myeloid inammation is associated with
high expression of IL-6, prostaglandins, and the CXCL8 family of
chemokines, which suppress the anti-tumor immunity. The
improved clinical outcome associated with atezolizumab +
bevacizumab compared with atezolizumab monotherapy in this
subgroup suggests that the addition of bevacizumab to
atezolizumab may overcome innate inammation-mediated
resistance in these tumors (104).
Based on the analysis of the angiogenic prole in comparison
with the immunological prole of the study IMmotion151, it is
possible to dene subgroups of tumors that benet from different
treatment strategies. Given the new associations, it would be
interesting to evaluate these aspects in other combinations of IO/
TKI and see if, for example, the addition of TKI modies the
immunogenicity of these tumors.
CONCLUSIONS
Considering the continuously evolving scenario in the treatment
of patients with mRCC, the future goal will be to better
characterize renal neoplasia in all its complexity, from the trunk
to the last of its branches. However, to outline the most
appropriate treatment path for each patient, we cannot deny
that only clinical criteria are very likely to understand the needs
of patients. Given the signicant improvement in therapeutic
options, prospective studies are needed that would elucidate:
what will be the most effective therapeutic algorithm and how
patients will be selected to hit more targets; will it be more effective
to use therapeutic agents in sequence or by focusing completely on
the rst therapeutic line; will it be more effective to use a
combination strategy from the beginning of mRCC treatment?
Further studies are required to answer these questions.
AUTHOR CONTRIBUTIONS
All authors have read and agreed to the published version of the
manuscript. All authors listed havemadeasubstantial,direct,and
intellectual contribution to the work and approved it for publication.
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 65763910
REFERENCES
1. Aiom. Linee Guida Tumori DEL Rene. (2019).
2. Pal SK, Ghate SR, Li N, Swallow E, Peeples M, Zichlin ML, et al. Real-World
Survival Outcomes and Prognostic Factors Among Patients Receiving First
Targeted Therapy for Advanced Renal Cell Carcinoma: A SeerMedicare
Database Analysis. Clin Genitourin Cancer (2017) 15:e57382. doi: 10.1016/
j.clgc.2016.12.005
3. Mekhail TM, Abou-Jawde RM, Boumerhi G, Malhi S, Wood L, Elson P, et al.
Validation and Extension of the Memorial Sloan-Kettering Prognostic
Factors Model for Survival in Patients With Previously Untreated
Metastatic Renal Cell Carcinoma. JClinOncol(2005) 23(4):83241.
doi: 10.1200/JCO.2005.05.179
4. Heng DYC, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al.
Prognostic Factors for Overall Survival in Patients With Metastatic Renal
Cell Carcinoma Treated With Vascular Endothelial Growth Factor-
Targeted Agents: Results From a Large, Multicenter Study. J Clin Oncol
(2009) 27(34):57949. doi: 10.1200/JCO.2008.21.4809
5. Choueiri TK, Escudier B, Powles T, Mainwaring PN, Rini BI, Donskov F,
et al. Cabozantinib Versus Everolimus in Advanced Renal-Cell Carcinoma.
N Engl J Med (2015) 373(19):181423. doi: 10.1056/NEJMoa1510016
6. Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK,
et al. Cabozantinib Versus Sunitinib as Initial Targeted Therapy for Patients
With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The
Alliance A031203 CABOSUN Trial. JClinOncol(2017) 35(6):5917.
doi: 10.1200/JCO.2016.70.7398
7. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S,
et al. Nivolumab Versus Everolimus in Advanced Renal-Cell Carcinoma.
N Engl J Med (2015) 373:111. doi: 10.1056/NEJMoa1510665
8. Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B,
Choueiri TK, et al. Nivolumab Plus Ipilimumab Versus Sunitinib in
Advanced Renal-Cell Carcinoma. N Engl J Med (2018) 378(14):127790.
doi: 10.1056/NEJMoa1712126
9. Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, et al. The
Cancer Genome Atlas Comprehensive Molecular Characterization of Renal
Cell Carcinoma. Cell Rep (2018) 23(12):3698. doi: 10.1016/
j.celrep.2018.06.032
10. Lindgren D, Sjölund J, Axelson H. Tracing Renal Cell Carcinomas Back to
the Nephron. Trends Cancer (2018) 4(7):47284. doi: 10.1016/
j.trecan.2018.05.003
11. Hakimi AA, Tickoo SK, Jacobsen A, Sarungbam J, Sfakianos JP, Sato Y, et al.
TCEB1-Mutated Renal Cell Carcinoma: A Distinct Genomic and
Morphological Subtype. Mod Pathol (2015) 28(6):84553. doi: 10.1038/
modpathol.2015.6
12. Hakimi AA, Pham CG, Hsieh JJ. A Clear Picture of Renal Cell Carcinoma.
Nat Genet (2013) 45(8):84950. doi: 10.1038/ng.2708
13. Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, et al.
Integrated Molecular Analysis of Clear-Cell Renal Cell Carcinoma. Nat
Genet (2013) 45(8):8607. doi: 10.1038/ng.2699
14. Kaelin WG. The Von Hippel-Lindau Tumour Suppressor Protein: O2 Sensing
and Cancer. Nat Rev Cancer (2008) 8(11):86573. doi: 10.1038/nrc2502
15. Zhang J, Wu T, Simon J, Takada M, Saito R, Fan C, et al. VHL Substrate
Transcription Factor ZHX2 as an Oncogenic Driver in Clear Cell Renal Cell
Carcinoma. Science (2018) 361(6399):2905. doi: 10.1126/science.aap8411
16. Beksac AT, Paulucci DJ, Blum KA, Yadav SS, Sfakianos JP, Badani KK.
Heterogeneity in Renal Cell Carcinoma. Urol Oncol Semin Orig Investig
(2017) 35(8):50715. doi: 10.1016/j.urolonc.2017.05.006
17. Turajlic S, Xu H, Litcheld K, Rowan A, Horswell S, Chambers T, et al.
Deterministic Evolutionary Trajectories Inuence Primary Tumor Growth:
TRACERx Renal. Cell (2018) 173(3):595610. doi: 10.1016/j.cell.2018.03.043
18. Creighton CJ, Morgan M, Gunaratne PH, Wheeler DA, Gibbs RA,
Robertson G, et al. Comprehensivemolecular Characterization of Clear
Cell Renal Cell Carcinoma. Nature (2013) 499(7456):439. doi: 10.1038/
nature12222
19. Chang Y, An H, Xu L, Zhu Y, Yang Y, Lin Z, et al. Systemic Inammation
Score Predicts Postoperative Prognosis of Patients With Clear-Cell Renal
Cell Carcinoma. Br J Cancer (2015) 113(4):62633. doi: 10.1038/
bjc.2015.241
20. Li J, Byrne KT, Yan F, Yamazoe T, Chen Z, Baslan T, et al. Tumor Cell-
Intrinsic Factors Underlie Heterogeneity of Immune Cell Inltration and
Response to Immunotherapy. Immunity (2018) 49(1):17893. doi: 10.1016/
j.immuni.2018.06.006
21. Grivennikov SI, Greten FR, Karin M. Immunity, Inammation, and Cancer.
Cell (2010) 140(6):88399. doi: 10.1016/j.cell.2010.01.025
22. Wellenstein MD, de Visser KE. Cancer-Cell-Intrinsic Mechanisms Shaping
the Tumor Immune Landscape. Immunity (2018) 48(3):399416.
doi: 10.1016/j.immuni.2018.03.004
23. Meylan E, Dooley AL, Feldser DM, Shen L, Turk E, Ouyang C, et al.
Requirement for NF-kappaB Signalling in a Mouse Model of Lung
Adenocarcinoma. Nature (2009) 462(7269):1047. doi: 10.1038/
nature08462
24. Ancrile B, Lim K-H, Counter CM. Oncogenic Ras-induced Secretion of IL6
is Required for Tumorigenesis. Genes Dev (2007) 21(14):17149.
doi: 10.1101/gad.1549407
25. Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic
Kras-induced Gm-CSF Production Promotes the Development of Pancreatic
Neoplasia. Cancer Cell (2012) 21(6):83647. doi: 10.1016/j.ccr.2012.04.024
26. Schwitalla S, Ziegler PK, Horst D, Becker V, Kerle I, Begus-Nahrmann Y,
et al. Loss of p53 in Enterocytes Generates an Inammatory
Microenvironment Enabling Invasion and Lymph Node Metastasis of
Carcinogen-Induced Colorectal Tumors. Cancer Cell (2013) 23(1):93106.
doi: 10.1016/j.ccr.2012.11.014
27. Nishida J, Momoi Y, Miyakuni K, Tamura Y, Takahashi K, Koinuma D, et al.
Epigenetic Remodelling Shapes Inammatory Renal Cancer and Neutrophil-
Dependent Metastasis. Nat Cell Biol (2020) 22(4):46575. doi: 10.1038/
s41556-020-0491-2
28. Choueiri TK, Figueroa DJ, Fay AP, Signoretti S, Liu Y, Gagnon R, et al.
Correlation of PD-L1 Tumor Expression and Treatment Outcomes in
Patients With Renal Cell Carcinoma Receiving Sunitinib or Pazopanib:
Results From COMPARZ, a Randomized Controlled Trial. Clin Cancer Res
(2015) 21(5):10717. doi: 10.1158/1078-0432.CCR-14-1993
29. Callea M, Albiges L, Gupta M, Cheng SC, Genega EM, Fay AP, et al.
Differential Expression of PD-L1 Between Primary and Metastatic Sites in
Clear-Cell Renal Cell Carcinoma. Cancer Immunol Res (2015) 3(10):1158
64. doi: 10.1158/2326-6066.CIR-15-0043
30. Burrell RA, McGranahan N, Bartek J, Swanton C. The Causes and
Consequences of Genetic Heterogeneity in Cancer Evolution. Nature
(2013) 501(7467):33845. doi: 10.1038/nature12625
31. De LX, Hoang A, Zhou L, Kalra S, Yetil A, Sun M, et al. Resistance to
Antiangiogenic Therapy is Associated With an Immunosuppressive Tumor
Microenvironment in Metastatic Renal Cell Carcinoma. Cancer Immunol
Res (2015) 3(9):101729. doi: 10.1158/2326-6066.CIR-14-0244
32. . scheda AIFA cabozantinib.
33. Powles T, Motzer RJ, Escudier B, Pal S, Kollmannsberger C, Pikiel J, et al.
Outcomes Based on Prior Therapy in the Phase 3 METEOR Trial of
Cabozantinib Versus Everolimus in Advanced Renal Cell Carcinoma. Br J
Cancer (2018) 119(6):6639. doi: 10.1038/s41416-018-0164-0
34. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S,
et al. Nivolumab Versus Everolimus in Advanced Renal-Cell Carcinoma.
N Engl J Med (2015) 373(19):180313. doi: 10.1056/NEJMoa1510665
35. Larkin J, Chiarion-Sileni V, Gonzalez R, Jacques Grob J, Lao CD,
Schadendorf D, et al. Combined Nivolumab and Ipilimumab or
Monotherapy in Previously Untreated Melanoma Corresponding Authors.
N Engl J Med (2015). doi: 10.1056/NEJMoa1504030.Combined
36. Cella D, Grünwald V, Escudier B, Hammers HJ, George S, Nathan P, et al.
Patient-Reported Outcomes of Patients With Advanced Renal Cell
Carcinoma Treated With Nivolumab Plus Ipilimumab Versus Sunitinib
(CheckMate 214): A Randomised, Phase 3 Trial. Lancet Oncol (2019) 20
(2):297310. doi: 10.1016/S1470-2045(18)30778-2
37. Escudier B, Motzer RJ, Tannir NM, Porta C, Tomita Y, Maurer MA, et al.
Efcacy of Nivolumab Plus Ipilimumab According to Number of IMDC Risk
Factors in CheckMate 214. Eur Urol (2020) 77(4):44953. doi: 10.1016/
j.eururo.2019.10.025
38. McDermott DF, Choueiri TK, Motzer RJ, Aren OR, George S, Powles T, et al.
CheckMate 214 Post-Hoc Analyses of Nivolumab Plus Ipilimumab or
Sunitinib in IMDC Intermediate/Poor-Risk Patients With Previously
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 65763911
Untreated Advanced Renal Cell Carcinoma With Sarcomatoid Features.
JClinOncol(2019) 37(15_suppl):4513. doi: 10.1200/JCO.2019.
37.15_suppl.4513
39. Vitale MG, Scagliarini S, Galli L, Pignata S, Lo Re G, Berruti A, et al. Efcacy
and Safety Data in Elderly Patients With Metastatic Renal Cell Carcinoma
Included in the Nivolumab Expanded Access Program (EAP) in Italy. PloS
One (2018) 13(7):e0199642. doi: 10.1371/journal.pone.0199642
40. Donskov F, Motzer RJ, Voog E, Hovey E, Grüllich C, Nott LM, et al.
Outcomes Based on Age in the Phase III METEOR Trial of Cabozantinib
Versus Everolimus in Patients With Advanced Renal Cell Carcinoma. Eur J
Cancer (2020) 126:110. doi: 10.1016/j.ejca.2019.10.032
41. Hale P, Hahn AW, Rathi N, Pal SK, Haaland B, Agarwal N. Treatment
of Metastatic Renal Cell Carcinoma in Older Patients: A Network Meta-
Analysis. J Geriatr Oncol (2019) 10(1):14954. doi: 10.1016/j.jgo.2018.05.010
42. Chen DS, Mellman I. Chen and Mellman (2017) Elements of Cancer
Immunity and the CancerImmune Set Point, Nature. Nature (2017) 541
(7637):32130. doi: 10.1038/nature21349
43. Ott PA, Stephen Hodi F, Buchbinder EI. Inhibition of Immune Checkpoints
and Vascular Endothelial Growth Factor as Combination Therapy for
Metastatic Melanoma: An Overview of Rationale, Preclinical Evidence,
and Initial Clinical Data. Front Oncol (2015) 5:202(SEP). doi: 10.3389/
fonc.2015.00202
44. Amin A, Plimack ER, Ernstoff MS, Lewis LD, Bauer TM, McDermott DF,
et al. Safety and Efcacy of Nivolumab in Combination With Sunitinib or
Pazopanib in Advanced or Metastatic Renal Cell Carcinoma: The
CheckMate 016 Study. J Immunother Cancer (2018) 6(1):109.
doi: 10.1186/s40425-018-0420-0
45. Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C,
et al. Atezolizumab Plus Bevacizumab Versus Sunitinib in Patients
With Previously Untreated Metastatic Renal Cell Carcinoma
(Immotion151): A Multicentre, Open-Label, Phase 3, Randomised
Controlled Trial. Lancet (2019) 393(10189):240415. doi: 10.1016/S0140-
6736(19)30723-8
46. Motzer RJ, Penkov K, Haanen J, et al. Avelumab Plus Axitinib Versus
Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med Overseas Ed
(2019). 38:110315. doi: 10.1056/NEJMoa1816047
47. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al.
Pembrolizumab Plus Axitinib Versus Sunitinib for Advanced Renal-Cell
Carcinoma. NEnglJMed(2019) 380(12):111627. doi: 10.1056/
NEJMoa1816714
48. Rini BI, Motzer RJ, Powles T, McDermott DF, Escudier B, Donskov F, et al.
Atezolizumab Plus Bevacizumab Versus Sunitinib for Patients With
Untreated Metastatic Renal Cell Carcinoma and Sarcomatoid Features: A
Prespecied Subgroup Analysis of the IMmotion151 Clinical Trial. Eur Urol
(2020). doi: 10.1016/j.eururo.2020.06.021
49. Choueiri TK, Motzer RJ, Rini BI, Haanen J, Campbell MT, Venugopal B,
et al. Updated Efcacy Results From the JAVELIN Renal 101 Trial: First-
Line Avelumab Plus Axitinib Versus Sunitinib in Patients With Advanced
Renal Cell Carcinoma. Ann Oncol Off J Eur Soc Med Oncol (2020) 31
(8):10309. doi: 10.1016/j.annonc.2020.04.010
50. Powles T, Plimack ER, Soulières D, Waddell T, Stus V, Gafanov R, et al.
Pembrolizumab Plus Axitinib Versus Sunitinib Monotherapy as First-Line
Treatment of Advanced Renal Cell Carcinoma (KEYNOTE-426): Extended
Follow-Up From a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol
(2020) 21(12):156373. doi: 10.1016/S1470-2045(20)30436-8
51. Procopio G, Nichetti F, Verzoni E. Pembrolizumab Plus Axitinib: Another
Step Ahead in Advanced Renal Cell Carcinoma. Lancet Oncol (2020) 21
(12):15389. doi: 10.1016/S1470-2045(20)30482-4
52. Kfoury M, Oing C. Esmo20 YO for YO: Highlights on Metastatic Renal Cell
Carcinoma-the CheckMate-9ER Trial. ESMO Open (2021) 6(1):100025.
doi: 10.1016/j.esmoop.2020.100025
53. Choueiri TK, Powles T, Burotto M, Bourlon MT, Zurawski B, Oyervides
Juarez VM, et al. 696o_Pr Nivolumab + Cabozantinib vs Sunitinib in First-
Line Treatment for Advanced Renal Cell Carcinoma: First Results From the
Randomized Phase III CheckMate 9ER Trial. Ann Oncol (2020) 31:S1159.
doi: 10.1016/j.annonc.2020.08.2257
54. Rini BI, Pal SK, Escudier BJ, Atkins MB, Hutson TE, Porta C, et al. Tivozanib
Versus Sorafenib in Patients With Advanced Renal Cell Carcinoma (TIVO-
3): A Phase 3, Multicentre, Randomised, Controlled, Open-Label Study.
Lancet Oncol (2020) 21(1):95104. doi: 10.1016/S1470-2045(19)30735-1
55. Albiges L, Barthelemy P, Gross-Goupil M, Negrier S, Needle MN, Escudier
B. TiNivo: Safety and Efcacy of Tivozanib-Nivolumab Combination
Therapy in Patients With Metastatic Renal Cell Carcinoma. Ann Oncol Off
J Eur Soc Med Oncol (2021) 32(1):97102. doi: 10.1016/j.annonc.2020.09.021
56. Powles T. Recent eUpdate to the ESMO Clinical Practice Guidelines on
Renal Cell Carcinoma on Cabozantinib and Nivolumab for First-Line Clear
CellRenalCancer:RenalCellCarcinoma: ESMO Clinical Practice
Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol (2021) 32
(3):4223. doi: 10.1016/j.annonc.2020.11.016
57. Wallis CJD, Klaassen Z, Bhindi B, Ye XY, Chandrasekar T, Farrell AM, et al.
First-Line Systemic Therapy for Metastatic Renal Cell Carcinoma: A
Systematic Review and Network Meta-Analysis [Figure Presented]. Eur
Urol (2018). doi: 10.1016/j.eururo.2018.03.036
58. Heng DYC, Xie W, Regan MM, Harshman LC, Bjarnason GA,
Vaishampayan UN, et al. External Validation and Comparison With
Other Models of the International Metastatic Renal-Cell Carcinoma
Database Consortium Prognostic Model: A Population-Based Study.
Lancet Oncol (2013) 14(2):1418. doi: 10.1016/S1470-2045(12)70559-4
59. Santini D, Stellato M, De Giorgi U, Pignata S, Pantano F, De Lisi D, et al.
Clinical Outcomes of Metastatic Renal Carcinoma Following Disease
Progression to Programmed Death (PD)-1 or PD-L1 Inhibitors (I-O): A
Meet-URO Group Real-World Study (Meet-Uro 7). J Clin Oncol (2020) 38
(6_suppl):6911. doi: 10.1200/jco.2020.38.6_suppl.691
60. Iacovelli R, Ciccarese C, Facchini G, Milella M, Urbano F, Basso U, et al.
Cabozantinib After a Previous Immune Checkpoint Inhibitor in Metastatic
Renal Cell Carcinoma: A Retrospective Multi-Institutional Analysis. Target
Oncol (2020) 17. doi: 10.1007/s11523-020-00732-y
61. Mejean A, Ravaud A, Thezenas S, Colas S, Beauval JB, Bensalah K, et al.
Sunitinib Alone or After Nephrectomy in Metastatic Renal-Cell Carcinoma.
N Engl J Med (2018). doi: 10.1056/NEJMoa1803675
62. Rogers W, Thulasi Seetha S, Refaee TAG, Refaee TAG, Lieverse RIY,
Granzier RWY, et al. Radiomics: From Qualitative to Quantitative
Imaging. Br J Radiol (2020) 93(1108):20190948. doi: 10.1259/bjr.20190948
63. Karlo CA, Di Paolo PL, Chaim J, Hakimi AA, Ostrovnaya I, Russo P, et al.
Radiogenomics of Clear Cell Renal Cell Carcinoma: Associations Between
CT Imaging Features and Mutations. Radiology (2014) 270(2):46471.
doi: 10.1148/radiol.13130663
64. Shinagare AB, Vikram R, Jaffe C, Akin O, Kirby J, Huang E, et al.
Radiogenomics of Clear Cell Renal Cell Carcinoma: Preliminary Findings
of The Cancer Genome Atlas-Renal Cell Carcinoma (Tcga-Rcc) Imaging
Research Group. Abdom Imaging (2015) 40(6):168492. doi: 10.1007/
s00261-015-0386-z
65. Nathan PD, Vinayan A, Stott D, Juttla J, Goh V. CT Response Assessment
Combining Reduction in Both Size and Arterial Phase Density Correlates
With Time to Progression in Metastatic Renal Cancer Patients Treated With
Targeted Therapies. Cancer Biol Ther (2010) 9(1):159. doi: 10.4161/
cbt.9.1.10340
66. Smith AD, Lieber ML, Shah SN. Assessing Tumor Response and Detecting
Recurrence in Metastatic Renal Cell Carcinoma on Targeted Therapy:
Importance of Size and Attenuation on Contrast-Enhanced CT. Am J
Roentgenol (2010) 194(1):15765. doi: 10.2214/AJR.09.2941
67. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, et al.
iRECIST: Guidelines for Response Criteria for Use in Trials Testing
Immunotherapeutics. Lancet Oncol (2017) 18(3):e14352. doi: 10.1016/
S1470-2045(17)30074-8
68. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al.
New Response Evaluation Criteria in Solid Tumours: Revised RECIST
Guideline (Version 1.1). Eur J Cancer (2009) 45(2):22847. doi: 10.1016/
j.ejca.2008.10.026
69. Tozuka T, Kitazono S, Sakamoto H, Yoshida H, Amino Y, Uematsu S, et al.
Dissociated Responses At Initial Computed Tomography Evaluation is a
Good Prognostic Factor in non-Small Cell Lung Cancer Patients Treated
With Anti-Programmed Cell death-1/ligand 1 Inhibitors. BMC Cancer
(2020) 20(1):18. doi: 10.1186/s12885-020-6704-z
70. Thompson RH, Dong H, Kwon ED. Implications of B7-H1 Expression in
Clear Cell Carcinoma of the Kidney for Prognostication and Therapy. Clin
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 65763912
Cancer Res (2007) 13(2 Pt 2):709s15s. doi: 10.1158/1078-0432.CCR-06-
1868
71. Ueda K, Suekane S, Kurose H, Chikui K, Nakiri M, Nishihara K, et al. Prognostic
Value of PD-1 and PD-L1 Expression in Patients With Metastatic Clear Cell
Renal Cell Carcinoma. Urol Oncol (2018) 36(11):499.e916. doi: 10.1016/
j.urolonc.2018.07.003
72. Kahlmeyer A, Stöhr CG, Hartmann A, Goebell PJ, Wullich B, Wach S, et al.
Expression of PD-1 and CTLA-4 are Negative Prognostic Markers in Renal
Cell Carcinoma. J Clin Med (2019) 8(5). doi: 10.3390/jcm8050743
73. Flaifel A, Xie W, Braun DA, Ficial M, Bakouny Z, Nassar AH, et al. Pd-L1
Expression and Clinical Outcomes to Cabozantinib, Everolimus, and
Sunitinib in Patients With Metastatic Renal Cell Carcinoma: Analysis of
the Randomized Clinical Trials METEOR and CABOSUN. Clin Cancer Res
(2019) 25(20):60808. doi: 10.1158/1078-0432.CCR-19-1135
74. Shin S-J, Jeon YK, Cho YM, Lee J-L, Chung DH, Park JY, et al. The Association
Between PD-L1 Expression and the Clinical Outcomes to Vascular Endothelial
Growth Factor-Targeted Therapy in Patients With Metastatic Clear Cell Renal
Cell Carcinoma. Oncologist (2015) 20(11):125360. doi: 10.1634/
theoncologist.2015-0151
75. Iacovelli R, Nolè F, Verri E, Renne G, Paglino C, Santoni M, et al. Prognostic
Role of PD-L1 Expression in Renal Cell Carcinoma. A Systematic Review
and Meta-Analysis. Target Oncol (2016) 11(2):1438. doi: 10.1007/s11523-
015-0392-7
76. Patel SP, Kurzrock R. Pd-L1 Expression as a Predictive Biomarker in Cancer
Immunotherapy. Mol Cancer Ther (2015) 14(4):84756. doi: 10.1158/1535-
7163.MCT-14-0983
77. Zhu J, Armstrong AJ, Friedlander TW, et al. Biomarkers of Immunotherapy
in Urothelial and Renal Cell Carcinoma: PD-L1, Tumor Mutational Burden,
and Beyond. J Immunother Cancer (2018) 6(1):4. doi: 10.1186/s40425-018-
0314-1
78. Kluger HM, Zito CR, Turcu G, Baine MK, Zhang H, Adeniran A, et al. Pd-L1
Studies Across Tumor Types, Its Differential Expression and Predictive
Value in Patients Treated With Immune Checkpoint Inhibitors. Clin Cancer
Res (2017) 23(15):42709. doi: 10.1158/1078-0432.CCR-16-3146
79. Kammerer-Jacquet S-F, Deleuze A, Saout J, Mathieu R, Laguerre B, Verhoest
G, et al. Targeting the PD-1/PD-L1 Pathway in Renal Cell Carcinoma. Int J
Mol Sci (2019) 20(7). doi: 10.3390/ijms20071692
80. Jilaveanu LB, Shuch B, Zito CR, Parisi F, Barr M, Kluger Y, et al. Pd-L1
Expression in Clear Cell Renal Cell Carcinoma: An Analysis of Nephrectomy
and Sites of Metastases. J Cancer (2014) 5(3):16672. doi: 10.7150/jca.8167
81. Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and
Response Rate to PD-1 Inhibition. N Engl J Med (2017) 377(25):25001.
doi: 10.1056/NEJMc1713444
82. Hellmann MD, Ciuleanu T-E, Pluzanski A, Lee JS, Otterson GA, Audigier-
Valette C, et al. Nivolumab Plus Ipilimumab in Lung Cancer With a High
Tumor Mutational Burden. NEnglJMed(2018) 378(22):2093104.
doi: 10.1056/NEJMoa1801946
83. de Velasco G, Miao D, Voss MH, Hakimi AA, Hsieh JJ, Tannir NM, et al.
Tumor Mutational Load and Immune Parameters Across Metastatic Renal
Cell Carcinoma Risk Groups. Cancer Immunol Res (2016) 4(10):8202.
doi: 10.1158/2326-6066.CIR-16-0110
84. Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian
YY, et al. Tumor Mutational Load Predicts Survival After Immunotherapy
Across Multiple Cancer Types. Nat Genet (2019) 51(2):2026. doi: 10.1038/
s41588-018-0312-8
85. Zhang C, Li Z, Qi F, Hu X, Luo J. Exploration of the Relationships Between
Tumor Mutation Burden With Immune Inltrates in Clear Cell Renal Cell
Carcinoma. Ann Transl Med (2019) 7(22):648. doi: 10.21037/atm.2019.10.84
86. Labriola MK, Zhu J, Gupta RT, McCall S, Jackson J, Kong EF, et al.
Characterization of Tumor Mutation Burden, PD-L1 and DNA Repair
Genes to Assess Relationship to Immune Checkpoint Inhibitors Response
in Metastatic Renal Cell Carcinoma. J Immunother Cancer (2020) 8(1).
doi: 10.1136/jitc-2019-000319
87. Yao J, Xi W, Zhu Y, Wang H, Hu X, Guo J. Checkpoint Molecule PD-1-
assisted Cd8(+) T Lymphocyte Count in Tumor Microenvironment Predicts
Overall Survival of Patients With Metastatic Renal Cell Carcinoma Treated
With Tyrosine Kinase Inhibitors. Cancer Manag Res (2018) 10:341931.
doi: 10.2147/CMAR.S172039
88. Senbabaoglu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco G,
et al. Tumor Immune Microenvironment Characterization in Clear Cell
Renal Cell Carcinoma Identies Prognostic and Immunotherapeutically
Relevant Messenger RNA Signatures. Genome Biol (2016) 17(1):231.
doi: 10.1186/s13059-016-1092-z
89. Zhang S, Zhang E, Long J, Hu Z, Peng J, Liu L, et al. Immune Inltration in
Renal Cell Carcinoma. Cancer Sci (2019) 110(5):156472. doi: 10.1111/
cas.13996
90. Kawashima A, Uemura M, Nonomura N. Importance of Multiparametric
Evaluation of Immune-Related T-Cell Markers in Renal-Cell Carcinoma. Clin
Genitourin Cancer (2019) 17(6):e114752. doi: 10.1016/j.clgc.2019.07.021
91. Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, Aizawa M, et al.
Proliferative Activity of Intratumoral CD8(+) T-Lymphocytes as a
Prognostic Factor in Human Renal Cell Carcinoma: Clinicopathologic
Demonstration of Antitumor Immunity. Cancer Res (2001) 61(13):51326.
92. Giraldo NA, Becht E, Pagès F, Skliris G, Verkarre V, Vano Y, et al.
Orchestration and Prognostic Signicance of Immune Checkpoints in the
Microenvironment of Primary and Metastatic Renal Cell Cancer. Clin
Cancer Res (2015) 21(13):303140. doi: 10.1158/1078-0432.CCR-14-2926
93. Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ,
et al. The Intratumoral Balance Between Metabolic and Immunologic Gene
Expression is Associated With Anti-PD-1 Response in Patients With Renal
Cell Carcinoma. Cancer Immunol Res (2016) 4(9):72633. doi: 10.1158/
2326-6066.CIR-16-0072
94.WallinJJ,BendellJC,FunkeR,SznolM,KorskiK,JonesS,etal.
Atezolizumab in Combination With Bevacizumab Enhances Antigen-
Specic T-cell Migration in Metastatic Renal Cell Carcinoma. Nat
Commun (2016) 7:12624. doi: 10.1038/ncomms12624
95. Feng G, Ye X, Fang F, Pu C, Huang H, Li G. Quantication of Plasma Cell-
Free DNA in Predicting Therapeutic Efcacy of Sorafenib on Metastatic
Clear Cell Renal Cell Carcinoma. Dis Markers (2013) 34(2):10511.
doi: 10.3233/DMA-120950
96. Pal SK, Sonpavde G, Agarwal N, Vogelzang NJ, Srinivas S, Haas NB, et al.
Evolution of Circulating Tumor DNA Prole From First-line to Subsequent
Therapy in Metastatic Renal Cell Carcinoma. Eur Urol (2017) 72(4):55764.
doi: 10.1016/j.eururo.2017.03.046
97. Dizman N, Bergerot P, Bergerot C, Lanman RB, Raymond VM, Banks KC,
et al. Exceptional Response to Nivolumab Rechallenge in Metastatic Renal
Cell Carcinoma With Parallel Changes in Genomic Prole. Eur Urol (2018)
73(2):30810. doi: 10.1016/j.eururo.2017.08.006
98. Hahn AW, Gill DM, Maughan B, Agarwal A, Arjyal L, Gupta S, et al.
Correlation of Genomic Alterations Assessed by Next-Generation
Sequencing (NGS) of Tumor Tissue DNA and Circulating Tumor DNA
(ctDNA) in Metastatic Renal Cell Carcinoma (mRCC): Potential Clinical
Implications. Oncotarget (2017) 8(20):3361420. doi: 10.18632/
oncotarget.16833
99. Zurita AJ, Jonasch E, Wang X, Khajavi M, Yan S, Du DZ, et al. A Cytokine
and Angiogenic Factor (CAF) Analysis in Plasma for Selection of Sorafenib
Therapy in Patients With Metastatic Renal Cell Carcinoma. Ann Oncol Off J
Eur Soc Med Oncol (2012) 23(1):4652. doi: 10.1093/annonc/mdr047
100. Voss MH, Chen D, Marker M, Hakimi AA, Lee C-H, Hsieh JJ, et al.
Circulating Biomarkers and Outcome From a Randomised Phase II Trial
of Sunitinib vs Everolimus for Patients With Metastatic Renal Cell
Carcinoma. Br J Cancer (2016) 114(6):6429. doi: 10.1038/bjc.2016.21
101. Deprimo SE, Bello CL, Smeraglia J, Baum CM, Spinella D, Rini BI, et al.
Circulating Protein Biomarkers of Pharmacodynamic Activity of Sunitinib in
Patients With Metastatic Renal Cell Carcinoma: Modulation of VEGF and
VEGF-related Proteins. JTranslMed(2007) 5:32. doi: 10.1186/1479-5876-5-32
102. Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin A-M, et al.
Prognostic or Predictive Plasma Cytokines and Angiogenic Factors for
Patients Treated With Pazopanib for Metastatic Renal-Cell Cancer: A
Retrospective Analysis of Phase 2 and Phase 3 Trials. Lancet Oncol (2012)
13(8):82737. doi: 10.1016/S1470-2045(12)70241-3
103. Gigante M, Li G, Ferlay C, Perol D, Blanc E, Paul S, et al. Prognostic Value of
Serum CA9 in Patients With Metastatic Clear Cell Renal Cell Carcinoma
Under Targeted Therapy. Anticancer Res (2012) 32(12):544751.
104. McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B,
et al. Clinical Activity and Molecular Correlates of Response to Atezolizumab
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 65763913
Alone or in Combination With Bevacizumab Versus Sunitinib in Renal Cell
Carcinoma. Nat Med (2018) 24(6):74957. doi: 10.1038/s41591-018-0053-3
105. Joseph RW, Millis SZ, Carballido EM, Bryant D, Gatalica Z, Reddy S, et al.
PD-1 and PD-L1 Expression in Renal Cell Carcinoma With Sarcomatoid
Differentiation. Cancer Immunol Res (2015) 3(12):13037. doi: 10.1158/
2326-6066.CIR-15-0150
106. Hakimi AA, Voss MH, Kuo F, Sanchez A, Liu M, Nixon BG, et al.
Transcriptomic Proling of the Tumor Microenvironment Reveals
Distinct Subgroups of Clear Cell Renal Cell Cancer: Data From a
Randomized Phase III Trial. Cancer Discovery (2019) 9(4):51025.
doi: 10.1158/2159-8290.CD-18-0957
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2021 Roberto, Botticelli, Panebianco, Aschelter, Gelibter, Ciccarese,
Minelli, Nuti, Santini, Laghi, Tomao and Marchetti. This is an open-access article
distributed under the terms of the Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other forums is permitted, provided the
original author(s) and the copyright owner(s) are credited and that the original
publicationin this journal iscited, in accordance with accepted academic practice. No
use, distribution or reproduction is permitted which does not comply with these terms.
Roberto et al. Metastatic Renal Cell Carcinoma Management
Frontiers in Oncology | www.frontiersin.org April 2021 | Volume 11 | Article 65763914
... mRCC can be treated with Tyrosine Kinase Inhibitors (TKI) or Immunotherapy [5]. Recently, a combination of Immune checkpoint inhibitors like Ipilimumab plus Nivolumab has been approved for Metastatic Renal Cell Carcinoma management [6]. However, intrinsic and therapy-induced heterogeneity, and changes in the tumor microenvironment often yield to multiple cross-resistance mechanisms in non-responder patients. ...
... Phylogenic studies showed that subclonal populations with different mutations arise from the trunk mutations that are present in all regions of the tumor before formation of the founder cancer cells. Those subclones are often responsible of metastasis and are not many to be detected by bulk sequencing [6][7][8][9]. This leads to mRCC patients with a therapeutic response that differs significantly independently of their trunk mutation status [10,11]. ...
Article
Full-text available
Clear cell Renal Cell Carcinoma (ccRCC) is one of the most prevalent kidney cancers, which is often asymptomatic and thus discovered at a metastatic state (mRCC). mRCC are highly heterogeneous tumors composed of subclonal populations that lead to poor treatment response rate. Several recent works explored the potential of ccRCC tumoroids culture derived from patients. However, these models were produced following a scaffold-based method using collagen I or Matrigel that exhibit lot variability and whose complexity could induce treatment response modifications and phenotypic alterations. Following the observation that ccRCC tumoroids can create their own niche by secreting extracellular matrix components, we developed the first scaffold-free tumoroid model of ccRCC tumors. Tumoroids from mice as well as from human tumors were generated with high success rate (≥90%) using a magnetic suspension method and standard culture media. Immunofluorescence analysis revealed their self-organization capacities to maintain multiple tumor-resident cell types, including endothelial progenitor cells. Transcriptomic analysis showed the reproducibility of the method highlighting that the majority of gene expression patterns was conserved in tumoroids compared to their matching tumor tissue. Moreover, this model enables to evaluate drug effects and invasiveness of renal cancer cells in a 3D context, providing a robust preclinical tool for drug screening and biomarker assessment in line with alternative ex vivo methods like tumor tissue slice culture or in vivo xenograft models.
... Most cases of ccRCC are in an advanced stage when they are first diagnosed, and nearly 30% of cases present with metastasis at first diagnosis [3]. In recent years, the clinical application of antiangiogenic drugs has significantly improved the prognosis of patients with advanced ccRCC [4]. However, longterm use of antiangiogenic drugs reduces the sensitivity of ccRCC patients to these drugs [5]. ...
Article
Full-text available
Background The combined use of CDK4/6 inhibitors and mTOR inhibitors has achieved some clinical success in ccRCC. Exploring the underlying mechanism of the CDK4/6 pathway in cancer cells and the drug interactions of CDK4/6 inhibitors in combination therapy could help identify new therapeutic strategies for ccRCC. Notably, CDK4/6 inhibitors inactivate the mTOR pathway by increasing the protein levels of TSC1, but the mechanism by which CDK4/6 inhibitors regulate TSC1 is still unclear. Methods Mass spectrometry analysis, coimmunoprecipitation analysis, GST pull-down assays, immunofluorescence assays, Western blot analysis and RT‒qPCR analysis were applied to explore the relationships among CDK4, RNF26 and TSC1. Transwell assays, tube formation assays, CCK-8 assays, colony formation assays and xenograft assays were performed to examine the biological role of RNF26 in renal cancer cells.TCGA-KIRC dataset analysis and RT‒qPCR analysis were used to examine the pathways affected by RNF26 silencing. Results CDK4/6 inhibitors stabilized TSC1 in cancer cells. We showed that CDK4 enhances the interaction between TSC1 and RNF26 and that RNF26 activates the mTOR signaling pathway in ccRCC, contributes to ccRCC progression and angiogenesis, and promotes tumorigenesis. We then found that RNF26 functions as an E3 ligase of TSC1 to regulate CDK4-induced TSC1. This finding suggested that RNF26 promotes ccRCC progression and angiogenesis to some extent by negatively regulating TSC1. Conclusion Our results revealed a novel CDK4/RNF26/TSC1 axis that regulates the anticancer efficacy of CDK4/6 inhibitors and mTOR inhibitors in ccRCC.
... However, survival is heavily influenced by the stage of diagnosis. Approximately one-third of patients with RCC have metastatic disease, with a five-year survival rate of only 12% [4][5][6][7]. ...
Article
Full-text available
The management of renal cell carcinoma (RCC) has been revolutionized over the past two decades with several practice-changing treatments. Treatment for RCC often requires a multimodal approach: Local treatment, such as surgery or ablation, is typically recommended for patients with localized tumors, while stage IV cancers often require both local and systemic therapy. The treatment of advanced RCC heavily relies on immunotherapy and targeted therapy, which are highly contingent upon histological subtypes. Despite years of research on biomarkers for RCC, the standard of care is to choose systemic therapy based on the risk profile according to the International Metastatic RCC Database Consortium and Memorial Sloan Kettering Cancer Centre models. However, many questions still need to be answered. Should we consider metastatic sites when deciding on treatment options for metastatic RCC? How do we choose between dual immunotherapy and combinations of immunotherapy and tyrosine kinase inhibitors? This review article aims to answer these unresolved questions surrounding the concept of personalized medicine.
... Thus, TKIs, which prevent cell division and growth of new blood vessels, seem to be the most effective therapeutic option (Thomson et al., 2023;Pal et al., 2012). The drugs precisely target the genetic mechanisms based on oncogenesis and proliferation of renal cancer cells (Roberto et al., 2021). ...
Article
Full-text available
Objective: This study aimed to compare the safety profile of tyrosine kinase inhibitors (TKIs) approved for use as monotherapy or combination therapy for the first-line treatment of adult patients with metastatic clear cell renal cell carcinoma (RCC). Methods: A systematic review with frequentist network meta-analysis (NMA) was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We included randomized controlled trials (RCTs) investigating the use of: cabozantinib, pazopanib, sorafenib, sunitinib, tivozanib, cabozantinib + nivolumab, lenvatinib + pembrolizumab, axitinib + avelumab, and axitinib + pembrolizumab in previously untreated adult patients with metastatic clear cell RCC. Eligible studies were identified by two reviewers in MEDLINE (via PubMed), EMBASE, and Cochrane Library. The risk of bias for RCTs was assessed using the Cochrane Collaboration tool. The P score was used to determine the treatment ranking. The mean probability of an event along with the relative measures of the NMA was considered with the treatment rankings. Results: A total of 13 RCTs were included in the systematic review and NMA. Sorafenib and tivozanib used as monotherapy were the best treatment options. Sorafenib achieved the highest P score for treatment discontinuation due to adverse events (AEs), fatigue, nausea, vomiting of any grade, and hypertension of any grade or grade ≥3. Tivozanib achieved the highest P score for AEs, grade ≥3 AEs, dose modifications due to AEs, and grade ≥3 diarrhea. Sunitinib was the best treatment option in terms of diarrhea and dysphonia of any grade, while cabozantinib, pazopanib, and axitinib + pembrolizumab–in terms of grade ≥3 fatigue, nausea, and vomiting. TKIs used in combination were shown to have a poorer safety profile than those used as monotherapy. Lenvatinib + pembrolizumab was considered the worst option in terms of any AEs, grade ≥3 AEs, treatment discontinuation due to AEs, dose modifications due to AEs, fatigue of any grade, nausea, vomiting, and grade ≥3 nausea. Axitinib + avelumab was the worst treatment option in terms of dysphonia, grade ≥3 diarrhea, and hypertension, while cabozantinib + nivolumab was the worst option in terms of grade ≥3 vomiting. Interestingly, among the other safety endpoints, cabozantinib monotherapy had the lowest P score for diarrhea and hypertension of any grade. Conclusion: The general safety profile, including common AEs, is better when TKIs are used as monotherapy vs. in combination with immunological agents. To confirm these findings, further research is needed, including large RCTs.
... Lors du suivi radiologique en septembre 2013, trois nouveaux foyers ronds pulmonaires sont apparus, qui, en raison de l'absence d' enrichissement au TEP/TDM, ont été interprétés comme étant des métastases du carcinome rénal [4]. Celles-ci -il avait été convenu d'attendre la dynamique de l' évolution -présentaient une nette progression en mars 2014, de sorte qu'un nouveau traitement médicamenteux de la tumeur a dû être initié. ...
... In der radiologischen Verlaufskontrolle vom September 2013 fielen drei neue Lungenrundherde auf, die aufgrund fehlender Anreicherung im PET-Scan als Metastasen des Nierenzellkarzinoms [4] bewertet wurden. Diese -es war ein Abwarten der Dynamik vereinbart worden -wiesen im März 2014 einen deutlichen Progress auf, sodass eine neue medikamentöse Tumorbehandlung gestartet werden mussten. ...
... Oligo-metastatic RCC for instance, can be managed with local therapies such as metastasectomy or radiation, which can delay the requirement for systemic therapy and improve survival outcomes [4,5]. More extensive recurrence, or de-novo metastatic disease is more typically managed with systemic therapy such as tyrosine kinase inhibitors, immunotherapy or their combination [6]. Computerised tomography (CT) scan is routinely used for delineation of extent of disease, or staging, and assessing response to treatment. ...
Article
Full-text available
Purpose: There is an emerging role of the use of Prostate-Specific Membrane Antigen (PSMA) Positron Emission Tomography (PET) in renal cell carcinoma. Herein, we report our experience in use of PSMA PET in recurrent or metastatic renal cell carcinoma (RCC). Methods: A retrospective analysis of all patients who underwent PSMA PET for suspected recurrent or de-novo metastatic RCC between 2015 and 2020 at three institutions was performed. The primary outcome was change in management (intensification or de-intensification) following PSMA PET scan. Secondary outcomes included histopathological correlation of PSMA avid sites, comparison of sites of disease on PSMA PET to diagnostic CT and time to systemic treatment.
... Although KIRP has less invasiveness and better prognostic performance compared with ccRCC, it is reported that around 25-35% of patients had distant metastasis in their initial diagnosis. Moreover, previous studies have shown that KIRP patients tend to have worse survival outcomes than ccRCC patients after metastases [2], and the 5-year survival rate of those patients is barely around 12% [3,4]. Therefore, a robust, accurate, and efficient clinical model is highly demanded to predict and evaluate patients' prognoses and identify potentially high-risk individuals. ...
Article
Full-text available
Background: Many studies have demonstrated the crucial roles of 5-methylcytosine (m5C) RNA methylation in cancer pathogenesis. Methods: Two datasets, including TCGA-KIRP and ICGC, and related clinical information were downloaded, where the expression of 13 m5C regulators was examined. We applied LASSO regression to construct a multi-m5C-regulator-based signature in the TCGA cohort, which was further validated using the ICGC cohort. Univariate and multivariate Cox regressions were applied to evaluate the independent prognostic value of our model. The differences in biological functions and immune characterizations between high and low-risk groups divided based on the risk scores were also investigated via multiple approaches, such as enrichment analyses, mutation mining, and immune scoring. Finally, the sensitivities of commonly used targeted drugs were tested, and the connectivity MAP (cMAP) was utilized to screen potentially effective molecules for patients in the high-risk group. Experimental validation was done following qPCR tests in Caki-2 and HK-2 cell lines. Results: 3 m5C regulators, including ALYREF, DNMT3B and YBX1, were involved in our model. Survival analysis revealed a worse prognosis for patients in the high-risk group. Cox regression results indicated our model's superior predictive performance compared to single-factor prognostic evaluation. Functional enrichment analyses indicated a higher mutation frequency and poorer tumor microenvironment of patients in the high-risk group. qPCR-based results revealed that ALYREF, DNMT3B, and YBX1 were significantly up-regulated in Caki-2 cell lines compared with HK-2 cell lines. Molecules like BRD-K72451865, Levosimendan, and BRD-K03515135 were advised by cMAP for patients in the high-risk group. Conclusion: Our study presented a novel predictive model for KIRP prognosis. Furthermore, the results of our analysis provide new insights for investigating m5C events in KIRP pathogenesis.
Article
Renal cell carcinoma (RCC) stands among the top 10 malignant neoplasms with the highest fatality rates. It exhibits pronounced heterogeneity and robust metastatic behavior. Patients with RCC may present with solitary or multiple metastatic lesions at various anatomical sites, and their prognoses are contingent upon the site of metastasis. When deliberating the optimal therapeutic approach for a patient, thorough evaluation of significant risk factors such as the feasibility of complete resection, the presence of oligometastases, and the patient’s functional and physical condition is imperative. Recognizing the nuanced differences in RCC metastasis to distinct organs proves advantageous in contemplating potential treatment modalities aimed at optimizing survival outcomes. Moreover, discerning the metastatic site holds promise for enhancing risk stratification in individuals with metastatic RCC. This review summarizes the recent data pertaining to the current status of different RCC metastatic sites and elucidates their role in informing clinical management strategies across diverse metastatic locales of RCC.
Article
Full-text available
Background Treatment with tivozanib, a highly selective and potent vascular endothelial growth factor receptor tyrosine kinase inhibitor, has demonstrated single-agent efficacy in advanced renal cell carcinoma (RCC) along with minimal off-target toxicities and a favorable adverse event (AE) profile. We report final results from TiNivo, a phase Ib/II study of tivozanib combined with nivolumab. Patients and methods In phase Ib, patients with metastatic RCC received tivozanib 1.0 mg once daily (QD) for 21 days followed by 7 days off treatment (n = 3) or tivozanib 1.5 mg QD (n = 3) plus nivolumab 240 mg every 2 weeks. The maximum tolerated dose was determined to be tivozanib 1.5 mg, and 22 additional patients were enrolled at the maximum tolerated dose for phase II. Primary end points included safety and tolerability, with secondary end points of objective response rate, disease control rate, and progression-free survival. Results In total, 25 patients were treated with tivozanib 1.5 mg QD [12 (48%) treatment-naïve; 13 (52%) previously treated]. Treatment-related grade 3/4 AEs were reported in 20 patients (80%); 4 patients (17%) experienced AEs that led to dose reduction, and 8 (32%) discontinued due to AEs. The objective response rate was 56% (including one complete response) and disease control rate was 96%, with a median time to best response of 7.9 weeks. Twenty patients (80%) had tumor shrinkage. With a median follow-up of 19.0 months (range, 12.6–22.8), median progression-free survival was 18.9 months (95% confidence interval 16.4–not reached) in all patients and was similar in treatment-naïve and previously treated patients. Conclusions Tivozanib plus nivolumab combination therapy showed a generally tolerable AE profile and promising antitumor efficacy. These results support further development of tivozanib combined with nivolumab as a treatment option in patients with treatment-naïve or previously treated metastatic RCC. Clinical Trial number NCT03136627.
Article
Full-text available
Background The phase 3 JAVELIN Renal 101 trial (NCT02684006) demonstrated significantly improved progression-free survival (PFS) with first-line avelumab plus axitinib vs sunitinib in advanced renal cell carcinoma (aRCC). We report updated efficacy data from the second interim analysis. Patients and methods Treatment-naïve patients with aRCC were randomized (1:1) to receive avelumab (10 mg/kg) intravenously every 2 weeks plus axitinib (5 mg) orally twice daily or sunitinib (50 mg) orally once daily for 4 weeks (6-week cycle). The two independent primary endpoints were PFS and overall survival (OS) among patients with PD-L1+ tumors. Key secondary endpoints were OS and PFS in the overall population. Results Of 886 patients, 442 were randomized to the avelumab plus axitinib arm and 444 to the sunitinib arm; 270 and 290 had PD-L1+ tumors, respectively. After a minimum follow-up of 13 months (data cutoff Jan 28, 2019), PFS was significantly longer in the avelumab plus axitinib arm than in the sunitinib arm (PD-L1+ population: hazard ratio [HR] 0.62 [95% CI, 0.490–0.777]; 1-sided P < 0.0001; median 13.8 [95% CI, 10.1–20.7] vs 7.0 months [95% CI, 5.7–9.6]; overall population: HR 0.69 [95% CI, 0.574–0.825]; 1-sided P < 0.0001; median 13.3 [95% CI, 11.1–15.3] vs 8.0 months [95% CI, 6.7–9.8]). OS data were immature (PD-L1+ population: HR 0.828 [95% CI, 0.596–1.151]; 1-sided P = 0.1301; overall population: HR 0.796 [95% CI, 0.616–1.027]; 1-sided P = 0.0392). Conclusion Among patients with previously untreated aRCC, treatment with avelumab plus axitinib continued to result in a statistically significant improvement in PFS vs sunitinib; OS data were still immature.
Article
Objectives: The aim of our study was to collect data about of the outcome of metastatic renal cell carcinoma patients who progressed after immune checkpoint inhibitors in order to enhance data about efficacy and safety of treatment beyond immune-oncology (IO). Materials and methods: A total of 162 eligible patients, progressing to IO, were enrolled from 16 Italian referral centers adhering to the Meet-Uro association. Baseline characteristics, outcome data and toxicities were retrospectively collected. Descriptive analysis was made using median values and ranges. Kaplan-Meier method and Mantel-Haenszel log-rank test were performed to compare differences between groups. Results: A total of 111 patients (68.5%) were treated after IO progression. In all, 51 patients (31.5%) did not receive further treatment for clinical deterioration. Median IO progression free survival (PFS) was 4 months (95% confidence interval [CI]: 3.1-4.8). IO-PFS tends to be longer in patients reporting adverse events (AE) of any grade (5.03 [95% CI: 3.8-6.1] vs. 2.99 [95% CI: 2.4-3.5] months P=0.004). Subsequent therapies included cabozantinib (n=79, 48%), everolimus (n=11, 6.7%), and others (n=21, 12.9%).Median PFS post-IO was 6.5 months (95% CI: 5.1-7.8). Cabozantinib showed longer PFS compared with everolimus (7.6 mo [95% CI: 5.2-10.1] vs. 3.2 mo [95% CI: 1.8-4.5]) (hazard ratio: 0.2; 95% CI: 0.1026-0.7968) and other drugs (4.3 mo [95% CI: 1.3-7.4]) (hazard ratio: 0.6; 95% CI: 0.35-1.23). All grade AE were reported in 83 patients (74%) and G3 to G4 AE in 39 patients (35%). Target therapies post-IO showed median overall survival of 14.7 months (95% CI: 0.3-21.4). Conclusions: In our real world experience after progression to IO, vascular endotelial groth factor-tyrosine kinase inhibitors, given to patients, proved to be active and safe choices. Cabozantinib was associated with a better outcome in terms of median PFS.
Article
Background The first interim analysis of the KEYNOTE-426 study showed superior efficacy of pembrolizumab plus axitinib over sunitinib monotherapy in treatment-naive, advanced renal cell carcinoma. The exploratory analysis with extended follow-up reported here aims to assess long-term efficacy and safety of pembrolizumab plus axitinib versus sunitinib monotherapy in patients with advanced renal cell carcinoma. Methods In the ongoing, randomised, open-label, phase 3 KEYNOTE-426 study, adults (≥18 years old) with treatment-naive, advanced renal cell carcinoma with clear cell histology were enrolled in 129 sites (hospitals and cancer centres) across 16 countries. Patients were randomly assigned (1:1) to receive 200 mg pembrolizumab intravenously every 3 weeks for up to 35 cycles plus 5 mg axitinib orally twice daily or 50 mg sunitinib monotherapy orally once daily for 4 weeks per 6-week cycle. Randomisation was done using an interactive voice response system or integrated web response system, and was stratified by International Metastatic Renal Cell Carcinoma Database Consortium risk status and geographical region. Primary endpoints were overall survival and progression-free survival in the intention-to-treat population. Since the primary endpoints were met at the first interim analysis, updated data are reported with nominal p values. This study is registered with ClinicalTrials.gov, NCT02853331. Findings Between Oct 24, 2016, and Jan 24, 2018, 861 patients were randomly assigned to receive pembrolizumab plus axitinib (n=432) or sunitinib monotherapy (n=429). With a median follow-up of 30·6 months (IQR 27·2–34·2), continued clinical benefit was observed with pembrolizumab plus axitinib over sunitinib in terms of overall survival (median not reached with pembrolizumab and axitinib vs 35·7 months [95% CI 33·3–not reached] with sunitinib); hazard ratio [HR] 0·68 [95% CI 0·55–0·85], p=0·0003) and progression-free survival (median 15·4 months [12·7–18·9] vs 11·1 months [9·1–12·5]; 0·71 [0·60–0·84], p<0·0001). The most frequent (≥10% patients in either group) treatment-related grade 3 or worse adverse events were hypertension (95 [22%] of 429 patients in the pembrolizumab plus axitinib group vs 84 [20%] of 425 patients in the sunitinib group), alanine aminotransferase increase (54 [13%] vs 11 [3%]), and diarrhoea (46 [11%] vs 23 [5%]). No new treatment-related deaths were reported since the first interim analysis. Interpretation With extended study follow-up, results from KEYNOTE-426 show that pembrolizumab plus axitinib continues to have superior clinical outcomes over sunitinib. These results continue to support the first-line treatment with pembrolizumab plus axitinib as the standard of care of advanced renal cell carcinoma. Funding Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc.
Article
Background Angiogenesis has been recognized as the most important factor for tumor invasion, proliferation, and progression in metastatic renal cell carcinoma (mRCC). However, few clinical data are available regarding the efficacy of cabozantinib following immunotherapy.Objective To describe the outcome of cabozantinib in patients previously treated with immunotherapy.Patients and methodsPatients with mRCC who received cabozantinib immediately after nivolumab were included. The primary endpoint was to assess the outcome in terms of efficacy and activity.ResultsEighty-four mRCC patients met the criteria to be included in the final analysis. After a median follow-up of 9.4 months, median overall survival was 17.3 months. According to the IMDC criteria, the rates of patients alive at 12 months in the good, intermediate, and poor prognostic groups were 100%, 74%, and 33%, respectively (p < 0.001). The median progression-free survival (PFS) was 11.5 months (95% CI 8.3–14.7); no difference was found based on duration of previous first-line therapy or nivolumab PFS. The overall response rate was 52%, stable disease was found as the best response in 25.3% and progressive disease in 22.7% of patients. Among the 35 patients with progressive disease on nivolumab, 26 (74.3%) patients showed complete/partial response or stable disease with cabozantinib as best response after nivolumab. The major limitations of this study are the retrospective nature and the short follow-up.Conclusions Cabozantinib was shown to be effective and active in patients previously receiving immune checkpoint inhibitors. Therefore, cabozantinib can be considered a valid therapeutic option for previously treated mRCC patients, irrespective of the type and duration of prior therapies.
Article
Patients with metastatic renal cell carcinoma with sarcomatoid features (sRCC) have a poor prognosis and have shown limited responsiveness to inhibition of the VEGF pathway. We conducted a prespecified analysis of the randomised, phase 3 IMmotion151 trial in previously untreated patients with advanced or metastatic RCC to assess the effectiveness of atezolizumab + bevacizumab versus sunitinib in a subgroup of patients with sarcomatoid features. Patients whose tumour had any component of sarcomatoid features were included and received atezolizumab + bevacizumab (n = 68) or sunitinib (n = 74). Baseline characteristics were similar between the groups. Median progression-free survival was significantly longer in the group receiving atezolizumab + bevacizumab overall (8.3 vs 5.3 mo; hazard ratio [HR] 0.52 95% confidence interval [CI] 0.34–0.79) and in the subset of patients with PD-L1–positive tumours (8.6 vs 5.6 mo; HR 0.45, 95% CI 0.26–0.77). More patients receiving atezolizumab + bevacizumab achieved an objective response (49% vs 14%), including complete responses (10% vs 3%), and reported greater symptom improvements versus sunitinib. Safety was consistent with the known profiles of each drug and with that reported in the overall safety-evaluable population of IMmotion151. This analysis supports enhanced activity of atezolizumab + bevacizumab in patients with sRCC. Patient summary In this report, we looked at patients with a specific type of kidney cancer (tumours with sarcomatoid features) that has been hard to treat. A treatment with two drugs (atezolizumab and bevacizumab) appeared to help patients live longer without the disease getting worse than another drug (sunitinib) that is often used. Patients who took the two drugs also said they were better able to carry out their everyday activities than patients who took sunitinib. The combination of these two drugs may work better in patients with this type of advanced kidney cancer.