Download PDF
Review  |  Open Access  |  26 Jun 2023

Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors

Views: 569 |  Downloads: 85 |  Cited:   2
Cancer Drug Resist 2023;6:416-29.
10.20517/cdr.2023.02 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of renal cell carcinoma. The prognosis for patients with ccRCC has improved over recent years with the use of combination therapies with an anti-programmed death-1 (PD-1) backbone. This has enhanced the quality of life and life expectancy of patients with this disease. Unfortunately, not all patients benefit; eventually, most patients will develop resistance to therapy and progress. Recent molecular, biochemical, and immunological research has extensively researched anti-angiogenic and immune-based treatment resistance mechanisms. This analysis offers an overview of the principles underpinning the resistance pathways related to immune checkpoint inhibitors (ICIs). Additionally, novel approaches to overcome resistance that may be considered for the trial context are discussed.

Keywords

Renal cell carcinoma, immunotherapy, treatment resistance, tumor microenvironment, intrinsic factors

INTRODUCTION

Renal cell carcinoma (RCC) accounts for 3%-5% of all malignancies in adults, with an incidence of about 400,000 cases yearly and mortality in approximately 175,000 cases worldwide in 2020[1-4]. Around one-fourth of patients present initially with metastasis, with the rest recurring after nephrectomy[5].

The most common histological pattern of renal tumors is clear cell renal cell carcinoma (ccRCC), which accounts for 70%-90% of the disease, followed by papillary (10%-15%) and chromophobe RCCs (3%-5%)[6,7].

In the past few decades, the mainstay of treatment of metastatic RCC (mRCC) involved vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKI)[8,9]. Monoclonal approaches to the same target, as characterized by bevacizumab, have been unsuccessful. A study by Motzer et al. showed no improvement in OS in patients with mRCC treated with atezolizumab plus bevacizumab[10]. When this immunologically “hot” tumor microenvironment is exposed to immune checkpoint inhibitors and/or inhibitors of VEGF as a combination, outcomes are significantly and clinically improved compared to VEGFR-TKI monotherapy alone[7,11]. Unfortunately, a cure is still not achievable for the vast majority of these patients presenting in the metastatic setting.

This review aims to summarize the molecular mechanisms that drive resistance to immune checkpoint inhibitors and their clinical implications in patients with mRCC.

Molecular mechanism of resistance to immune checkpoint inhibitors

Resistance to ICIs therapy has been classified into primary (initial) resistance and secondary (acquired) resistance[12]. Part of the classification is based on imaging response to ICI therapy, defined by the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 criteria[13]. Primary resistance is defined as the best response of progressive disease (PD) or stable disease (SD) for less than 6 months of ICI initiation. Patients with secondary resistance develop progressive disease after having a response or stable disease for greater than six months. Various mechanisms have been postulated for developing primary and secondary resistance in solid tumors, including mRCC, which can be broadly subcategorized into patient-related factors, tumor cell-related factors, or factors related to the tumor microenvironment (TME). The precise role of these mechanisms and a variety of others in primary and secondary resistance in the context of mRCC remains to be fully elucidated. However, a better understanding of these complex mechanisms is crucial to optimizing treatment approaches for those resistant to mRCC disease[14].

Patients’-intrinsic factors

Patients’-intrinsic factors play a vital role in the immune response to the therapy. Several factors are related to the immune response in cancer therapy and are summarized in Table 1.

Table 1

Molecular mechanism of resistance - patient’s intrinsic factors

FactorTheorized mechanismPotential outcomes
Gender[15-18]Exposure to mutagenic factors (i.e., Smoking, exposure is higher in men)
Differences in tumor histology
Estrogen modulation leads to:
(a) Increased expression of FoxP3
(b) Modulation of PD-1 expression
Increased regulatory T-cell, dendritic cell, and macrophage activity.
Obesity[19]Both pro-tumor and anti-tumor activityMixed outcomes in patients with RCC
Sarcopenia[95]Low skeletal mass indexWorse prognosticator
HLA genotype[25,26]Classical HLA-1 Molecules resulting in High Divergent ExpressionGreater response to ICI treatment
Gut microbiome[27,28]Fecal Microbiota Transplant resulting in upregulation of CD4+ T cell and PD-L1Improved ICI response
Antibiotic/Steroid use[29,33]Antibiotic Use causing dysbiosisDecreased ICI response
Steroids altering T-cell activation, altering gut microbiotaMay result in favorable disease course due to other factors

Gender

Gender-related factors may be attributed to confounding behaviors, such as male patients with increased exposure to smoking[15]. Another possible hypothesis is the role of estrogen modulation. Estrogen upregulates the activities of T-cells and PD-1 expression on cells[16,17]. A recent meta-analysis showed a significant disparity in overall survival (OS) between men and women with metastatic cancers with pooled OS in men was HR = 0.72 (95%CI: 0.65-0.79) and in women was HR = 0.86 (95%CI: 0.79-0.93), with a positive interaction value[15].

In a randomized trial in patients with ccRCC, treated with nivolumab or everolimus in a second-line setting, the hazard ratio (HR) for death in males was 0.70 (95%CI 0.50-0.90) compared to 0.84 (0.57-1.24) in women, which was similar to patients with melanoma or small cell lung carcinoma treated with ICIs[18].

Obesity

Obesity has shown variable modification capabilities in a human cell with both pro-tumor (increased VEGF, vascular endothelial growth factor, hypoxia, angiogenesis, plasmacytoid dendritic cells, and mast cell infiltration) and anti-tumor activities (increased T-cell and natural-killer cell response and decrease intratumor PDL-1 expression)[19]. Several studies have shown the beneficial effects of obesity on survival in patients with RCC[20,21]. Utilizing CT based body composition, correlations with specific phenotypes were found (i.e., Adipose, skeletal density vs. sarcopenia). While the exact mechanism to an increased BMI results in positive outcomes, theories include tumorigenic immune dysfunction vs. T-cell antigen cross-reactivity. This has the potential to improve sensitivity to ICI therapy. In contrast, an analysis of those with a similar phenotypic profile who were receiving Anti-PD-1 therapy showed a reduction in therapeutic response (lower PFS) as compared to their leaner counterparts[22-24].

Malnutrition and sarcopenia

Sarcopenia has been associated with adverse outcomes even before the immunotherapy era[22-24]. A retrospective multicenter real-world study found that sarcopenic patients had significantly worse OS (HR = 2.2, 95%CI: 1.3-3.6, P = 0.0026) when treated with ICIs. On multivariate analysis, low muscle mass was associated with an inferior OS[25]. Similarly, malnutrition has shown poor prognosis and quality of life in patients with lung cancer and is useful in predicting the response to the treatment[26,27].

Human leucocyte antigen genotype

Human leucocyte antigen (HLA) class 1 genes usually underlie the control of cancer with the diverged presentation of proteins that can influence response to ICIs[25,26]. This hypothesis suggests that patients with heterozygous divergent alleles have a more extensive presentation of peptides to assist T-cell response than less divergent HLA alleles. Patients with melanoma having high divergent alleles receiving ICIs respond better to the treatment. The outcomes were significantly different with 20 months in patients with high divergent alleles than 8 months in patients with low-divergent alleles (HR = 0.43, 95%CI: 0.2-0.8, P = 0.0094)[26]. Similar outcomes were observed in non-small cell lung cancer patients who received ICIs[25,26]. So far, there is a lack of specific studies for patients with RCC to understand the difference between high versus low divergent alleles and their response to the treatment[25,26].

Gut microbiota

Fecal microbiota transplantation (FMT) has been studied as a predictive factor for a response to ICIs treatment. Routy et al. found a positive correlation between response to ICI and Akkermansia muciniphila in mice receiving anti-PD-1 blockade[27]. It has also been noticed that the FMT from a patient who responded to ICIs in germ-free mice improves ICI efficiency but, unfortunately, not in non-responding patients. FMT upregulates helper T-cells (CD4+) and natural killer T- cells (NKT) in the spleen, resulting in improved response to ICIs. In a prospective randomized control trial, the gut microbiome modulation in patients with mRCC has been associated with improved PFS[28], which will be discussed later in this paper.

Antibiotics and use of steroids

The alteration in the gut microbiome predicts the response to immunotherapy. It has been observed that dysbiosis with antibiotics has been associated with poor outcomes in patients treated with ICIs[29]. Other retrospective studies by Lalani[30], Derosa et al.[31], and Tinsley et al.[32] also showed a negative association between antibiotic use and outcomes in patients with RCC, with a significant decrease in PFS[30-32]. Derosa et al.[31] and Tinsley et al.[32] also showed a reduction in OS. On multivariate analysis, antibiotic use was an independent predictive factor for worse outcomes in patients with RCC[31,32].

The use of steroids is routine in oncology patients for symptom management. A dose of > 10 mg prednisone or equivalent induces immunosuppression by altering the T cell activation with the expansion of M2 macrophages and thus altering the gut microenvironment. In a multicentre study by Arbour et al.[33], baseline higher steroid dose was associated with lower ORR (7% vs 18%), progression-free survival (PFS) (P < 0.001), and OS (P < 0.001). However, steroids used to manage immune-related adverse events have shown no negative impact on the efficiency of ICIs[34-36]. Indeed, patients that develop severe immune-related adverse events and need systemic steroids have a more favorable disease course in recent real-world data[37].

TUMOR CELL INTRINSIC FACTORS

Tumor cell-intrinsic factors are cell-related factors that can identify the response to the therapy and are summarized in Table 2.

Table 2

Molecular mechanism of resistance - tumor cell-intrinsic factor

FactorTheorized mechanismPotential outcomes
IFN-γ dysregulation[39-41]Loss of JAK-1&2 Function resulting in decreased PD-L1 expression Resistance to PD1 Inhibitors
Decreased immune cell recruitment with decreased MHC-1 antigen presentationIncreased proliferation and decreased apoptosis
MAPK[38,42]Increased production of VEGF, IL-6,8 and 10 inhibiting T-cell functionIncreased resistance
Downregulation of antigen/MHC expressionIncreased proliferation and decreased apoptosis
PI3K/AKT alteration[43,44]Loss of expression of PTEN resulting in:
Cytokine suppression
Inhibition of auto-phagosome activity
Decrease ICI response due to poor recruitment.
Wnt/β-catenin overexpression[45,46,48]Absence of T-cell expression and T-cell exclusionResistance to ICI therapy
Loss of MHC[96-99]Loss of MHC-1 and 2Resistance to ICI therapy

Signalling pathways

Interferon-gamma signaling pathway

The activation of interferon-gamma (IFN-γ) activates Janus kinase (JAK), and interferon regulatory factor 1 (IRF1), leading to the expression of PDL1. Patients with dysregulation of the IFN-γ pathway were shown to develop resistance to ICIs[38]. The IFN-γ recruits immune cells and enhances MHC-1 antigen presentation, which is essential for the antiproliferative and proapoptotic signals[39,40]. The dysregulation of JAK genes has been associated with a lack of response to the IFN-γ and, thus, with PD1 inhibitors[41].

Mitogen-activated protein kinases

Mitogen-activated protein kinases (MAPK) is associated with VEGF, IL-6, IL-8, and IL-10 production, inhibiting T-cell functions and immune cell recruitment. The MAPK pathways downregulate antigen presentation and MHC expression and decrease the sensitivity to antiproliferative effects of IFN-γ and TNF alpha[38,42].

PI3K/mTOR/AKT pathway

Patients with ccRCC have altered PI3K/AKT pathway along with alteration in the expression of PTEN[43]. These lead to the inhibition of immunosuppressive cytokines and auto phagosome, resulting in decreased T cell response, cell recruitment, and cell-mediated death. The loss of PTEN has been associated with the worse outcome in patients treated with ICIs therapy[44].

Wnt/β-catenin pathway

Wnt/β -catenin is involved with stem cell embryogenesis, immune regulation, and cell differentiation. In most cancers, it is overexpressed, reducing the expression of T cells and thus resulting in resistance to ICIs[45-48].

Insufficient tumor antigenicity

Investigations highlighted a correlation between poor antigenicity and decreased sensitivity to ICIs[49,50]. Repeat analysis supports the idea that these tumor neoantigens can be targeted to promote a positive response to checkpoint blockade. This concept can be applied to a variety of malignancies.

It is important to highlight that despite an understanding of the molecular patterns of resistance to ICIs in this context, no current biomarkers, molecular or otherwise, are validated for use in the clinic to help determine therapy selection.

TUMOR MICROENVIRONMENT

Tumor microenvironment (Tme) includes tumor cells and other cells interacting with them. These cells are crucial for tumor cells’ development, progression, and relapse. Factors that can affect the TME are summarized in Table 3.

Table 3

Molecular mechanism of resistance - tumor microenvironment

FactorTheorized mechanismPotential outcomes
Tumor-associated Macrophage activation[51-53]Greater M1 and M2 Macrophage activation, M2 (CD163+) subset correlated to poor survival outcomesNivolumab improved PFS in high M2 density activation
High T-cell density[56]Higher CD8+ infiltration associated with poorer clinical outcomes in ccRCC patientsPrognosticator for worse potential outcomes
B-cell and tertiary lymphoid structures[50,57,62]Increased B cell-related genes associated with strong memory responseGreat response to therapy
Tertiary lymphoid structures promote Regulatory T-cell activationPrognosticator for positive potential outcomes.
Hypoxia[64-67]Increased recruitment and infiltration of myeloid-driven suppressor cells, decreased function of cytotoxic T-cellsImmune evasion
Induction of HIF-1a and 2a, increasing PD-L1 expression, and VEGF generationImmune evasion

Tumor-associated macrophage

During inflammation, macrophages transform to M1 (classical) or M2 (alternative) activation. M1 produces inflammatory cytokines (ie. IL-6, IL-12, and IL-23), whereas the later correlates with the subsets of M2a, M2b, M2c, and M2d, respectively[51,52]. In 2011, a study found poor survival in patients with RCC with a high burden of M2 (CD163+)[53]. The diverse and extensive tumor-associated macrophages in the TME of patients with RCC indicate cancer progression and metastases. Strategies to direct therapeutic response to suppress tumor-associated macrophage recruitment have shown some improvement in the response[54], but macrophage-targeted therapies still need to be implemented in clinical settings.

The TME associated with abundant cells has shown an overall better response to ICIs (P < 0.001) as compared to TKI treatment (P = 0.15)[55]. Voss et al. found that the TME, with abundant infiltrative cell types, was associated with a good response to ICIs (P < 0.001), which was the opposite in patients treated with TKIs (P = 0.15)[55]. In another trial, patients treated with nivolumab having higher densities of M2 macrophages showed better PFS (HR = 0.69, P = 0.016), but no significant improvement in OS (P = 0.5)[56].

T cells

RCC is a tumor mainly enriched with T cells, particularly CD8+ T cells. The presence of CD8+ T cells is correlated with poor outcomes[57,58]. Choueiri et al. reported that high CD8 infiltration was associated with poor clinical outcomes in patients with ccRCC treated with sunitinib versus the avelumab-axitinib combination[59]. This was discordant from another phase II study, NIVOREN-GETUG AFU 26, which showed that patients with high CD8+ infiltration treated with Nivolumab have poor PFS (HR = 3.96, P < 0.0001) and OS (HR = 2.43, P = 0.04)[56]. Interestingly, Voss. et al. identified no correlation between the CD8+ cell infiltrations and survival in mRCC when treated with ICIs[55].

The Cancer Genome Atlas analysis showed that patients with RCC have more regulatory T-cells and are associated with poor clinical outcomes (HR = 1.59, 95%CI: 1.23-0.06; P < 0.01)[60].

B cells and tertiary lymphoid structures

B cells assist T-cell regulation and differentiation through different pathway activation, including interleukin (IL) and growth factors[49]. The memory of classical B cells has been associated with response against tumor-associated antigens[50]. The Microenvironment Cells Populations-counter (MCP-counter) analysis showed that responders have higher B cell densities in the TME than non-responders in melanoma and ccRCC[61].

Tertiary lymphoid structures are also associated with better outcomes in oncology patients. It assists in transforming cells and activating T regulatory cells[57,62].

Hypoxia

Tumors in the hypervascular environment led to insufficient nutrition and oxygen intake, resulting in hypoxia and progression[63]. Hypoxia induces upregulations of myeloid-driven suppressor cells that lead to decreased function of cytotoxic T cells[64,65]. Hypoxia also induces factor 1a (HIF-1a) and 2a (HIF-2a) to express PD-L1 in tumor cells[66,67]. Elevated levels of HIF are associated with generating VEGF, which acts as an immune escape mechanism

by upregulating CTLA4, TIM3, LAG3, and PD-L1 on dendritic cells[65,68]. Hypoxia causes tissue deposits of adenosine which suppresses the activity of T cells[69].

CLINICAL IMPLICATION OF ICIs RESISTANCE

ICI resistance in the clinical trial setting

Despite therapeutic advances, ICI resistance remains a significant issue in clinical trials and real-world settings due to a lack of validated clinical tools to identify patients at risk for primary or secondary resistance. Currently, there is no clinically accessible molecular classification of mRCC, and although clinical trials investigating molecular biomarkers in mRCC populations are promising, such approaches remain restricted to the research domain.

Molecular subtypes as biomarkers of ICI response

Using molecular markers to identify subgroups of mRCC patients who experience more robust responses to ICI therapy remains a promising avenue of study.

Transcriptomic analysis in a subset of primary resected ccRCC samples obtained from patients with metastatic disease identified a 35-gene signature capable of subgrouping metastatic clear-cell renal cell carcinoma (ccmRCC) into four groups (ccRCC 1-4). These subtypes were associated with differential response to sunitinib treatment, with ccRCC 1 and 4 tumors having a lower response rate and poorer survival outcomes than ccRCC 2 and ccRCC 3 subtypes. Molecular characterization of these tumor subtypes identified unique tumor microenvironments that may explain the relative response to sunitinib treatment. For example, ccRCC 4 cancers were associated with a suppressive immune microenvironment with overexpression of PD-L1 and PD-L2, while the ccRCC 2 subgroups demonstrated a more pro-angiogenic subtype[70]. These results lead to the hypothesis that these molecular subtypes could stratify patients into groups most likely to benefit from TKI therapy, ICI monotherapy, and combined ICI therapy approaches.

This hypothesis was tested in the phase II BIONIKK trial, the first to stratify patients into treatment groups based on their molecular subtype. Patients were divided into the ccRCC risk group using the aforementioned 35-gene transcriptomic signature and were prospectively allocated into treatment groups. Patients with ccRCC 1 or 4 tumors were assigned to anti-PD1 monotherapy with nivolumab alone or anti-PD1/anti-CTLA4 with a combination of nivolumab-ipilimumab. Those patients with ccRCC 2 or ccRCC 3 group tumors received TKI therapy and a combination of nivolumab-ipilimumab. The primary outcome of this study was the overall response rate per the RECIST criteria, with survival outcomes, tolerability, and duration of the response being secondary outcomes[71]. Meylan et al.[72] further identified the biomarkers for the efficacy of Nivolumab (N) +/- Ipilimumab (I) in mRCC patients with TLS > 2 treated with N or NI showed a response rate of 73% and 71%, respectively. Both TLS > 2 and higher densities of Ki67/PD1 correlated with better response rates (80% vs. 43% P < 0.01) and decreased incidence of progression events (5% vs 36%, P = 0.02).

Another example of molecular subtyping in a trial setting comes from the IMmotion 150 and 151 study and the ongoing phase II OPTIC trial. The IMmotion 150 trial compared treatment with atezolizumab alone or in combination with bevacizumab to sunitinib in a cohort of 305 patients with previously untreated metastatic renal cell carcinoma. Though treatment with a combination of atezolizumab alone or in combination with bevacizumab did not demonstrate improved PFS, the retrospective analysis did identify gene expression signatures associated with PFS involving angiogenesis, immunity (primarily T-effector presence and function), and myeloid inflammatory pathways. In particular, patients with a high angiogenesis signature score had an improved overall response rate and a longer PFS within the treatment sunitinib arm compared to those with low signature scores. Those with a high T-effector gene signature score experienced an improved overall response rate and longer PFS than those with a low score in the atezolizumab plus bevacizumab arm. In contrast, those with a high myeloid inflammation gene expression score experienced a shorter PFS in the atezolizumab plus bevacizumab and atezolizumab monotherapy treatment arms. These results suggest that such gene expression signatures may be useful tools in identifying patients more likely to benefit from various treatment regimens[73].

The findings of the IMmotion 150 study were validated and expanded upon in the phase 3 open-label IMmotion 151 study, which enrolled 915 patients and allocated them to receive either a combination of atezolizumab plus bevacizumab or sunitinib monotherapy. While this trial failed to demonstrate an overall survival benefit to combination therapy, it has provided rich biomarker data for mRCC molecular subtyping[74]. The IMmotion 151 trial retrospectively assigned patients to subgroups based on the previously identified gene expression signatures developed in the IMmotion 150 cohort. The authors again demonstrated that patients with a high angiogenesis signature score had improved PFS in the sunitinib arm, and those treated with atezolizumab plus bevacizumab had an improved PFS compared to sunitinib in subgroups with a high T-effector score or a low angiogenesis score. Based on these analyses, patients enrolled in the IMmotion 151 trial were further retrospectively classified into seven molecular subtypes - termed clusters - according to their unique genomic and transcriptomic enrichment pathways. Clusters 1 (angiogenic/stromal) and 2 (angiogenic) were enriched among the favorable risk groups as defined by the IMDC and Memorial Sloan-Kettering Cancer Center (MSKCC) scores, while clusters 4 (T-effector/proliferative), 5 (proliferative), and 6 (stromal/proliferative) were enriched among poor-risk patients.

Additionally, patients in clusters 1 and 2 demonstrated improved survival, while those in cluster 6 had poor PFS outcomes compared to other clusters, irrespective of treatment arm. Moreover, treatment with atezolizumab plus bevacizumab demonstrated improved overall response rates and longer PFS than sunitinib in clusters 4, 5, and 7 (small nucleolar RNA). Multivariable analysis of these identified molecular subtypes with clinical scores, including the MSKCC and IMDC scores, demonstrated an independent association with survival. These results suggested that these molecular subtypes may have the predictive capacity and could be incorporated with existing clinical tools to provide additional benefit, though additional prospective testing would be important prior to any clinical translation[75]. Of note, the phase II OPTIC trial is taking the first steps towards this goal, looking to utilize these molecular subgroups to prospectively assign patients to combination ICI treatment with ipilimumab plus nivolumab or ICI-TKI therapy with nivolumab and cabozantinib. Currently, in the recruitment phase, this trial will provide critical insights into the utility of molecular subtypes for treatment stratification in mRCC (NCT05361720).

Future perspectives

The above trials are a testament to the advancements in understanding the molecular underpinnings of mRCC and the role these insights can play in tailoring therapy in this patient population. Despite this progress, there are still several areas requiring further study. First, many unanswered questions remain regarding integrating these molecular subtypes with current clinical risk criteria. Further, these approaches are time-consuming and cost-prohibitive due to their reliance on multi-omic profiling. Therefore, studies on the cost-efficacy and accessibility of these tools will be critical before a transition into the clinical landscape. Additionally, molecular signature identification may benefit from targeted therapy, as there are an increasing number of TKI and PD-1/PD-L1 options available. Therefore, although these trials’ results are promising, further study is imperative to bring molecular subtypes into the clinical setting.

Clinical implication in real-world settings

Combination of lenvatinib plus everolimus for those with primary resistance

In a cohort of 7 patients that had shown resistance to VEGF-targeted TKI’s or ICI therapy, a combination of lenvatinib and everolimus as either second or third-line therapy resulted in a partial response in three patients and stable disease in three patients. Progression-free survival ranged from 3 to 15 months[76].

Combination therapy as second-line therapy active in distinct clinicopathological features

This combination retrospective study included 343 patients, with 123 receiving Cabozantinib and 220 receiving Nivolumab. Patients receiving Nivolumab and first treated with Pazopanib showed a non-statistically significant median overall survival of 26.8 vs. 11.6 months. The OS for patients with Cabozantinib was 25.7 months as compared to Sunitinib 21.7 months, but again, not statistically significant (P = 0.45). Notably, Cabozantinib exhibited activity in terms of progression-free survival, particularly in patients with Clear Cell histology (7.8 vs. 5.4, P = 0.026) and those with good risk features (12.3 vs. 5.7, P = 0.022)[77].

Another phase II trial, including a high dose of Cabozantinib with atezolizumab therapy (COSMIC 021), demonstrated an encouraging clinical response[78]. However, the CONTACT-03 study did not show any promising results in patients treated after disease progression during or after immune checkpoint inhibitor therapy (either combination or monotherapy) (NCT04338269).

APPROACH TO OVERCOME ICI RESISTANCE

Targeting the TME

Colony stimulating factor 1 receptor inhibitor

M2 macrophages promote tumor neoangiogenesis, and progression, which play a role in the treatment’s resistance. The expression of colony stimulating factor 1 receptor (CSF1R) allows switching the type 1 macrophages to the type II tumor associated-macrophages[79]. There are phase-1 trials currently undergoing to assess the effectiveness of combining treatment with CSF1R inhibitors and ICIs (NCT02718911, NCT02526017).

Indoleamine 2,3-dioxygenase 1 inhibitors

The indoleamine 2,3-dioxygenase 1 inhibitors deprives T cells of nutrients and can be the target for treatment. A study published in 2018 on patients with metastatic ccRCC treated with Nivolumab has shown that IDO-1 overexpression ( > 10%) was found in patients with an excellent response to the treatment and thus better PFS. This study suggested that IDO could be used as a biomarker for patients with RCC[80].

A phase I/II ECHO-202/KEYNOTE 037 trial combining oral IDO-1 enzyme inhibitors with pembrolizumab was associated with a 40% objective response (8 complete and 13 with stable disease)[81]. Unfortunately, in another phase III study, IDO-1 enzyme inhibitors failed to show a positive response in patients with melanoma, so their use was stopped after the study results[82].

Stimulators of interferon genes and retinoic acid-inducible gene 1 agonists

The stimulators of interferon genes (STING)pathway promotes the production of pro-inflammatory cytokines[83]. RIG-1 stimulates natural killer cells and CD8+ T cells[84]. There are a c ouple of trials assessing STING agonist and retinoic acid-inducible gene (RIG-1) agonist use as monotherapy or in combination with ICIs (NCT03010176 and NCT 03739138)

Targeting HIF-2α

Belzutifan is a first-in-class novel therapy to inhibit HIF-2α resulting in anti-tumor activity by impairing the hypoxic signal pathway in cancer cells. A phase 1 study with patients with pre-treated ccRCC showed a 25% response rate[85]. In a phase 2 study, patients who received prior treatments (immunotherapy or chemotherapy) were treated with Belzutifan plus Cabozantinib showed an objective response rate in 16 [30.8% (95%CI: 18.7-45.1)] of 52 patients[86].

Another inhibitor of HIF is PT2385, which was studied in combination with Nivolumab in patients with mRCC who previously had received up to three treatments. It showed an objective response rate of 22% with a PFS of 10 months among patients receiving therapeutic doses of PT2385 versus 4.7 months in the sub-therapeutic group[87].

Pegylated IL-2 and cytokines

IL-2 has shown anti-tumor potential by lysing tumor cells[88,89]. In a study of patients with mRCC, high-dose IL-2 combined with Pembrolizumab has shown an objective response rate (ORR) of 69%[90]. In a study in patients with previously untreated mRCC, Bempegaldesleukin (NKTR-214), combined with Nivolumab, showed an ORR of 54% in untreated mRCC[91].

Targeting patient’s intrinsic factor

Modulation of the gut microbiome

CBM58 is a bifidogenic live bacterium that can augment the effect of ICI by modulating the gut microbiome. The single-center randomized study (NCT03829111) in mRCC patients assessed nivolumab and ipilimumab with or without daily oral CBM588. The abundance of the bifidogenic bacterium was not seen. However, patients who received nivolumab-ipilimumab with CBM 588 had a significantly lower PFS (12.7 months vs. 2.5 months, HR 0.15, 95%CI: 0.05-0.47, P = 0.001) and a higher response rate (58% vs. 20%, P = 0.06) compared to those without CBM 588[92].

Recently Fecal microbiota transplantation (FMT) has been shown in several studies to augment the effect of ICI and overcome resistance, particularly in patients with melanoma[27,93,94]. Clinical trial in patients with RCC is still recruiting to assess the role of FMT in improving the efficacy of ICI (NCT04758507).

CONCLUSION

The outcomes of patients with metastatic RCC have changed significantly over the past few decades. The new therapeutic options, particularly with ICIs, have survival benefits and a durable response rate either used as a monotherapy or in combination with other therapies. However, patients may have resistance initially reflecting primary resistance or initial response to the treatment and then develop secondary resistance. Resistance to ICIs is influenced by three significant components: patient intrinsic factor, tumor cell-intrinsic factor, and contributions from the tumor microenvironment. Many innovative approaches have been studied and investigated in clinical trials to assess ICI resistance mechanisms in patients with mRCC.

DECLARATIONS

Acknowledgments

Graphical abstract: created with Biorender.com

Authors’ contributions

Conceptualization: Samnani S, Navani V

Writing - Original Draft Preparation: Samnani S, Sachedina F, Gupta M, Guo E

Figures and Tables: Samnani S, Sachedina F, Guo E

Writing - Review and Editing: Navani V, Samnani S, Sachedina F, Gupta M, Guo E

All authors have approved the final version of the manuscript.

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69:7-34.

2. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209-49.

3. Ferlay J, Colombet M, Soerjomataram I, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer 2018;103:356-87.

4. Capitanio U, Bensalah K, Bex A, et al. Epidemiology of renal cell carcinoma. Eur Urol 2019;75:74-84.

5. Dabestani S, Thorstenson A, Lindblad P, Harmenberg U, Ljungberg B, Lundstam S. Renal cell carcinoma recurrences and metastases in primary non-metastatic patients: a population-based study. World J Urol 2016;34:1081-6.

6. Rosenblatt J, McDermott DF. Immunotherapy for renal cell carcinoma. Hematol Oncol Clin North Am 2011;25:793-812.

7. Choueiri TK, Motzer RJ. Systemic therapy for metastatic renal-cell carcinoma. N Engl J Med 2017;376:354-66.

8. Clark JI, Wong MKK, Kaufman HL, et al. Impact of sequencing targeted therapies with high-dose interleukin-2 immunotherapy: an analysis of outcome and survival of patients with metastatic renal cell carcinoma from an on-going observational IL-2 clinical trial: PROCLAIMSM. Clin Genitourin Cancer 2017;15:31-41.e4.

9. Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol 2013;31:3791-9.

10. Motzer RJ, Powles T, Atkins MB, et al. Final overall survival and molecular analysis in immotion151, a phase 3 trial comparing atezolizumab plus bevacizumab vs. sunitinib in patients with previously untreated metastatic renal cell carcinoma. JAMA Oncol 2022;8:275-80.

11. Navani V, Heng DYC. Treatment selection in first-line metastatic renal cell carcinoma-the contemporary treatment paradigm in the age of combination therapy: a review. JAMA Oncol 2022;8:292-9.

12. Kluger HM, Tawbi HA, Ascierto ML, et al. Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce. J Immunother Cancer 2020;8:e000398.

13. Schwartz LH, Litière S, de Vries E, et al. RECIST 1.1-update and clarification: from the recist committee. Eur J Cancer 2016;62:132-7.

14. Navani V, Graves MC, Mandaliya H, et al. Melanoma: an immunotherapy journey from bench to bedside. Cancer Treat Res 2022;183:49-89.

15. Conforti F, Pala L, Bagnardi V, et al. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol 2018;19:737-46.

16. Polanczyk MJ, Hopke C, Vandenbark AA, Offner H. Estrogen-mediated immunomodulation involves reduced activation of effector T cells, potentiation of Treg cells, and enhanced expression of the PD-1 costimulatory pathway. J Neurosci Res 2006;84:370-8.

17. Polanczyk MJ, Hopke C, Vandenbark AA, Offner H. Treg suppressive activity involves estrogen-dependent expression of programmed death-1 (PD-1). Int Immunol 2007;19:337-43.

18. Motzer RJ, Tykodi SS, Escudier B, et al. Final analysis of the CheckMate 025 trial comparing nivolumab (NIVO) versus everolimus (EVE) with > 5 years of follow-up in patients with advanced renal cell carcinoma (aRCC). J Clin Oncol 2020;38:617-617.

19. Farag KI, Makkouk A, Norian LA. Re-evaluating the effects of obesity on cancer immunotherapy outcomes in renal cancer: what do we really know? Front Immunol 2021;12:668494.

20. Sanchez A, Furberg H, Kuo F, et al. Transcriptomic signatures related to the obesity paradox in patients with clear cell renal cell carcinoma: a cohort study. Lancet Oncol 2020;21:283-93.

21. Labadie BW, Liu P, Bao R, et al. BMI, irAE, and gene expression signatures associate with resistance to immune-checkpoint inhibition and outcomes in renal cell carcinoma. J Transl Med 2019;17:386.

22. Bergerot PG, Bergerot CD, Philip EJ, et al. Targeted therapy and immunotherapy: effect of body mass index on clinical outcomes in patients diagnosed with metastatic renal cell carcinoma. Kidney Cancer 2019;3(1):63-70.

23. De Giorgi U, Procopio G, Giannarelli D, et al. Association of systemic inflammation index and body mass index with survival in patients with renal cell cancer treated with nivolumab. Clin Cancer Res 2019;25:3839-46.

24. Boi SK, Orlandella RM, Gibson JT, et al. Obesity diminishes response to PD-1-based immunotherapies in renal cancer. J Immunother Cancer 2020;8:e000725.

25. Chowell D, Morris LGT, Grigg CM, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018;359:582-7.

26. Chowell D, Krishna C, Pierini F, et al. Evolutionary divergence of HLA class I genotype impacts efficacy of cancer immunotherapy. Nat Med 2019;25:1715-20.

27. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91-7.

28. Dizman N, Hsu J, Bergerot PG, et al. Randomized trial assessing impact of probiotic supplementation on gut microbiome and clinical outcome from targeted therapy in metastatic renal cell carcinoma. Cancer Med 2021;10:79-86.

29. Elkrief A, Derosa L, Kroemer G, Zitvogel L, Routy B. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: a new independent prognostic factor? Ann Oncol 2019;30:1572-9.

30. Lalani AA, Xie W, Braun DA, et al. Effect of antibiotic use on outcomes with systemic therapies in metastatic renal cell carcinoma. Eur Urol Oncol 2020;3:372-81.

31. Derosa L, Hellmann MD, Spaziano M, 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;29:1437-44.

32. Tinsley N, Zhou C, Tan G, et al. Cumulative antibiotic use significantly decreases efficacy of checkpoint inhibitors in patients with advanced cancer. Oncologist 2020;25:55-63.

33. Arbour KC, Mezquita L, Long N, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol 2018;36:2872-8.

34. Santini FC, Rizvi H, Plodkowski AJ, et al. Safety and efficacy of re-treating with immunotherapy after immune-related adverse events in patients with NSCLC. Cancer Immunol Res 2018;6:1093-9.

35. Horvat TZ, Adel NG, Dang TO, et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at memorial sloan kettering cancer center. J Clin Oncol 2015;33:3193-8.

36. Weber JS, Hodi FS, Wolchok JD, et al. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J Clin Oncol 2017;35:785-92.

37. Watson AS, Goutam S, Stukalin I, et al. Association of immune-related adverse events, hospitalization, and therapy resumption with survival among patients with metastatic melanoma receiving single-agent or combination immunotherapy. JAMA Netw Open 2022;5:e2245596.

38. Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20:25-39.

39. Restifo NP, Esquivel F, Kawakami Y, et al. Identification of human cancers deficient in antigen processing. J Exp Med 1993;177:265-72.

40. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol 2005;5:375-86.

41. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819-29.

42. Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res 2010;70:5213-9.

43. Ricketts CJ, De Cubas AA, Fan H, et al. Cancer genome atlas research network. The cancer genome atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep 2018;23:313-326.e5.

44. Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov 2016;6:202-16.

45. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015;523:231-5.

46. Sweis RF, Spranger S, Bao R, et al. Molecular drivers of the non-T-cell-inflamed tumor microenvironment in urothelial bladder cancer. Cancer Immunol Res 2016;4:563-8.

47. Seiwert TY, Zuo Z, Keck MK, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res 2015;21:632-41.

48. Jiménez-Sánchez A, Memon D, Pourpe S, et al. Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian cancer patient. Cell 2017;170:927-938.e20.

49. Sarvaria A, Madrigal JA, Saudemont A. B cell regulation in cancer and anti-tumor immunity. Cell Mol Immunol 2017;14:662-74.

50. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity 2015;42:607-12.

51. Mier JW. The tumor microenvironment in renal cell cancer. Curr Opin Oncol 2019;31:194-9.

52. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014;6:1670-90.

53. Komohara Y, Hasita H, Ohnishi K, et al. Macrophage infiltration and its prognostic relevance in clear cell renal cell carcinoma. Cancer Sci 2011;102:1424-31.

54. Santoni M, Massari F, Amantini C, et al. Emerging role of tumor-associated macrophages as therapeutic targets in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother 2013;62:1757-68.

55. Voss MH, Buros Novik J, Hellmann MD, et al. Correlation of degree of tumor immune infiltration and insertion-and-deletion (indel) burden with outcome on programmed death 1 (PD1) therapy in advanced renal cell cancer (RCC). J Clin Oncol 2018;36:4518-4518.

56. Vano Y, Rioux-leclercq N, Dalban C, et al. NIVOREN GETUG-AFU 26 translational study: association of PD-1, AXL, and PBRM-1 with outcomes in patients (pts) with metastatic clear cell renal cell carcinoma (mccRCC) treated with nivolumab (N). J Clin Oncol 2020;38:618-618.

57. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012;12:298-306.

58. Becht E, Giraldo NA, Lacroix L, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol 2016;17:218.

59. Choueiri TK, Escudier B, Powles T, et al. METEOR investigators. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol 2016;17:917-27.

60. Zhang S, Zhang E, Long J, et al. Immune infiltration in renal cell carcinoma. Cancer Sci 2019;110:1564-72.

61. Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020;577:549-55.

62. Finkin S, Yuan D, Stein I, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol 2015;16:1235-44.

63. Stubbs M, McSheehy PM, Griffiths JR, Bashford CL. Causes and consequences of tumour acidity and implications for treatment. Mol Med Today 2000;6:15-9.

64. Sormendi S, Wielockx B. Hypoxia pathway proteins as central mediators of metabolism in the tumor cells and their microenvironment. Front Immunol 2018;9:40.

65. Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol 2018;15:310-24.

66. Garcia-Lora A, Algarra I, Garrido F. MHC class I antigens, immune surveillance, and tumor immune escape. J Cell Physiol 2003;195:346-55.

67. Yu W, Guo Y. Prognostic significance of programmed death ligand-1 immunohistochemical expression in esophageal cancer: A meta-analysis of the literature. Medicine 2018;97:e11614.

68. Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. FEBS J 2019;286:4160-75.

69. Romero-Garcia S, Moreno-Altamirano MM, Prado-Garcia H, Sánchez-García FJ. Lactate contribution to the tumor microenvironment: mechanisms, effects on immune cells and therapeutic relevance. Front Immunol 2016;7:52.

70. Beuselinck B, Job S, Becht E, 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.

71. Epaillard N, Simonaggio A, Elaidi R, et al. BIONIKK: A phase 2 biomarker driven trial with nivolumab and ipilimumab or VEGFR tyrosine kinase inhibitor (TKI) in naïve metastatic kidney cancer. Bull Cancer 2020;107:eS22-7.

72. Meylan M, Sun C, Elaidi R, et al. 1451MO In-situ immune markers predict nivolumab (N)+/-ipilimumab (I) efficacy in frontline metastatic clear cell renal cell carcinoma (mccRCC): Key ancillary analyses from the BIONIKK randomized trial. Annals of Oncolog 2022;33:S1207.

73. McDermott DF, Huseni MA, Atkins MB, 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.

74. Rini BI, Powles T, Atkins MB, et al. IMmotion151 study group. Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet 2019;393:2404-15.

75. Motzer RJ, Banchereau R, Hamidi H, et al. Molecular subsets in renal cancer determine outcome to checkpoint and angiogenesis blockade. Cancer Cell 2020;38:803-817.e4.

76. Hamieh L, Beck RL, Le VH, Hsieh JJ. The Efficacy of lenvatinib plus everolimus in patients with metastatic renal cell carcinoma exhibiting primary resistance to front-line targeted therapy or immunotherapy. Clin Genitourin Cancer 2020;18:252-257.e2.

77. Santoni M, Aurilio G, Massari F, et al. Nivolumab VERSUS cabozantinib as second-line therapy in patients with advanced renal cell carcinoma: a real-world comparison. Clin Genitourin Cancer 2022;20:285-95.

78. Pal SK, McGregor B, Suárez C, et al. Cabozantinib in combination with atezolizumab for advanced renal cell carcinoma: results from the COSMIC-021 study. J Clin Oncol 2021;39:3725-36.

79. Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Rüttinger D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer 2017;5:53.

80. Seeber A, Klinglmair G, Fritz J, et al. High IDO-1 expression in tumor endothelial cells is associated with response to immunotherapy in metastatic renal cell carcinoma. Cancer Sci 2018;109:1583-91.

81. Mitchell TC, Hamid O, Smith DC, et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase i results from a multicenter, open-label phase i/ii trial (ECHO-202/KEYNOTE-037). J Clin Oncol 2018;36:3223-30.

82. Long GV, Dummer R, Hamid O, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol 2019;20:1083-97.

83. Corrales L, Glickman LH, McWhirter SM, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep 2015;11:1018-30.

84. Poeck H, Besch R, Maihoefer C, et al. 5'-Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma. Nat Med 2008;14:1256-63.

85. Choueiri TK, Bauer TM, Papadopoulos KP, et al. Inhibition of hypoxia-inducible factor-2α in renal cell carcinoma with belzutifan: a phase 1 trial and biomarker analysis. Nat Med 2021;27:802-5.

86. Choueiri TK, McDermott DF, Merchan J, et al. Belzutifan plus cabozantinib for patients with advanced clear cell renal cell carcinoma previously treated with immunotherapy: an open-label, single-arm, phase 2 study. Lancet Oncol 2023;24:553-62.

87. Rini BI, Appleman LJ, Figlin RA, et al. Results from a phase I expansion cohort of the first-in-class oral HIF-2α inhibitor PT2385 in combination with nivolumab in patients with previously treated advanced RCC. J Clin Oncol 2019;37:558-558.

88. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med 1982;155:1823-41.

89. Gillis S. Interleukin 2: biology and biochemistry. J Clin Immunol 1983;3:1-13.

90. Chatzkel JA, Swank J, Ludlow S, et al. Overall responses with coordinated pembrolizumab and high dose IL-2 (5-in-a-row schedule) for therapy of metastatic clear cell renal cancer: A single center, single arm trial. J Clin Oncol 2019;37:7_suppl, 657-657.

91. Diab A, Hurwitz ME, Cho DC, 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;36:3006-3006.

92. Dizman N, Meza L, Bergerot P, et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat Med 2022;28:704-12.

93. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018;359:97-103.

94. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018;359:104-8.

95. Cortellini A, Bozzetti F, Palumbo P, et al. Weighing the role of skeletal muscle mass and muscle density in cancer patients receiving PD-1/PD-L1 checkpoint inhibitors: a multicenter real-life study. Sci Rep 2020;10:1456.

96. Jerby-Arnon L, Shah P, Cuoco MS, et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 2018;175:984-997.e24.

97. Deng J, Wang ES, Jenkins RW, et al. CDK4/6 Inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov 2018;8:216-33.

98. Sade-Feldman M, Jiao YJ, Chen JH, et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun 2017;8:1136.

99. Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist 2019;2:141-60.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Samnani S, Sachedina F, Gupta M, Guo E, Navani V. Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors. Cancer Drug Resist 2023;6:416-29. http://dx.doi.org/10.20517/cdr.2023.02

AMA Style

Samnani S, Sachedina F, Gupta M, Guo E, Navani V. Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors. Cancer Drug Resistance. 2023; 6(2): 416-29. http://dx.doi.org/10.20517/cdr.2023.02

Chicago/Turabian Style

Samnani, Sunil, Faraz Sachedina, Mehul Gupta, Edward Guo, Vishal Navani. 2023. "Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors" Cancer Drug Resistance. 6, no.2: 416-29. http://dx.doi.org/10.20517/cdr.2023.02

ACS Style

Samnani, S.; Sachedina F.; Gupta M.; Guo E.; Navani V. Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors. Cancer Drug Resist. 2023, 6, 416-29. http://dx.doi.org/10.20517/cdr.2023.02

About This Article

Special Issue

This article belongs to the Special Issue Resistance to Systemic Therapies in Renal Cell Carcinoma
© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
569
Downloads
85
Citations
2
Comments
0
5

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 3 clicks
Like This Article 5 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/