1,904
Views
1
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

Immune approaches beyond traditional immune checkpoint inhibitors for advanced renal cell carcinoma

ORCID Icon, ORCID Icon, , , , & ORCID Icon show all
Article: 2276629 | Received 05 Aug 2023, Accepted 25 Oct 2023, Published online: 10 Nov 2023

ABSTRACT

Renal cell carcinoma (RCC), especially clear cell RCC, is generally considered an immunotherapy-responsive cancer. Recently, the prognosis for patients with locally advanced and metastatic RCC has significantly improved with the regulatory approvals of anti-PD-1/PD-L1/CTLA-4 immune checkpoint inhibitor (ICI)-based regimens. Yet in most cases, RCC will remain initially unresponsive to treatment or will develop resistance over time. Hence, there remains an unmet need to understand what leads to ICI resistance and to develop novel immune and nonimmune treatments to enhance the response to ICIs. In this review, we highlight recently published studies and the latest clinical studies investigating the next generation of immune approaches to locally advanced and metastatic RCC beyond traditional ICIs. These trials include cytokines, gut microbiota-based therapies, novel immune checkpoint agents, vaccines, and chimeric antigen receptor T cells. These agents are being evaluated as monotherapy or in combination with traditional ICIs and will hopefully provide improved outcomes to patients with RCC soon.

This article is part of the following collections:
Immunotherapy of Renal Cell Carcinoma

Introduction

Renal cell carcinoma (RCC), especially of the clear cell subtype, has long been considered a cancer susceptible to immune-based treatments. Prior to the year 2000, high-dose systemic interleukin-2 (IL-2) and interferon alfa were both approved in the setting of metastatic RCC (mRCC) because some patients had complete responses to cytokine infusions for several decades.Citation1 However, these cases were not common, and the associated side effects from cytokines were numerous. Starting in the year 2015 with the approval of nivolumab as second-line therapy in mRCC, immune checkpoint inhibitors (ICIs) (specifically anti-CTLA-4, PD-1 and PD-L1 monoclonal antibodies) have revolutionized the management of RCC including in the frontline metastatic setting and as adjuvant therapy in the localized setting (specifically pembrolizumab for intermediate-high or high-risk of recurrence).Citation2 Unfortunately, many patients with RCC either do not respond to anti-CTLA-4/PD-1/PD-L1 ICIs or develop ICI resistance. Hence, more novel methods and agents are needed. This review provides an overview of the promising upcoming immunotherapeutic agents for locally advanced and mRCC, focusing on cytokine-based therapies, the gut microbiota, novel immune checkpoint agents, vaccines, and chimeric antigen receptor (CAR) T cells that are currently being evaluated in clinical trials.

Cytokines

Cytokines continue to remain of interest in mRCC after the success of systemic interleukin-2 and interferon-alfa for some patients despite the high toxicities. The cytokines that are under current clinical investigation are engineered to minimize toxicities seen with systemic high-dose IL-2 while preserving antitumor immunity. These include IL-2, IL-12, IL-15, and IL-27. summarizes the latest clinical trials involving these cytokines.

Table 1. Current active and recruiting clinical trials evaluating engineered interleukins in metastatic RCC.

IL-2

While systemic IL-2 continues to be used in clinical trials, such as in combination with pembrolizumab or nivolumab (e.g., NCT03991130, NCT05155033, NCT03260504, NCT02964078), novel engineered cytokines using the IL-2 pathway while mitigating toxicity are being explored. The engineered cytokines being evaluated in mRCC include bempegaldesleukin (NKTR-214) and nemvaleukin-alfa (ALKS 4230).

Bempegaldesleukin consists of an IL-2 protein bound to releasable polyethylene glycol (PEG) chains; when IL-2 is bound to PEG, it remains inactive. Bempegaldesleukin selectively activates the IL-2 receptors of cytotoxic T cells over helper T regulatory cells, theoretically maximizing antitumor effects and minimizing toxicities.Citation3 The combination of bempegaldesleukin and nivolumab was evaluated in 557 patients with various solid tumors in the PIVOT-02 phase 1/2 trial (NCT02983045). PIVOT-02 involved a cohort of 49 patients with mRCC receiving this combination as frontline therapy, and results for this cohort were published.Citation4 After a median follow-up of 32.7 months, the objective response rate (ORR) was 34.7% (17/49 patients) and the median progression-free survival (PFS) was 7.7 months (95% CI, 3.8–13.9). Every patient except for one had a treatment-emergent adverse event (TEAE), including 38.8% with grade 3/4 events. The combination overall demonstrated safety and efficacy, leading to the PIVOT-09 open-label phase 3 study (NCT03729245) that randomized 623 patients to either bempegaldesleukin/nivolumab or the investigator’s choice of sunitinib or cabozantinib as frontline therapy in mRCC. Unfortunately, the experimental arm did not meet the prespecified boundary for statistical significance compared to the control arm, so the trial was discontinued.Citation5 Overall, the combination of bempegaldesleukin with nivolumab has been a disappointment, as the PIVOT IO 001 trial for frontline metastatic melanoma also did not meet the primary endpoint.Citation6 As a result, most clinical trials involving this combination for various cancers have been discontinued. While doublet treatments were ineffective, triplet therapy is being evaluated in the PIVOT IO 011 phase 1 trial (NCT04540705). This trial assesses 30 patients with previously untreated mRCC to receive the combination of bempegaldesleukin and nivolumab with either cabozantinib or axitinib.Citation7

Nemvaleukin-alfa binds with intermediate affinity to the IL-2 receptor while sparing high-affinity IL-2 receptors to minimize toxicities.Citation8 ARTISTRY-1 is a phase 1/2 trial (NCT02799095) involving 243 patients with advanced solid tumors including RCC refractory to or intolerant of approved therapies, receiving either nemvaleukin-alfa as monotherapy or in combination with pembrolizumab. Preliminary data presented at the 2022 ASCO Annual Meeting showed tolerability and durable antitumor activity for both nemvaleukin-alfa monotherapy and the combination with pembrolizumab.Citation9 Nemvaleukin-alfa monotherapy was also shown to induce a robust expansion of cytotoxic T cells and NK cells with minimal effect on regulatory T cells. The data is promising and warrants further investigation of nemvaleukin-alfa with ICIs.

IL-12

IL-12 is a pro-inflammatory cytokine involved in cell-mediated immunity with the potential to reverse tumor-induced immunosuppression, but it previously failed in clinical trials due to systemic toxicities including death.Citation10 Localized and targeted IL-12 therapies are being designed. There is an ongoing phase 1 trial (NCT04235777) of bintrafusp alfa and NHS-IL12 with or without stereotactic body radiation therapy for 66 patients with metastatic non-prostate genitourinary cancers.Citation11 Bintrafusp alfa is a bifunctional antibody that inhibits both PD-L1 and tumor growth factor-beta, which are both immunosuppressive.Citation12 NHS-IL12 is a fusion protein of two molecules of IL-12 with an antibody (NHS76) to single- and double-stranded DNA, targeting necrotic tumor with DNA exposure.Citation13 In preclinical models, these two agents had synergistic activity, inducing both adaptive and innate tumor immunity and generating tumor antigen-specific immune memory.Citation14

IL-15

IL-15 is another novel cytokine target being investigated in RCC. It is structurally like IL-2 due to sharing the common gamma chain.Citation15 It has been shown to stimulate cytotoxic T cells and NK cells and promotes memory cytotoxic T-cell survival, while having no effect on regulatory T cells.Citation16 IL-15 itself has a half-life of less than one hour, so its direct use as a cancer therapy is limited, but its components and receptor are being investigated in clinical trials. Nanrilkefusp alfa (SO-C101, SOT101) is an IL-15 superagonist being evaluated for safety and preliminary efficacy as monotherapy or in combination with pembrolizumab in the AURELIO-03 phase 1/1b trial (NCT04234113) in 200 patients with relapsed/refractory metastatic solid tumors including RCC. Preliminary data reported at the 2022 ASCO Annual Meeting showed a favorable safety profile and early signs of efficacy for nanrilkefusp alfa as monotherapy and in combination with ICIs.Citation17 N-803, an IL-15 superagonist fusion protein, is being evaluated in QUILT-3.055, a phase 2 trial (NCT03228667), in combination with different anti-PD-1/PD-L1 ICIs and PD-L1 targeting high-affinity NK cells in 147 patients with metastatic solid tumors including RCC who have progressed on prior anti-PD-1/PD-L1 ICIs. Preliminary data suggests N-803 has minimal toxicity and provides promising efficacy.Citation18 PF-07209960, an anti-PD-1 targeting IL-15 fusion protein, is being evaluated as monotherapy in a phase 1 trial (NCT04628780) in 37 patients with metastatic solid tumors including RCC who have progressed on two prior lines of therapy.

IL-27

IL-27 is a member of the IL-12 family of cytokinesCitation19; however, it has primarily been shown to be anti-inflammatory and represses anti-tumor immunity.Citation20 Increased IL-27 transcript expression is associated with a poor prognosis in RCC.Citation21 SRF388 is a monoclonal antibody that blocks the interaction of IL-27 with its receptor. SRF388 is being evaluated in a phase 1/1b trial (NCT04374877) as either monotherapy or in combination with pembrolizumab in 220 patients with metastatic solid tumors, including patients with RCC who have previously received vascular endothelial growth factor-targeted therapy and ICI. Preliminary results reported at the 2022 ASCO Annual Meeting suggested SRF388 as monotherapy had good tolerability with preliminary evidence of antitumor activity, including 1 of 10 patients with RCC achieving a partial response, warranting further evaluation in a phase 2 trial.Citation22

STING

Stimulator of Interferon Genes (STING) is a cytosolic DNA-sensing protein critical for activating the release of inflammatory cytokines, which upregulate the innate immunity to destroy pathogens.Citation23 STING agonists are being designed as novel agents to stimulate their release to activate the innate immune response against tumor cells.Citation24 STING agonists are being combined with traditional ICIs in clinical trials to upregulate both the adaptive and innate immunity and to provide synergy to destroy tumor cells. TAK-500 is a STING agonist being evaluated as monotherapy and in combination with pembrolizumab in a phase 1 trial (NCT05070247) for patients with metastatic solid tumors including mRCC. IMSA101, a STING agonist injected intratumorally, is being assessed with an ICI and personalized ultra-fractionated stereotactic adaptive radiotherapy (PULSAR) in patients with oligometastatic RCC compared to a control arm of ICI and PULSAR in a phase 2 trial (NCT05846646).

Gut microbiota and traditional immune checkpoint inhibitors

There is growing evidence that the gut microbiota, which refers to the trillions of microbes (bacteria, fungi, viruses, archaea) that colonize the human gut, are one of the key host factors associated with response to ICI therapies in a variety of solid tumors including RCC.Citation25–31 This observation was first made by noting a correlation between a poor response to ICIs when cancer patients were exposed to antibiotics.Citation32 Specific bacteria have been demonstrated to be potential biomarkers of response to ICIs. However, only recently have we begun to understand the mechanisms by which the gut microbiota directly enhances the ICI anti-tumor response through direct/indirect interactions between microbes and the immune system, the tumor microenvironment, and tumor cells themselves.Citation33 Therefore, there are now clinical trials investigating microbiota-derived therapies (such as probiotics and fecal microbiota transplants [FMT]) to enhance ICI efficacy in a variety of solid tumors including RCC.

A prospective cohort biomarker study of patients with solid tumors treated with either nivolumab or pembrolizumab also had their short chain fatty acids (SCFAs) concentration analyzed from fecal and plasma samples, and high fecal SCFA concentrations were associated with a statistically significantly longer PFS.Citation34 In addition, recent antibiotic use, which disturbs the gut flora, has been associated with a decreased overall survival (OS), PFS, and response rate for cancer patients on ICIs.Citation35 For RCC, bacterial species such as Akkermansia muciniphila and Bifidobacterium spp. have been positively associated with ICI response, while the Enterocloster genus has been linked to lack of ICI response.Citation36

The promising gut microbiota-based therapy under investigation for RCC is CBM588. CBM588 is a live bacterial product that contains the probiotic Clostridium butyricum, which produces butyric and acetic acidsCitation37 and promotes expansion of Bifidobacterium spp. which have been shown to correlate with response to ICI in patients with RCC.Citation38 CBM588 was previously demonstrated in a retrospective study to improve both PFS and OS for patients with metastatic non-small cell lung cancer (NSCLC) receiving ICIs compared to patients who did not receive C. butyricum.Citation39 In RCC, a phase 1 trial (NCT03829111) enrolled 30 patients with metastatic disease and no prior ICIs to receive either CBM588 (taken 80 mg orally twice daily) with ipilimumab/nivolumab versus ipilimumab/nivolumab alone in a 2:1 ratio.Citation40 Although the primary endpoint of the change in Bifidobacterium spp. at baseline to 12 weeks was not met, the patients receiving CBM588 had a significantly prolonged median PFS (12.7 months vs. 2.5 months, HR 0.15; 95% CI, 0.05–0.47; p = 0.001) and a trend toward higher response rate (58% vs. 20%, p = 0.06). This trial suggests the importance of incorporating the gut microbiota into future clinical trials involving ICIs. In a similar trial design, CBM588 is being assessed in another phase 1 trial (NCT05122546), as 30 patients received CBM588 with nivolumab/cabozantinib versus nivolumab/cabozantinib alone in a 2:1 ratio. Preliminary results reported at the 2023 ASCO Annual Meeting showed benefit of the CBM588 cohort, with the median PFS not reached compared to 5.8 months in the nivolumab/cabozantinib cohort (p = 0.03) and response rates of 63% versus 33%, respectively.Citation41

Outside of probiotics, FMT are currently under clinical investigation to improve ICI response in RCC. FMT-based clinical trials utilize donor stool from patients who have responded to ICI and transplant them into patients about to receive ICI. This has shown remarkable efficacy in refractory metastatic melanoma patientsCitation42,Citation43 and has even been used to treat ICI-induced colitis.Citation44 In RCC, the TACITO phase 1/2 trial (NCT04758507) is evaluating whether FMT from donors who have had excellent responses to ICIs can enhance ICI efficacy in treatment-naïve patients. In this quadruple-blinded placebo-controlled trial, a total of 50 patients with RCC being treated with ICIs will be randomized to receive either donor FMT versus placebo FMT via colonoscopy infusions and then oral frozen fecal or placebo capsules at 90 or 180 days after the first FMT.Citation45 Another phase 1 trial (NCT05354102) is enrolling 12 patients with either NSCLC, melanoma, or RCC who have progressed on prior PD-1/PD-L1 ICIs to receive BMC128 (oral capsule composed of a live bacterial consortium of four bacterial strains) with nivolumab.Citation46 Outside of probiotics and FMT, there are several promising microbiota-based therapeutics in the preclinical pipeline which will be evaluated in the clinical trial setting soon.

Novel immune checkpoint targets

The first ICIs targeting the PD-1/PD-L1 or CTLA-4 proteins on T cells have revolutionized the care of cancer patients. However, there are other novel immune checkpoints under clinical investigation.Citation47 These novel targets are not only expressed on CD4 or CD8 T cells but also aim to enhance ICI efficacy by targeting other cells within the tumor microenvironment such as myeloid cells. This section focuses on the checkpoint proteins, both inhibitory and stimulatory, with antibodies under investigation in clinical trials that include patients with RCC. A summary of the active and recruiting novel immune checkpoint clinical trials described in this section is provided in .

Table 2. Current active and recruiting clinical trials evaluating novel immune checkpoint targets involving patients with metastatic RCC.

LAG-3

Lymphocyte-activation gene 3 (LAG-3) is an inhibitory coreceptor that becomes expressed on T cells and other immune cells upon antigen stimulation, and it binds to major histocompatibility complex (MHC) II. By binding MHC II, LAG-3 prevents presentation of tumor antigens to T cells leading to diminished anti-tumor immunity.Citation48 There are two monoclonal antibodies targeting LAG-3 under clinical investigation in RCC, relatlimab (BMS-986016) and ieramilimab (LAG525). Of note, relatlimab was recently approved by the United States Food and Drug Administration in March 2022 for frontline therapy of metastatic melanoma in combination with nivolumab, after this combination showed a greater PFS benefit compared to nivolumab alone in RELATIVITY-047 (NCT03470922).Citation49 The FRACTION-RCC phase 2 trial (NCT02996110) evaluated various arms of combination ICIs, including relatlimab/nivolumab, with ipilimumab/nivolumab as the control arm for patients who had progressed on or after prior ICI; the results from the relatlimab/nivolumab arm have not yet been reported.

A phase 1/2 study (NCT02460224) evaluated ieramilimab with spartalizumab (PDR001, an anti-PD-1 agent) in 255 patients with metastatic solid tumors with progression on prior therapy, including 11 patients with mRCC (7 received ieramilimab alone and 4 received combination therapy).Citation50 Overall, ieramilimab alone or with spartalizumab was well tolerated in this patient population, with no specific details reported for RCC. Antitumor activity was stronger in the combination arm over ieramilimab alone, as 10% of patients had either a complete or partial response to the treatment combination.

In addition, RO7247669 is an anti-PD-1/anti-LAG-3 bispecific antibody delivering dual checkpoint inhibition. In a phase 2 trial (NCT05805501), RO7247669 is being combined with axitinib alone or with axitinib/tiragolumab (another novel ICI, as explained later) as frontline options in 210 patients with mRCC, compared to the control arm of pembrolizumab/axitinib.

TIM-3

T-cell immunoglobulin mucin-3 (TIM-3) is expressed on many immune cells and plays an important role in the inhibition of T cell responses. High levels of TIM-3 expression are correlated with T-cell exhaustion.Citation51 There was a phase 1/2 trial (NCT02608268) evaluating sabatolimab (MBG453), an anti-TIM-3 monoclonal antibody, with and without spartalizumab in metastatic solid tumors including RCC that progressed or were intolerant to standard therapies. The phase 1/1b portion included 219 patients (6 RCC) and results showed that sabatolimab alone did not result in tumor response, and the combination of sabatolimab/spartalizumab resulted in some patients with partial responses.Citation52 The trial was terminated due to business reasons.

BGB-A425 is another anti-TIM-3 monoclonal antibody. Tislelizumab is a novel anti-PD-1 antibody designed to minimize binding to macrophages in order to minimize potential resistance to anti-PD-1 therapy.Citation53 LBL-007 is an anti-LAG-3 monoclonal antibody. These three drugs, in various combinations, are being evaluated in a phase 1/2 trial (NCT03744468) for patients with advanced solid tumors. In the phase 2 dose expansion arm, this triplet combination is being evaluated in a cohort of multiple cancers including RCC.

TIGIT

T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is another novel immune checkpoint protein expressed on both T cells and natural killer (NK) cells. It plays an important role by directly inhibiting T- and NK-cell effector function and elimination of tumor cells by binding to CD155 on tumor cells.Citation54 Tiragolumab is an anti-TIGIT antibody, and it received breakthrough designation in February 2021 for its improved PFS and ORR for previously untreated metastatic NSCLC in combination with atezolizumab compared to placebo/atezolizumab in the CITYSCAPE phase 2 trial (NCT03563716).Citation55 A phase 2 trial (NCT03977467) is evaluating tiragolumab/atezolizumab in 80 patients with advanced malignancies (including approximately 15 patients with RCC) with progression after frontline PD-1 inhibition.

ILT2–4

Immunoglobulin-like transcripts 2, 3, and 4 (ILT2, ILT3, and ILT4) are expressed on immune cells, and when they bind to their ligands, they contribute to immune evasion by tumor cells.Citation56 Both ILT2 and ILT4 bind to human leukocyte antigen-G on tumor cells, assisting with tumor escape.Citation57 NGM707 is a humanized monoclonal antibody against both ILT2 and ILT4.Citation58 NGM707 is being evaluated with and without pembrolizumab in a phase 1/2 trial (NCT04913337) involving 179 patients with metastatic solid tumors including RCC that progressed or were intolerant to all prior available therapies. ILT3 is expressed on select myeloid antigen-presenting cells,Citation59 including myeloid-derived suppressor cells (MDSCs) which accumulate in the tumor microenvironment, inhibiting the antitumor immune response and encouraging tumor growth.Citation60 The interaction of ILT3 with fibronectin was found to promote an immunosuppressive state.Citation61 NGM831, an ILT3 antibody, is being evaluated as monotherapy and in combination with pembrolizumab in a phase 1/2 trial (NCT05215574) involving 179 patients with metastatic solid tumors including RCC.

TREM2

Triggering receptor expressed on myeloid cells-2 (TREM2) is expressed on some myeloid cells including macrophages and dendritic cells (DCs), and high expression of TREM2-positive cells leads to decreased antitumor immunity including the inhibition of T-cell proliferation.Citation62 TREM2 is expressed on tumor-associated macrophages, which are linked to tumor growth, immune evasion, and overall poor prognosis for cancer patients.Citation63 TREM2 has also been associated as an oncogenic factor in the development of RCC, as TREM2 expression was abnormally high in RCC tumor tissues in vivo, and silencing TREM2 inhibited tumor cell growth.Citation64 PY314 is a TREM2 monoclonal antibody, and it is being assessed either alone or in combination with pembrolizumab in a phase 1 trial (NCT04691375) in 288 patients with refractory/relapsed metastatic solid tumors including RCC to standard therapies.Citation65

VISTA

V-domain Ig suppressor of T-cell activation (VISTA) is expressed on both myeloid cells and T cells, and it generally acts as an inhibitory immune checkpoint for many cancers and downregulates T-cell activity.Citation66 VISTA has been found to be highly expressed in clear cell RCC tumors,Citation67 and high expression of VISTA on tumor-associated immune cells has been associated with a worse prognosis for patients with clear cell RCC with venous tumor thrombus.Citation68 Therefore, this makes VISTA an attractive target for patients with RCC.

CA-170 is an oral PD-L1, PD-L2, and VISTA antagonist that rescued the proliferation and effector function of T cells resulting in significant antitumor activity in preclinical models.Citation69 In a phase 1 trial of CA-170 for patients with advanced solid tumors including three patients with mRCC that were naïve to ICI therapy, the drug was well tolerated with evidence of antitumor activity.Citation70

SNS-101 is a conditionally active antibody that selectively targets the active form of VISTA with potent tumor inhibition but also minimizes the risk of cytokine release syndrome. A phase 1/2 trial (NCT05864144) is evaluating the safety and efficacy of SNS-101 as monotherapy or in combination with cemiplimab in advanced solid tumors refractory or intolerant to standard of care therapies.

BTLA

B- and T-lymphocyte attenuator (BTLA), a member of the CD28 superfamily and is structurally similar to PD-1 and CTLA-4, is an inhibitory immune checkpoint that is expressed on many immune cells including lymphocytes, macrophages, and DCs.Citation71 Icatolimab (JS004) is an anti-BTLA monoclonal antibody being evaluated for safety and tolerability as monotherapy in a phase 1 trial (NCT04773951) for patients with advanced solid tumors, including RCC, refractory to standard of care therapies. HFB200603 is another anti-BTLA antibody being evaluated as monotherapy and in combination with tislelizumab in a phase 1 trial (NCT05789069) for patients with advanced solid tumors including RCC refractory to at least two lines of therapy.

OX40 and 4–1BB

OX40 (CD134, also known as tumor necrosis factor receptor superfamily [TNFRSF], member 4) is a co-stimulatory molecule expressed on activated T cells, and it induces expression of anti-apoptotic proteins and cell-cycle progression, upregulating the immune system against tumor cells.Citation72 Similarly, 4–1BB (CD37, also known as TNFRSF, member 9) is expressed on the surface of T cells after activation and promotes anti-tumor activity.Citation73 Hence, antibodies that stimulate OX40 and 4–1BB signaling may enhance the immune response against tumors.

INCAGN01949, an anti-OX40 agonist, was evaluated in a phase 1/2 trial (NCT02923349) as monotherapy for patients with 87 metastatic solid tumors including RCC previously treated with available therapies. INCAGN01949 monotherapy had no major safety concerns; however, just one patient achieved a partial response, and 23 had stable disease.Citation74 INCAGN01949 did not enhance proliferation or activation of T cells and did not reduce circulation of regulatory T cells. A phase 1/2 trial (NCT02737475) of BMS-986178 (anti-OX40 agonist) monotherapy (20 patients) or in combination with ipilimumab and/or nivolumab (165 patients) was reported, which included eight patients with mRCC (with one to at least three lines of standard therapy and no prior ICI exposure) who received BMS-986178/nivolumab/ipilimumab every 3 weeks for four cycles followed by maintenance BMS-986178/nivolumab.Citation75 One patient with RCC had a partial response, three had stable disease, and three had progressive disease. Five of the eight patients with RCC experienced treatment-related adverse events (none grade 3/4). Ivuxolimab (PF-04518600, another anti-OX40 agonist) was evaluated in combination with axitinib against axitinib alone in a phase 2 randomized double-blind trial (NCT03092856) for patients with mRCC with prior ICI exposure. Unfortunately, preliminary data presented at the 2022 ASCO Annual Meeting, which analyzed 29 patients receiving the axitinib/ivuxolimab combination and 30 patients receiving axitinib/placebo, revealed no difference in the median PFS (13.1 vs. 8.5 months, HR 0.85, p = 0.61), and the study was closed to further accrual.Citation76

More novel anti-OX40 agonistic antibodies with possibly better efficacy are being actively evaluated in phase 1 clinical trials. HFB301001 is a second-generation anti-OX40 agonist that binds to a unique epitope on OX40 that does not bind to the endogenous OX40 ligand (OX40L), allowing for a more agonistic effect compared to prior OX40 agonists which often do interact with OX40L.Citation77 HFB301001 is being evaluated as monotherapy in a phase 1 dose-escalation trial (NCT05229601) in 84 patients with metastatic solid tumors, including patients with RCC, who have received at least two lines of therapy. Similarly, BGB-A445 is an anti-OX40 agonist that also does not interact with the OX40–OX40L axis directly.Citation78 BGB-A445 is being evaluated as monotherapy or in combination with tislelizumab (anti-PD-1 antibody) in a phase 1/2 trial (NCT05661955) that has two cohorts (monotherapy and combination) enrolling patients with previously treated RCC (1–3 prior lines). INBRX-106, a hexavalent OX40 agonist which has superior activity compared to bivalent agonists,Citation79 is being evaluated as either monotherapy or in combination with pembrolizumab in a phase 1/2 trial (NCT04198766) enrolling 333 patients with metastatic tumors who have progressed on all standard therapies including ICIs. SL-279252 (PD1-Fc-OX40L), a PD-1 and OX40L bifunctional fusion protein, is being evaluated in a phase 1 trial (NCT03894618) for 87 patients with advanced solid tumors including RCC. Preliminary results showed SL-279252 was well tolerated with some observed durable anti-tumor activity.Citation80

Utomilumab (PF-05082566) is a 4–1BB agonist that was evaluated in a dose-escalation phase 1b trial (NCT02179918) in combination with pembrolizumab in 23 patients with metastatic solid tumors who progressed on standard therapies, including five patients with RCC.Citation81 Overall, the combination was safe and tolerable. For the patients who responded to the combination, there was a trend toward high levels of activated memory and effector cytotoxic T cells. Ivuxolimab as monotherapy and in combination with utomilumab was evaluated in a phase 1 trial (NCT02315066) involving 174 patients with metastatic solid tumors including RCC who progressed or were intolerant to standard therapies including traditional ICIs. An analysis of the ivuxolimab monotherapy cohort (52 patients, including 9 with RCC) showed ivuxolimab to be well tolerated and have increased immune cell infiltrate on tumor biopsies.Citation82 An analysis of the dose-escalation and dose-expansion cohorts of the combination (total of 87 patients) was reported, which did not include any patients with RCC.Citation83 The combination was found to be well tolerated with some antitumor activity, warranting further investigation of various combinations of these immune checkpoint approaches.

INBRX-105 is a bispecific antibody targeting PD-L1 and 4–1BB. It is being evaluated in a phase 2 trial (NCT03809624) as monotherapy and in combination with pembrolizumab for different metastatic solid tumors including RCC. Patients with tumors that have progressed on traditional ICIs as well as tumors that are ICI-naïve are being enrolled.

CD27 and CD70

CD27 (also known as TNFRSF7), like OX40 and 4–1BB, is expressed on naïve lymphocytes and plays a role in B- and T-cell costimulation when activated.Citation84 The natural ligand of CD27 is CD70, which is highly expressed on some activated immune cells. Normally, CD27/70 binding activates the immune system. However, CD70 is also frequently expressed in malignancies including RCC.Citation85 Paradoxically, the CD27/CD70 interaction allows for tumor cells to evade the immune system, leading to tumor cell proliferation.Citation85 CD27/CD70 may also play a role in the apoptosis of lymphocytes in patients with RCC.Citation86 Hence, both the tumor-promoting and tumor-evading effects of CD27/CD70 make these targets of significant clinical interest.

Varlilumab is a CD27 agonist, and it was evaluated in combination with nivolumab in 175 patients with refractory metastatic solid tumors who have received up to five lines of treatment without prior anti-PD-1/PD-L1 agent in a phase 1/2 study (NCT02386111).Citation87 A phase 2 cohort evaluating ORR included 14 patients with RCC. Nine patients developed TEAEs. Of 12 patients who were evaluable for objective response, none had a complete or partial response, while five had stable disease. Overall, the combination of varlilumab and nivolumab was well tolerated, but the treatment efficacy of the combination was not much better than expected compared to nivolumab alone. There are currently no active trials with other CD27 agonists in patients with RCC.

Because of the high expression of CD70 on the surface of RCC tumor cells, there are various RCC trials targeting CD70 as an RCC tumor marker for tumor cell destruction. These trials mostly involve antibody-drug conjugates (beyond the scope of this review) and CAR T cells. The CAR T cells targeting CD70 will be discussed in a later section.

A2AR and CD73

When adenosine binds to adenosine-A2A receptor (A2AR), it triggers a complex signaling cascade that protects the normal cells from autoimmunity, and tumor cells can use this pathway to inhibit the immune response and to promote tumor immune escape.Citation88 CD73 is a checkpoint protein found on the surface of cells including endothelium, lymphocytes, and some tumors. CD73 has many roles, including the synthesis of adenosine, angiogenesis in the tumor microenvironment, and cancer cell proliferation.Citation89

In a phase 1/1b trial (NCT02655822) of 502 refractory solid tumor patients including 68 patients with refractory RCC (including with anti-PD-1/PD-L1 ICIs), ciforadenant (CPI-444, an A2AR small molecule antagonist) was evaluated as monotherapy or in combination with atezolizumab.Citation90 One in 33 receiving monotherapy and 4 of 35 patients receiving combination achieved a partial response, and the median PFS was 4.1 months and 5.8 months, respectively. The trial noted an association with cytotoxic T-cell infiltration into the tumor with prolonged disease control. Patients after treatment also had an increase in T-cell receptor diversity. Importantly, patients who responded to treatment had an adenosine-regulated gene expression signature in pre-treatment tumor biopsies, suggesting a possible predictive biomarker for A2AR inhibition.

Taminadenant (PBF509/NIR178) is an oral selective A2AR antagonist. It is being combined with spartalizumab and DFF332 (a hypoxia-inducible factor 2 alpha inhibitor) in a phase 1 trial (NCT04895748) for patients with RCC and other solid tumors refractory to all approved standard therapies. Similarly, TT-10 is another oral A2AR antagonist being evaluated in a phase 1/2 trial (NCT04969315) as monotherapy for patients with metastatic solid tumors including RCC refractory to standard of care therapies.

Mupadolimab (CPI-006) is an anti-CD73 antagonist that has been shown to activate B cells and stimulate antibody production.Citation91 Mupadolimab is being evaluated as monotherapy or with ciforadenant alone, pembrolizumab alone, and the combination of ciforadenant and pembrolizumab in 378 patients with incurable solid tumors including RCC in a phase 1/1b trial (NCT03454451). Preliminary data presented at the 2019 ASCO Annual Meeting for 17 patients showed mupadolimab monotherapy induces rapid lymphocyte redistribution and early evidence of antitumor activity.Citation92

Summary of novel immune checkpoint targets

Novel immune checkpoints including both activators (particularly the tumor necrosis factor receptor superfamily) and inhibitors of the immune system are of major interest. Many of the trials have evaluated these drugs in combination with traditional ICIs. Some of these trials have been completed, which have not overwhelmingly shown to be of major benefit for patients over current ICIs. None of the agents have reached phase 3 clinical trials yet for mRCC. However, the clinical trials of these novel immune checkpoints show enough promising results for further studies to continue. In addition, novel immune checkpoints are also of interest in the localized setting. For example, the NESCIO phase 2 trial (NCT05148546) is evaluating relatlimab/nivolumab in one experimental arm (other arms include ipilimumab/nivolumab and nivolumab alone) as neoadjuvant therapy in high-risk localized RCC.

With these novel immune checkpoints, there is an urgent need to determine predictive biomarkers. Currently, high tumor mutational burden (TMB), microsatellite instability, deficient mismatch repair systems, and PD-L1 expression all have their limits in accurately predicting tumor response to traditional ICIs.Citation93 Furthermore, mRCC frequently has low TMB and low microsatellite instability.Citation94 Genes on chromosome 3p that are frequently mutated in clear cell RCC, including BAP1, PBRM1, and SETD2, have been studied as potential predictive biomarkers. BRCA-1-associated protein-1 (BAP1) is a tumor suppressor, and its loss is associated with PD-L1 expression, inflammation, and abundant infiltration of immune cells.Citation95,Citation96 Mutations in PBRM1, which encodes a protein important in chromatin remodeling, have been shown to be associated with positive outcomes in pan-cancer patients receiving ICIs,Citation97 although there is also conflicting data suggesting PBRM1 mutations may lead to ICI resistance.Citation98 SETD2 encodes a histone methyltransferase critical to maintaining genomic integrity and stability, and mutations in SETD2 have been linked to higher TMB and microsatellite instability and therefore more favorable clinical outcomes to ICIs.Citation99 The association of these gene mutations with ICI response needs to be further validated. Because definitive predictive biomarkers for ICIs remain elusive, evaluating the genomic profile of tumors that have a good response to novel immune checkpoint agents is necessary.

Vaccines

Tumor vaccines that aim to enhance intrinsic immune responses against tumor cells have been studied for at least the past 20 y in patients with RCC. Although they were determined to be safe and tolerable, most were not very effective, so none led to regulatory approval.Citation100 However, cancer vaccine technology has continued to improve and remains of significant interest for RCC because of the inherent immunogenicity of RCC tumors. Currently, more vaccine trials for mRCC are underway, which include peptides and personalized neoantigens, DCs, and viruses. summarizes the clinical trials involving RCC vaccines.

Table 3. Current active and recruiting clinical trials evaluating vaccines in metastatic RCC.

Peptides and neoantigens

Peptide vaccines are generally designed to be 20–30 amino acids long containing specific immunogenic epitopes. Peptide vaccines are overall safer and cheaper than the other types of cancer vaccines. To improve efficacy, peptide vaccines can include epitopes that stimulate cytotoxic T cells and other epitopes that stimulate helper T cells to promote both a humoral and cell-mediated immune response.Citation101 The vaccines have also been given in combination with traditional ICIs to prevent T-cell exhaustion. As the technology of next-generation sequencing has improved, there are opportunities to identify tumor-specific mutations in different patients and to develop personalized neoantigen-based therapeutic vaccines. Advantages of this vaccine over a traditional peptide vaccine include a more tumor-specific T-cell response sparing normal cells and a decrease in a T-cell central tolerance, while disadvantages include higher financial costs and production delays.Citation102

IMA901 is a peptide-based vaccine consisting of 10 different tumor-associated peptides (nine binding to HLA class I and one binding to HLA class II) that were strongly overexpressed in RCC compared to normal cells. In the phase 3 trial IMPRINT (NCT01265901), the combination of IMA901 with sunitinib was compared to sunitinib alone in 339 treatment-naïve mRCC, and patients were randomly assigned in a 2:1 ratio to combination versus monotherapy.Citation103 The study did not meet its primary endpoint, as the median OS for the combination therapy did not differ significantly between the two groups (HR 1.34; 95% CI, 0.96–1.86; p = 0.087). More grade 3 or higher TEAEs were present in the combination group (57% vs. 47%).

Modi-1 is a peptide combination vaccine based on citrullinated vimentin and enolase peptides that stimulates tumor-infiltrating helper T cells.Citation104 The Modi-1 peptides are linked to a toll-like receptor ligand 1/2 adjuvant to enhance the immune response by several fold. ModiFY (NCT05329532) is a phase 1/2 trial evaluating Modi-1 alone or in combination with pembrolizumab or nivolumab in 144 patients with advanced solid tumors including RCC. RCC inclusion criteria in the study includes prior anti-angiogenic therapy and favorable/intermediate risk disease. Primary endpoints include adverse events and cellular immune response measured on the IFN-gamma ELISpot assay.

There are two phase 1 trials evaluating personalized RCC protein neoantigen cancer vaccines (NeoVax and IVAC-RCC-001), recruiting patients with advanced mRCC who have undergone resection of their tumors. NeoVax is being used in combination with ipilimumab in 19 patients (NCT02950766). Patients may not have received prior immunotherapy; prior systemic therapy excluding immunotherapy is allowed if given prior to six months before the first injection NeoVax. The primary endpoint is dose-limiting toxicity. Similarly, IVAC-RCC-001 will be used in combination with ipilimumab/nivolumab in 10 patients (NCT05641545). Patients may not have received prior systemic therapy. The primary endpoint is treatment success, defined as a vaccine-specific T-cell responses without unacceptable toxicities. PNeoVCA (NCT05269381) is another phase 1 trial using a personalized neoantigen peptide-based vaccine in combination with pembrolizumab for patients with advanced solid tumors.

Autogene cevumeran (RO7198457), a personalized mRNA-based neoantigen vaccine delivered via lipoplex nanoparticles intravenously, is being evaluated in several metastatic solid tumors in a phase 1 trial (NCT03289962) either as monotherapy or in combination with atezolizumab.Citation105 Preliminary results reported an ICI-naïve cohort of nine patients with mRCC with one prior line of therapy receiving the combination of autogene cevumeran with atezolizumab had an ORR of 22% (2 patients) in the dose-expansion phase.

Dendritic cell vaccines

DCs are the professional antigen-presenting cells of the immune system. DCs express high amounts of the costimulatory molecules CD40, CD80, and CD86 as well as high levels of MHCII, and it is becoming increasingly recognized that DCs are essential for anti-tumor immunity by initiating T-cell priming and through production of key cytokines such as interferon-gamma and IL-12.Citation106 Autologous DC vaccines expose tumor antigens to DCs in hopes of stimulating a tumor-specific immune response.

Rocapuldencel-T is a mature monocyte-derived DC that is coelectroporated with amplified autologous RCC tumor RNA (isolated from nephrectomy) and CD40L RNA. In the pre-ICI era, it was tested in combination with standard-of-care sunitinib in the phase 3 trial ADAPT (NCT01582672), which enrolled 462 patients with mRCC to receive either rocapuldencel-T with sunitinib or sunitinib alone.Citation107 The combination was unsuccessful, as the median OS was 27.7 months compared to 32.4 months with sunitinib alone (HR 1.10, 95% CI, 0.83–1.40), and the trial was terminated early. The study did show that patients with high numbers of regulatory T cells and IL-12 levels could be survival-predictive biomarkers for patients receiving rocapuldencel-T.

Now in the ICI era, a phase 2 trial (NCT01876212) for patients with ICI-refractory metastatic melanoma determined that a DC vaccine targeting tumor blood vessel antigens in combination with dasatinib could improve clinical outcomes.Citation108 A phase 2 trial (NCT04203901) is evaluating CMN-001 (an autologous, tumor antigen-loaded DC vaccine) in combination with ipilimumab/nivolumab against the control arm of ipilimumab/nivolumab in intermediate/poor risk patients with mRCC. Patients who progress in either cohort will be switched to lenvatinib/everolimus. Notably, the primary outcome measure of this trial is OS.

DCs and tumor cells can be fused together to become a heterokaryon that expresses both MHC Class I and II molecules, co-stimulatory molecules, and tumor-associated antigens. DC-cell tumor fusions can become a potent activator of the immune system and can induce strong anti-tumor immune responses over tumor cells alone as tumor cells themselves lack immune costimulatory molecules.Citation109 There is an active phase 1/2 trial (NCT00458536) for 38 patients with mRCC who have undergone a debulking nephrectomy and no prior systemic therapy, testing the safety of a DC-tumor fusion vaccine with granulocyte macrophage colony stimulating factor (GM-CSF). Patients receive three subcutaneous vaccinations of fused cells every 3 weeks. Six patients received only the vaccine, and the other patients received the study vaccine with GM-CSF.

Notably, there is a phase 2 trial (NCT05127824) for patients with localized RCC, in which 21 patients will be enrolled to receive neoadjuvant autologous tumor blood vessel antigen DC vaccine injections intradermally with cabozantinib prior to surgical resection, and they will be compared with 21 controls without RCC that will receive no intervention. The primary outcome measure is the probability of a CD8 T-cell immune response for the combination treatment.

Oncolytic viruses

Oncolytic viruses are designed to cause selective replication and lysis of cancer cells while sparing normal cells, and the release of immunogenic tumor proteins ideally provides tumor-associated antigens for APCs to stimulate the immune system to further destroy cancer cells.Citation110 Talimogene laherparepvec (T-VEC), the only regulatory-approved oncolytic immunotherapy which selectively replicates within tumors and secretes GM-CSF, is used to treat advanced melanoma, based on the findings from the successful phase 3 trial OPTiM (NCT00769704).Citation111 A phase 1/2 trial (NCT03294083) has enrolled 89 patients with mRCC (both frontline and refractory to anti-PD-1/PD-L1 therapy settings allowed) to evaluate safety and efficacy of Pexastimogene devacirepvec (Pexa-Vec, a modified version of the vaccinia virus with the GM-CSF gene and deletion of thymidine kinase gene) in combination with cemiplimab. Both intravenous infusion as well as direct intratumoral injection of Pexa-Vec are used.Citation112

The p53–expressing modified vaccinia Ankara (p53MVA) virus delivers the full-length p53 to antigen-presenting cells which can reactivate or generate de novo effector and memory T cell responses against numerous antigenic p53 epitopes, as many cancer cells express p53 variants on the cell surface while normal cells do not.Citation113 Aphase 1 trial (NCT02432963) is evaluating p53MVA in combination with pembrolizumab for patients with advanced solid tumors including RCC. Preliminary results of this trial of 11 patients (RCC not included) suggested this combination is feasible and safe.Citation113

CAR T-cell therapy

CAR T-cell therapy has received regulatory approval in various hematologic malignancies (in the autologous setting), and its use has begun to expand to metastatic solid tumors. There remain several challenges and setbacks to CAR T-cell therapy, not limited to on-target/off-tumor effects, immune effector cell-associated neurotoxicity, cytokine release syndrome, financial costs, and complex manufacturing requirements.Citation114 However, the main setback for CAR T cells in solid tumors is the lack of a ubiquitously expressed tumor antigen that the CAR T cells can target. CAR T-cell therapies for RCC have failed in the past, using the targets carbonic anhydrase IX (CAIX) due to liver toxicity (as CAIX is expressed on the bile duct epithelium creating on-target/off tumor effects),Citation115 and vascular endothelial growth factor receptor 2 (VEGFR2) due to no objective responses resulting in termination (NCT01218867). Despite these setbacks, there are several ongoing trials of CAR T cells designed to treat mRCC refractory to approved therapies. Some of the CAR T cells being used in RCC trials involve allogeneic CAR T cells, which decreases the manufacturing time for patients compared to autologous CAR T cells. RCC targets in current clinical trials include CAIX, CD70, ROR2, and AXL. A summary of these clinical trials is provided in .

Table 4. Current active and recruiting clinical trials CAR T-cell therapy in metastatic RCC.

CAIX

Carbonic anhydrase IX (CAIX) is a transmembrane protein that normally is upregulated in hypoxic stress in normal kidney cells. CAIX can become highly expressed on the surface of clear cell RCC tumor cells even in normal oxygen conditions, and its downstream effects ultimately lead to tumor progression.Citation116 CAIX is being actively studied as a target not just for CAR T-cell therapy but also in the field of theranostics. Girentuximab is a monoclonal antibody targeting CAIX. 89Zr-DFO-girentuximab (TLX250–CDx) PET/CT imaging is being evaluated in the phase 3 trial ZIRCON (NCT03849118), and data reported at the 2023 ASCO Genitourinary Cancers Symposium suggests it can accurately detect RCC for patients with indeterminate renal lesions.Citation117 The currently recruiting phase 2 trial STARLITE 2 (NCT05239533) is evaluating the combination of the radiopharmaceutical 177Lu-girentuximab and nivolumab in patients with mRCC who have failed prior treatment with an anti-PD-1/PD-L1 ICI.Citation118

As mentioned above, a prior experience with CAIX-directed CAR T cells for 12 patients with CAIX-expressing mRCC was disappointing because of liver toxicity.Citation119 A phase 1 trial (NCT04969354) is enrolling 20 patients with mRCC refractory to prior therapies (including up to two regimens of systemic treatments including anti-PD-1/PD-L1 ICIs) to receive CAIX-targeted CAR T cells, with primary outcome measures of adverse events and ORR. Prior to CAR T-cell infusion, patients receive a CAIX monoclonal antibody injected into the hepatic artery to minimize liver toxicity.

CD70

As mentioned in a prior section, CD70 interacts with CD27 to activate B and T cells in normal circumstances. However, for cancer cells, CD70 can be aberrantly expressed, which paradoxically facilitates immune evasion and tumor growth. CD70 is known to be highly expressed in some malignancies including RCC while having limited expression in normal tissue,Citation120 making it a promising target for CAR T-cell therapy. There are three trials dedicated to evaluating CD70–targeting CAR T cells for patients with RCC, and more that are evaluating CD70–targeting CAR T cells in solid tumors expressing CD70.

CTX130 is an allogeneic CAR T cell that was evaluated in the phase 1 trial COBALT-RCC (NCT04438083), which is enrolling 107 patients with mRCC who have failed all standard of care treatments. Patients receive the CTX130 infusion after lymphodepleting chemotherapy. Preliminary data presented at the 2022 SITC Annual Meeting showed that for 13 evaluable patients, the disease control rate (DCR) was 77%.Citation121 The ORR was 8% (one patient), but this patient developed a complete response by 3 months that was maintained at 18 months. Overall, the results were encouraging with a good safety profile and encouraging antitumor activity.

Similarly, ALLO-316 is an allogeneic CAR T cell that was evaluated in the phase 1 trial TRAVERSE (NCT04696731), which is enrolling 120 patients with mRCC who have progressed on prior ICI and tyrosine kinase inhibitor. Patients received a lymphodepleting deconditioning regimen prior to infusion with ALLO-316. Preliminary results presented at the 2023 AACR Annual Meeting revealed that for 18 evaluable patients, the ORR was 17%, DCR was 89%, while for 10 patients with CD70+ RCC, three patients achieved a partial response and the DCR was 100%.Citation122 Importantly, the safety profile was consistent with autologous CAR T-cell therapy. ALLO-316 also eliminated CD70+ host T cells, which prevented allorejection and preserved treatment efficacy.

Another phase 1 trial (NCT05420519) is enrolling 24 patients with mRCC who have failed all standard of care treatments to receive CD70–targeted CAR T cells, with primary endpoints of a maximum tolerated dose and to determine the incidence of adverse events. There are other phase 1 trials underway enrolling patients with CD70+ metastatic solid tumors including RCC (e.g., NCT05518253, NCT05468190, NCT05420545). The primary outcome measures of all these studies are adverse events and maximum tolerated dose. Two phase 1/2 trials (NCT02830724 and NCT05795595) are enrolling patients with relapsed and refractory CD70+ metastatic solid tumors to receive allogeneic CAR T cells, with primary outcome measures to include adverse events and ORR.

ROR2 and AXL

Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a tumor-associated kinase that has been associated with RCC tumor growth and migration, with high expression associated with poor outcomes.Citation123,Citation124 AXL is a receptor tyrosine kinase overexpressed in many cancers, and its expression on RCC is much higher than for normal kidney tissue,Citation125 and high expression of AXL in patients with RCC treated with sunitinib led to worse outcomes.Citation126 A phase 1/2 trial (NCT03393936) is evaluating autologous CAR T cells after lymphodepletion in 66 patients with mRCC refractory to standard therapies. Patients with RCC expressing ROR2 will receive CCT301–59 T cells, and patients with RCC expressing AXL will receive CCT301–38 T cells. The primary outcome measures are dose-limiting toxicity and ORR.

Conclusions

RCC continues to be a difficult malignancy to treat despite all the success of traditional immune checkpoint inhibitor therapies targeting CTLA-4, PD-1 and PD-L1. Many promising clinical trials are underway for RCC, evaluating novel immune approaches for both frontline and refractory/relapsed patients as either monotherapy or in combination with already approved agents. Given the adverse effects of the currently approved ICIs for solid tumors, the safety of these immunotherapies will be heavily scrutinized. Fortunately, many agents being evaluated that have demonstrated efficacy also appear to be safely tolerated. With more experience, the design of these novel therapies will further improve the clinical outcomes of patients with advanced RCC and will hopefully become more widely available to patients in the near future.

Disclosure statement

Jonathan E. Shoag received personal fees from Fortec Medical and grants from BMS Foundation. Jorge A. Garcia received personal fees from Pfizer, Aptitude Health, and the US Food and Drug Administration. Tian Zhang received research funding from Acerta, Novartis, Merrimack, AbbVie/StemCentrx, Merck, Regeneron, Mirati Therapeutics, Janssen, AstraZeneca, Pfizer, OmniSeq, Personal Genome Diagnostics, Astellas, and Eli Lilly, served as advisory board member and consultant for Merck, Exelixis, Sanofi-Aventis, Janssen, AstraZeneca, Pfizer, Amgen, BMS, Pharmacyclics, SeaGen, Calithera, QED Therapeutics, Eisai, Aveo, Eli Lilly, Bayer, and Aravive, and received honoraria from MJH Associates, Vaniam, Aptitude Health, and PeerView. Pedro C. Barata received personal fees from BMS, Merck, and EMD Serono, grants from Roche, personal fees from Eisai, Exelixis, Pfizer, Caris Life Sciences, Myovant, and UroToday, and nonfinancial support from Guardant360. All the other authors declare no conflict of interest.

Additional information

Funding

The author(s) reported that there is no funding associated with the work featured in this article.

References

  • Rini BI, McDermott DF, Hammers H, Bro W, Bukowski RM, Faba B, Faba J, Figlin RA, Hutson T, Jonasch E, et al. Society for immunotherapy of cancer consensus statement on immunotherapy for the treatment of renal cell carcinoma. J Immunother Cancer. 2016;4(1):81. doi:10.1186/s40425-016-0180-7.
  • Jang A, Adler DM, Rauterkus GP, Bilen MA, Barata PC. immunotherapies in genitourinary oncology: where are we now? Where are we going? Cancers Basel. 2021;13(20):5065. doi:10.3390/cancers13205065.
  • Sharma M, Khong H, Fa’ak F, Bentebibel SE, Janssen LME, Chesson BC, Creasy CA, Forget M-A, Kahn LMS, Pazdrak B, et al. Bempegaldesleukin selectively depletes intratumoral tregs and potentiates T cell-mediated cancer therapy. Nat Commun. 2020;11(1):661. doi:10.1038/s41467-020-14471-1.
  • Tannir NM, Cho DC, Diab A, Sznol M, Bilen MA, Balar AV, Grignani G, Puente E, Tang L, Chien D, et al. Bempegaldesleukin plus nivolumab in first-line renal cell carcinoma: results from the PIVOT-02 study. J Immunother Cancer. 2022;10(4):10. doi:10.1136/jitc-2021-004419.
  • Nektar and Bristol Myers Squibb announce update on clinical development program for bempegaldesleukin (BEMPEG) in combination with opdivo (nivolumab). Bristol Myers Squibb; 2022. https://news.bms.com/news/details/2022/Nektar-and-Bristol-Myers-Squibb-Announce-Update-on-Clinical-Development-Program-for-Bempegaldesleukin-BEMPEG-in-Combination-with-Opdivo-nivolumab/default.aspx#:~:text=(PRINCETON%2C%20N.J.%2C%20%26%20SAN,nivolumab)%20in%20renal%20cell%20carcinoma%20(.
  • Bristol Myers Squibb and Nektar announce update on phase 3 PIVOT IO-001 trial evaluating bempegaldesleukin (BEMPEG) in combination with opdivo (nivolumab) in previously untreated unresectable or metastatic melanoma. Bristol Myers Squibb; 2022. https://news.bms.com/news/details/2022/Bristol-Myers-Squibb-and-Nektar-Announce-Update-on-Phase-3-PIVOT-IO-001-Trial-Evaluating-Bempegaldesleukin-BEMPEG-in-Combination-with-Opdivo-nivolumab-in-Previously-Untreated-Unresectable-or-Metastatic-Melanoma/default.aspx.
  • Voss MH, Tykodi SS, Grimm M-O, Hammers HJ, Rini BI, Tannir NM, Qureshi AH, Tsipouri V, Hodari M, Ravimohan S, et al. PIVOT IO 011: a phase 1/2 study of bempegaldesleukin (BEMPEG; NKTR-214) plus nivolumab (NIVO) and tyrosine kinase inhibitor (TKI) versus NIVO and TKI alone in patients (pts) with previously untreated advanced or metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2022;40(6_suppl):TPS403–15. doi:10.1200/JCO.2022.40.6_suppl.TPS403.
  • Lopes JE, Fisher JL, Flick HL, Wang C, Sun L, Ernstoff MS, Alvarez JC, Losey HC. ALKS 4230: a novel engineered IL-2 fusion protein with an improved cellular selectivity profile for cancer immunotherapy. J Immunother Cancer. 2020;8(1):8. doi:10.1136/jitc-2020-000673.
  • Vaishampayan UN, Tomczak P, Muzaffar J, Winer IS, Rosen SD, Hoimes CJ, Chauhan A, Spreafico A, Lewis KD, Bruno DS, et al. Nemvaleukin alfa monotherapy and in combination with pembrolizumab in patients (pts) with advanced solid tumors: ARTISTRY-1. J Clin Oncol. 2022;40(16_suppl):2500. doi:10.1200/JCO.2022.40.16_suppl.2500.
  • Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA, Zaharoff DA. Localized interleukin-12 for cancer immunotherapy. Front Immunol. 2020;11:575597. doi:10.3389/fimmu.2020.575597.
  • Niglio SA, Girardi DdM, Cordes LM, Ley L, Mallek M, Sierra Ortiz O, Cadena J, Diaz C, Chalfin H, Kydd A, et al. A phase I study of bintrafusp alfa (M7824) and NHS-IL12 (M9241) alone and in combination with stereotactic body radiation therapy (SBRT) in adults with metastatic non-prostate genitourinary malignancies. J Clin Oncol. 2021;39(15_suppl):TPS4599–TPS. doi:10.1200/JCO.2021.39.15_suppl.TPS4599.
  • Strauss J, Heery CR, Schlom J, Madan RA, Cao L, Kang Z, Lamping E, Marté JL, Donahue RN, Grenga I, et al. Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFβ, in advanced solid tumors. Clin Cancer Res. 2018;24(6):1287–95. doi:10.1158/1078-0432.CCR-17-2653.
  • Fallon J, Tighe R, Kradjian G, Guzman W, Bernhardt A, Neuteboom B, Lan Y, Sabzevari H, Schlom J, Greiner JW, et al. The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget. 2014;5(7):1869–84. doi:10.18632/oncotarget.1853.
  • Xu C, Marelli B, Qi J, Qin G, Yu H, Wang H, Jenkins MH, Lo KM, Lan Y. NHS-IL12 and bintrafusp alfa combination therapy enhances antitumor activity in preclinical cancer models. Transl Oncol. 2022;16:101322. doi:10.1016/j.tranon.2021.101322.
  • Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, O’Shea MA, Fauci AS. The common γ-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol (Baltimore, Md : 1950). 2008;181(10):6738–46. doi: 10.4049/jimmunol.181.10.6738.
  • Zhou Y, Husman T, Cen X, Tsao T, Brown J, Bajpai A, Li M, Zhou K, Yang L. Interleukin 15 in cell-based cancer immunotherapy. Int J Mol Sci. 2022;23(13):23. doi:10.3390/ijms23137311.
  • Garralda E, Naing A, Galvao V, LoRusso P, Grell P, Cassier PA, Gomez-Roca CA, Korakis I, Bechard D, Palova Jelinkova L, et al. Interim safety and efficacy results from AURELIO-03: a phase 1 dose escalation study of the IL-2/IL-15 receptor βγ superagonist SOT101 as a single agent and in combination with pembrolizumab in patients with advanced solid tumors. J Clin Oncol. 2022;40(16_suppl):2502. doi:10.1200/JCO.2022.40.16_suppl.2502.
  • Wrangle JM, Awad MM, Badin FB, Rubinstein MP, Bhar P, Garner C, Reddy SK, Soon-Shiong P. Preliminary data from QUILT 3.055: a phase 2 multi-cohort study of N803 (IL-15 superagonist) in combination with checkpoint inhibitors (CPI). J Clin Oncol. 2021;39(15_suppl):2596. doi:10.1200/JCO.2021.39.15_suppl.2596.
  • Hunter CA. New IL-12-family members: IL-23 and IL-27, cytokines with divergent functions. Nat Rev Immunol. 2005;5(7):521–31. doi:10.1038/nri1648.
  • Fabbi M, Carbotti G, Ferrini S. Dual roles of IL-27 in cancer biology and immunotherapy. Mediators Inflamm. 2017;2017:3958069. doi:10.1155/2017/3958069.
  • Rausch M, Hua J, Moodley D, White KF, Walsh KH, Miller CE, Tan G, Lee BH, Cousineau I, Lattouf J-B, et al. Increased IL-27 is associated with poor prognosis in renal cell carcinoma and supports use of SRF388, a first-in-class IL-27p28 blocking antibody, to counteract IL-27-mediated immunosuppression in this setting. Cancer Res. 2020;80(16_Supplement):4550. doi:10.1158/1538-7445.AM2020-4550.
  • Naing A, Mantia C, Morgensztern D, Kim T-Y, Li D, Kang Y-K, Marron TU, Tripathi A, George S, Rini BI, et al. First-in-human study of SRF388, a first-in-class IL-27 targeting antibody, as monotherapy and in combination with pembrolizumab in patients with advanced solid tumors. J Clin Oncol. 2022;40(16_suppl):2501. doi:10.1200/JCO.2022.40.16_suppl.2501.
  • Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M, Hayakawa Y, Vance RE. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478(7370):515–8. doi:10.1038/nature10429.
  • Amouzegar A, Chelvanambi M, Filderman JN, Storkus WJ, Luke JJ. STING agonists as cancer therapeutics. Cancers Basel. 2021;13(11):2695. doi:10.3390/cancers13112695.
  • Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Man Lei Y, Jabri B, Alegre M-L, et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–9. doi:10.1126/science.aac4255.
  • Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–79. doi:10.1093/annonc/mdx108.
  • Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia (New York, NY). 2017;19(10):848–55. doi:10.1016/j.neo.2017.08.004.
  • Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi:10.1126/science.aan4236.
  • Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7. doi:10.1126/science.aan3706.
  • Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8. doi:10.1126/science.aao3290.
  • Choi Y, Lichterman JN, Coughlin LA, Poulides N, Li W, Del Valle P, Palmer SN, Gan S, Kim J, Zhan X, et al. Immune checkpoint blockade induces gut microbiota translocation that augments extraintestinal antitumor immunity. Sci Immunol. 2023;8(81):eabo2003. doi:10.1126/sciimmunol.abo2003.
  • Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, Long N, Plodkowski AJ, Arbour KC, Chaft JE, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol. 2018;29(6):1437–44. doi:10.1093/annonc/mdy103.
  • Xavier JB, Young VB, Skufca J, Ginty F, Testerman T, Pearson AT, Macklin P, Mitchell A, Shmulevich I, Xie L, et al. The cancer microbiome: distinguishing direct and indirect effects requires a systemic view. Trends Cancer. 2020;6(3):192–204. doi:10.1016/j.trecan.2020.01.004.
  • Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4):e202895. doi:10.1001/jamanetworkopen.2020.2895.
  • Tsikala-Vafea M, Belani N, Vieira K, Khan H, Farmakiotis D. Use of antibiotics is associated with worse clinical outcomes in patients with cancer treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Int J Infect Dis. 2021;106:142–54. doi:10.1016/j.ijid.2021.03.063.
  • Meza L, Feng M, Lee K, Sperandio R, Pal SK. The gut microbiome and metastatic renal cell carcinoma. J Clin Med. 2023;12(4):12. doi:10.3390/jcm12041502.
  • Stoeva MK, Garcia-So J, Justice N, Myers J, Tyagi S, Nemchek M, McMurdie PJ, Kolterman O, Eid J. Butyrate-producing human gut symbiont, clostridium butyricum, and its role in health and disease. Gut Microbes. 2021;13(1):1–28. doi:10.1080/19490976.2021.1907272.
  • Salgia NJ, Bergerot PG, Maia MC, Dizman N, Hsu J, Gillece JD, Folkerts M, Reining L, Trent J, Highlander SK, et al. Stool microbiome profiling of patients with metastatic renal cell carcinoma receiving anti–PD-1 immune checkpoint inhibitors. Eur Urol. 2020;78(4):498–502. doi:10.1016/j.eururo.2020.07.011.
  • Tomita Y, Ikeda T, Sakata S, Saruwatari K, Sato R, Iyama S, Jodai T, Akaike K, Ishizuka S, Saeki S, et al. Association of probiotic clostridium butyricum therapy with survival and response to immune checkpoint blockade in patients with lung cancer. Cancer Immunol Res. 2020;8(10):1236–42. doi:10.1158/2326-6066.CIR-20-0051.
  • Dizman N, Meza L, Bergerot P, Alcantara M, Dorff T, Lyou Y, Frankel P, Cui Y, Mira V, Llamas M, et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat Med. 2022;28(4):704–12. doi:10.1038/s41591-022-01694-6.
  • Meza LA, Lee K, Malhotra J, Alcantara M, Zengin ZB, Dizman N, Dizman N, Govindarajan A, Hsu J, Llamas-Quitiquit M, et al. Effect of CBM588 in combination with cabozantinib plus nivolumab for patients (pts) with metastatic renal cell carcinoma (mRCC): a randomized clinical trial. J Clin Oncol. 2023;41(17_suppl):LBA104–LBA. doi:10.1200/JCO.2023.41.17_suppl.LBA104.
  • Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–9. doi:10.1126/science.abb5920.
  • Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science. 2021;371(6529):595–602. doi:10.1126/science.abf3363.
  • Halsey TM, Thomas AS, Hayase T, Ma W, Abu-Sbeih H, Sun B, Parra ER, Jiang Z-D, DuPont HL, Sanchez C, et al. Microbiome alteration via fecal microbiota transplantation is effective for refractory immune checkpoint inhibitor–induced colitis. Sci Transl Med. 2023;15(700):eabq4006. doi:10.1126/scitranslmed.abq4006.
  • Porcari S, Ciccarese C, Pinto F, Quaranta G, Giorgi SD, Rondinella D, Settanni CR, Cortesi E, Roberto M, Primi F, et al. Fecal microbiota transplantation to improve efficacy of immune checkpoint inhibitors in renal cell carcinoma (TACITO trial). J Clin Oncol. 2022;40(6_suppl):TPS407–TPS. doi:10.1200/JCO.2022.40.6_suppl.TPS407.
  • Maurice-Dror C, Tirosh O, Ben-Shabat SK, Eshar S, Plolonsky O, Davis N, Haber E, Meshner S, Perets R. BMC128: a rationally designed live bacterial consortium for the potentiation of immune checkpoint therapy in solid tumors. J Clin Oncol. 2022;40(16_suppl):TPS2685–TPS. doi:10.1200/JCO.2022.40.16_suppl.TPS2685.
  • Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 2021;14(1):45. doi:10.1186/s13045-021-01056-8.
  • Maruhashi T, Sugiura D, Okazaki IM, Okazaki T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer. 2020;8(2):8. doi:10.1136/jitc-2020-001014.
  • Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Castillo Gutiérrez E, Rutkowski P, Gogas HJ, Lao CD, De Menezes JJ, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022;386(1):24–34. doi:10.1056/NEJMoa2109970.
  • Schöffski P, Tan DSW, Martín M, Ochoa-de-Olza M, Sarantopoulos J, Carvajal RD, Kyi C, Esaki T, Prawira A, Akerley W, et al. Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) ± anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J Immunother Cancer. 2022;10(2):10. doi:10.1136/jitc-2021-003776.
  • Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017;276(1):97–111. doi:10.1111/imr.12520.
  • Curigliano G, Gelderblom H, Mach N, Doi T, Tai D, Forde PM, Sarantopoulos J, Bedard PL, Lin C-C, Hodi FS, et al. Phase I/Ib clinical trial of sabatolimab, an anti–TIM-3 antibody, alone and in combination with Spartalizumab, an anti–PD-1 antibody, in advanced solid tumors. Clin Cancer Res. 2021;27(13):3620–9. doi:10.1158/1078-0432.CCR-20-4746.
  • Zhang L, Geng Z, Hao B, Geng Q. Tislelizumab: a modified anti-tumor programmed death receptor 1 antibody. Cancer Control. 2022;29:10732748221111296. doi:10.1177/10732748221111296.
  • Harjunpää H, Guillerey C. TIGIT as an emerging immune checkpoint. Clin Exp Immunol. 2020;200(2):108–19. doi:10.1111/cei.13407.
  • Cho BC, Abreu DR, Hussein M, Cobo M, Patel AJ, Secen N, Lee KH, Massuti B, Hiret S, Yang JCH, et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022;23(6):781–92. doi:10.1016/S1470-2045(22)00226-1.
  • Kang X, Kim J, Deng M, John S, Chen H, Wu G, Phan H, Zhang CC. Inhibitory leukocyte immunoglobulin-like receptors: immune checkpoint proteins and tumor sustaining factors. Cell Cycle. 2016;15(1):25–40. doi:10.1080/15384101.2015.1121324.
  • Rouas-Freiss N, LeMaoult J, Verine J, Tronik-Le Roux D, Culine S, Hennequin C, Desgrandchamps F, Carosella ED. Intratumor heterogeneity of immune checkpoints in primary renal cell cancer: focus on HLA-G/ILT2/ILT4. Oncoimmunology. 2017;6(9):e1342023. doi:10.1080/2162402X.2017.1342023.
  • Mondal K, Song C, Tian J, Ho C, Rivera L, Huang J, Chen P, Crawley S, Lin V, Sitrin J, et al. Preclinical evaluation of NGM707, a novel anti-ILT2/anti-ILT4 dual antagonist monoclonal antibody. Cancer Res 2021;81(13_Supplement):LB156. doi:10.1158/1538-7445.AM2021-LB156.
  • Cella M, Döhring C, Samaridis J, Dessing M, Brockhaus M, Lanzavecchia A, Colonna M. A novel inhibitory receptor (ILT3) expressed on monocytes, macrophages, and dendritic cells involved in antigen processing. J Exp Med. 1997;185(10):1743–51. doi:10.1084/jem.185.10.1743.
  • Singh L, Muise ES, Bhattacharya A, Grein J, Javaid S, Stivers P, Zhang J, Qu Y, Joyce-Shaikh B, Loboda A, et al. ILT3 (LILRB4) promotes the immunosuppressive function of tumor-educated human monocytic myeloid-derived suppressor cells. In: Molecular cancer research. Vol. 19. MCR; 2021. p. 702–16. doi:10.1158/1541-7786.MCR-20-0622.
  • Paavola KJ, Roda JM, Lin VY, Chen P, O’Hollaren KP, Ventura R, Crawley SC, Li B, Chen HIH, Malmersjö S, et al. The fibronectin–ILT3 interaction functions as a stromal checkpoint that suppresses myeloid cells. Cancer Immunol Res. 2021;9(11):1283–97. doi:10.1158/2326-6066.CIR-21-0240.
  • Qiu H, Shao Z, Wen X, Jiang J, Ma Q, Wang Y, Huang L, Ding X, Zhang L. TREM2: keeping pace with immune checkpoint inhibitors in cancer immunotherapy. Front Immunol. 2021;12:716710. doi:10.3389/fimmu.2021.716710.
  • Khantakova D, Brioschi S, Molgora M. Exploring the impact of TREM2 in tumor-associated macrophages. Vaccines. 2022;10(6):10. doi:10.3390/vaccines10060943.
  • Zhang H, Sheng L, Tao J, Chen R, Li Y, Sun Z, Qian W. Depletion of the triggering receptor expressed on myeloid cells 2 inhibits progression of renal cell carcinoma via regulating related protein expression and PTEN-PI3K/Akt pathway. Int J Oncol. 2016;49(6):2498–506. doi:10.3892/ijo.2016.3740.
  • Patnaik A, Hamilton EP, Winer IS, Tan W, Hubbard JM, Schenk EL, Sonbol MB, Jahchan N, Pierce K, Li Y, et al. A phase 1a dose-escalation study of PY314, a TREM2 (Triggering Receptor Expressed on Macrophages 2) targeting monoclonal antibody. J Clin Oncol. 2022;40(16_suppl):2648. doi:10.1200/JCO.2022.40.16_suppl.2648.
  • Wang L, Rubinstein R, Lines JL, Wasiuk A, Ahonen C, Guo Y, Lu L-F, Gondek D, Wang Y, Fava RA, et al. VISTA, a novel mouse ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208(3):577–92. doi:10.1084/jem.20100619.
  • Hong S, Yuan Q, Xia H, Zhu G, Feng Y, Wang Q, Zhang Z, He W, Lu J, Dong C, et al. Analysis of VISTA expression and function in renal cell carcinoma highlights VISTA as a potential target for immunotherapy. Protein Cell. 2019;10(11):840–5. doi:10.1007/s13238-019-0642-z.
  • Zapała Ł, Kunc M, Sharma S, Pęksa R, Popęda M, Biernat W, Radziszewski P. Immune checkpoint receptor VISTA on immune cells is associated with expression of T-cell exhaustion marker TOX and worse prognosis in renal cell carcinoma with venous tumor thrombus. J Cancer Res Clin Oncol. 2023;149(7):4131–9. doi:10.1007/s00432-022-04329-y.
  • Sasikumar PG, Sudarshan NS, Adurthi S, Ramachandra RK, Samiulla DS, Lakshminarasimhan A, Ramanathan A, Chandrasekhar T, Dhudashiya AA, Talapati SR, et al. PD-1 derived CA-170 is an oral immune checkpoint inhibitor that exhibits preclinical anti-tumor efficacy. Commun Biol. 2021;4(1):699. doi:10.1038/s42003-021-02191-1.
  • Bang Y, Sosman J, Daud A, Meric-Bernstam F, Garcia-Corbacho J, Patel M, Lee J, Kim KP, Brody J, Rha SY, et al. Phase 1 study of CA-170, a first-in-class, orally available, small molecule immune checkpoint inhibitor (ICI) dually targeting VISTA and PD-L1, in patients with advanced solid tumors or lymphomas. J Immunother Cancer. 2018;6:114.
  • Wojciechowicz K, Spodzieja M, Lisowska KA, Wardowska A. The role of the BTLA-HVEM complex in the pathogenesis of autoimmune diseases. Cell Immunol. 2022;376:104532. doi:10.1016/j.cellimm.2022.104532.
  • Alves Costa Silva C, Facchinetti F, Routy B, Derosa L. New pathways in immune stimulation: targeting OX40. Vol. 5. ESMO Open; 2020. doi:10.1136/esmoopen-2019-000573.
  • Kim AMJ, Nemeth MR, Lim SO. 4-1BB: a promising target for cancer immunotherapy. Front Oncol. 2022;12:968360. doi:10.3389/fonc.2022.968360.
  • Davis EJ, Martin-Liberal J, Kristeleit R, Cho DC, Blagden SP, Berthold D, Cardin DB, Vieito M, Miller RE, Hari Dass P, et al. First-in-human phase I/II, open-label study of the anti-OX40 agonist INCAGN01949 in patients with advanced solid tumors. J Immunother Cancer. 2022;10(10):10. doi:10.1136/jitc-2021-004235.
  • Gutierrez M, Moreno V, Heinhuis KM, Olszanski AJ, Spreafico A, Ong M, Chu Q, Carvajal RD, Trigo J, Ochoa de Olza M, et al. OX40 agonist BMS-986178 alone or in combination with nivolumab and/or ipilimumab in patients with advanced solid tumors. Clin Cancer Res. 2021;27(2):460–72. doi:10.1158/1078-0432.CCR-20-1830.
  • Sadeghi S, Parikh RA, Tsao-Wei DD, Groshen SG, Li M, Appleman LJ, Tagawa ST, Nanus DM, Molina AM, Kefauver C, et al. Phase II randomized double blind trial of axitinib (Axi) +/- PF-04518600, an OX40 antibody (PFOX) after PD1/PDL1 antibody (IO) therapy (Tx) in metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2022;40(16_suppl):4529–. doi:10.1200/JCO.2022.40.16_suppl.4529.
  • Fulton R, Gan J, Lu Y-Y, Crivello J, Duda Z, Wang Z, Silver R, Staskus A, Ortega C, Ellouze S, et al. Clinical approach and biomarker strategy for HFB301001, a novel OX40 agonistic antibody. Cancer Res. 2021;81(13_Supplement):1882. doi:10.1158/1538-7445.AM2021-1882.
  • Liu Y, Jiang B, Zhang T, Wang Z, Feng Y, Li H, Gong W, Wang X, Gao Y, Zhou X, et al. 699 A differentiated anti-OX40 agonist BGB-A445 does not block OX40-OX40L interaction and reveals remarkable anti-tumor efficacy in preclinical models. J Immunother Cancer. 2020;8:A420–A.
  • Rowell E, Kinkead H, Torretti E, Becklund B, Sulzmaier F, Crago W, Jones K, Timmer J, Deveraux Q, Eckelman B, et al. 856 INBRX-106: a novel hexavalent anti-OX40 agonist for the treatment of solid tumors. J Immunother Cancer. 2021;9:A897–A. doi:10.1136/jitc-2021-SITC2021.856.
  • Johnson M, Siu L, Hong D, Schoffski P, Galvao V, Rangwala F, Hernandez R, Gonzalez L, Ma B, Pandite L, et al. 494 Phase 1 dose escalation and dose expansion study of an agonist redirected checkpoint (ARC) fusion protein, SL-279252 (PD1-Fc-OX40L), in subjects with advanced solid tumors or lymphomas. J Immunother Cancer. 2021;9:A526–A. doi:10.1136/jitc-2021-SITC2021.494.
  • Tolcher AW, Sznol M, Hu-Lieskovan S, Papadopoulos KP, Patnaik A, Rasco DW, Di Gravio D, Huang B, Gambhire D, Chen Y, et al. Phase Ib study of utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in combination with pembrolizumab (MK-3475) in patients with advanced solid tumors. Clin Cancer Res. 2017;23(18):5349–57. doi:10.1158/1078-0432.CCR-17-1243.
  • Diab A, Hamid O, Thompson JA, Ros W, Eskens F, Doi T, Hu-Lieskovan S, Klempner SJ, Ganguly B, Fleener C, et al. A phase I, open-label, dose-escalation study of the OX40 agonist ivuxolimab in patients with locally advanced or metastatic cancers. Clin Cancer Res. 2022;28(1):71–83. doi:10.1158/1078-0432.CCR-21-0845.
  • Hamid O, Chiappori AA, Thompson JA, Doi T, Hu-Lieskovan S, Eskens F, Ros W, Diab A, Spano J-P, Rizvi NA, et al. First-in-human study of an OX40 (ivuxolimab) and 4-1BB (utomilumab) agonistic antibody combination in patients with advanced solid tumors. J Immunother Cancer. 2022;10(10):10. doi:10.1136/jitc-2022-005471.
  • Starzer AM, Berghoff AS. New emerging targets in cancer immunotherapy: CD27 (TNFRSF7). Vol. 4. ESMO Open; 2020. p. e000629. doi:10.1136/esmoopen-2019-000629.
  • Flieswasser T, Van den Eynde A, Van Audenaerde J, De Waele J, Lardon F, Riether C, de Haard H, Smits E, Pauwels P, Jacobs J, et al. The CD70-CD27 axis in oncology: the new kids on the block. J Exp Clin Cancer Res. 2022;41(1):12. doi:10.1186/s13046-021-02215-y.
  • Diegmann J, Junker K, Loncarevic IF, Michel S, Schimmel B, von Eggeling F. Immune escape for renal cell carcinoma: CD70 mediates apoptosis in lymphocytes. Neoplasia (New York, NY). 2006;8(11):933–8. doi:10.1593/neo.06451.
  • Sanborn RE, Pishvaian MJ, Callahan MK, Weise A, Sikic BI, Rahma O, Cho DC, Rizvi NA, Sznol M, Lutzky J, et al. Safety, tolerability and efficacy of agonist anti-CD27 antibody (varlilumab) administered in combination with anti-PD-1 (nivolumab) in advanced solid tumors. J Immunother Cancer. 2022;10(8):10. doi:10.1136/jitc-2022-005147.
  • Sun C, Wang B, Hao S. Adenosine-A2A receptor pathway in cancer immunotherapy. Front Immunol. 2022;13:837230. doi:10.3389/fimmu.2022.837230.
  • Chen S, Wainwright DA, Wu JD, Wan Y, Matei DE, Zhang Y, Zhang B. CD73: an emerging checkpoint for cancer immunotherapy. Immunotherapy. 2019;11(11):983–97. doi:10.2217/imt-2018-0200.
  • Fong L, Hotson A, Powderly JD, Sznol M, Heist RS, Choueiri TK, George S, Hughes BGM, Hellmann MD, Shepard DR, et al. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov. 2020;10(1):40–53. doi:10.1158/2159-8290.CD-19-0980.
  • Miller RA, Luke JJ, Hu S, Mahabhashyam S, Jones WB, Marron T, Merchan JR, Hughes BGM, Willingham SB. Anti-CD73 antibody activates human B cells, enhances humoral responses and induces redistribution of B cells in patients with cancer. J Immunother Cancer. 2022;10(12):10. doi:10.1136/jitc-2022-005802.
  • Luke JJ, Powderly JD, Merchan JR, Barve MA, Hotson AN, Mobasher M, Kwei L, Luciano G, Buggy JJ, Piccione E, et al. Immunobiology, preliminary safety, and efficacy of CPI-006, an anti-CD73 antibody with immune modulating activity, in a phase 1 trial in advanced cancers. J Clin Oncol. 2019;37(15_suppl):2505–. doi:10.1200/JCO.2019.37.15_suppl.2505.
  • Bai R, Lv Z, Xu D, Cui J. Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark Res. 2020;8(1):34. doi:10.1186/s40364-020-00209-0.
  • Labriola MK, Zhu J, Gupta RT, McCall S, Jackson J, Kong EF, White JR, Cerqueira G, Gerding K, Simmons JK, 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):e000319. doi:10.1136/jitc-2019-000319.
  • Joseph RW, Kapur P, Serie DJ, Parasramka M, Ho TH, Cheville JC, Frenkel E, Parker AS, Brugarolas J. Clear cell renal cell carcinoma subtypes identified by BAP1 and PBRM1 expression. J Urol. 2016;195(1):180–7. doi:10.1016/j.juro.2015.07.113.
  • Kapur P, Rajaram S, Brugarolas J. The expanding role of BAP1 in clear cell renal cell carcinoma. Hum Pathol. 2023;133:22–31. doi:10.1016/j.humpath.2022.07.022.
  • Dai J, Cui Y, Liang X, Xu J, Li J, Chen Y, Zhang E, Guo R. PBRM1 mutation as a predictive biomarker for immunotherapy in multiple cancers. Front Genet. 2022;13:1066347. doi:10.3389/fgene.2022.1066347.
  • Liu XD, Kong W, Peterson CB, McGrail DJ, Hoang A, Zhang X, Lam T, Pilie PG, Zhu H, Beckermann KE, et al. PBRM1 loss defines a nonimmunogenic tumor phenotype associated with checkpoint inhibitor resistance in renal carcinoma. Nat Commun. 2020;11(1):2135. doi:10.1038/s41467-020-15959-6.
  • Lu M, Zhao B, Liu M, Wu L, Li Y, Zhai Y, Shen X. Pan-cancer analysis of SETD2 mutation and its association with the efficacy of immunotherapy. NPJ Precis Oncol. 2021;5(1):51. doi:10.1038/s41698-021-00193-0.
  • Sönmez MG, Sönmez L. New treatment modalities with vaccine therapy in renal cell carcinoma. Urol Ann. 2019;11(2):119–25. doi:10.4103/UA.UA_166_17.
  • Abd-Aziz N, Poh CL, Ding X. Development of peptide-based vaccines for Cancer. J Oncol. 2022;2022:1–17. doi:10.1155/2022/9749363.
  • Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215–29. doi:10.1038/s41571-020-00460-2.
  • Rini BI, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, Weikert S, Bracarda S, Crabb SJ, Bedke J, et al. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17(11):1599–611. doi:10.1016/S1470-2045(16)30408-9.
  • Brentville VA, Metheringham RL, Daniels I, Atabani S, Symonds P, Cook KW, Vankemmelbeke M, Choudhury R, Vaghela P, Gijon M, et al. Combination vaccine based on citrullinated vimentin and enolase peptides induces potent CD4-mediated anti-tumor responses. J Immunother Cancer. 2020;8(1):8. doi:10.1136/jitc-2020-000560.
  • Lopez JS, Camidge R, Iafolla M, Rottey S, Schuler M, Hellmann M, Balmanoukian A, Dirix L, Gordon M, Sullivan R, et al. Abstract CT301: a phase Ib study to evaluate RO7198457, an individualized neoantigen specific immunoTherapy (iNeST), in combination with atezolizumab in patients with locally advanced or metastatic solid tumors. Cancer Res. 2020;80(16_Supplement):CT301. doi:10.1158/1538-7445.AM2020-CT301.
  • Marciscano AE, Anandasabapathy N. The role of dendritic cells in cancer and anti-tumor immunity. Semin Immunol. 2021;52:101481. doi:10.1016/j.smim.2021.101481.
  • Figlin RA, Tannir NM, Uzzo RG, Tykodi SS, Chen DYT, Master V, Kapoor A, Vaena D, Lowrance W, Bratslavsky G, et al. Results of the ADAPT phase 3 study of rocapuldencel-T in combination with sunitinib as first-line therapy in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2020;26(10):2327–36. doi:10.1158/1078-0432.CCR-19-2427.
  • Storkus WJ, Maurer D, Lin Y, Ding F, Bose A, Lowe D, Rose A, DeMark M, Karapetyan L, Taylor JL, et al. Dendritic cell vaccines targeting tumor blood vessel antigens in combination with dasatinib induce therapeutic immune responses in patients with checkpoint-refractory advanced melanoma. J Immunother Cancer. 2021;9(11):e003675. doi:10.1136/jitc-2021-003675.
  • Koido S, Homma S, Okamoto M, Namiki Y, Takakura K, Uchiyama K, Kajihara M, Arihiro S, Imazu H, Arakawa H, et al. Fusions between dendritic cells and whole tumor cells as anticancer vaccines. Oncoimmunology. 2013;2(5):e24437. doi:10.4161/onci.24437.
  • Bartlett DL, Liu Z, Sathaiah M, Ravindranathan R, Guo Z, He Y, Guo ZS. Oncolytic viruses as therapeutic cancer vaccines. Mol Cancer. 2013;12(1):103. doi:10.1186/1476-4598-12-103.
  • Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33(25):2780–8. doi:10.1200/JCO.2014.58.3377.
  • Rha SY, Merchan J, Oh SY, Kim C, Bae WK, Lee HW, Dillon M, Deering R, Kroog G, Ha KS, et al. Abstract CT121: a phase Ib study of recombinant vaccinia virus in combination with immune checkpoint inhibition (ICI) in advanced renal cell carcinoma (RCC). Cancer Res. 2020;80(16_Supplement):CT121–CT. doi:10.1158/1538-7445.AM2020-CT121.
  • Chung V, Kos FJ, Hardwick N, Yuan Y, Chao J, Li D, Waisman J, Li M, Zurcher K, Frankel P, et al. Evaluation of safety and efficacy of p53MVA vaccine combined with pembrolizumab in patients with advanced solid cancers. Clin Transl Oncol. 2019;21(3):363–72. doi:10.1007/s12094-018-1932-2.
  • Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69. doi:10.1038/s41408-021-00459-7.
  • Lamers CH, Klaver Y, Gratama JW, Sleijfer S, Debets R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells–a completed study overview. Biochem Soc Trans. 2016;44(3):951–9. doi:10.1042/BST20160037.
  • Courcier J, de la Taille A, Nourieh M, Leguerney I, Lassau N, Ingels A. Carbonic anhydrase IX in renal cell carcinoma, implications for disease management. Int J Mol Sci. 2020;21(19):21. doi:10.3390/ijms21197146.
  • Shuch BM, Pantuck AJ, Bernhard J-C, Morris MA, Master VA, Scott AM, Van Praet C, Bailly C, Aksoy T, Merkx R, et al. Results from phase 3 study of 89Zr-DFO-girentuximab for PET/CT imaging of clear cell renal cell carcinoma (ZIRCON). J Clin Oncol. 2023;41(6_suppl):LBA602–LBA. doi:10.1200/JCO.2023.41.6_suppl.LBA602.
  • Feldman DR, Motzer RJ, Knezevic A, Lee C-H, Voss MH, Lyashchenko SK, Park H, Larson SM, Pandit-Taskar N. STARLITE 2: Phase 2 study of nivolumab plus 177Lutetium-labeled anti-carbonic anhydrase IX (CAIX) monoclonal antibody girentuximab (177Lu-girentuximab) in patients (pts) with advanced clear cell renal cell carcinoma (ccRCC). J Clin Oncol. 2022;40(16_suppl):TPS4603–TPS. doi:10.1200/JCO.2022.40.16_suppl.TPS4603.
  • Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, Vulto A, den Bakker M, Oosterwijk E, Debets R, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. 2013;21(4):904–12. doi:10.1038/mt.2013.17.
  • Jilaveanu LB, Sznol J, Aziz SA, Duchen D, Kluger HM, Camp RL. CD70 expression patterns in renal cell carcinoma. Hum Pathol. 2012;43(9):1394–9. doi:10.1016/j.humpath.2011.10.014.
  • Pal S, Tran B, Haanen J, Hurwitz M, Sacher A, Agarwal N, Tannir N, Budde E, Harrison S, Klobuch S, et al. 558 CTX130 allogeneic CRISPR-Cas9–engineered chimeric antigen receptor (CAR) T cells in patients with advanced clear cell renal cell carcinoma: results from the phase 1 COBALT-RCC study. J Immunother Cancer. 2022;10:A584–A.
  • Srour S, Kotecha R, Curti B, Chahoud J, Drakaki A, Tang L, Goyal L, Prashad S, Szenes V, Norwood K, et al. Abstract CT011: a phase 1 multicenter study (TRAVERSE) evaluating the safety and efficacy of ALLO-316 following conditioning regimen in pts with advanced or metastatic clear cell renal cell carcinoma (ccRCC). Cancer Res. 2023;83(8_Supplement):CT011–CT. doi:10.1158/1538-7445.AM2023-CT011.
  • Rasmussen NR, Debebe Z, Wright TM, Brooks SA, Sendor AB, Brannon AR, Hakimi AA, Hsieh JJ, Choueiri TK, Tamboli P, et al. Expression of Ror2 mediates invasive phenotypes in renal cell carcinoma. PLoS One. 2014;9(12):e116101. doi:10.1371/journal.pone.0116101.
  • Yang CM, Ji S, Li Y, Fu LY, Jiang T, Meng FD. Ror2, a developmentally regulated kinase, is associated with tumor growth, apoptosis, migration, and invasion in renal cell carcinoma. Oncol Res. 2017;25(2):195–205. doi:10.3727/096504016X14732772150424.
  • Yu H, Liu R, Ma B, Li X, Yen HY, Zhou Y, Krasnoperov V, Xia Z, Zhang X, Bove AM, et al. Axl receptor tyrosine kinase is a potential therapeutic target in renal cell carcinoma. Br J Cancer. 2015;113(4):616–25. doi:10.1038/bjc.2015.237.
  • Zucca LE, Morini Matushita MA, da Silva Oliveira RJ, Scapulatempo-Neto C, de Lima MA, Ribeiro GG, Viana CR, Cárcano FM, Reis RM. Expression of tyrosine kinase receptor AXL is associated with worse outcome of metastatic renal cell carcinomas treated with sunitinib. Urol Oncol. 2018;36(1):11.e3–.e21. doi:10.1016/j.urolonc.2017.09.003.