Renal cell carcinoma stands out as one of the most immune-infiltrated tumors in pan-cancer comparisons. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy. With evolving frontline options in the metastatic setting, several immune checkpoint blockade regimens have emerged as efficacious, and there is growing interest in characterizing features of tumor biology that can reproducibly prognosticate patients and/or predict the likelihood of their deriving therapeutic benefit. Herein, we review pertinent characteristics of the tumor microenvironment with dedicated attention to candidate prognostic and predictive signatures as well as possible targets for future drug development.

Significance:

Tumor microenvironment features broadly characterizing angiogenesis and inflammatory signatures have shown striking differences in response to immune checkpoint blockade and antiangiogenic agents. Integration of stromal and immune biomarkers may hence produce predictive and prognostic signatures to guide management with existing regimens as well as future drug development.

Renal cell carcinoma (RCC) is the most common form of kidney cancer, claiming more than 14,000 lives per year in the United States (1). Clinically, one third of patients will present with metastatic RCC (mRCC; ref. 2). Coupled with relapsing metastases in a quarter of patients with “local” disease following curative nephrectomy, mRCC constitutes a substantial medical burden (2). Although all RCCs arise from the nephrons and receive similar clinical regimens, histologic subtypes are highly heterogeneous in their biology and therapeutic outcomes (3). The most common (70%–80%) subtype, clear-cell RCC (ccRCC), is also one of the most aggressive (3).

Generally insensitive to cytotoxic chemotherapy and uniquely dependent on tumor angiogenesis, ccRCC emerged early as the prototype solid-tumor malignancy for application of molecularly targeted agents, particularly tyrosine kinase inhibitors (TKI) targeting the VEGFR pathway (2). Additionally, ccRCC has long been categorized as “immunotherapy responsive,” with cytokine-based regimens having been a standard before the advent of antiangiogenic therapy (2). More recently, immune checkpoint inhibitors (ICI) which block PD-1/PD-L1 or CTLA4 T cell–inhibitory receptors have proved highly efficacious in this disease and are now considered standards in treatment-naïve and pretreated patients (4, 5).

Recent data from large registration studies confirm that pairing VEGF-directed therapies with ICIs adds value over TKI monotherapy (6, 7), and this combination is now FDA-approved. This wealth of treatment choices will provide treating oncologists with multiple FDA-approved options in the first- and second-line setting. With no standard predictive biomarker available to help guide treatment selection, clinicians often rely on validated prognostic risk models—the International Metastatic RCC Database Consortium (IMDC) or Memorial Sloan Kettering Cancer Center (MSKCC) criteria (8, 9). These tools integrate clinical and laboratory variables and risk-stratify patients with mRCC into favorable-, intermediate-, or poor-risk disease. Poor prognostic factors include poor performance status, high serum calcium, low hemoglobin concentration, less than 1 year interval from diagnosis to treatment, high serum lactate dehydrogenase (MSKCC only), high neutrophil count (IMDC only), and high platelet count (IMDC only), with 0, >2, or >3 of these factors defining good-, intermediate-, and poor-risk groups, respectively (8, 9). As expected, recent data confirm that these categories serve as surrogates for underlying disease biology (10).

Originally developed for prognostication and stratification on clinical trials, these criteria have recently emerged as tools to help with treatment selection. Specifically, combination ICI therapy with ipilimumab plus nivolumab has shown superiority over TKI monotherapy with sunitinib in patients with intermediate- and poor-risk disease, while being similar in efficacy with long-term follow-up in the favorable-risk setting (11). With the approval of combination TKI plus ICI strategies for ccRCC, IMDC risk stratification is no longer sufficient for treatment selection, as benefit over TKI monotherapy was apparent across all three risk groups (6, 7). Rather, the field awaits biomarker data to recapitulate how these agents synergize to manipulate the stromal and immune tumor microenvironment (TME) to elicit an antitumor response. With that in mind, and through a series of larger-scale analyses from retrospective cohorts and clinical trials, it has become apparent that expression-based analyses are more likely to prove useful in identifying predictive signatures than tumor genomics.

In this review, we first describe the relatively unique and heterogeneous TME of ccRCC. To understand the potential mechanisms of synergy between VEGF TKIs and ICIs, we summarize how these therapies alter the immune TME and highlight specific features important for systemic therapy decision-making.

The truncal event in 90% of ccRCCs is biallelic loss of the Von Hippel-Lindau (VHL) tumor suppressor, which functions as a negative regulator of the HIF1α/2α transcription factors (12). HIF accumulation drives the cellular hypoxic response, leading to increased angiogenesis, glycolysis, and aberrant fatty-acid metabolism, thus bestowing ccRCC with its characteristic glycogen- and lipid-rich cytoplasmic deposits and hypervascularity (13).

It is well appreciated that angiogenesis and immunity are closely interlinked, and targeting VEGF can promote immune surveillance by normalizing dysfunctional tumor vessels (14). In line with this, validated RNA sequencing (RNA-seq)–derived gene signatures showed that ccRCC has the highest angiogenesis score and is also highly immune-infiltrated when compared with 18 epithelial cancer types (15, 16). More specifically, ccRCC has the highest overall T-cell, CD8+, T helper 1 (Th1), dendritic cell (DC), neutrophil, and cytotoxic cell and relatively low Th2 and regulatory T-cell (Treg) scores, suggesting an overall proinflammatory profile (15). However, such pooled analyses can be misleading, as accumulating data continue to highlight the heterogeneity within ccRCC, with distinct subgroups differing in expression of angiogenesis programs, immune infiltration patterns, and clinical outcomes (Fig. 1; refs. 10, 15, 17, 18).

Figure 1.

Emerging model of targeted therapy efficacy within distinct ccRCC subgroups. Transcriptomically distinct prognostic and predictive subgroups of ccRCC can be broadly defined as: (i) immune-silent, characterized by low immune infiltration, high angiogenesis, and altered metabolism;(ii) immune-activated, characterized by proinflammatory immune infiltrates; and (iii) immune-suppressed, characterized by an overall anti-inflammatory cell milieu. Anti-VEGF (α-VEGF), ICI, and combination therapy of α-VEGF and ICI are most effective in highly angiogenic immune-silent tumors, immune-activated tumors, and immune-suppressed tumors, respectively (represented by triangles at top). Anti-VEGF treatment can promote immune infiltration and activation, whereas combination treatment with α-VEGF and ICI may reinvigorate the active antitumor immune response (dashed blue arrows). White boxes below each ccRCC subgroup depict their respective TMEs and potential cellular targets of α-VEGF or ICI therapies. Generally accepted targets for ICIs include inhibiting CTL downregulatory signals derived from Tregs, tumor cells, or other immune cells. Beyond inhibiting angiogenesis, anti-VEGF therapies can also promote CTL recruitment and activation by unclear mechanisms which may include normalizing abnormal tumor vessels, inhibiting MDSCs, promoting Th1 and DC activation, and potentially by inhibiting anti–PD-L1-efficacy-limiting myeloid cells. Th1, T helper 1; Th2, T helper 2; α-PD-1, anti–programmed cell death-1; α-PD-L1, anti–programmed cell death-ligand 1. Created with Biorender.com.

Figure 1.

Emerging model of targeted therapy efficacy within distinct ccRCC subgroups. Transcriptomically distinct prognostic and predictive subgroups of ccRCC can be broadly defined as: (i) immune-silent, characterized by low immune infiltration, high angiogenesis, and altered metabolism;(ii) immune-activated, characterized by proinflammatory immune infiltrates; and (iii) immune-suppressed, characterized by an overall anti-inflammatory cell milieu. Anti-VEGF (α-VEGF), ICI, and combination therapy of α-VEGF and ICI are most effective in highly angiogenic immune-silent tumors, immune-activated tumors, and immune-suppressed tumors, respectively (represented by triangles at top). Anti-VEGF treatment can promote immune infiltration and activation, whereas combination treatment with α-VEGF and ICI may reinvigorate the active antitumor immune response (dashed blue arrows). White boxes below each ccRCC subgroup depict their respective TMEs and potential cellular targets of α-VEGF or ICI therapies. Generally accepted targets for ICIs include inhibiting CTL downregulatory signals derived from Tregs, tumor cells, or other immune cells. Beyond inhibiting angiogenesis, anti-VEGF therapies can also promote CTL recruitment and activation by unclear mechanisms which may include normalizing abnormal tumor vessels, inhibiting MDSCs, promoting Th1 and DC activation, and potentially by inhibiting anti–PD-L1-efficacy-limiting myeloid cells. Th1, T helper 1; Th2, T helper 2; α-PD-1, anti–programmed cell death-1; α-PD-L1, anti–programmed cell death-ligand 1. Created with Biorender.com.

Close modal

Early efforts at molecular characterization of RCC by development of a 34-gene expression signature derived from microarray data allowed separation of patients into two broad prognostic groups, ccA and ccB (17, 19). These were characterized by high angiogenesis and improved prognosis or high lymphocytes and macrophages and worse survival, respectively (20). However, when performing simple unsupervised clustering of microarray data in 53 ccRCCs, four molecular groups could be resolved including an immune-infiltrated group and three noninfiltrated groups. The noninfiltrated groups seemed to outline a spectrum of disease progression from normal tissue-like to upregulation of hypoxia and glycolysis followed by MYC upregulation (21).

Larger analyses using single-sample gene set enrichment analysis of 415 primary tumor samples from patients with metastatic ccRCC within The Cancer Genome Atlas (TCGA) database allowed further subcategorization of these immune-infiltrated tumors (15). Again, a non–immune-infiltrated subgroup resembling normal kidney and a poorly immune-infiltrated group with high angiogenesis resembling ccA tumors were identified. Interestingly, within the T cell–enriched cluster, two distinct subclusters emerged, termed TCa and TCb. Although patients with TCa had similar survival to noninfiltrated and poorly infiltrated groups, TCb had the overall poorest survival and was significantly enriched with macrophages, stromal cells, and immunosuppressive Treg and Th2 cell signatures (15).

Similar molecular subgroups were validated at the protein level by Giraldo and colleagues when combining gene-expression data with detailed flow cytometry phenotyping of tumor-infiltrating lymphocytes (TIL). Analysis of tumors from 40 localized ccRCC tumors identified three distinct groups: immune silent, immune activated, and immune regulated (22). Whereas TILs in the immune-silent group resembled nonmalignant kidney resident TILs, the immune-activated and immune-regulated groups were infiltrated by activated CD8+ T cells. However, T cells in the immune-regulated group were less clonal and poorly cytotoxic, with more Tregs and dysfunctional DCs, compared with immune-activated tumors. The immune-regulated group, analogous with TCb tumors, was enriched with high-grade tumors (74% grades 3–4) and had the highest risk of relapse. More recent studies also support that inflamed tumors have worse survival compared with noninflamed but highly angiogenic tumors, when patients were categorized using a novel deconvolution method utilizing RCC-specific immune and stromal genes (23). It would be useful to validate and refine the existence of immune-activated and immune-regulated clusters using these RCC-specific signatures.

In summary, these data suggest that highly immune-infiltrated tumors are more aggressive than angiogenic tumors, consistent with observations across IMDC risk groups (10). However, T-cell activation state within infiltrated tumors is a stronger prognostic indicator. Unlike most other cancer types, overall high CD8+ T-cell infiltration in ccRCC is associated with worse prognosis, suggesting that the infiltrating CD8+ T-cell pool may be dominated by suppressed and dysfunctional cells. Indeed, defective T-cell function has been reported in many ccRCC studies (24–31). Thus, simple measurement of single populations such as CD8+ T cells is unlikely to provide sufficient prognostic or predictive information.

Tumor Immunogenicity

It is generally thought that tumor-specific mutations give rise to immunogenic neoantigens that can drive immune infiltration, a prerequisite for ICI efficacy. Accordingly, tumor mutation burden (TMB) and genomic instability serve as robust predictors of ICI response in many malignancies (32). ccRCC is a historically immunogenic cancer, but the precise nature of the T cell–activating neoantigens still eludes us. Interestingly, among other highly infiltrated cancer types such as lung adenocarcinoma and melanoma, ccRCC stands out as having only a modest mutation burden (15). TMB is similar between MSKCC prognostic risk groups (33), and immune infiltration is independent of predicted neoantigen affinity (15). Ultimately, TMB alone does not predict RCC response to ICI (18, 34–36).

Although nonsynonymous single-nucleotide variants (nsSNV) are the most established readout of TMB, frameshifts caused by small nucleotide insertions or deletions (indels) are predicted to generate more immunogenic neoantigens than nsSNVs. In pan-cancer comparisons, indels are strikingly abundant in ccRCC, perhaps explaining the disparity between low SNVs and high immune infiltration (34). However, in the IMmotion-150 phase II trial, indels and frameshift burdens were not significantly associated with response to atezolizumab (anti–PD-L1) alone or combined with bevacizumab (anti-VEGF; ref. 18).

Reactivated expression of the germline-encoded gene CSAG2 (a cancer testis antigen) and expression of 7 different endogenous retroviruses (ERV) in ccRCC are positively associated with cytolytic activity score [a GZMB- and PRF1-based gene-expression metric strongly associated with cytotoxic T lymphocyte abundance (CTL); ref. 37]. Whether CSAG2- and ERV-derived neoantigens can elicit T-cell responses and influence ICI efficacy is unknown, but these alterations occur relatively infrequently. Ongoing efforts to identify immunogenic RCC antigens are critical for future development of novel vaccination or adoptive cell transfer approaches.

Tumor immunogenicity is also dependent on successful presentation of tumor neoantigens by antigen-presenting cells. High levels of somatic copy-number alterations (SCNA) are independently associated with immune evasion across cancer types, including ccRCC, and predict worst response to ICI in melanoma (35). Interestingly, reduction of cytotoxic CD8+ and natural killer (NK) cells was predicted only by chromosomal and arm-level but not focal SCNA events, despite a significant overlap in genes. The authors speculate that the protein imbalance may impair key signals, such as neoantigen peptide presentation, which are required for immunogenicity.

Moreover, high SCNA ccRCC tumors were associated with significantly decreased ratios of pro- and anti-inflammatory cells and cytokines, and lower ratios of proinflammatory M1/anti-inflammatory M2 macrophages compared with low SCNA tumors, together suggesting that high tumor SCNA promotes both immune exclusion and an immunosuppressive microenvironment (35). Given these significant effects on the inflammatory milieu and the recurrent patterns of SCNA occurring in ccRCC (38), interrogation of the predictive power of SCNA in ICI therapy is warranted.

T-cell Suppression

In highly infiltrated ccRCC tumors, T-cell activation state is a pivotal determinant of ccRCC prognosis and likely of immunotherapy response (discussed below). To determine which cell types may mediate T-cell suppression, coinfiltration analyses were performed on over 7,000 tumors from the TCGA database spanning 23 different cancer types (39). Consistent with the TCa and TCb groups observed in ccRCC (15), in 5 other cancer types (lung adenocarcinoma, glioma, pancreatic, ovarian, and bladder), tumors with high CD8+ T-cell infiltrates could be subdivided into prognostic groups in which low macrophage levels dictated longer survival times (39). This is also true of ovarian and lung cancers with low CD8+ infiltrates, highlighting the T cell–dependent and –independent tumor-promoting effects of macrophages, which are well reviewed elsewhere (40).

Recently, mass cytometry was used to comprehensively phenotype tumor resident T cells and tumor-associated macrophages (TAM) from 73 patients with ccRCC spanning stage I to IV and metastatic disease, including adjacent normal kidney (29). T cells were the most prevalent immune subset (50%) followed by TAMs (25%), NK cells (9%), B cells (4%), and other low-frequency cells including plasma cells, DCs, and neutrophils. The authors teased out 20 T-cell and 17 macrophage clusters, highlighting the sheer heterogeneity of cell subtypes and substates.

In agreement with other data, tumor-infiltrating T cells (abundant in tumors but not normal or early-stage disease) were predominantly CD8+ cells expressing high levels of coinhibitory receptors including PD-1 and low levels of the proliferation marker Ki-67, indicative of a near-exhausted phenotype. Tregs also comprised a significant proportion, together depicting a largely exhausted and immunosuppressive T-cell landscape in advanced ccRCC tumors.

In contrast to T cells, human macrophage subsets are far less characterized and exhibited more heterogeneous marker expression across clusters. Interestingly, three prevalent mature TAM populations distinct from normal kidney resident macrophages were identified, termed M5, M11, and M13, and tended not to co-occur. Instead M5 TAMs, characterized by high CD38 expression, were positively associated with the number of Tregs or exhausted T cells. CD38 is upregulated in human monocytes and macrophages in response to classic proinflammatory stimulus (lipopolysaccharide) but not the alternative macrophage stimulus IL4 (41) and has been demonstrated to be critical for macrophage FcγR-mediated phagocytosis, albeit only in mice (42). This suggests that the CD38+ M5 macrophage subset may be proinflammatory in nature, promoting initial T-cell activation and eventual exhaustion in ccRCC, but further functional characterization is required.

In contrast, the M11 and M13 TAMs were mutually exclusive with activated and exhausted T cells. In fact, M11 seems to be mutually exclusive with all T-cell clusters. It is tempting to speculate that these M11 TAMs may be involved in T-cell exclusion, a phenomenon recently reported in human lung tumors and mouse breast cancer models (43). Fitting with this hypothesis, low M5, high M11, and high M13 macrophages corresponded to poor survival in ccRCC, but validation is required in larger cohorts (29). Of note, three sizeable macrophage clusters expressed high levels of the commonly used protumor M2 macrophage marker CD206. These include M11 TAMs but also two potentially nonpathogenic tissue-resident macrophage populations. In sum, this crucial study highlights the complexity and need to cocharacterize TAM subsets and T-cell activation states in predictive and functional studies.

DCs can also modulate TIL function in ccRCC. Tertiary lymphoid structures (TLS) have been shown to exist in various human tumors including RCC and are sites of T-cell activation with immune organization analogous to lymph nodes (44). Localization of mature DCs within TLS was associated with better survival in patients with ccRCC with high levels of activated CD8+ T cells (22, 28). In contrast, more DCs located outside of TLS were associated with coinfiltration of dysfunctional CD8+ T cells and poor survival, together suggesting that local T-cell priming by DCs within TLS is a key component of effective tumor immune surveillance.

VEGF-Directed Effects on TME

Current FDA-approved antiangiogenesis agents have known immunomodulatory effects through both VEGF and their effects on other tyrosine kinase receptors. For instance, presurgical bevacizumab reduces total CD68+ macrophages (45). Bevacizumab treatment has also been shown to increase gene signatures related to Th1 responses, and high CD8+ T-cell infiltration has been shown with combination bevacizumab and atezolizumab (46). Presurgical pazopanib is associated with a reduction in CD8+ T-cell infiltration, increased PD-L1 expression, and enhanced DC activation proposed through downregulation of the pERK/β-catenin pathway (47, 48). Sunitinib is associated with a reduction in Tregs and myeloid-derived suppressor cells (MDSC) with an improved Th1 response (49, 50). This effect has therapeutic implications, as reductions in Tregs during sunitinib treatment are associated with prolonged survival (51). Sunitinib has also been shown to enhance overall T-cell infiltration, including Tregs, CD8+ cells, and CD4+ cells (52). This is accompanied by increased tumor cell PD-L1 expression and negative associations for survival, suggesting that sunitinib may enhance initial immune responses but cannot prevent subsequent immunosuppression. As each agent may demonstrate distinct immune compartmental changes and downstream impacts on survival (10), continued large-scale efforts which characterize these changes will be crucial in designing novel combination strategies.

Several studies suggest that RNA-based molecular signatures from tumor tissue can have predictive and prognostic implications for VEGFR TKI therapy, possibly more so than traditional IMDC risk stratification. For instance, Beuselinck and colleagues performed unsupervised cluster analysis to distinguish 4 subgroups of RCC tumors: ccrcc1–4 (21). Patients with ccrcc1/4 tumors had lower response rates and shorter progression-free survival (PFS) and overall survival (OS) when compared with ccrcc2/3 tumors to both sunitinib and pazopanib therapy (21, 53). Further analyses delineated broader differences between these tumor subgroups: ccrcc2 tumors were hallmarked by angiogenesis-high gene signatures, and ccrcc3 tumors had indolent disease courses. In contrast, ccrcc1 tumors had lower rates of T-cell infiltration and were typically “immune-cold,” and ccrcc4 tumors had high T-cell infiltration with high expression of inhibitory markers like PD-1, cognate ligands PD-L1/PD-L2, LAG3, and TGFβ, and high expression of activated myeloid cells with high CSF1 expression. Subsequent analyses suggest that these signatures may also inform surgical decision-making for patients considered for metastasectomy (54).

Large-scale correlative work from the COMPARZ trial, a phase III study of patients with treatment-naïve metastatic RCC treated with sunitinib or pazopanib, has uncovered several TME factors associated with outcomes on VEGFR TKI therapy (10). In this analysis, four biologically distinct groups were identified by uninformed RNA-based consensus clustering with significant differences in median PFS and OS. Among the groups, cluster 3 had the best OS and was associated with the highest angiogenesis scores. Forty-one percent of patients with ccRCC harbor PBRM1 loss-of-function mutations (55). PBRM1 mutations were enriched in cluster 3 and were also associated with improved response to sunitinib in a separate cohort (18), fitting with its known roles in augmenting the HIF-mediated hypoxia response signature of VHL-deficient tumors (56).

Cluster 4 had the worst OS and was enriched for BAP1 and TP53 mutations. In addition, cluster 4 demonstrated high enrichment for immune and particularly macrophage gene signatures with associated high PD-L1+ macrophage enrichment. High macrophage infiltration was associated with poor OS (HR, 1.54; P = 0.0018), and angiogenesis-low/macrophage-high signatures had the worst OS with TKI therapy (HR, 3.12; P = 0.000003). However, when considering the two TKIs separately, angiogenesis had only marginal impact on survival within the pazopanib cohort but remained significantly predictive for improved sunitinib response. These data remain in line with results from the phase II IMmotion-150 study in which sunitinib achieved improved PFS in angiogenesis-high tumors compared with angiogenesis-low tumors [HR, 0.31; 95% confidence interval (CI), 0.18–0.55; ref. 18]. In contrast, macrophages were significantly predictive for poor response to pazopanib but not sunitinib, emphasizing that different TKIs can modulate the TME in different ways. Supporting this, hepatocellular carcinoma models implanted into immunocompetent and immunodeficient mice show that the antitumor activity of the antiangiogenesis TKI lenvatinib but not sorafenib is dependent on adaptive immunity (57). In sum, these data support the characterization of stromal and cellular immune components of the TME in an effort to develop RNA-based biomarker signatures for VEGF-directed therapies (10).

Immune Checkpoint Blockade and TME

The differences in therapy response and survival when comparing combination ICI therapy and VEGFR TKI therapies are rooted in underlying tumor biology. In the phase III registration study of ipilimumab and nivolumab versus sunitinib, patients with intermediate- to poor-risk disease had superior survival and response compared with favorable-risk patients, setting this regimen apart from others (4). In IMmotion-151, a phase III study of atezolizumab plus bevacizumab versus sunitinib, angiogenesis-high signatures were more prevalent in MSKCC favorable-risk patients compared with intermediate- and poor-risk patients (P = 4.28e-06; ref. 58). TME may also significantly influence response differences seen across sarcomatoid dedifferentiated tumors. Subgroup analyses of patients with sarcomatoid treated with combination ICI or VEGF plus ICI therapy have shown impressive response rates, including a complete response in 18.3% for ipilimumab plus nivolumab (59), 11.8% for pembrolizumab plus axitinib (60), and 10% for atezolizumab plus bevacizumab (61). As these tumors have higher PD-L1 expression and T-effector gene signatures, with lower angiogenesis signatures, compared with their nonsarcomatoid counterparts (61), this may provide rationale for the robust responses seen to combination ICI therapy.

Specific TME features that correlate with combination VEGF plus ICI therapy response have been gleaned from the above-mentioned IMmotion-150 study, a phase II 3-arm randomized study of atezolizumab and bevacizumab, and atezolizumab and sunitinib monotherapy. Consistent with data from the COMPARZ cohort (10), sunitinib had improved PFS and response rate in those tumors harboring angiogenesis-high versus angiogenesis-low signatures. This difference in both response and survival rates was not seen in the atezolizumab monotherapy arm, suggesting that angiogenesis is not predictive of anti–PD-L1 response. When directly comparing outcomes for atezolizumab and sunitinib monotherapy, no difference was seen in PFS overall nor when stratified by levels of angiogenesis or T-effector cells. Overall, the combination arm was only significantly superior to sunitinib in patients with >1% PD-L1–expressing immune cells, emphasizing the necessity for prior immune activation in tumors for ICI combination efficacy. Further, angiogenesis-low tumors had improved PFS with combination ICI plus VEGF therapy when compared with sunitinib monotherapy. Interestingly, when stratifying T-effector–high tumors by myeloid signature abundance, the combination arm was superior to atezolizumab monotherapy in myeloid-high but not myeloid-low groups, suggesting that myeloid immunosuppression hinders single-agent ICI therapy, and that the immunomodulatory effects of concurrent VEGF inhibition can help overcome this (18). Validation of these molecular analyses was confirmed in the phase III IMmotion-151 study, where T-effector signatures were associated with superior PFS with combination atezolizumab plus bevacizumab when compared with sunitinib monotherapy (HR, 0.76; 95% CI, 0.59–0.99), and high T-effector gene signatures were associated with increased PD-L1 expression. Further, combination therapy is associated with superior PFS compared with sunitinib in angiogenesis-low tumors (HR, 0.68; 95% CI, 0.52–0.89; refs. 58, 62). Recently, PBRM1 was discovered to confer resistance to T cell–induced apoptosis, and deletion in a B16F10 melanoma mouse model increases susceptibility to anti–PD-1 and anti-CTLA4 (63). In small RCC cohorts, PBRM1 status was also suggested to be a positive predictor of anti–PD-1 efficacy (64), but this signal was not seen in the context of anti–PD-L1 therapy (18). Given these conflicting data, validation in larger cohorts is required, and PBRM1 mutations should not be used as biomarkers at present.

Novel Approaches to Manipulating the TME

With the identification of specific TME constituents and their recognized impact on treatment responses, combination strategies that target distinct cell populations may help overcome treatment resistance. As highlighted here, TAMs significantly contribute to VEGFR TKI and ICI therapy resistance, and targeting of macrophage-specific pathways may represent a novel treatment for patients with advanced RCC. Direct blockade of CSF1 and its cognate receptor CSF1R, responsible for macrophage recruitment and differentiation, and CD47/SIRPα, responsible for transmitting a protective “don't eat me” signal, may represent new therapeutic avenues for investigation for patients harboring macrophage-high tumors (65, 66). The institution of these new therapies may be facilitated by the integration of rapid biomarker assessment tools and incorporation of novel radiomic approaches. Ferumoxytol-enhanced MRI or hybrid MR/PET scans using alternative tracers can identify TAMs (67, 68), and combination imaging modalities have shown high concordance between imaging and RNA-seq data distinguishing angiogenesis-low or angiogenesis-high tumors (69). Application of these modalities may also yield similar classifying metrics; for example, hybrid PET/MRI has been shown to distinguish tumors into ccA and ccB classifications represented by either angiogenesis-high or TGFβ/epithelial-to-mesenchymal transition–high tumor differentiation (70). Integration of these tools will lead to future biomarker-driven studies which help select and stratify patients based on individual TME characteristics for response evaluation.

Agents that additionally target other immunosuppressive populations through alternative mechanisms serve as attractive strategies for paired or enhanced ICI response. Entinostat, a histone deacetylase inhibitor, has shown inhibitory effects on monocytes and neutrophils (71), and paired ICI therapies to target immunosuppressive MDSCs or TAMs using this approach are under way (NCT02526017; ref. 72). Early-phase studies of X4P-001, a CXCR4 inhibitor which downregulates HIF2α with preliminary efficacy in patients with advanced RCC, also may downregulate MDSC trafficking (73). Further, pegilodecakin, a pegylated IL10 which increases CD8+ T cells and reduces TGFβ, has shown preliminary efficacy when combined with an anti–PD-1 agent, but interestingly has shown hemophagocytic lymphohistiocytosis (HLH; 7.9% any grade, n = 38) as a reported treatment side effect. As HLH only occurred in this small cohort of patients with RCC and preliminarily not in other solid tumors, whether this effect represents a heightened macrophage activation state due to the high infiltration of TAMs in RCC specifically is unclear. The prodrug NKTR-214, a CD-122 agonist designed to provide sustained signaling through the IL2 receptor pathway, has been shown to activate and expand NK and CD8+ effector T cells over Tregs. In a phase I trial, serial TME analyses during treatment showed that NKTR-214 induces an increase in total and proliferating NK cells, CD8+ cells, and CD4+ cells, with correlations between peripheral immune cells and immune infiltration in the tumor (74). Preliminary results for the combination of NKTR-214 with ICI have shown clinical responses, and higher clinical benefit for those patients whose tumors were PD-L1+ at baseline or those tumors which converted to PD-L1+ during treatment (75).

Metabolic-based therapeutic strategies which alter the TME also remain under active investigation in early-phase studies. CB-839, a selective glutaminase inhibitor which blocks formation of glutamate metabolites and cell proliferation, may have additional effects as other constituents of the TME compete for nutrients (76). Combination strategies utilizing CB-839 with VEGFR TKI, mTOR inhibitors, and ICI agents are ongoing (NCT03428217, NCT03163667, NCT02771626). Metabolic changes with CB-839 and downstream oxidative stress may be captured radiographically, and new PET avid tracers of glutamine metabolism which capture glutamine and glutamate conversion may aid in monitoring treatment response with therapeutic use (77). In a small cohort of patients with advanced RCC, peripheral blood metabolic analysis highlights additional metabolic pathways including indoleamine 2,3-dioxygenase and adenosine and associations with ICI therapy response. Patients with no response to PD-1 monotherapy had significantly higher adenosine levels at baseline and while on-treatment, and high levels were associated with a significantly worse PFS (P = 0.004; ref. 78). The use of CPI-444, an A2A receptor antagonist, may overcome this dampening and promote immune responses when combined with ICIs (NCT02655822). On-treatment tumor biopsies also show high associations between CD8+ T-cell infiltration and response with CPI-444 treatment when compared with baseline samples (P < 0.016), and implementation of an adenosine gene signature composed of chemokines and cytokines associated with myeloid-derived suppression was also associated with response (P < 0.008; ref. 79). Interestingly, this adenosine signature is similar to the myeloid-high signature previously used for characterization in the IMmotion-150 trial, supporting the role of adenosine as an additional mechanism for tumor escape (80).

Molecular classification of RCC into tumors harboring high expression of angiogenic or inflammatory signatures is associated with differential responses to VEGF-directed and ICI-based therapies. Whether these distinct ccRCC subgroups exist along a spectrum of high angiogenesis followed by immune infiltration and eventual immunosuppression is not yet known, but given their associations with prognosis and stage, this would not be surprising.

TAMs play a significant role in both VEGFR TKI and ICI therapy resistance, and directed attention to macrophages and other immunosuppressive compartments in the RCC TME provides opportunity to salvage therapeutic response in refractory patients. Although several macrophage-depletion agents are under clinical evaluation, TAMs are highly heterogeneous and it is likely that targeting specific TAM subpopulations may lead to better outcomes than pan-macrophage ablation. Further comprehensive functional characterization of ccRCC TAMs and their interplay with T cells is greatly needed and will likely be guided by emerging high-resolution technologies such as single-cell RNA-seq (scRNA-seq).

We have discussed potentially treatment-altering insights made possible by large-scale genomic analyses of clinical trial specimens, but it is important to note the limitations of current genomic tools. Conflicting reports on the prognostic effects of macrophages in ccRCC (10, 22, 39) may be confounded by the high degree of macrophage heterogeneity and highlight the inherent challenge this poses for the generation of cell type–specific gene signatures for accurate quantification of immune populations from bulk RNA-seq data. Indeed, existing immune gene signatures were not sufficiently representative of RCC-specific transcripts and immune populations (23). Development of RCC-specific signatures could greatly improve the accuracy of ccRCC immune deconvolution, but further refinement of these signatures is needed, as a small proportion of leukocytes were still misidentified (23).

Finally, deep sequencing of multiple regions of primary and metastatic tumors from more than 100 patients with ccRCC revealed that single-region biopsies relied on to date are unlikely to capture the full spectrum of tumor subclones and that metastases are significantly more clonal than primary tumors (81, 82). This has fundamental implications for the interpretation of the majority of published studies that infer TME characteristics and therapeutic outcomes of metastatic clones from the phenotypes of fully excised primary tumors. Moreover, important spatial or coinfiltration patterns can be missed when relying fully on genomics and protein cytometry (28, 83). Going forward, multiregion assessments and integrative genomic and imaging-based models will be critical for robust clarification of the relationships between tumor genomics, the TME, and their impact on immunotherapy outcomes.

M.H. Voss reports receiving commercial research grants from BMS and Genentech and is a consultant/advisory board member for Alexion, Bayer, Calithera Biosciences, Corvus Pharmaceuticals, Exelixis, Eisai, GSK, Natera, Novartis, and Pfizer. No potential conflicts of interest were disclosed by the other authors.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2018
.
CA Cancer J Clin
2018
;
68
:
7
30
.
2.
Choueiri
TK
,
Motzer
RJ
. 
Systemic therapy for metastatic renal-cell carcinoma
.
N Engl J Med
2017
;
376
:
354
66
.
3.
Linehan
WM
. 
Genetic basis of kidney cancer: role of genomics for the development of disease-based therapeutics
.
Genome Res
2012
;
22
:
2089
100
.
4.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
90
.
5.
Motzer
RJ
,
Fukasawa
S
,
Shinohara
N
,
Kitamura
H
,
Oya
M
,
Eto
M
, et al
Nivolumab versus everolimus in advanced renal-cell carcinoma
.
N Engl J Med
2015
;
373
:
1803
13
.
6.
Motzer
RJ
,
Penkov
K
,
Haanen
J
,
Rini
B
,
Albiges
L
,
Campbell
MT
, et al
Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
2019
;
380
:
1103
15
.
7.
Rini
BI
,
Plimack
ER
,
Stus
V
,
Gafanov
R
,
Hawkins
R
,
Nosov
D
, et al
Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
2019
;
380
:
1116
27
.
8.
Motzer
RJ
,
Mazumdar
M
,
Bacik
J
,
Berg
W
,
Amsterdam
A
,
Ferrara
J
. 
Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma
.
J Clin Oncol
1999
;
17
:
2530
40
.
9.
Heng
DY
,
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
:
5794
9
.
10.
Hakimi
AA
,
Voss
MH
,
Kuo
F
,
Sanchez
A
,
Liu
M
,
Nixon
BN
, et al
Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer - data from a randomized phase III trial
.
Cancer Discov
2019
;
9
:
510
25
.
11.
Tannir
NM
,
Frontera
OA
,
Hammers
HJ
,
Carducci
MA
,
McDermott
DF
,
Salman
P
, et al
Thirty-month follow-up of the phase III CheckMate 214 trial of first-line nivolumab + ipilimumab (N+I) or sunitinib (S) in patients (pts) with advanced renal cell carcinoma (aRCC)
.
J Clin Oncol
2019
;
37
:
547
.
12.
Hsieh
JJ
,
Le
VH
,
Oyama
T
,
Ricketts
CJ
,
Ho
TH
,
Cheng
EH
. 
Chromosome 3p loss-orchestrated VHL, HIF, and epigenetic deregulation in clear cell renal cell carcinoma
.
J Clin Oncol
2018
;
JCO2018792549
.
13.
Hakimi
AA
,
Reznik
E
,
Lee
CH
,
Creighton
CJ
,
Brannon
AR
,
Luna
A
, et al
An integrated metabolic atlas of clear cell renal cell carcinoma
.
Cancer Cell
2016
;
29
:
104
16
.
14.
Fukumura
D
,
Kloepper
J
,
Amoozgar
Z
,
Duda
DG
,
Jain
RK
. 
Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges
.
Nat Rev Clin Oncol
2018
;
15
:
325
40
.
15.
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 identifies prognostic and immunotherapeutically relevant messenger RNA signatures
.
Genome Biol
2016
;
17
:
231
.
16.
Yoshihara
K
,
Shahmoradgoli
M
,
Martínez
E
,
Vegesna
R
,
Kim
H
,
Torres-Garcia
W
, et al
Inferring tumour purity and stromal and immune cell admixture from expression data
.
Nat Commun
2013
;
4
:
2612
.
17.
Brooks
SA
,
Brannon
AR
,
Parker
JS
,
Fisher
JC
,
Sen
O
,
Kattan
MW
, et al
ClearCode34: a prognostic risk predictor for localized clear cell renal cell carcinoma
.
Eur Urol
2014
;
66
:
77
84
.
18.
McDermott
DF
,
Huseni
MA
,
Atkins
MB
,
Motzer
RJ
,
Rini
BI
,
Escudier
B
, et al
Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma
.
Nat Med
2018
;
24
:
749
57
.
19.
de Velasco
G
,
Culhane
AC
,
Fay
AP
,
Hakimi
AA
,
Voss
MH
,
Tannir
NM
, et al
Molecular subtypes improve prognostic value of international metastatic renal cell carcinoma database consortium prognostic model
.
Oncologist
2017
;
22
:
286
92
.
20.
Iglesia
MD
,
Parker
JS
,
Hoadley
KA
,
Serody
JS
,
Perou
CM
,
Vincent
BG
. 
Genomic analysis of immune cell infiltrates across 11 tumor types
.
J Natl Cancer Inst
2016
;
108
.
21.
Beuselinck
B
,
Job
S
,
Becht
E
,
Karadimou
A
,
Verkarre
V
,
Couchy
G
, et al
Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting
.
Clin Cancer Res
2015
;
21
:
1329
39
.
22.
Giraldo
NA
,
Becht
E
,
Vano
Y
,
Petitprez
F
,
Lacroix
L
,
Validire
P
, et al
Tumor-infiltrating and peripheral blood T-cell immunophenotypes predict early relapse in localized clear cell renal cell carcinoma
.
Clin Cancer Res
2017
;
23
:
4416
28
.
23.
Wang
T
,
Lu
R
,
Kapur
P
,
Jaiswal
BS
,
Hannan
R
,
Zhang
Z
, et al
An empirical approach leveraging tumorgrafts to dissect the tumor microenvironment in renal cell carcinoma identifies missing link to prognostic inflammatory factors
.
Cancer Discov
2018
;
8
:
1142
55
.
24.
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
:
5132
6
.
25.
Thompson
RH
,
Dong
H
,
Lohse
CM
,
Leibovich
BC
,
Blute
ML
,
Cheville
JC
, et al
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma
.
Clin Cancer Res
2007
;
13
:
1757
61
.
26.
Wang
QJ
,
Hanada
K
,
Robbins
PF
,
Li
YF
,
Yang
JC
. 
Distinctive features of the differentiated phenotype and infiltration of tumor-reactive lymphocytes in clear cell renal cell carcinoma
.
Cancer Res
2012
;
72
:
6119
29
.
27.
Siska
PJ
,
Beckermann
KE
,
Mason
FM
,
Andrejeva
G
,
Greenplate
AR
,
Sendor
AB
, et al
Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma
.
JCI Insight
2017
;
2:pii:93411
.
28.
Giraldo
NA
,
Becht
E
,
Pagès
F
,
Skliris
G
,
Verkarre
V
,
Vano
Y
, et al
Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer
.
Clin Cancer Res
2015
;
21
:
3031
40
.
29.
Chevrier
S
,
Levine
JH
,
Zanotelli
VRT
,
Silina
K
,
Schulz
D
,
Bacac
M
, et al
An immune atlas of clear cell renal cell carcinoma
.
Cell
2017
;
169
:
736
49
.
30.
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
:
3698
.
31.
Matsushita
H
,
Sato
Y
,
Karasaki
T
,
Nakagawa
T
,
Kume
H
,
Ogawa
S
, et al
Neoantigen load, antigen presentation machinery, and immune signatures determine prognosis in clear cell renal cell carcinoma
.
Cancer Immunol Res
2016
;
4
:
463
71
.
32.
Havel
JJ
,
Chowell
D
,
Chan
TA
. 
The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy
.
Nature Rev Cancer
2019
;
19
:
133
50
.
33.
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
:
820
2
.
34.
Turajlic
S
,
Litchfield
K
,
Xu
H
,
Rosenthal
R
,
McGranahan
N
,
Reading
JL
, et al
Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis
.
Lancet Oncol
2017
;
18
:
1009
21
.
35.
Davoli
T
,
Uno
H
,
Wooten
EC
,
Elledge
SJ
. 
Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy
.
Science
2017
;
355:pii:eaaf8399
.
36.
Samstein
RM
,
Lee
CH
,
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
:
202
6
.
37.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
38.
Beroukhim
R
,
Brunet
JP
,
Di Napoli
A
,
Mertz
KD
,
Seeley
A
,
Pires
MM
, et al
Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney
.
Cancer Res
2009
;
69
:
4674
81
.
39.
Varn
FS
,
Wang
Y
,
Mullins
DW
,
Fiering
S
,
Cheng
C
. 
Systematic pan-cancer analysis reveals immune cell interactions in the tumor microenvironment
.
Cancer Res
2017
;
77
:
1271
82
.
40.
Noy
R
,
Pollard
JW
. 
Tumor-associated macrophages: from mechanisms to therapy
.
Immunity
2014
;
41
:
49
61
.
41.
Amici
SA
,
Young
NA
,
Narvaez-Miranda
J
,
Jablonski
KA
,
Arcos
J
,
Rosas
L
, et al
CD38 is robustly induced in human macrophages and monocytes in inflammatory conditions
.
Front Immunol
2018
;
9
:
1593
.
42.
Kang
J
,
Park
K-H
,
Kim
J-J
,
Jo
E-K
,
Han
M-K
,
Kim
UH
. 
The role of CD38 in Fcgamma receptor (FcgammaR)-mediated phagocytosis in murine macrophages
.
J Biol Chem
2012
;
287
:
14502
14
.
43.
Peranzoni
E
,
Lemoine
J
,
Vimeux
L
,
Feuillet
V
,
Barrin
S
,
Kantari-Mimoun
C
, et al
Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment
.
PNAS
2018
;
115
:
E4041
50
.
44.
Dieu-Nosjean
MC
,
Goc
J
,
Giraldo
NA
,
Sautes-Fridman
C
,
Fridman
WH
. 
Tertiary lymphoid structures in cancer and beyond
.
Trends Immunol
2014
;
35
:
571
80
.
45.
Tamma
R
,
Rutigliano
M
,
Lucarelli
G
,
Annese
T
,
Ruggieri
S
,
Cascardi
E
, et al
Microvascular density, macrophages, and mast cells in human clear cell renal carcinoma with and without bevacizumab treatment
.
Urol Oncol
2019
;
37
:
355.
e11–355.e19
.
46.
Wallin
JJ
,
Bendell
JC
,
Funke
R
,
Sznol
M
,
Korski
K
,
Jones
S
, et al
Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma
.
Nat Commun
2016
;
7
:
12624
.
47.
Powles
T
,
Sarwar
N
,
Stockdale
A
,
Sarker
SJ
,
Boleti
E
,
Protheroe
A
, et al
Safety and efficacy of pazopanib therapy prior to planned nephrectomy in metastatic clear cell renal cancer
.
JAMA Oncol
2016
;
2
:
1303
9
.
48.
Zizzari
IG
,
Napoletano
C
,
Botticelli
A
,
Caponnetto
S
,
Calabrò
F
,
Gelibter
A
, et al
TK inhibitor pazopanib primes DCs by downregulation of the beta-catenin pathway
.
Cancer Immunol Res
2018
;
6
:
711
22
.
49.
Finke
JH
,
Rini
B
,
Ireland
J
,
Rayman
P
,
Richmond
A
,
Golshayan
A
, et al
Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients
.
Clin Cancer Res
2008
;
14
:
6674
82
.
50.
Ko
JS
,
Zea
AH
,
Rini
BI
,
Ireland
JL
,
Elson
P
,
Cohen
P
, et al
Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients
.
Clin Cancer Res
2009
;
15
:
2148
57
.
51.
Adotevi
O
,
Pere
H
,
Ravel
P
,
Haicheur
N
,
Badoual
C
,
Merillon
N
, et al
A decrease of regulatory T cells correlates with overall survival after sunitinib-based antiangiogenic therapy in metastatic renal cancer patients
.
J Immunother
2010
;
33
:
991
8
.
52.
Liu
XD
,
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
:
1017
29
.
53.
Verbiest
A
,
Couchy
G
,
Job
S
,
Zucman-Rossi
J
,
Caruana
L
,
Lerut
E
, et al
Molecular subtypes of clear cell renal cell carcinoma are associated with outcome during pazopanib therapy in the metastatic setting
.
Clinical Genitourin Cancer
2018
;
16
:
e605
e612
.
54.
Verbiest
A
,
Couchy
G
,
Job
S
,
Caruana
L
,
Lerut
E
,
Oyen
R
, et al
Molecular subtypes of clear-cell renal cell carcinoma are prognostic for outcome after complete metastasectomy
.
Eur Urol
2018
;
74
:
474
80
.
55.
Varela
I
,
Tarpey
P
,
Raine
K
,
Huang
D
,
Ong
CK
,
Stephens
P
, et al
Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma
.
Nature
2011
;
469
:
539
42
.
56.
Gao
W
,
Li
W
,
Xiao
T
,
Liu
XS
,
Kaelin
WG
 Jr
. 
Inactivation of the PBRM1 tumor suppressor gene amplifies the HIF-response in VHL-/- clear cell renal carcinoma
.
PNAS
2017
;
114
:
1027
32
.
57.
Kimura
T
,
Kato
Y
,
Ozawa
Y
,
Kodama
K
,
Ito
J
,
Ichikawa
K
, et al
Immunomodulatory activity of lenvatinib contributes to antitumor activity in the Hepa1-6 hepatocellular carcinoma model
.
Cancer Sci
2018
;
109
:
3993
4002
.
58.
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 (London, England)
2019
;
393
:
2404
15
.
59.
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 untreated advanced renal cell carcinoma with sarcomatoid features
.
J Clin Oncol
2019
;
37
:
4513
.
60.
Rini
BI
,
Plimack
ER
,
Stus
V
,
Gafanov
R
,
Hawkins
R
,
Nosov
D
, et al
Pembrolizumab (pembro) plus axitinib (axi) versus sunitinib as first-line therapy for metastatic renal cell carcinoma (mRCC): outcomes in the combined IMDC intermediate/poor risk and sarcomatoid subgroups of the phase 3 KEYNOTE-426 study
.
J Clin Oncol
2019
;
37
:
4500
.
61.
Rini
BI
,
Plimack
ER
,
Stus
V
,
Gafanov
R
,
Hawkins
R
,
Nosov
D
, et al
Atezolizumab (atezo) + bevacizumab (bev) versus sunitinib (sun) in pts with untreated metastatic renal cell carcinoma (mRCC) and sarcomatoid (sarc) histology: IMmotion151 subgroup analysis
.
J Clin Oncol
2019
;
37
:
4512
.
62.
Rini
BI
,
Huseni
M
,
Atkins
MB
,
McDermott
DF
,
Powles
T
,
Escudier
B
, et al
Molecular correlates differentiate response to atezolizumab + bevacizumab vs sunitinib: results from a phase III study (IMmotion151) in untreated metastatic renal cell carcinoma
.
Ann Oncol
2018
;
29
:
Suppl_8
,
Abstract LBA31
.
63.
Pan
D
,
Kobayashi
A
,
Jiang
P
,
Ferrari de Andrade
L
,
Tay
RE
,
Luoma
AM
, et al
A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing
.
Science
2018
;
359
:
770
5
.
64.
Miao
D
,
Margolis
CA
,
Gao
W
,
Voss
MH
,
Li
W
,
Martini
DJ
, et al
Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma
.
Science
2018
;
359
:
801
6
.
65.
Advani
R
,
Flinn
I
,
Popplewell
L
,
Forero
A
,
Bartlett
NL
,
Nilanjan Ghosh
N
, et al
CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin's lymphoma
.
N Engl J Med
2018
;
379
:
1711
21
.
66.
Pathria
P
,
Louis
TL
,
Varner
JA
. 
Targeting tumor-associated macrophages in cancer
.
Trends Immunol
2019
;
40
:
310
27
.
67.
Locke
LW
,
Mayo
MW
,
Yoo
AD
,
Williams
MB
,
Berr
SS
. 
PET imaging of tumor associated macrophages using mannose coated 64Cu liposomes
.
Biomaterials
2012
;
33
:
7785
93
.
68.
Iv
M
,
Samghabadi
P
,
Holdsworth
S
,
Gentles
A
,
Rezaii
P
,
Harsh
G
, et al
Quantification of macrophages in high-grade gliomas by using ferumoxytol-enhanced MRI: a pilot study
.
Radiology
2019
;
290
:
198
206
.
69.
Yin
Q
,
Hung
SC
,
Wang
L
,
Lin
W
,
Fielding
JR
,
Rathmell
WK
, et al
Associations between tumor vascularity, vascular endothelial growth factor expression and PET/MRI radiomic signatures in primary clear-cell-renal-cell-carcinoma: proof-of-concept study
.
Sci Rep
2017
;
7
:
43356
.
70.
Yin
Q
,
Hung
SC
,
Rathmell
WK
,
Shen
L
,
Wang
L
,
Lin
W
, et al
Integrative radiomics expression predicts molecular subtypes of primary clear cell renal cell carcinoma
.
Clin Radiol
2018
;
73
:
782
91
.
71.
Orillion
A
,
Hashimoto
A
,
Damayanti
N
,
Shen
L
,
Adelaiye-Ogala
R
,
Arisa
S
, et al
Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma
.
Clin Cancer Res
2017
;
23
:
5187
201
.
72.
Neubert
NJ
,
Schmittnaegel
M
,
Bordry
N
,
Nassiri
S
,
Wald
N
,
Martignier
C
, et al
T cell–induced CSF1 promotes melanoma resistance to PD1 blockade
.
Sci Transl Med
2018
;
10
:
eaan3311
.
73.
Atkins
M
,
Joseph
R
,
Ho
T
,
Vaishampayan
U
,
Ali
S
,
Matrana
M
, et al
Abstract B201: a phase 1 dose-finding study of X4P-001 (an oral CXCR4 inhibitor) and axitinib in patients with advanced renal cell carcinoma (RCC)
.
Mol Cancer Ther
2018
;
17
:
B201
.
74.
Hurwitz
ME
,
Diab
A
,
Bernatchez
C
,
Haymaker
CL
,
Kluger
HM
,
Tetzlaff
MT
, et al
Effect of NKTR-214 on the number and activity of CD8+ tumor infiltrating lymphocytes in patients with advanced renal cell carcinoma
.
J Clin Oncol
2017
;
35
:
454
.
75.
Diab
A
,
Hurwitz
ME
,
Cho
DC
,
Papadimitrakopoulou
V
,
Curti
BD
,
Tykodi
SS
, et al
NKTR-214 (CD122-biased agonist) plus nivolumab in patients with advanced solid tumors: preliminary phase 1/2 results of PIVOT
.
J Clin Oncol
2018
;
36
:
3006
.
76.
Tannir
NM
,
Fan
AC
,
Lee
RJ
,
Carthon
BC
,
Iliopoulos
O
,
Mier
JW
, et al
Phase 1 study of glutaminase (GLS) inhibitor CB-839 combined with either everolimus (E) or cabozantinib (Cabo) in patients (pts) with clear cell (cc) and papillary (pap) metastatic renal cell cancer (mRCC)
.
J Clin Oncol
2018
;
36
:
603
.
77.
Abu Aboud
O
,
Habib
SL
,
Trott
J
,
Stewart
B
,
Liang
S
,
Chaudhari
AJ
, et al
Glutamine addiction in kidney cancer suppresses oxidative stress and can be exploited for real-time imaging
.
Cancer Res
2017
;
77
:
6746
58
.
78.
Giannakis
M
,
Li
H
,
Jin
C
,
Gopal
S
,
Desai
K
,
Horak
C
, et al
Metabolomic correlates of response in nivolumab-treated renal cell carcinoma and melanoma patients
.
J Clin Oncol
2017
;
35
:
3036
.
79.
Lawrence Fong
JP
,
Luke
J
,
Hotson
D
,
Sznol
M
,
George
S
,
Choueiri
T
, et al
O45 refractory renal cell cancer (RCC) exhibits high adenosine A2A receptor (A2AR) expression and prolonged survival following treatment with the A2AR antagonist, CPI-444
. In:
Proceedings of the 33rd Annual Meeting & Pre-Conference Programs of the Society for Immunotherapy of Cancer (SITC 2018); 2018 Nov 10
; 
2018
.
80.
Hotson
AN
,
Hotson
AN
,
Laport
G
,
Kwei
L
,
Fong
L
,
Sznol
M
, et al
1137PDIdentification of adenosine pathway genes associated with response to therapy with the adenosine receptor antagonist CPI-444
.
Ann Oncol
2018
;
29
:
mdy288.010
.
81.
Turajlic
S
,
Xu
H
,
Litchfield
K
,
Rowan
A
,
Horswell
S
,
Chambers
T
, et al
Deterministic evolutionary trajectories influence primary tumor growth: TRACERx Renal
.
Cell
2018
;
173
:
595
610
.
82.
Turajlic
S
,
Xu
H
,
Litchfield
K
,
Rowan
A
,
Chambers
T
,
Lopez
JI
, et al
Tracking cancer evolution reveals constrained routes to metastases: TRACERx Renal
.
Cell
2018
;
173
:
581
94
.
83.
Keren
L
,
Bosse
M
,
Marquez
D
,
Angoshtari
R
,
Jain
S
,
Varma
S
, et al
A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging
.
Cell
2018
;
174
:
1373
87
.