Download PDF
Review  |  Open Access  |  29 May 2023

Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma

Views: 612 |  Downloads: 190 |  Cited:   1
Cancer Drug Resist 2023;6:314-26.
10.20517/cdr.2023.09 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Renal-cell carcinoma (RCC) remains a leading cause of cancer-related mortality worldwide. Though newer therapeutic combinations of immune checkpoint inhibitors and targeted therapies have greatly improved outcomes, resistance to these therapies is becoming a challenge for long-term control. Mechanisms of resistance have been explored in a variety of solid tumors, including RCC. Based upon our review of the current literature on the mechanisms of resistance to immunotherapies for the management of metastatic clear-cell renal cell carcinomas (mccRCC), the ensuing conclusions have been made:

The management of mccRCC has progressed substantially with the advent of checkpoint inhibitors and targeted oral therapies, alone and/or in combination.

Nevertheless, innate or developed resistance to these therapies remains an ongoing challenge, particularly to immune checkpoint inhibitors (ICIs).

Several of the known mechanisms of resistance have been well defined, but recent progression in cellular therapies helps to expand the armamentarium of potential combination options that may overcome these modes of resistance and improve long-term disease control and survival for an otherwise dismal disease.

In the ensuing review and update of the literature on the mechanisms of resistance to immunotherapies in mccRCC, we have revisited the known resistance mechanisms of immunotherapies in metastatic clear-cell RCC and explored ongoing and future strategies to overcome them.

Keywords

Metastatic clear-cell renal-cell carcinoma, immune therapy, checkpoint inhibitor resistance, chimeric antigen receptor T-cell therapy

INTRODUCTION

Renal-cell carcinoma (RCC) remains a common cause of morbidity and mortality worldwide. It is the eighth most common cancer diagnosed in the United States, and in 2022 there were an estimated 79,000 new cases diagnosed, accounting for 4% of new cancer diagnoses in the country[1]. Though the relative 5-year survival is nearly 77%, the prognosis of advanced RCC remains dismal with an estimated 5-year survival of 15%[1]. Clear-cell renal-cell carcinoma (ccRCC) still accounts for the majority of RCC, with non-clear-cell histologies making up about 25%[2].

Historically, standard chemotherapy and radiation have been ineffective for ccRCC. Clinical studies suggested an immunologic role in the growth and control of RCC, particularly the presence of tumor-infiltrating lymphocytes (TILs) within the tumor and the process of immune evasion[3-5]. Immune therapies were then explored as potential therapeutic options, particularly immune cytokines such as interferons and interleukin-2 (IL-2) and later immune checkpoint inhibitors (ICIs). A better understanding of the role of programmed death 1 (PD-1), programmed death ligand 1 (PDL1), and cytotoxic T-lymphocyte–associated protein 4 (CTLA4) led to the eventual use of ICIs in metastatic renal-cell carcinoma (mRCC).

The discovery of a defective von Hippel-Lindau (VHL) gene as a major molecular alteration in the pathogenesis of ccRCC was another leap forward. VHL alterations resulted in upregulation of several growth factors involved in angiogenesis (platelet-derived growth factor receptor-beta, vascular endothelial growth factor [VEGF], and transforming growth factor alpha) using the hypoxia-inducible factor (HIF) pathway. This led to the development of newer therapies specifically targeting these factors[6]. VEGF-receptor tyrosine-kinase inhibitors, such as sunitinib, pazopanib, and sorafenib, quickly became the standard of care given their improved response rates, more convenient administration, and manageable toxicity profiles[7-9].

Single-agent use of tyrosine-kinase inhibitors and ICIs was effective but with limited responses and long-term control. Resistance, particularly to ICIs, remains a barrier to achieving and maintaining a durable response to these therapies. It is also important to note that ICIs have several potential adverse effects which must be taken into consideration, especially with long-term use[10]. Efforts have been made to combine immunotherapies and anti-angiogenic agents with each other and with other drugs. Other therapies include chemotherapy and radiation for potential synergistic and immunomodulatory effects and to overcome this resistance. Recent reviews summarized potential mechanisms but primarily focused on anti-angiogenic drug resistance[11]. In a prior review by Moreira et al. published in this journal, resistance mechanisms to immunotherapies in the management of metastatic RCC were explored[12]. Here, we revisit these mechanisms and discuss updated ongoing and future strategies for overcoming resistance, particularly adoptive cellular therapies.

Aim

To review and update the literature on mechanisms of resistance to immunotherapies in mccRCC.

Methods

Various internet databases were searched, including: PUBMED, Yahoo, Google, and Google Scholar. The search words that were used included: metastatic clear-cell renal-cell carcinoma, immune therapy, immune checkpoint inhibitor, immune checkpoint inhibitor resistance, and chimeric antigen receptor T-cell therapy. One hundred and six (106) references were identified, which were used to write the review and update the literature on the mechanisms of resistance to immunotherapies in mccRCC.

CURRENT THERAPEUTIC LANDSCAPE

The current treatment landscape for ccRCC in the first and second lines reflects the efficacy seen with immune and anti-angiogenic agents, both alone and in combination, over previously standard cytokine therapies. Choice of treatment is in part led by risk as determined by the International mRCC Database Consortium and Memorial Sloan Kettering Cancer Center/Motzer risk-stratification criteria[13,14]. Individual patient factors, including comorbid conditions, concomitant medications, and socioeconomics, also play a large role in the choice of therapy.

Current preferred first-line treatments for favorable risk metastatic ccRCC include: pembrolizumab/axitinib, nivolumab/cabozantinib, and pembrolizumab/lenvatinib, with response rates ranging from 55% to 71%[15-17]. Current preferred first-line treatment options for intermediate/poor-risk disease include: pembrolizumab/axitinib, nivolumab/cabozantinib, nivolumab/ipilimumab, pembrolizumab/lenvatinib, and cabozantinib[18].

Second-line treatments vary and include anti-angiogenic and immune-therapy agents that the patient may not have previously received. Inhibitors of the PI3K/AKT/mTOR pathway, such as everolimus and temsirolimus, currently have a role in later lines of therapy[19,20].

MECHANISMS OF RESISTANCE

ICIs have now become essential in the therapeutic armamentarium for several malignancies, including urothelial carcinoma, melanoma, and non-small-cell lung cancer. In addition to approved combinations for metastatic ccRCC, ICI was recently integrated into the adjuvant treatment of ccRCC with the approval of pembrolizumab post-nephrectomy for high-risk disease, based on results from KEYNOTE-564, noting a disease-free survival benefit of nearly 10% at 24 months, and 30 months disease-free survival HR of 0.63[21,22]. Though ICIs have been very effective, we now see evidence of resistance to these therapies, which limits the durability of response.

The complex and intricate interaction between the immune system and the cancer cell has been described by the various host- and tumor-specific characteristics that have an impact on this interaction and are visually depicted by Blank et al., where potential and confirmed biomarkers at these levels are also noted [Figure 1][23]. However, this interface is complex, and many parts of this interaction remain undiscovered. In general, immune resistance can be innate, acquired, intrinsic, or extrinsic[24]. Innate or primary resistance is an immediate lack of response caused by the presence of resistant clones before starting treatment. Acquired or secondary resistance occurs while on active therapy after an initial response to treatment. Intrinsic resistance occurs when the tumor cells interfere with internal processes, such as cell signaling, gene expression, DNA damage response, and immune recognition, whereas extrinsic resistance occurs through T-cell activation and other processes outside the cell[24]. Moreira et al. divided potential factors contributing to resistance into three major groups - patient, tumor cell, and tumor microenvironment (TME)[12].

Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma

Figure 1. The cancer immunogram - parameters that characterize aspects of cancer-immune interactions where biomarkers have been or may be identified[23]. Reprinted with permission from the American Association for the Advancement of Science.

Patient-associated factors of resistance

Patient-related factors include sex/gender, HLA genotype, sarcopenia, gut microbiome, and antibiotic and corticosteroid use. Recent data have continued to show trends toward sex-related differences in response to ICI, favoring males over females[25-27]. Over recent years, there has been tremendous interest in the relationship between the gut microbiome and ICI response. Several studies in melanoma and renal-cell carcinoma have shown that increased diversity in intestinal bacteria is associated with better responses to checkpoint blockade and that certain species may improve or diminish response[28,29]. Mouse-model studies exhibited that the microbiome may alter the amount of tumor dendritic cells, antigen-presenting cells, and cytokines[30]. Species associated with improved response in mRCC include Akkermansia spp. and Bifidobacterium spp[28]. In a recent phase I study, Dizman et al. investigated the use of nivolumab and ipilimumab with or without CBM588, a bifidogenic live bacterial product, in mRCC and found enhanced outcomes among patients receiving combination therapy with the probiotic agent[31]. Moreover, antibiotic therapy may alter ICI response because it dysregulates the microbiome. Studies found poor ICI response associated with antibiotic use among patients with mRCC on immunotherapy[32]. This was also evaluated in a recent meta-analysis of 10 studies which found decreased progression-free survival (PFS), overall survival (OS), and objective response rate (ORR)[33]. In a recent population-based study of patients over 65 years of age, antibiotic use within 1 year of ICI therapy, in particular fluoroquinolones, was associated with worse OS[34]. Additionally, steroid use while receiving ICIs has been associated with poor outcomes (PFS and OS). In a recent meta-analysis of 16 studies including non-small cell lung cancer and melanoma, steroid use for supportive care or brain metastases was associated with reduced OS rather than use for adverse effects from ICIs[35]. Ongoing studies are evaluating ways of modifying the gut microbiome and factors of antibiotics and steroid use that may be modifiable and affect response to therapy.

Tumor cell-associated factors of resistance

Tumor cell-related factors for immune evasion and resistance include altered methods of antigen presentation and T-cell exhaustion. By reducing the expression of tumor antigens and downregulating MHC class I, tumor cells may avoid immune surveillance and destruction[36]. Alternatively, chronic exposure to an antigen may lead to upregulation of PD-1 or the expression of other inhibitory receptors, such as TIM3, LAG3, BTLA, TIGIT, and VISTA[24,37,38].

This interaction between tumor antigens and immune response led to the theory of combining immunotherapies and stereotactic body radiation therapy (SBRT) as a therapeutic option. Radiation causes tumor necrosis and may release more tumor antigens, which in turn may allow ICIs to work more effectively both locally and potentially beyond what is irradiated. Several studies evaluated the efficacy of this approach, including NIVES, RADVAX RCC, and a high-dose IL2 + SBRT study. NIVES was a phase II study evaluating the role of nivolumab and SBRT in a pretreated patient with mRCC. Sixty-nine patients were enrolled with ORR of 17%, disease control rate of 55%, and OS of 20 months. However, the authors concluded that this combination did not result in improved outcomes among pretreated patients but could be studied further and considered in an oligometastatic population[39]. RADVAX RCC evaluated dual-checkpoint blockade with ipilimumab and nivolumab and SBRT. Twenty-five patients were enrolled and initial analysis noted an ORR of 56%[40]. Interleukin-2 with SBRT has been studied in metastatic melanoma and mRCC with promising antitumor activity[41,42].

TME-associated factors of resistance

The TME includes factors extrinsic to the cancer cell and plays a substantial role in regulating T-lymphocytes. The balance between Tregs and Teff cells is an important factor in the response or resistance to ICI, with greater Tregs resulting in a diminished response. Moreover, myeloid-derived suppressor cells are another regulatory mechanism that allows for continued tumor growth through immune regulation[24]. Tumor-associated macrophages, TGF-beta, VEGF, and cytokines are all involved in these regulatory processes and may alter the response to ICI therapy.

Chronic antigen exposure can result in upregulation of PD-1 expression and ultimately T-cell exhaustion, a hypofunctional state associated with decreased Teff function[43,44]. Exhausted T cells in cancer are similar to those in chronic viral infections, and upregulation of immune checkpoints is a hallmark feature[45]. This exhaustion in turn may alter antigen presentation and can be a potential mechanism of resistance to checkpoint blockade in multiple tumor types, including mRCC[46-48].

TILs are strong tumor-defense mechanisms regulating growth and spread. In most malignancies, higher amounts of TILs have been associated with better prognosis and response to ICIs[49-51]. However, there are some tumor types in which more TILs are not necessarily better. Though RCC is a heavily T-cell - enriched tumor with high numbers of CD8+ TILs, these are mostly dysfunctional or exhausted[52-55]. They also express more inhibitory receptors, such as LAG3 and Tim-3, co-expressed with PD-1, which have been associated with more aggressive phenotypes exhibited by higher TNM staging and a higher Fuhrman grade[49,56].

Hypoxia is a major feature in the TME of RCC that may contribute to immune dysregulation and tumor progression through several different mechanisms, as was previously described by Moreira et al. and others[12,38,57,58]. Hypoxia results in the release of hypoxia-inducible factors 1 and 2 (HIF-1a and HIF-2a), which stimulate the expression of inhibitory signals, such as VEGF, CTLA4, and LAG3, and suppress T-cell activation and function[12]. Belzutifan, a selective HIF-2a inhibitor, was studied in a phase I trial with promising antitumor activity in heavily pretreated patients with mccRCC[59]. It is currently FDA-approved for VHL disease - associated tumors, including RCC, CNS hemangioblastomas, and pancreatic neuroendocrine tumors[60]. The use of belzutifan in other settings of RCC and in combination with other therapies, including ICI, is under investigation. Tumor-intrinsic factors and the TME and their interplay are illustrated in Figure 2 as originally developed by Ballesteros et al.[61].

Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma

Figure 2. Tumor-intrinsic factors and the TME to describe potential mechanisms of resistance to immune therapies[61]. Reprinted with permission from Multidisciplinary Digital Publishing Institute. Abbreviation: TME, tumor microenvironment. Reproduced from[60] under the Creative Commons Agreement License.

FUTURE DIRECTIONS

Proposed strategies to overcome resistance to ICIs include novel combination approaches, including checkpoint inhibitors with cytokine or chemokine therapy, adoptive cellular therapies, or oncolytic viruses. Biomarkers remain elusive in mRCC as most, including PD-L1, have failed to predict therapeutic response[62-64]. Cell-free circulating tumor DNA (ctDNA) may have some utility in mRCC but is limited by lower levels in RCC compared to other solid tumors[65,66]. Those with higher tumor volume appear to shed more ctDNA[67]. Tumor mutation burden may also be a useful predictor of response to ICIs because a higher tumor mutation burden is associated with a favorable response, though this does not appear to hold for combined checkpoint inhibitors[68]. Recently, transcriptomics, metabolomics, and metabolic profiling of RCC cells have allowed for a potential avenue of tumor and resistance detection based on common metabolic features of the cancer cell. Lower OS and greater ICI resistance have been noted in tumor cells with an altered kynurenine/tryptophan ratio and higher hypoxia and Wnt/beta-catenin signaling[69-72].

Revisiting cytokine therapy

The success of ICIs is limited by resistance reinvigorated interest in cytokine therapy, which had originally been effective in RCC, though without robust responses (around 20%) and their use was limited by challenges in administration and potential toxicities[73-75]. In a retrospective analysis, Buchbinder et al. evaluated response among patients with metastatic melanoma and mRCC who had received HD IL-2 after prior treatment with PD-1/PD-L1 therapies through the PROCLAIM database. Of the 57 total patients, 17 had mRCC and ORR was 24% with two complete responses[76]. In the PIVOT-02 phase I/II study, bempegaldesleukin, a pegylated form of IL-2, was tested with nivolumab as the first line in mccRCC. Initial tumor activity was noted with an ORR of 35% and a complete response of 6%[77]. This is being evaluated further in PIVOT-09, the phase III study, though enrollment was recently terminated as of November 2022[78,79]. Another recent early-phase trial studied pembrolizumab with HD IL-2 in mRCC and noted an ORR of 70% and no worsening toxicities suggesting further exploration[80].

Chimeric antigen receptor T-cell (CAR-T) therapy

CAR-T therapy is an adoptive cellular treatment that has revolutionized the management of refractory hematologic malignancies. In essence, a patient’s T cells are removed and modified against a specific cellular target and then infused back into the patient (autologous therapy)[81]. This began with the FDA approval of axicabtagene ciloleucel (axi-cel), an autologous anti-CD-19 CAR-T, in October 2017 for relapsed/refractory (R/R) large B-cell lymphoma after two or more prior lines of therapy based on results of the phase I Zuma-1 study[82,83]. Since then, axi-cel has been approved for second-line management of large B-cell lymphoma based on results from Zuma-9[84]. Other agents and indications have received approval - brexucabtagene autoleucel (brexu-cel) for R/R B-cell precursor acute lymphoblastic leukemia (ZUMA-3); tisagenlecleucel (tisa-cel) for R/R B-ALL (NCT02435849) and R/R large B-cell lymphoma (JULIET); idecabtagene vicleucel (ide-cel) for R/R multiple myeloma (KarMMa); abi-cel for R/R follicular lymphoma (FL) (ZUMA-5); and lisocabtagene maraleucel (liso-cel) for R/R large B-cell lymphomas (TRANSCEND). However, axi-cel’s utility in solid tumors remains under investigation with modest results to date, partly because of tumor-suppressing molecules in the TME and tumor heterogeneity[85-91].

Several preclinical studies assessed potential targets for CAR-T in RCC, with CD70 as a promising site because it appears highly expressed in clear-cell and sarcomatoid tumors[92-94]. Early findings from the COBALT-RCC trial (NCT04438083) were recently presented at the 2022 Society for Immunotherapy of Cancer Annual Meeting with safety and clinical activity for CTX130, an allogeneic anti-CD70 CAR-T that has been CRISP/Cas9 gene-edited, in the 13 evaluable patients so far. The ORR was 8% and the disease control rate 77%, with 1 partial response resulting in a CR maintained at 18 months[95]. TRAVERSE is an ongoing clinical trial studying the safety and efficacy of ALLO-360, an allogeneic CAR targeting CD70, in mRCC with preliminary data pending (NCT04696731). Another CAR-T product directed against carboxy-anhydrase-IX (CAIX) was promising in preclinical studies, though in an early phase trial, anti-CAR-T antibodies developed with immune responses, and no clinical benefit was noted, and the study was terminated[96]. This agent has again been evaluated now in combination with sunitinib, though in mouse models, with some synergistic response seen[97]. Ongoing studies are investigating other potential targets and methods of overcoming resistance and suppressive mechanisms of CAR-T by potentially combining it with other agents.

TIL therapy

TIL therapy has been investigated in mRCC for decades but with limited success, often because of suppressive features of the TME limiting retrieval of large amounts of tumor-reactive TILs[98-100]. Figlan et al. studied radical nephrectomy with IL-2 and TIL therapy among patients with mRCC with favorable results[101]. In a recent commentary, Andersen et al. highlighted several prior TIL studies in RCC with variable ORR ranging from 0% to 35%[99]. Ongoing studies for TIL therapy are limited though may benefit from combination-based strategies as previously considered, particularly with ICIs to help overcome TME-suppression coinhibitory signals[102]. Newer methods of TIL retrieval and expansion are also being explored to help overcome these limiting features[99,103]. Table 1 summarizes ongoing cellular therapy trials in mRCC.

Table 1

Summary of ongoing cellular therapy clinical trials in metastatic/advanced RCC. Information obtained from clinicaltrials.gov

NameIdentifierModalityPrimary SiteStatus
Clinical study of CAIX-targeted CAR-T Cells in the Treatment of Advanced Renal Cell CarcinomaNCT04969354CAR-TAffiliated Hospital of Xuzhou Medical UniversityRecruiting (last updated June 2021)
Clinical study of CD-70 targeted CAR-T therapy in advanced renal cancer
NCT05420519CAR-TThe Second People’s Hospital of Shandong ProvinceRecruiting (updated June 2022)
Aldesleukin and pembrolizumab in treating patients with advanced or metastatic renal cell carcinomaNCT03260504CAR-TFred Hutch/ University of Washington Cancer ConsortiumRecruiting (updated October 2022)
TIL Therapy for Metastatic Renal Cell CarcinomaNCT02926053TILCenter for Cancer Immune Therapy, DenmarkRecruiting/ unknown (updated December 2019)
Safety and efficacy of ALLO-316 in subjects with advanced or metastatic clear-cell renal cell carcinoma (TRAVERSE)NCT04696731CAR-TCity of Hope, UCLA Medical Center, Moffitt Cancer Center, Memorial Sloan Kettering Cancer Center, Providence Portland Medical Center, MD Anderson Cancer CenterRecruiting (updated March 2022)
P-MUC1C-ALLO1 Allogeneic CAR-T cells in the treatment of subjects with advanced or metastatic solid tumorsNCT05239143CAR-TUniversity of California San Francisco, Sarah Cannon Research Institute at HealthOne, University of Kansas Cancer Center, MD Anderson Cancer Center, NEXT OncologyRecruiting (updated October 2022)
A clinical research about CD70-positive advanced/ metastatic solid tumors treated by CD70-targeted CAR-TNCT05468190CAR-THenan Cancer HospitalRecruiting (updated September 2022)
A clinical study of CD70-targeted CAR-T in the treatment of CD70-positive advanced/ metastatic solid tumorsNCT05518253CAR-TFirst Affiliated Hospital, Zhejiang UniversityRecruiting (updated September 2022)
Safety and Efficacy of CCT301 CAR-T in adult subjects with recurrent or refractory stage IV renal cell carcinomaNCT03393936CAR-TShanghai Public Health Clinical CenterActive, not recruiting (updated October 2021)
A safety and efficacy study evaluating CTX130 in subjects with relapse or refractory renal cell carcinoma (COBALT-RCC)NCT04438083CAR-TMultiple sites including the United States, Australia, Canada, and the NetherlandsRecruiting (updated May 2022)
HERV-E TCR Transduced Autologous T cells in People with Metastatic Clear-Cell RCCNCT03354390TCRNational Institutes of Health Clinical CenterRecruiting (updated December 2022)
Administering Peripheral Blood Lymphocytes Transduced with a CD70-Binding Chimeric Antigen Receptor to People with CD70 Expressing CancersNCT02830724CAR-TNational Institutes of Health Clinical CenterRecruiting (updated January 2023)

Oncolytic virus-based therapy

Another avenue to overcome resistance is the combination of ICIs with oncolytic viruses, which elicit antitumor immunity[104]. Their use seems limited when used alone but may be more beneficial when combined with other agents targeting specific tumor-suppressive signals in the TME. In a phase II open-label study, patients with metastatic melanoma received either talimogene laherparepvec with ipilimumab or ipilimumab alone, and higher antitumor activity with a greater response rate was seen in the combination without increased toxicities (ORR 39%)[105]. Recently, a phase I study evaluating NeoVax, a neo-antigen cancer vaccine, with ipilimumab in RCC is actively recruiting (NCT02950766).

Other targeted approaches

As discussed previously, HIF-2a inhibitors are now being investigated in mRCC, and favorable outcomes led to the approval of belzutifan for VHL-associated tumors, including RCC. The role of HIF-2a inhibitors is being expanded and its use in combination with other agents, including ICIs, is being tested. Preclinical studies for belzutifan used in mouse models without HIF-2a expression found no efficacy for single-agent use, but there was potential synergy with checkpoint inhibitors[106].

Cyclin-dependent kinase 4/6 (CDK4/6) are signaling molecules that promote the progression of the cell cycle by overcoming the tumor suppressor activity of retinoblastoma. Inhibitors of CDK4/6 are currently FDA-approved in the management of breast and ovarian cancers with favorable response rates. Recent preclinical data show promise for CDK4/6 inhibition in RCC to potentiate response with ICIs and chemotherapy[107].

CONCLUSIONS

The management of mccRCC has progressed substantially with the advent of checkpoint inhibitors and targeted oral therapies, alone and/or in combination. However, innate or developed resistance to these therapies remains an ongoing challenge, particularly to ICIs. Several of the known mechanisms of resistance have been well defined, but recent progression in cellular therapies helps to expand the armamentarium of potential combination options that may overcome these modes of resistance and improve long-term disease control and survival for an otherwise dismal disease.

DECLARATIONS

Acknowledgment

Editorial assistance was provided by Moffitt Cancer Center’s Office of Scientific Publishing by Daley Drucker and Gerard Hebert; no compensation was given beyond their regular salaries. We would like to thank the American Association for the Advancement of Science (AAAS) and Multidisciplinary Digital Publishing Institute (MDPI) for granting us permission to reprint the included figures.

Author’s contributions

Conceived and organized the design of the manuscript, performed the literature review and evaluation, and wrote the body of the manuscript: Chatwal MS

Provided administrative, technical, and material support and assisted in editing and reviewing the manuscript: Spiess PE

Performed the literature review, provided material and technical support, and helped to review and edit the manuscript: Chahoud J

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

Monica Chatwal has no conflicts of interest to report. Jad Chahoud reports advisory board consultation for Pfizer, Aveo, and Exelixis. Philippe Spiess has no conflicts of interest to report.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. SEER*Explorer: An interactive website for SEER cancer statistics [Internet]. Surveillance Research Program, National Cancer Institute; 2023 Apr 19. Available from: https://seer.cancer.gov/statistics-network/explorer/. [Last accessed on 2 Jun 2023].

2. Lopez-Beltran A, Scarpelli M, Montironi R, Kirkali Z. 2004 WHO classification of the renal tumors of the adults. Eur Urol 2006;49:798-805.

3. Finke JH, Rayman P, Hart L, et al. Characterization of tumor-infiltrating lymphocyte subsets from human renal cell carcinoma: specific reactivity defined by cytotoxicity, interferon-gamma secretion, and proliferation. J Immunother Emphasis Tumor Immunol 1994;15:91-104.

4. Giraldo NA, Becht E, Pagès F, et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res 2015;21:3031-40.

5. Jian Y, Yang K, Sun X, et al. Current advance of immune evasion mechanisms and emerging immunotherapies in renal cell carcinoma. Front Immunol 2021;12:639636.

6. Gnarra JR, Tory K, Weng Y, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet 1994;7:85-90.

7. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115-24.

8. Casper J, Schumann-Binarsch S, Köhne CH. Pazopanib versus sunitinib in renal cancer. N Engl J Med 2013;369:1968-70.

9. Escudier B, Eisen T, Stadler WM et al. Sorafenib in advanced clear-cell renal-cell carcinoma. NEJM 2007;356:125-34.

10. Wu Z, Chen Q, Qu L, et al. Adverse events of immune checkpoint inhibitors therapy for urologic cancer patients in clinical trials: a collaborative systematic review and meta-analysis. Eur Urol 2022;81:414-25.

11. Makhov P, Joshi S, Ghatalia P, Kutikov A, Uzzo RG, Kolenko VM. Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies. Mol Cancer Ther 2018;17:1355-64.

12. Moreira M, Pobel C, Epaillard N, Simonaggio A, Oudard S, Vano YA. Resistance to cancer immunotherapy in metastatic renal cell carcinoma. Cancer Drug Resist 2020;3:454-71.

13. Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A, Ferrara J. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol 1999;17:2530-40.

14. Heng DY, Xie W, Regan MM, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol 2009;27:5794-9.

15. Powles T, Plimack ER, Soulières D et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol 2020;21:1563-73.

16. Motzer RJ, Powles T, Burotto M et al. Nivolumab plus cabozantinib versus sunitinib in first-line treatment for advanced renal cell carcinoma (CheckMate 9ER): long-term follow-up results from an open-label, randomised, phase 3 trial. Lancet Oncol 2022;23:888-98.

17. Motzer R, Alekseev B, Rha SY, et al. CLEAR Trial Investigators. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma. N Engl J Med 2021;384:1289-300.

18. Motzer RJ, Jonasch E, Agarwal N, et al. Kidney cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2022;20:71-90.

19. Motzer RJ, Escudier B, Oudard S, et al. RECORD‐1 Study Group. Phase 3 trial of everolimus for metastatic renal cell carcinoma : final results and analysis of prognostic factors. Cancer 2010;116:4256-65.

20. Hudes G, Carducci M, Tomczak P, et al. Global ARCC Trial. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271-81.

21. Choueiri TK, Tomczak P, Park SH, et al. KEYNOTE-564 Investigators. Adjuvant pembrolizumab after nephrectomy in renal-cell carcinoma. N Engl J Med 2021;385:683-94.

22. Powles T, Tomczak P, Park SH et al. Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for clear cell renal cell carcinoma (KEYNOTE-564): 30-month follow-up analysis of a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:1133-44.

23. Blank CU, Haanen JB, Ribas A, Schumacher TN. The cancer immunogram. Science 2016;352:658-60.

24. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of resistance to immune checkpoint blockade: why does checkpoint inhibitor immunotherapy not work for all patients? Am Soc Clin Oncol Educ Book 2019;39:147-64.

25. Wallis CJD, Butaney M, Satkunasivam R, et al. Association of patient sex with efficacy of immune checkpoint inhibitors and overall survival in advanced cancers: a systematic review and meta-analysis. JAMA Oncol 2019;5:529-36.

26. Jang SR, Nikita N, Banks J, et al. Association between sex and immune checkpoint inhibitor outcomes for patients with melanoma. JAMA Netw Open 2021;4:e2136823.

27. Scott SC, Shao XM, Niknafs N, et al. Sex-specific differences in immunogenomic features of response to immune checkpoint blockade. Front Oncol 2022;12:945798.

28. Derosa L, Routy B, Fidelle M, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol 2020;78:195-206.

29. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015;350:1084-9.

30. 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.

31. 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.

32. Ueda K, Yonekura S, Ogasawara N, et al. The impact of antibiotics on prognosis of metastatic renal cell carcinoma in japanese patients treated with immune checkpoint inhibitors. Anticancer Res 2019;39:6265-71.

33. Luo Z, Hao S, Li Y, et al. The negative effect of antibiotics on RCC patients with immunotherapy: A systematic review and meta-analysis. Front Immunol 2022;13:1065004.

34. Eng L, Sutradhar R, Niu Y, et al. Impact of antibiotic exposure before immune checkpoint inhibitor treatment on overall survival in older adults with cancer: a population-based study. J Clin Oncol 2023:JCO2200074.

35. Petrelli F, Signorelli D, Ghidini M, et al. Association of steroids use with survival in patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers 2020;12:546.

36. Dutcher JP, Flippot R, Fallah J, Escudier B. On the shoulders of giants: the evolution of renal cell carcinoma treatment-cytokines, targeted therapy, and immunotherapy. Am Soc Clin Oncol Educ Book 2020;40:1-18.

37. Barrueto L, Caminero F, Cash L, Makris C, Lamichhane P, Deshmukh RR. Resistance to checkpoint inhibition in cancer immunotherapy. Transl Oncol 2020;13:100738.

38. Weverwijk A, de Visser KE. Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer 2023;23:193-215.

39. Masini C, Iotti C, De Giorgi U, et al. Nivolumab in combination with stereotactic body radiotherapy in pretreated patients with metastatic renal cell carcinoma. results of the phase II NIVES study. Eur Urol 2022;81:274-82.

40. Hammers HJ, Vonmerveldt D, Ahn C, et al. Combination of dual immune checkpoint inhibition (ICI) with stereotactic radiation (SBRT) in metastatic renal cell carcinoma (mRCC) (RADVAX RCC). JCO 2020;38:614-614.

41. Seung SK, Curti BD, Crittenden M et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med 2012;4:137ra74-ra74.

42. Curti B, Crittenden M, Seung SK, et al. Randomized phase II study of stereotactic body radiotherapy and interleukin-2 versus interleukin-2 in patients with metastatic melanoma. J Immunother Cancer 2020;8:e000773.

43. Chow A, Perica K, Klebanoff CA, Wolchok JD. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol 2022;19:775-90.

44. Thommen DS, Schumacher TN. T cell dysfunction in cancer. Cancer Cell 2018;33:547-62.

45. Wherry EJ, Ha SJ, Kaech SM, et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007;27:670-84.

46. Thompson RH, Dong H, Lohse CM, et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res 2007;13:1757-61.

47. Kang MJ, Kim KM, Bae JS, et al. Tumor-infiltrating PD1-positive lymphocytes and FoxP3-positive regulatory T cells predict distant metastatic relapse and survival of clear cell renal cell carcinoma. Transl Oncol 2013;6:282-9.

48. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis 2015;6:e1792.

49. Giraldo NA, Becht E, Vano Y, et al. Tumor-infiltrating and peripheral blood T-cell immunophenotypes predict early relapse in localized clear cell renal cell carcinoma. Clin Cancer Res 2017;23:4416-28.

50. 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.

51. Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 2011;105:93-103.

52. Nakano O, Sato M, Naito Y et al. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res 2001;61:5132-6.

53. Braun DA, Bakouny Z, Hirsch L, et al. Beyond conventional immune-checkpoint inhibition - novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol 2021;18:199-214.

54. Verma NK, Wong BHS, Poh ZS, et al. Obstacles for T-lymphocytes in the tumour microenvironment: therapeutic challenges, advances and opportunities beyond immune checkpoint. EBioMedicine 2022;83:104216.

55. Alkhouli MA, Bazargan S, Pilon-Thomas S, Poch M, Chahoud J. Current state of cell therapies for genitourinary malignancies. Cancer J 2022;28:294-300.

56. Granier C, Dariane C, Combe P, et al. Tim-3 expression on tumor-infiltrating PD-1+CD8+ T cells correlates with poor clinical outcome in renal cell carcinoma. Cancer Res 2017;77:1075-82.

57. Castillo-Rodríguez RA, Trejo-Solís C, Cabrera-Cano A, Gómez-Manzo S, Dávila-Borja VM. Hypoxia as a modulator of inflammation and immune response in cancer. Cancers 2022;14:2291.

58. Pietrobon V, Marincola FM. Hypoxia and the phenomenon of immune exclusion. J Transl Med 2021;19:9.

59. 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.

60. FDA approves belzutifan for cancers associated with von Hippel-Lindau disease. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-belzutifan-cancers-associated-von-hippel-lindau-disease. [Last accessed on 30 May 2023].

61. Ballesteros PÁ, Chamorro J, Román-Gil MS, et al. Molecular mechanisms of resistance to immunotherapy and antiangiogenic treatments in clear cell renal cell carcinoma. Cancers 2021;13:5981.

62. Carretero-González A, Lora D, Martín Sobrino I, et al. The value of PD-L1 expression as predictive biomarker in metastatic renal cell carcinoma patients: a meta-analysis of randomized clinical trials. Cancers 2020;12:1945.

63. Zhu J, Armstrong AJ, Friedlander TW, et al. Biomarkers of immunotherapy in urothelial and renal cell carcinoma: PD-L1, tumor mutational burden, and beyond. J Immunother Cancer 2018;6:4.

64. Choueiri TK, Albiges L, Haanen JBAG, et al. Biomarker analyses from JAVELIN Renal 101: Avelumab + axitinib (A+Ax) versus sunitinib (S) in advanced renal cell carcinoma (aRCC). JCO 2019;37:101-101.

65. Geertsen L, Koldby KM, Thomassen M, Kruse T, Lund L. Circulating tumor DNA in patients with renal cell carcinoma. A systematic review of the literature. Eur Urol Open Sci 2022;37:27-35.

66. Kim YJ, Kang Y, Kim JS, et al. Potential of circulating tumor DNA as a predictor of therapeutic responses to immune checkpoint blockades in metastatic renal cell carcinoma. Sci Rep 2021;11:5600.

67. Hahn AW, Nussenzveig RH, Maughan BL, Agarwal N. Cell-free circulating tumor DNA (ctDNA) in metastatic renal cell carcinoma (mRCC): current knowledge and potential uses. KCA 2019;3:7-13.

68. Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 2017;16:2598-608.

69. di Meo NA, Lasorsa F, Rutigliano M, et al. Renal cell carcinoma as a metabolic disease: an update on main pathways, potential biomarkers, and therapeutic targets. Int J Mol Sci 2022;23:14360.

70. Mock A, Zschäbitz S, Kirsten R, et al. Serum very long-chain fatty acid-containing lipids predict response to immune checkpoint inhibitors in urological cancers. Cancer Immunol Immunother 2019;68:2005-14.

71. Hoefflin R, Harlander S, Schäfer S, et al. HIF-1α and HIF-2α differently regulate tumour development and inflammation of clear cell renal cell carcinoma in mice. Nat Commun 2020;11:4111.

72. Wang G, Zhang ZJ, Jian WG, et al. Novel long noncoding RNA OTUD6B-AS1 indicates poor prognosis and inhibits clear cell renal cell carcinoma proliferation via the Wnt/β-catenin signaling pathway. Mol Cancer 2019;18:15.

73. Medical Research Council Renal Cancer Collaborators. Interferon-alpha and survival in metastatic renal carcinoma: early results of a randomised controlled trial. Lancet 1999;353:14-7.

74. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 1995;13:688-96.

75. Parton M, Gore M, Eisen T. Role of cytokine therapy in 2006 and beyond for metastatic renal cell cancer. J Clin Oncol 2006;24:5584-92.

76. Buchbinder EI, Dutcher JP, Daniels GA, et al. Therapy with high-dose Interleukin-2 (HD IL-2) in metastatic melanoma and renal cell carcinoma following PD1 or PDL1 inhibition. J Immunother Cancer 2019;7:49.

77. Tannir NM, Cho DC, Diab A, et al. Bempegaldesleukin plus nivolumab in first-line renal cell carcinoma: results from the PIVOT-02 study. J Immunother Cancer 2022;10:e004419.

78. Tannir NM, Agarwal N, Pal SK, et al. PIVOT-09: A phase III randomized open-label study of bempegaldesleukin (NKTR-214) plus nivolumab versus sunitinib or cabozantinib (investigator’s choice) in patients with previously untreated advanced renal cell carcinoma (RCC). JCO 2020;38:TPS763-TPS763.

79. Clinicaltrials.gov. A study of bempegaldesleukin (NKTR-214: BEMPEG) in combination with nivolumab compared with the investigator's choice of a Tyrosine Kinase Inhibitor (TKI) therapy (either sunitinib or cabozantinib monotherapy) for advanced metastatic renal cell carcinoma (RCC). Available from: https://clinicaltrials.gov/ct2/show/NCT03729245. [Last accessed on 30 May 2023].

80. Chatzkel J, Schell MJ, Chahoud J, et al. Coordinated pembrolizumab and high dose IL-2 (5-in-a-row schedule) for therapy of metastatic clear cell renal cancer. Clin Genitourin Cancer 2022;20:252-9.

81. June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med 2018;379:64-73.

82. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017;377:2531-44.

83. FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-car-t-cell-therapy-treat-adults-certain-types-large-b-cell-lymphoma. [Last accessed on 30 May 2023].

84. Locke FL, Miklos DB, Jacobson CA, et al. All ZUMA-7 investigators and contributing kite members. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med 2022;386:640-54.

85. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439-48.

86. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet 2021;398:491-502.

87. Schuster SJ, Bishop MR, Tam CS, et al. JULIET Investigators. Tisagenlecleucel in adult relapsed or refractory diffuse large b-cell lymphoma. N Engl J Med 2019;380:45-56.

88. Jacobson CA, Chavez JC, Sehgal AR, et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol 2022;23:91-103.

89. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet 2020;396:839-52.

90. Munshi NC, Anderson LD Jr, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med 2021;384:705-16.

91. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J 2021;11:69.

92. Ye H, Huang RR, Shuch BM, et al. CD70 is a promising CAR-T cell target in patients with advanced renal cell carcinoma. JCO 2022;40:384-384.

93. Panowski SH, Srinivasan S, Tan N, et al. Preclinical development and evaluation of allogeneic CAR T cells targeting CD70 for the treatment of renal cell carcinoma. Cancer Res 2022;82:2610-24.

94. Wang QJ, Yu Z, Hanada KI, et al. Preclinical evaluation of chimeric antigen receptors targeting CD70-expressing cancers. Clin Cancer Res 2017;23:2267-76.

95. Pal SK, Tran B, Hannen JB, et al. CTX130 allogeneic CRISPR-Cas9 - engineered chimeric antigen receptor (CAR) T cells in patients with advanced clear cell renal cell carcinoma: results from the phase 1 COBALT-RCC study. Presented at: 2022 SITC Annual Meeting; November 8-12, 2022; Boston, MA. Available from: http://ir.crisprtx.com/static-files/0941e694-f38e-41d3-b13d-1f7e15523054. [Last accessed on 30 May 2023].

96. Lamers CH, Klaver Y, Gratama JW, Sleijfer S, Debets R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem Soc Trans 2016;44:951-9.

97. Li H, Ding J, Lu M, et al. CAIX-specific CAR-T cells and sunitinib show synergistic effects against metastatic renal cancer models. J Immunother 2020;43:16-28.

98. Markel G, Cohen-Sinai T, Besser MJ et al. Preclinical evaluation of adoptive cell therapy for patients with metastatic renal cell carcinoma. Anticancer Res 2009;29:145-54. PMID: 19331144.

99. Andersen R, Donia M, Westergaard MC, Pedersen M, Hansen M, Svane IM. Tumor infiltrating lymphocyte therapy for ovarian cancer and renal cell carcinoma. Hum Vaccin Immunother 2015;11:2790-5.

100. Andersen R, Westergaard MCW, Kjeldsen JW, et al. T-cell responses in the microenvironment of primary renal cell carcinoma-implications for adoptive cell therapy. Cancer Immunol Res 2018;6:222-35.

101. Figlin RA, Pierce WC, Kaboo R, et al. Treatment of metastatic renal cell carcinoma with nephrectomy, interleukin-2 and cytokine-primed or CD8(+) selected tumor infiltrating lymphocytes from primary tumor. J Urol 1997;158:740-5.

102. Yin H, Guo W, Sun X, Li R, Feng C, Tan Y. TILs and anti-PD1 therapy: an alternative combination therapy for PDL1 negative metastatic cervical cancer. J Immunol Res 2020;2020:8345235.

103. Baldan V, Griffiths R, Hawkins RE, Gilham DE. Efficient and reproducible generation of tumour-infiltrating lymphocytes for renal cell carcinoma. Br J Cancer 2015;112:1510-8.

104. Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P, Guo ZS. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer 2022;21:196.

105. Chesney J, Puzanov I, Collichio F, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J Clin Oncol 2018;36:1658-67.

106. Han G, Stevens C, Cao Z, et al. Abstract 4022: PT2385, a novel HIF-2α antagonist, combines with checkpoint inhibitor antibodies to inhibit tumor growth in preclinical models by modulating myeloid cells and enhancing T cell infiltration. Cancer Research 2016;76:4022-4022.

107. Chen D, Sun X, Zhang X, Cao J. Inhibition of the CDK4/6-Cyclin D-Rb Pathway by ribociclib augments chemotherapy and immunotherapy in renal cell carcinoma. Biomed Res Int 2020;2020:9525207.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Chatwal MS, Chahoud J, Spiess PE. Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma. Cancer Drug Resist 2023;6:314-26. http://dx.doi.org/10.20517/cdr.2023.09

AMA Style

Chatwal MS, Chahoud J, Spiess PE. Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma. Cancer Drug Resistance. 2023; 6(2): 314-26. http://dx.doi.org/10.20517/cdr.2023.09

Chicago/Turabian Style

Chatwal, Monica Sheila, Jad Chahoud, Philippe E. Spiess. 2023. "Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma" Cancer Drug Resistance. 6, no.2: 314-26. http://dx.doi.org/10.20517/cdr.2023.09

ACS Style

Chatwal, MS.; Chahoud J.; Spiess PE. Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma. Cancer Drug Resist. 2023, 6, 314-26. http://dx.doi.org/10.20517/cdr.2023.09

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
612
Downloads
190
Citations
1
Comments
0
9

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 9 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/