Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1021-335X Online ISSN: 1791-2431
Journal Cover
December-2024 Volume 52 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2024 Volume 52 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review

Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review)

  • Authors:
    • Raquel González‑Garza
    • Adrián Gutiérrez‑González
    • Mario César Salinas‑Carmona
    • Manuel Mejía‑Torres
  • View Affiliations / Copyright

    Affiliations: Immunology Service, University Hospital ‘José Eleuterio González’, Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, México, Urology Service, University Hospital ‘José Eleuterio González’, Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, México
  • Article Number: 164
    |
    Published online on: October 14, 2024
       https://doi.org/10.3892/or.2024.8823
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Renal cell carcinoma (RCC) is a highly aggressive neoplastic disease of the renal parenchyma that is characterized by an intrinsic resistance to cytotoxic chemotherapy; for this reason, curative treatment is only achieved through surgical intervention in its early stages. The successful treatment of advanced or metastatic RCC will require the combined use of novel targeted therapies such as tyrosine kinase inhibitors, vascular endothelial growth factor blockers and immune checkpoint blockade therapies. Unfortunately, not all patients are candidates for such treatments, and at present, it is not possible to predict a patient's therapeutic response or likelihood to develop treatment‑associated complications. The present review described the literature focusing on the use of biomarkers for predicting patients' responses to therapies that induce immune checkpoint blockade in RCC.

Introduction

Renal cell carcinoma (RCC) is a group of neoplastic diseases that affect the renal parenchyma and represent 2–3% of all cancer diagnoses (1). In 2022, the GLOBOCAN database estimated the global incidence of RCC as 434,419 cases, which were associated with 155,702 deaths (2). RCC is more commonly diagnosed in men (ratio men to women, 2:1), and in the sixth decade of life (3). Up to 70% of patients are incidentally diagnosed with RCC, 50% of them with metastatic disease (4); although localized RCC is curable, in up to 30% of patients this will progress to metastatic disease, which has a median survival time of 6–10 months (5). Among the different types of RCCs, 80% of all diagnoses consist of the clear cell histological variety (ccRCC), which is followed in frequency by the papillary variant (10–15% of cases), the chromophobe cell variant (4–5% of cases), and other molecularly defined phenotypes (<1% of cases) (6).

The high rates of mortality that are observed in RCC have been associated with factors such as the high prevalence of advanced disease at diagnosis (7) and the intrinsic chemoresistance of RCC tumors, which is mostly related to apoptotic resistance, the upregulation of xenobiotic excretion systems (8–10) and the metabolic dependence of such tumors on the Warburg effect (11). In total, <10% of patients with RCC will respond to cytotoxic chemotherapy (12), which makes it necessary to use novel approaches for which little clinical information is available. Current RCC treatments can be grouped into the following categories: cytotoxic chemotherapy, surgery, local therapy, recombinant-cytokine therapy, targeted anti-angiogenic therapy, immunotherapy [immune checkpoint blockade (ICB)], and other therapies that include the use of inhibitors of the mechanistic target of rapamycin pathway (Table I). Despite the broad diversity and availability of RCC treatments, the need for a prognostic tool for use in selecting the treatment of choice and evaluating the clinical responses of patients remains a matter of controversy (13). Treatment selection depends on factors such as the histological variety, cellular grade and clinical stage of the disease, as well as the failure of previous treatments (14–16). In this regard, there are currently several scales that have been validated for risk classification and clinical decision-making in patients with RCC; among these, the Heng score (International Metastatic RCC Database Consortium or IMDC), the MSKCC scale (Memorial Sloan-Kettering Cancer Center), and the general physical status according to ECOG-PS criteria (the Eastern Cooperative Oncology Group performance score) (17–19) stand out (Table II).

Table I.

Available treatments for renal cell carcinoma.

Table I.

Available treatments for renal cell carcinoma.

CategoryDescription
Cytotoxic chemotherapyCurrently not considered given the high intrinsic resistance
SurgeryNephrectomy (simple, radical, cytoreductive, metastasectomy)
Local TherapySBRT, EBRT, arterial embolization, cryotherapy, thermal ablation
Cytokine TherapyInterferon-α, Interleukin-2
Antiangiogenic targeted therapiesAnti-VEGF antibodies, multikinase inhibitors
ImmunotherapyImmune checkpoint inhibitors
OthermTOR inhibitors

[i] SBRT, stereotactic body radiation therapy; EBRT, external-beam radiation therapy; VEGF, vascular endothelial growth factor; mTOR, mechanistic target of rapamycin.

Table II.

Renal cell carcinoma guide for treatment according to the American Joint Committee on Cancer Stages.

Table II.

Renal cell carcinoma guide for treatment according to the American Joint Committee on Cancer Stages.

AJCC stageTNM classificationaFirst-line therapybSecond-line therapybThird- and fourth-line therapiesb
IT1, N0, M0 Nephrectomyc
Local Therapyd
IIT2, N0, M0Nephrectomy +/-adjuvant ITe
Palliative Local Therapy
IIIT1, N1, M0Nephrectomy +/-adjuvant
T2, N1, M0IT or TTf
T3, N0, M0Palliative Therapy
T3, N1, M0
IVT4, Any N, M0Radical or CytoreductiveAxitinibTivozanib
Any T, Any N, M1 Nephrectomyj
Ipilimumab + NivolumabkSorafenib
Pembrolizumab + Axitinib orPalliative care
Lenvatinib(external-beam
Avelumab + Axitinibradiation therapy)
Nivolumab + Cabozantinib Nivolumabi
Bevacizumab +/-Interferon-αLenvatinib +
Cabozantinib or Sunitinib or Everolimusi
Pazopanib or Sorafenib Cabozantinibi
Temsirolimusg Everolimusi
Interferon-α or Interleukin-2h
Palliative care

a T1, tumor <7 cm; T2, tumor >7 cm, limited to the kidney; T3, tumor extension to perinephric tissues, but not Gerota's fascia; T4, tumor invasion beyond Gerota's fascia; N0, no regional lymph node metastasis; N1, metastasis in regional lymph nodes; M0, no distant metastasis; M1, distant metastasis.

b There is always the possibility for the patient to be enrolled in an appropriate clinical trial. Most clinical trials available are for AJCC Stage IV and second- or subsequent line therapies.

c Includes partial, simple and radical nephrectomies.

d Includes stereotactic body radiation therapy (SBRT), cryotherapy/thermal ablation and palliative external beam radiation therapy (EBRT).

e IT or immune checkpoint inhibitors, including the following antibodies anti-PD-1 (nivolumab, pembrolizumab), anti-CTLA-4 (ipilimumab), and anti-PD-L1 (avelumab, atezolizumab).

f Anti-angiogenic TT, including anti-VEGF antibody (bevacizumab), and multitarget TKIs.

g Mechanistic target of rapamycin inhibitors.

h Cytokine therapy, including IFN-α and IL-2.

i Second-line therapies after no response to first-line TKIs' monotherapy.

j Metastasectomy or cytoreductive therapy in patients with low-risk according to MSKCC/ECOG.

k In patients with intermediate to poor prognostic profile. IT, immunotherapy; TT, targeted-therapy; TKI, tyrosine kinase inhibitors.

In the last decade, the use of monoclonal antibodies for reversing the ‘exhaustion’ state in tumor infiltrating lymphocytes (TILs) has proven to be a valuable treatment for numerous solid tumors and hematologic cancers. In 2015, the ChekMate-025 study demonstrated the efficacy of nivolumab (which was initially approved for treating melanoma and non-small cell lung cancer) in treating patients with RCC (20). In addition to nivolumab [which blocks the programmed cell death protein 1 (PD-1) receptor], four other monoclonal antibodies are currently approved for use in RCC, all of which reverse the inhibition of lymphocyte immune effectors: Pembrolizumab, which is also a PD-1 blocker; ipilimumab, which blocks the cytotoxic T-lymphocyte associated protein 4; and atezolizumab/avelumab, which blocks PD-1 ligand in myeloid and tumor cells and prevents PD-1 inhibitory signals in lymphocytes (Table III) (20–30).

Table III.

Clinical studies availing the use of immune checkpoint blockade for treatment in RCC.

Table III.

Clinical studies availing the use of immune checkpoint blockade for treatment in RCC.

First author, yearStudyPhaseDesignInterventionResults(Refs.)
Motzer et al, 2015CheckMate 025IIIRandomized, open-label, N= 821, mRCCNivolumab vs. EverolimusORR: 25 vs.5%(20)
Motzer et al, 2018CheckMate 214IIIRandomized, open-label, N=1096, mRCC Nivolumab/Ipilimumab vs. SunitinibORR: 42% vs. 27%(21)
Rini et al, 2019Keynote-426IIIOpen label, randomized, N= 861, mRCC Pembrolizumab/Axitinib vs. SunitinibORR: 59.3 vs. 35.7%(24)
Vogelzang et al, 2020CheckMate 374aIVMulticenter, open-label, N=44, mRCCNivolumabORR: 22.7%(26)
Choueiri et al, 2021CheckMate 9ERIIIRandomized, open label, N=651, mRCC Nivolumab/Cabozantinib vs. SunitinibORR: 55.7 vs. 27.1%(29)
Rini et al, 2019IMmotion151IIIMulticenter, open-label, N=915, mRCC Atezolizumab/Bevacizumab vs. SunitinibORR: 37 vs. 33%(25)
Motzer et al, 2021CLEARIIIRandomized, N=1069, mRCC Lenvatinib/Pembrolizumab vs. Lenvatinib/Everolimus vs. SunitinibORR: 71 vs. 53 vs. 36%(22)
Choueiri et al, 2021Keynote-564IIIDouble-blind, N=994, locRCCAdjuvant Pembrolizumab vs. PlaceboDFS 24-month: 77.3 vs. 68.1%(30)
Tykodi et al, 2022CheckMate 920aIVMulticohort, N=52, mRCC Nivolumab/IpilimumabORR: 19.6%(27)
Pal et al, 2023Contact-03IIIMulticenter, randomized, open-label, N=522, locRCC/mRCC Atezolizumab/Cabozantinib vs. CabozantinibPFS: 10.6 vs. 10.8 months(28)
Motzer et al, 2019JAVELIN Renal 101IIIMulticenter, randomized, open-label, N=886, mRCCAvelumab/Axitinib vs. SunitinibORR: 55.2 vs. 25.5%(23)

{ label (or @symbol) needed for fn[@id='tfn13-or-52-6-08823'] } RCC, renal cell carcinoma; mRCC, metastatic RCC; locRCC, localized RCC; PFS, progression-free survival; DFS, disease-free survival; ORR, objective response rate.

a Studies evaluating non-clear cell RCC.

In the following section, the current knowledge of the cellular mechanisms that lead to lymphocyte exhaustion within the highly dynamic tumor-immune microenvironment (TIME) was reviewed. It was concluded with a short description of experimental biomarkers that have been used to predict the patients' responses to ICB therapy.

The TIME

The TIME comprises a network of interacting elements within the tumor tissue that can be categorized as follows: Cells (tumor, stroma and infiltrating immune cells), small soluble elements (proteins, cytokines, growth factors, metabolites and chemokines), and the extracellular matrix (31). The growth of tumors requires two conditions: Cell transformation, whether genetic or acquired (32) and immune dysfunction (33). This second requirement explains the association between immunodeficiency status and cancer development (34). The clearance of transformed cells from tissues requires the activation of cytotoxic immunity that is achieved through the infiltration of CD8+ cytotoxic lymphocytes and natural killer cells into a tumor (35). Antitumor immunity is so efficient that even though transformation and cell damage occur over the course of every life, cancer develops only in a small proportion of patients; the occurrence of cancer is promoted by immune dysfunction.

Tumor cells proliferate at higher rates compared with normal cells. This accelerated proliferation, which is linked to metabolic changes that take place within the tumor bed, results in dysfunctional local immunity and further selection of the best-adapted tumor cells (36–38). Cytotoxic chemotherapy exploits this increased proliferative rate, and cytotoxic treatments are widely used in most neoplastic diseases other than RCC. The peculiarities of RCC include its intrinsic aggressive nature, its increased infiltration with lipids, its high rate of metastasis, and its resistance to cytotoxic chemotherapy (39). This chemo-resistance has numerous causal factors, both intrinsic (or genetic) and acquired. It is important to note that the epithelial renal cells normally have secretion systems for xenobiotics and intrinsic antioxidant mechanisms protecting the cells from toxic damage (40). Given that most RCC subtypes arise from renal epithelia, it is not surprising that such mechanisms are enhanced in renal carcinoma. In addition, renal tumor cells have increased expression of anti-apoptotic proteins Bcl-2, Bcl-XL, ARC (apoptosis repressor with a caspase recruitment domain) and XIAP (X-linked inhibitor of apoptosis) while pro-apoptotic proteins such as Bim are decreased. Anti-apoptotic and pro-apoptotic proteins are regulated by NF-κB and von Hippel-Lindau (VHL) pathways, respectively (8,10,41).

A total of up to 2/3 of patients with RCC have mutations in VHL gene (42). Among its numerous functions, VHL targets proteins for proteasomal degradation, including the pro-apoptotic protein Bim, whose levels are increased in patients with RCC (41). Conversely, the increase in anti-apoptotic pathways has also been implicated in chemoresistance, specifically in the increased expression of anti-apoptotic proteins Bcl-2, ARC and XIAP (9). Concomitant to these mechanisms, renal cancer cells are highly dependent on aerobic glycolysis (Warburg effect), a phenotype associated to a lack of response after ICB therapy (43), and a recent putative target for enhancing combined treatments for RCC (11).

In addition, although the presence of TILs is a favorable prognostic factor for most cancers, such infiltrative cells are associated with poor outcomes in RCC (44,45). Consequently, a hostile TIME develops that leads to the selection of tumors that are resistant to hypoxia, acidosis and the low availability of nutrients. In addition, a hostile TIME negatively affects lymphocyte-dependent antitumor immunity (Fig. 1 and Table IV) (31,46–51). Previous studies suggested that dysbiosis is an important element to be considered when evaluating either the responses of tumors to ICB or the general prognosis of patients with neoplastic diseases (52,53). The molecular basis for lymphocyte dysfunction involves several mechanisms that are associated with peripheral tolerance, namely anergy, suppression and exhaustion (54). Immune cell exhaustion is reversed by ICB, which blocks interactions between the inhibitory receptors of lymphocytes and their ligands (55). The PD-1/programmed death-ligand 1 (PD-L1) axis is the most studied of these interactions within TILs (56,57).

Figure 1.

Tumor immune microenvironment in RCC. (1) Risk and causal factors converge into tumor formation in renal cell epithelia. (2) In early disease, immunity drives the control of tumor growth through several mechanisms, namely cell cytotoxicity, cytokine-mediated inflammation and ligand-induced apoptosis. This stage of antitumor immunity is mediated mostly by Tc, NK and NKT. (3) As cancer turns into a chronic disease, the antitumor immunity is negatively regulated by tumor cells (direct inhibition) or by infiltrated leukocytes and metabolic factors produced by tumor cells within the tumor microenvironment (indirect regulation). Direct inhibition is mediated by the tumor expression of PD-L1 and B7 ligands. Indirect regulation is more complex and involves the synthesis and secretion of IL-10 by Treg and MDSCs, the expression of the ligands PD-L1 and B7 by TAMs, and the hostile tumor microenvironment characterized by the low availability of nutrients, tissue acidosis and hypoxia. Inset: Antitumor immunity is antigen-specific, requiring lymphocyte activation through TCR ligation + co-stimulation. Co-stimulatory signals could be either activating or inhibitory, favoring the latter in advanced disease. Excessive negative co-stimulation leads to lymphocyte exhaustion, a functional negative state therapeutically reverted by immunotherapy (also known as immune checkpoint blockade). In RCC, two pathways are successfully reverted by immunotherapy: The pathway PD-1/PD-L1 (blocked by pembrolizumab and atezolizumab, respectively) and the pathway CTLA-4/B7 (blocked by ipilimumab). Other proteins involved in lymphocyte exhaustion but currently lacking approved monoclonal antibodies for treating RCC disease are the receptors LAG-3 and TIM-3 on the surface of lymphocytes. Green arrows, perforin release; red arrows, upregulation of lymphocyte's exhaustion ligands on myeloid cells and stromal cells; block red arrows, direct lymphocyte inhibition mediated by the B7 family of ligands. Figure was constructed using BioRender (www.Biorender.com). RCC, renal cell carcinoma; Tc, cytotoxic T lymphocytes; NK, natural killer cells; NKT, CD3+ natural killer cells; PD-L1, programmed death-ligand 1 (also known as B7-H1); Treg, T regulatory lymphocytes; MDSCs, myeloid-derived suppressor cells; TAM, tumor-associated macrophages; TCR, T cell receptor; PD-1, programmed cell death protein 1; CTLA-4, T-lymphocyte associated protein 4; LAG-3, lymphocyte-activation gene 3; T cell immunoglobulin and mucin-domain containing-3; CAF, cancer-associated fibroblasts; MHC I, MHC-I, major histocompatibility complex class I.

Table IV.

Tumor-immune microenvironment elements driving lymphocyte exhaustion in RCC.

Table IV.

Tumor-immune microenvironment elements driving lymphocyte exhaustion in RCC.

First author, yearElementAssociated dysfunctionClinical implications(Refs.)
Kawashima et al, 2020LymphocytesCytotoxic T and NK lymphocytes are dysfunctional despite their increased infiltrationIn RCC, lymphocyte infiltrates correlate with poor overall prognosis and decreased response to ICB(46)
Wang et al, 2021MacrophagesTumor-supporting macrophages (M2 cells or TAM) enhances the tumor expression of ‘exhausting’ ligands for lymphocytesInfiltration with M2-like TAM is associated with poor prognosis and resistance to therapy(47)
Sabrina et al, 2023Suppressor CellsMDSC and Treg cells antagonize the antitumor cytotoxic immunity. Suppressor cells increase the production of anti-inflammatory cytokinesIncreased numbers of MDSC and/or Treg lymphocytes correlate with RCC progression and resistance to immunotherapy(48)
Liu et al, 2021FibroblastsCAF cells support the ECM remodeling, promoting in turn the tumor growth and metastasisAbundance of CAF is associated with poor prognosis and resistance to cancer therapy(49)
Zhang et al, 2021HypoxiaHypoxic microenvironment promotes tumor progression, angiogenesis, and immune suppression. Drives selection of cancer resistant cellsHypoxia selects tumor clones resistant to chemotherapy leading to poor prognosis(50)
Ballesteros et al, 2021CytokinesIL-10 and TGF-β lead to suppression of cytotoxic immunityImmunosuppressive cytokines contribute to immune evasion and resistance to treatment(51)
Monjarás-Avila et al, 2023Low nutrient supplyActivated cytotoxic cells are highly dependent on glucose supply. Selects resistant cancer cellsAnaerobic metabolism supports the viability of tumor cells, a condition detrimental to cytotoxic lymphocytes(31)

[i] RCC, renal cell carcinoma; ICB, immune checkpoint blockade; TAM, tumor-associated macrophages; MDSC, myeloid-derived suppressor cells; Treg, regulatory T lymphocytes; CAF, cancer-associated fibroblasts; ECM, extracellular matrix; IL-10, interleukin-10; TGF-β, transforming growth factor beta.

Even though numerous patients achieve complete clinical response to ICB therapy, a significant proportion of patients show only a partial response or no response at all, which is occasionally accompanied by signs of systemic toxicity (58). This situation explains why the search for prognostic markers for ICB in patients with RCC is a highly active area of research. Most prognostic markers for RCC can be divided into those addressing disease progression and those addressing responses to treatments, in particular immunotherapy and directed therapies. Thus, predicting the ICB responsiveness will impact in selecting the patients who will benefit the most and have the lowest probability of developing adverse events after receiving immunotherapy. There are currently no standardized and validated methods for properly assessing the risk/benefit ratio of using ICB therapy. Therefore, the goal of the present review was to synthesize the reported findings of the studies that have focused on this important topic.

Predictors of clinical response to the use of immune checkpoint inhibitors

ICB therapy is associated with an average mortality rate of 1%, with death mostly caused by immune-related adverse events (58). After the reporting of successful results from the CheckMate-025 study, and given the low predictability and consistency of ICB responsiveness (59), interest has been increasing in the search for reliable markers of ICB responsiveness. Among the biomarkers that have been studied, three stand out for their consistency: the level of C-reactive protein (CRP), the number of TILs and the basal expression levels of exhaustion markers in tumor tissue. CRP is an acute phase reactant that is frequently used for evaluating systemic inflammation, making it a logical putative marker for ICB responsiveness given the close association between inflammation and cancer. Previous studies have indicated that low basal CRP levels or low normalized levels of CRP measured after patients received their first ICB doses were predictive of their ICB responsiveness (60–63). Studies investigating TILs found that a patient's prognosis is associated with the nature of his or her infiltrating cells; infiltration with inflammatory leukocytes was associated with the best responses to ICB therapy (64–66). In peripheral blood, a recent study observed that increased numbers of circulating eosinophils were associated with an improved response to ICB, which is an intriguing result given the regulatory role that eosinophils play in systemic inflammation (67). Interestingly, the basal expression level of PD-L1 has not been consistently predictive of ICB responsiveness (68–70), whereas the basal expression level of T cell immunoglobulin and mucin-domain containing-3 (TIM-3) appears to be (71).

Several additional markers have been studied for predicting the response to ICB in patients with RCC. For example, longer responses to ICB have been associated with decreased levels of circulating tumor DNA, increased levels of chemokine CXCL14, increased levels of circulating miR-22 and miR-24, and the presence of immunogenic transcriptional signatures (72–75). In this context, several markers have been associated with poor responses after ICB therapy (increased levels of interleukin-8) or predictive of the development of immune-related adverse events (decreased levels of miR-146a) (76,77). All the studies described in this section are summarized in Table V.

Table V.

Predictive biomarkers for clinical response to immune checkpoint blockade in patients with RCC.

Table V.

Predictive biomarkers for clinical response to immune checkpoint blockade in patients with RCC.

First author, yearBiomarkerPredictor → ResponseRCC typeTissueTechniqueStudy phaseTreatmentNo. of patients(Refs.)
Ishihara et al, 2020CRP↓CRP → increased OS/PFSmRCCSerumELISAINivolumab70(60)
Noguchi et al, 2020CRP↑CRP → poor responsemRCCBloodBio-PlexINivolumab64(61)
Yano et al, 2022CRP↑CRP → decreased OSmRCCSerumELISAIIIDual ICB74(62)
Koh et al, 2022ctDNA↓ctDNA → longer PFSmRCCPlasma, tumorDNA-seqIDual ICB14(72)
Pan et al, 2023CXCL14↑CXCL14 → longer OSmRCCKidney, tumorIHCIIINivolumab120(74)
Schalper et al, 2020IL-8↑IL-8 → poor outcomemRCCSerumELISAIIINivolumab392(76)
Incorvaia et al, 2020miRsa↑miRsa → long responseRCCBloodqPCRINivolumab23(73)
Ivanova et al, 2022miRsb↓miRsb → irAEs to ICBRCCBloodqPCRPilot/PoCNivolumab86(77)
Atkins et al, 2022PD-L1↓PD-L1 → poor responsemRCCTumorIHCIIDual ICB123(68)
Pabla et al, 2021GES↑TIGS → improved responsemRCCTumorRNA-seqIIINivolumab54(75)
Motzer et al, 2022PD-L1PD-L1>1% → ↑PFS, no association in OSmRCCTumor, bloodIHC/GESIIIDual ICB498(69)
Kim et al, 2022TIL↑Th, Tc, and M1 cells → longer PFSmRCCTumorIHCPilot/PoCDual ICB24(64)
Sammarco et al, 2024TIL↑163+ macrophages → poor responsemRCCTumorIHCPilot/PoCDual ICB/ICB+TKI28(65)
Herrmann et al, 2021Eosino-phils↑AEC → improved response to ICBmRCCBloodCBCRetros-pectiveNivolumab64(67)
Kato et al, 2020TIM-3↑TIM-3 → response to ICBmRCCTumorIHC/IFRetrospectiveDual ICB25(71)
Brown et al, 2022PD-L1, CTLA-4Failed to predict ICBmRCCTumorIHC/qPCRRetrospectiveDual ICB62(70)
Kazama et al, 2022TIL↑CD8/↑CD68 → response to ICBmRCCTumorIHCPilot/PoCNot specified60(66)
Tomita et al, 2022CRPCRP normalization predicts responsemRCCSerumELISAPilot/PoCAvelumab + TKI789(63)

[i] a, miR-22, −24, −99a, −194, −214, −335, −339, −798.

[ii] b, miR-146a. PoC, proof of concept; mRCC, metastatic/advanced RCC; Dual ICB, dual immune checkpoint blockade (nivolumab + ipilimumab); CRP, C-reactive protein; ctDNA, circulating tumor DNA; miRs, micro-RNA molecules; PD-L1, programmed cell death receptor-ligand 1; TIL, tumor infiltrating leukocytes; ELISA, enzyme-linked immunosorbent assay; DNA-seq, DNA sequencing; RNA-seq, RNA sequencing; IHC/IF, immunohistochemistry/immunofluorescence; qPCR, quantitative polymerase chain reaction; WB, western blotting; OS, overall survival; PFS, progression-free survival; GES, Gene Expression Signature; TIGS, Tumor Immunogenic Signature; irAEs, immune-related adverse events; ICB, immune checkpoint blockade; CBC, complete blood count; AEC, absolute eosinophils count; TKI, tyrosine kinase inhibitors.

From a technical perspective, the evaluation of leukocytes' exhaustion markers require the performance of histopathology after the direct sampling of tissue, which is not feasible for all diagnoses (78). In this context, the studies assessing the suitability of peripheral biomarkers for predicting ICB responsiveness are encouraging (79,80).

Future directions and conclusions

Several limitations currently prevent the formal use of markers to predict ICB responsiveness. First, the inconsistency of reported outcomes is mostly related to heterogeneity in the target population and the low number of patients that have been studied. In addition, a lack of standardization in the use of methods and reagents is complicating the replication of pioneering studies. Furthermore, the relationship between the expression of such potential biomarkers and the underlying mechanisms of resistance to ICB is not fully understood, making it difficult not only to predict ICB responsiveness but also to know the clinical safety of using ICB. Finally, the intrinsic resistance of RCCs to chemotherapy and the complex combinations required for treatment make it difficult to determine a logical approach for studying the expression of putative cancer biomarkers. In the future, by integrating multi-omics data and machine-learning approaches, it should be possible to create more accurate prediction models that will help in the identification of novel biomarkers for ICB responsiveness.

Acknowledgements

The authors are grateful to Dr. Susana Chávez (Department for Research Assistance, University Hospital ‘José Eleuterio González’, Autonomous University of Nuevo León) for reviewing the English language in this manuscript.

Funding

The present study was supported by CONAHCYT México (grant no. 783446).

Availability of data and materials

Not applicable.

Authors' contributions

RGG and MMT performed the literature review, wrote the manuscript, made the illustrations and constructed the tables. AGG, MCSC and MMT revised the manuscript. RGG and MMT were involved in the conception of the study. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PD-1

programmed cell death protein 1

PD-L1

programmed death-ligand 1

References

1 

Padala SA and Barsouk A, Thandra KC, Saginala K, Mohammed A, Vakiti A, Rawla P and Barsouk A: Epidemiology of renal cell carcinoma. World J Oncol. 11:79–87. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Bukavina L, Bensalah K, Bray F, Carlo M, Challacombe B, Karam JA, Kassouf W, Mitchell T, Montironi R and O'Brien T: Epidemiology of renal cell carcinoma: 2022 update. Eur Urol. 82:529–542. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Musaddaq B, Musaddaq T, Gupta A, Ilyas S and von Stempel C: Renal cell carcinoma: The evolving role of imaging in the 21st century. Semin Ultrasound CT MR. 41:344–350. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Scosyrev E, Messing EM, Sylvester R and Van Poppel H: Exploratory subgroup analyses of renal function and overall survival in european organization for research and treatment of cancer randomized trial of Nephron-sparing surgery versus radical nephrectomy. Eur Urol Focus. 3:599–605. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Goswami PR, Singh G, Patel T and Dave R: The WHO 2022 classification of renal neoplasms (5th Edition): Salient updates. Cureus. 16:e584702024.PubMed/NCBI

7 

Didwaniya N, Edmonds RJ, Fang X, Silberstein PT and Subbiah S: Survival outcomes in metastatic renal carcinoma based on histological subtypes: SEER database analysis. J Clin Oncol. 29:381. 2011. View Article : Google Scholar

8 

Morais C, Gobe G, Johnson DW and Healy H: Inhibition of nuclear factor kappa B transcription activity drives a synergistic effect of pyrrolidine dithiocarbamate and cisplatin for treatment of renal cell carcinoma. Apoptosis. 15:412–425. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Toth C, Funke S, Nitsche V, Liverts A, Zlachevska V, Gasis M, Wiek C, Hanenberg H, Mahotka C, Schirmacher P and Heikaus S: The role of apoptosis repressor with a CARD domain (ARC) in the therapeutic resistance of renal cell carcinoma (RCC): The crucial role of ARC in the inhibition of extrinsic and intrinsic apoptotic signalling. Cell Commun Signal. 15:162017. View Article : Google Scholar : PubMed/NCBI

10 

Yang WZ, Zhou H and Yan Y: XIAP underlies apoptosis resistance of renal cell carcinoma cells. Mol Med Rep. 17:125–130. 2018.PubMed/NCBI

11 

Wang L, Fu B, Hou DY, Lv YL, Yang G, Li C, Shen JC, Kong B, Zheng LB, Qiu Y, et al: PKM2 allosteric converter: A self-assembly peptide for suppressing renal cell carcinoma and sensitizing chemotherapy. Biomaterials. 296:1220602023. View Article : Google Scholar : PubMed/NCBI

12 

Diamond E, Molina AM, Carbonaro M, Akhtar NH, Giannakakou P, Tagawa ST and Nanus DM: Cytotoxic chemotherapy in the treatment of advanced renal cell carcinoma in the era of targeted therapy. Crit Rev Oncol Hematol. 96:518–526. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Saliby RM, Saad E, Kashima S, Schoenfeld DA and Braun DA: Update on biomarkers in renal cell carcinoma. Am Soc Clin Oncol Educ Book. 44:e4307342024. View Article : Google Scholar : PubMed/NCBI

14 

Lam JS, Pantuck AJ, Belldegrun AS and Figlin RA: Protein expression profiles in renal cell carcinoma: Staging, prognosis, and patient selection for clinical trials. Clin Cancer Res. 13((2 Pt 2)): 703s–708s. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Lee JN, Chun SY, Ha YS, Choi KH, Yoon GS, Kim HT, Kim TH, Yoo ES, Kim BW and Kwon TG: Target molecule expression profiles in metastatic renal cell carcinoma: Development of individual targeted therapy. Tissue Eng Regen Med. 13:416–427. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Patard JJ, Leray E, Rioux-Leclercq N, Cindolo L, Ficarra V, Zisman A, De La Taille A, Tostain J, Artibani W, Abbou CC, et al: Prognostic value of histologic subtypes in renal cell carcinoma: A multicenter experience. J Clin Oncol. 23:2763–2771. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Heng DY, Xie W, Regan MM, Harshman LC, Bjarnason GA, Vaishampayan UN, Mackenzie M, Wood L, Donskov F, Tan MH, et al: External validation and comparison with other models of the International Metastatic Renal-Cell Carcinoma Database Consortium prognostic model: A population-based study. Lancet Oncol. 14:141–148. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Kattan MW, Reuter V, Motzer RJ, Katz J and Russo P: A Postoperative prognostic nomogram for renal cell carcinoma. J Urol. 166:63–67. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET and Carbone PP: Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 5:649–655. 1982. View Article : Google Scholar : PubMed/NCBI

20 

Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al: Nivolumab versus everolimus in advanced Renal-cell carcinoma. N Engl J Med. 373:1803–1813. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al: Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 378:1277–1290. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Kopyltsov E, Méndez-Vidal MJ, et al: Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma. N Engl J Med. 384:1289–1300. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, et al: Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 380:1103–1115. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, et al: Pembrolizumab plus axitinib versus sunitinib for advanced Renal-cell carcinoma. N Engl J Med. 380:1116–1127. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, Bracarda S, Stadler WM, Donskov F, Lee JL, et al: Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): A multicentre, open-label, phase 3, randomised controlled trial. The Lancet. 393:2404–2415. 2019. View Article : Google Scholar

26 

Vogelzang NJ, Olsen MR, McFarlane JJ, Arrowsmith E, Bauer TM, Jain RK, Somer B, Lam ET, Kochenderfer MD, Molina A, et al: Safety and efficacy of nivolumab in patients with advanced Non-clear cell renal cell carcinoma: Results from the Phase IIIb/IV CheckMate 374 study. Clin Genitourin Cancer. 18:461–468.e3. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Tykodi SS, Gordan LN, Alter RS, Arrowsmith E, Harrison MR, Percent I, Singal R, Van Veldhuizen P, George DJ, Hutson T, et al: Safety and efficacy of nivolumab plus ipilimumab in patients with advanced non-clear cell renal cell carcinoma: Results from the phase 3b/4 CheckMate 920 trial. J Immunother Cancer. 10:e0038442022. View Article : Google Scholar : PubMed/NCBI

28 

Pal SK, Albiges L, Tomczak P, Suárez C, Voss MH, de Velasco G, Chahoud J, Mochalova A, Procopio G, Mahammedi H, et al: Atezolizumab plus cabozantinib versus cabozantinib monotherapy for patients with renal cell carcinoma after progression with previous immune checkpoint inhibitor treatment (CONTACT-03): A multicentre, randomised, Open-label, phase 3 trial. The Lancet. 402:185–195. 2023. View Article : Google Scholar

29 

Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, Oyervides Juárez VM, Hsieh JJ, Basso U, Shah AY, et al: Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 384:829–841. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Chang YH, Hajek J, Symeonides SN, Lee JL, Sarwar N, et al: Adjuvant pembrolizumab after nephrectomy in renal-cell carcinoma. N Engl J Med. 385:683–694. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Monjaras-Avila CU, Lorenzo-Leal AC, Luque-Badillo AC, D'Costa N, Chavez-Muñoz C and Bach H: The tumor immune microenvironment in clear cell renal cell carcinoma. Int J Mol Sci. 24:79462023. View Article : Google Scholar : PubMed/NCBI

32 

Karras P, Black JRM, McGranahan N and Marine JC: Decoding the interplay between genetic and Non-genetic drivers of metastasis. Nature. 629:543–554. 2024. View Article : Google Scholar : PubMed/NCBI

33 

Ucche S and Hayakawa Y: Immunological aspects of cancer cell metabolism. Int J Mol Sci. 25:52882024. View Article : Google Scholar : PubMed/NCBI

34 

Bucciol G, Delafontaine S, Meyts I and Poli C: Inborn errors of immunity: A field without frontiers. Immunol Rev. 322:15–27. 2024. View Article : Google Scholar : PubMed/NCBI

35 

Kramer G, Blair T, Bambina S, Kaur AP, Alice A, Baird J, Friedman D, Dowdell AK, Tomura M, Grassberger C, et al: Fluorescence tracking demonstrates T cell recirculation is transiently impaired by radiation therapy to the tumor. Sci Rep. 14:119092024. View Article : Google Scholar : PubMed/NCBI

36 

Tan D, Miao D, Zhao C, Shi J, Lv Q, Xiong Z, Yang H and Zhang X: Comprehensive analyses of A 12-metabolism-associated gene signature and its connection with tumor metastases in clear cell renal cell carcinoma. BMC Cancer. 23:2642023. View Article : Google Scholar : PubMed/NCBI

37 

Wu Y and Li X: Senescence gene expression in clear cell renal cell carcinoma: Role of tumor immune microenvironment and senescence-associated survival prediction. Medicine (Baltimore). 102:e352222023. View Article : Google Scholar : PubMed/NCBI

38 

Zhang Q, Lin B, Chen H, Ye Y, Huang Y, Chen Z and Li J: Lipid Metabolism-related gene expression in the immune microenvironment predicts prognostic outcomes in renal cell carcinoma. Front Immunol. 14:13242052023. View Article : Google Scholar : PubMed/NCBI

39 

Bahadoram S, Davoodi M, Hassanzadeh S, Bahadoram M, Barahman M and Mafakher L: Renal cell carcinoma: An overview of the epidemiology, diagnosis, and treatment. G Ital Nefrol. 39:2022–vol3. 2022.PubMed/NCBI

40 

Mickisch G, Bier H, Bergler W, Bak M, Tschada R and Alken P: P-170 glycoprotein, glutathione and associated enzymes in relation to chemoresistance of primary human renal cell carcinomas. Urol Int. 45:170–176. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Guo Y, Schoell MC and Freeman RS: The von Hippel-lindau protein sensitizes renal carcinoma cells to apoptotic stimuli through stabilization of BIMEL. Oncogene. 28:18642009. View Article : Google Scholar : PubMed/NCBI

42 

Büscheck F, Fraune C, Simon R, Kluth M, Hube-Magg C, Möller-Koop C, Sarper I, Ketterer K, Henke T, Eichelberg C, et al: Prevalence and clinical significance of VHL mutations and 3p25 deletions in renal tumor subtypes. Oncotarget. 11:237–249. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ, Xu H, Pritchard TS, Fan J, Cheadle C, et al: The intratumoral balance between metabolic and immunologic gene expression is associated with Anti-PD-1 response in patients with renal cell carcinoma. Cancer Immunol Res. 4:726–733. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Rooney MS, Shukla SA, Wu CJ, Getz G and Hacohen N: Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 160:48–61. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Möller K, Fraune C, Blessin NC, Lennartz M, Kluth M, Hube-Magg C, Lindhorst L, Dahlem R, Fisch M, Eichenauer T, et al: Tumor cell PD-L1 expression is a strong predictor of unfavorable prognosis in immune checkpoint Therapy-naive clear cell renal cell cancer. Int Urol Nephrol. 53:2493–2503. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Kawashima A, Kanazawa T, Kidani Y, Yoshida T, Hirata M, Nishida K, Nojima S, Yamamoto Y, Kato T, Hatano K, et al: Tumour grade significantly correlates with total dysfunction of tumour Tissue-infiltrating lymphocytes in renal cell carcinoma. Sci Rep. 10:62202020. View Article : Google Scholar : PubMed/NCBI

47 

Wang Y, Yin C, Geng L and Cai W: Immune infiltration landscape in clear cell renal cell carcinoma implications. Front Oncol. 10:4916212021. View Article : Google Scholar : PubMed/NCBI

48 

Sabrina S, Takeda Y, Kato T, Naito S, Ito H, Takai Y, Ushijima M, Narisawa T, Kanno H, Sakurai T, et al: Initial myeloid cell status is associated with clinical outcomes of renal cell carcinoma. Biomedicines. 11:12962023. View Article : Google Scholar : PubMed/NCBI

49 

Liu B, Chen X, Zhan Y, Wu B and Pan S: Identification of a gene signature for renal cell Carcinoma-associated fibroblasts mediating cancer progression and affecting prognosis. Front Cell Dev Biol. 8:6046272021. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Y, Chen X, Fu Q, Wang F, Zhou X, Xiang J, He N, Hu Z and Jin X: Comprehensive analysis of pyroptosis regulators and tumor immune microenvironment in clear cell renal cell carcinoma. Cancer Cell Int. 21:6672021. View Article : Google Scholar : PubMed/NCBI

51 

Ballesteros PÁ, Chamorro J, Román-Gil MS, Pozas J, Gómez Dos Santos V, Granados ÁR, Grande E, Alonso-Gordoa T and Molina-Cerrillo J: Molecular mechanisms of resistance to immunotherapy and antiangiogenic treatments in clear cell renal cell carcinoma. Cancers. 13:59812021. View Article : Google Scholar : PubMed/NCBI

52 

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. 29:1437–1444. 2018. View Article : Google Scholar : PubMed/NCBI

53 

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. 359:91–97. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Xing Y and Hogquist KA: T-cell tolerance: Central and peripheral. Cold Spring Harb Perspect Biol. 4:a0069572012. View Article : Google Scholar : PubMed/NCBI

55 

Braun DA, Street K, Burke KP, Cookmeyer DL, Denize T, Pedersen CB, Gohil SH, Schindler N, Pomerance L, Hirsch L, et al: Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 39:632–648.e8. 2021. View Article : Google Scholar : PubMed/NCBI

56 

McKay RR, Bossé D, Xie W, Wankowicz SAM, Flaifel A, Brandao R, Lalani AA, Martini DJ, Wei XX, Braun DA, et al: The clinical activity of PD-1/PD-L1 inhibitors in metastatic non-clear cell renal cell carcinoma. Cancer Immunol Res. 6:758–765. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Pichler R, Siska PJ, Tymoszuk P, Martowicz A, Untergasser G, Mayr R, Weber F, Seeber A, Kocher F, Barth DA, et al: A chemokine network of T cell exhaustion and metabolic reprogramming in renal cell carcinoma. Front Immunol. 14:10951952023. View Article : Google Scholar : PubMed/NCBI

58 

Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, Zhao S, Das S, Beckermann KE, Ha L, et al: Fatal toxic effects associated with immune checkpoint inhibitors. JAMA Oncol. 4:1721–1728. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Lin E, Liu X, Liu Y, Zhang Z, Xie L, Tian K, Liu J and Yu Y: Roles of the dynamic tumor immune microenvironment in the individualized treatment of advanced clear cell renal cell carcinoma. Front Immunol. 12:6533582021. View Article : Google Scholar : PubMed/NCBI

60 

Ishihara H, Takagi T, Kondo T, Fukuda H, Tachibana H, Yoshida K, Iizuka J, Okumi M, Ishida H and Tanabe K: Predictive impact of an early change in serum C-reactive protein levels in nivolumab therapy for metastatic renal cell carcinoma. Urol Oncol Semin Orig Investig. 38:526–532. 2020.

61 

Noguchi G, Nakaigawa N, Umemoto S, Kobayashi K, Shibata Y, Tsutsumi S, Yasui M, Ohtake S, Suzuki T, Osaka K, et al: C-reactive protein at 1 month after treatment of nivolumab as a predictive marker of efficacy in advanced renal cell carcinoma. Cancer Chemother Pharmacol. 86:75–85. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Yano Y, Ohno T, Komura K, Fukuokaya W, Uchimoto T, Adachi T, Hirasawa Y, Hashimoto T, Yoshizawa A, Yamazaki S, et al: Serum C-reactive protein level predicts overall survival for clear cell and Non-clear cell renal cell carcinoma treated with ipilimumab plus nivolumab. Cancers. 14:56592022. View Article : Google Scholar : PubMed/NCBI

63 

Tomita Y, Larkin J, Venugopal B, Haanen J, Kanayama H, Eto M, Grimm MO, Fujii Y, Umeyama Y, Huang B, et al: Association of C-reactive protein with efficacy of avelumab plus axitinib in advanced renal cell carcinoma: Long-term follow-up results from JAVELIN renal 101. ESMO Open. 7:1005642022. View Article : Google Scholar : PubMed/NCBI

64 

Kim JH, Kim GH, Ryu YM, Kim SY, Kim HD, Yoon SK, Cho YM and Lee JL: Clinical implications of the tumor microenvironment using multiplexed immunohistochemistry in patients with advanced or metastatic renal cell carcinoma treated with nivolumab plus ipilimumab. Front Oncol. 12:9695692022. View Article : Google Scholar : PubMed/NCBI

65 

Sammarco E, Rossetti M, Salfi A, Bonato A, Viacava P, Masi G, Galli L and Faviana P: Tumor microenvironment and clinical efficacy of first line Immunotherapy-based combinations in metastatic renal cell carcinoma. Med Oncol. 41:1502024. View Article : Google Scholar : PubMed/NCBI

66 

Kazama A, Bilim V, Tasaki M, Anraku T, Kuroki H, Shirono Y, Murata M, Hiruma K and Tomita Y: Tumor-infiltrating immune cell status predicts successful response to immune checkpoint inhibitors in renal cell carcinoma. Sci Rep. 12:203862022. View Article : Google Scholar : PubMed/NCBI

67 

Herrmann T, Ginzac A, Molnar I, Bailly S, Durando X and Mahammedi H: Eosinophil counts as a relevant prognostic marker for response to nivolumab in the management of renal cell carcinoma: A retrospective study. Cancer Med. 10:6705–6713. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Atkins MB, Jegede OA, Haas NB, McDermott DF, Bilen MA, Stein M, Sosman JA, Alter R, Plimack ER, Ornstein M, et al: Phase II study of nivolumab and salvage Nivolumab/Ipilimumab in Treatment-naive patients with advanced clear cell renal cell carcinoma (HCRN GU16-260-Cohort A). J Clin Oncol. 40:2913–2923. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Motzer RJ, Choueiri TK, McDermott DF, Powles T, Vano YA, Gupta S, Yao J, Han C, Ammar R, Papillon-Cavanagh S, et al: Biomarker analysis from CheckMate 214: Nivolumab plus ipilimumab versus sunitinib in renal cell carcinoma. J Immunother Cancer. 10:e0043162022. View Article : Google Scholar : PubMed/NCBI

70 

Brown LC, Zhu J, Desai K, Kinsey E, Kao C, Lee YH, Pabla S, Labriola MK, Tran J, Dragnev KH, et al: Evaluation of tumor microenvironment and biomarkers of immune checkpoint inhibitor response in metastatic renal cell carcinoma. J Immunother Cancer. 10:e0052492022. View Article : Google Scholar : PubMed/NCBI

71 

Kato R, Jinnouchi N, Tuyukubo T, Ikarashi D, Matsuura T, Maekawa S, Kato Y, Kanehira M, Takata R, Ishida K and Obara W: TIM3 expression on tumor cells predicts response to anti-PD-1 therapy for renal cancer. Transl Oncol. 14:1009182020. View Article : Google Scholar : PubMed/NCBI

72 

Koh Y, Nakano K, Katayama K, Yamamichi G, Yumiba S, Tomiyama E, Matsushita M, Hayashi Y, Yamamoto Y, Kato T, et al: Early dynamics of circulating tumor DNA predict clinical response to immune checkpoint inhibitors in metastatic renal cell carcinoma. Int J Urol. 29:462–469. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Incorvaia L, Fanale D, Badalamenti G, Brando C, Bono M, De Luca I, Algeri L, Bonasera A, Corsini LR, Scurria S, et al: A ‘Lymphocyte MicroRNA Signature’ as predictive biomarker of immunotherapy response and plasma PD-1/PD-L1 expression levels in patients with metastatic renal cell carcinoma: Pointing towards epigenetic reprogramming. Cancers. 12:33962020. View Article : Google Scholar : PubMed/NCBI

74 

Pan Q, Liu R, Zhang X, Cai L, Li Y, Dong P, Gao J, Liu Y and He L: CXCL14 as a potential marker for immunotherapy response prediction in renal cell carcinoma. Ther Adv Med Oncol. 15:175883592312179662023. View Article : Google Scholar : PubMed/NCBI

75 

Pabla S, Seager RJ, Van Roey E, Gao S, Hoefer C, Nesline MK, DePietro P, Burgher B, Andreas J, Giamo V, et al: Integration of tumor inflammation, cell proliferation, and traditional biomarkers improves prediction of immunotherapy resistance and response. Biomark Res. 9:562021. View Article : Google Scholar : PubMed/NCBI

76 

Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, Walsh AM, Baxi V, Pandya D, Baradet T, et al: Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 26:688–692. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Ivanova E, Asadullina D, Rakhimov R, Izmailov A, Izmailov A, Gilyazova G, Galimov S, Pavlov V, Khusnutdinova E and Gilyazova I: Exosomal miRNA-146a is downregulated in clear cell renal cell carcinoma patients with severe immune-related adverse events. Noncoding RNA Res. 7:159–163. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Petitprez F, Ayadi M, de Reyniès A, Fridman WH, Sautès-Fridman C and Job S: Review of prognostic expression markers for clear cell renal cell carcinoma. Front Oncol. 11:6430652021. View Article : Google Scholar : PubMed/NCBI

79 

Lee A, Lee HJ, Huang HH, Tay KJ, Lee LS, Sim SPA, Ho SSH, Yuen SPJ and Chen K: Prognostic significance of inflammation-associated blood cell markers in nonmetastatic clear cell renal cell carcinoma. Clin Genitourin Cancer. 18:304–313. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Teishima J, Inoue S, Hayashi T and Matsubara A: Current status of prognostic factors in patients with metastatic renal cell carcinoma. Int J Urol. 26:608–617. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
González‑Garza R, Gutiérrez‑González A, Salinas‑Carmona MC and Mejía‑Torres M: Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review). Oncol Rep 52: 164, 2024.
APA
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M.C., & Mejía‑Torres, M. (2024). Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review). Oncology Reports, 52, 164. https://doi.org/10.3892/or.2024.8823
MLA
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M. C., Mejía‑Torres, M."Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review)". Oncology Reports 52.6 (2024): 164.
Chicago
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M. C., Mejía‑Torres, M."Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review)". Oncology Reports 52, no. 6 (2024): 164. https://doi.org/10.3892/or.2024.8823
Copy and paste a formatted citation
x
Spandidos Publications style
González‑Garza R, Gutiérrez‑González A, Salinas‑Carmona MC and Mejía‑Torres M: Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review). Oncol Rep 52: 164, 2024.
APA
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M.C., & Mejía‑Torres, M. (2024). Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review). Oncology Reports, 52, 164. https://doi.org/10.3892/or.2024.8823
MLA
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M. C., Mejía‑Torres, M."Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review)". Oncology Reports 52.6 (2024): 164.
Chicago
González‑Garza, R., Gutiérrez‑González, A., Salinas‑Carmona, M. C., Mejía‑Torres, M."Biomarkers for evaluating the clinical response to immune checkpoint inhibitors in renal cell carcinoma (Review)". Oncology Reports 52, no. 6 (2024): 164. https://doi.org/10.3892/or.2024.8823
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team