Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Review ArticleReview
Open Access

The Potential Impact of New Drug and Therapeutic Modalities on Drug Resistance to Renal Cell Carcinoma

PEDRO CAETANO PINTO
Anticancer Research March 2023, 43 (3) 983-991; DOI: https://doi.org/10.21873/anticanres.16242
PEDRO CAETANO PINTO
Department of Urology, University Medicine Greifswald, Greifswald, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: pintop@uni-greifswald.de
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

The treatment of advanced renal cell carcinoma has been substantially improved by the introduction of targeted and immune therapies and their respective combinations. Unleashing the activity of the immune system opened a new and successful front in the fight against cancers. Despite the benefits, drug resistance phenomena and adverse side effects can compromise efficacy. The development of new modalities of drugs and therapies with properties and mechanisms of action that break away from conventional medicines was expanded in recent years. This perspective discusses the prospects of these innovative and highly potent novel treatments in overcoming much of the current issues surrounding the resistance to approved renal cell carcinoma treatments and the challenges facing their introduction.

Key Words:
  • Renal cell carcinoma
  • targeted therapy
  • immune therapy
  • new drug modalities
  • drug-resistance
  • review

Undoubtedly, the treatment of renal cell carcinoma (RCC) has seen major advancements in the XXI century. Surgical options (i.e., nephrectomies) remain the treatment of choice for localized RCC with what could be considered a good clinical prognosis. However, advanced, or metastatic cancer (mRCC) is associated with a poor outcome. The introduction of tyrosine-kinase inhibitors (TKI; i.e., targeted therapy) and immune checkpoint inhibitors (ICI), has significantly improved the clinical prognosis of mRCC patients. These drugs specifically target the angiogenic and immunosuppressive activity of tumors, two cellular mechanisms that are pivotal for the survival and proliferation of RCC (1).

TKI are small-molecule drugs that selectively block the propagation of signals through cellular pathways that depend on the activity of protein kinases. In the treatment of mRCC, inhibiting the signaling cascades of membrane receptor tyrosine kinases (RTK) can effectively suppress the de novo vascularization of tumors (2). TKI preclude the activity of vascular endothelial growth factor receptors (VEGFR) and platelet-derived growth factor receptors (PDGFR), in both tumor and neighboring endothelial cells. By preventing the sprouting and integration of capillary vessels into the tumor microenvironment, driven by pro-angiogenic factors, TKI effectively deny mRCC access to the bloodstream, hence hampering their survival and growth (3).

ICI are an antibody-based immunotherapy that blocks specific immune response regulators known as checkpoints (CP). CP receptors, or ligands, are expressed by cytotoxic T-cells and act as a molecular switch that can activate or suppress their activity. Cells express CP on their surface as components of intricate immune tolerance pathways (4). RCC cells stimulate the expression of CP antigens on their surface to negatively impact T-cells and dampen their immunological activity. ICI can bind to CP receptors and effectively block the communication between the tumor and T-cells, which trigger a cytotoxic response directed at the RCC cells. Programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are two CP receptors in T-cells targeted by approved ICI. ICI also target programmed cell death ligand 1 (PDL-1), which is the PD-1 counterpart expressed by cancer (5).

These two classes of drugs have transformed the therapeutic options for RCC, and as relatively new additions to the arsenal, it can be expected that our growing understanding of their pharmacodynamics and pharmacological interactions could lead to more effective treatment regimens. Despite their enhanced efficacy, TKI and ICI can fall short of their desired clinical outcomes thanks to drug resistance (6, 7). Characteristic to virtually any cancer type, mechanisms of drug resistance are allegedly inherent to the nature of malignant cells and their ability to survive and proliferate. In general, tumors can resist a drug by diminishing its cellular disposition, by re-writing their regulatory pathways, or by sub-population selection, where a population of drug-sensitive cells is depleted in benefit of a marginal non-responsive sub-population (8, 9).

This short perspective offers a brief overview of some of the new classes of drugs currently under development that could have a significant impact on the treatment of RCC. With the potential for enhanced efficacies combined with limited side effects, these new molecules burrow deeper into the molecular machinery of both cancer and immune cells and could push the treatment of RCC beyond TKI and ICI.

Resistance to Targeted and Immune Therapy

As small molecules, TKI are subjected to the activity of membrane-drug transporters that can dictate their intracellular concentrations and impact their efficacy (10). RCC-approved TKI including sunitinib, sorafenib, and axitinib are substrates for efflux transporters known to the expressed, and active, in RCC cells, namely P-glycoprotein (P-gp) and the Breast cancer resistance protein (BCRP) (11-13). Drug transport activity is believed to be a contributing factor to intrinsic drug resistance, alongside a reduction in apoptotic activity, where TKI therapy fails to yield any effect. This coincidental effect is still poorly understood and is a feature of advanced RCC (14). Acquired resistance to TKI, which reverts the initial benefits of the therapy, can manifest in different forms. Tumor cells can switch the activation of their angiogenic pathways and rely on networks that bi-pass the pro-angiogenic TKI-targets such as VEGFR and PDGFR. Indirectly, RCC can recruit proangiogenic inflammatory cells (e.g., monocytes) that can stabilize the tumor vasculature by increasing pericyte density. This is facilitated by endothelial exposure to the hypoxic environment characteristic of RCC (15). Moreover, RCC cells can promote epithelial-to-mesenchymal transition (EMT) in healthy epithelial cells to further progress tumor proliferation, opening new fronts for drug resistance in the tumor (16). TKI-resistance illustrates the extent that RCC cells are equipped to secure their angiogenic capabilities, and consequently their proliferation potential.

ICI are large biological molecules, with distinct pharmacokinetic properties relative to small-molecule drugs. With a serum half-life measured in weeks instead of hours, efficacy and acquired drug resistance are of much greater concern than drug disposition (17). Nivolumab, pembrolizumab, ipilimumab, and avelumab are ICI approved for the treatment of mRCC (18). The inflammatory and hypoxic nature of the RCC tumor microenvironment (TME) represents a double-edged sword when it comes to the activity of ICI. The RCC microenvironment is prolific in infiltrated immune cells, including lymphocytes, macrophages, and T-cells. These tumor-infiltrating immune cells (TIIC) are believed to be recruited to the TME by the secretion of an array of soluble immune factors. TIIC play a role in containing RCC progression by modulating cellular proliferation (19). However, RCC can rely on intricate immune regulatory machinery, involving the interplay of tumor cells and TIIC, to secrete a cytokine barrier that suppresses the activity of T-cells. Together with an extensive display of immune checkpoints on their membranes, RCC cells become stealthy and can bypass the immune response (20). ICI counteract this mechanism. Nonetheless, the activity of the cytokine barrier and its up-regulation in response to external stresses can inhibit large swaths of T-cells populations in the TME, rendering ICI ineffective. Several intrinsic pathways have been identified in both the regulation of cytokine and checkpoint expression. The mitogen-activated protein kinases (MAPK) and the PI3K/AKT/mTOR pathways are associated with the release of immunosuppressive cytokines (21, 22). Cyclin-dependent kinase 4 (CKD4) is a cell-cycle regulator whose deregulation is related to the proliferation of tumor cells. The inhibition of CKD4 activity has been associated with enhanced T-cell activation and respective anti-tumor activity; however, the mechanisms behind this interaction and its relation to immune checkpoint regulation remain unclear. The loss of major histocompatibility complex (MHC) antigens is another factor that diminishes the ability of T-cells to recognize RCC cells. This deregulation is associated with mutations in MHC genes in tumor cells, as their malignant phenotype progresses (22). Arguably, the TME plays a major role in the resistance to ICI. TIIC, tumor vasculature, immunosuppressive cytokines, and pro-angiogenic factors all interact in a matrix with varying levels of fibrosis and in itself a hypoxic environment that further impacts TIIC activities (23, 24). Overall, to be effective, cytotoxic T-cells have to navigate the intricacies of a hostile TME to recognize RCC cells actively masking their identity. Hence, ICI are a piece of a wider puzzle.

Both TKI and ICI are approved as front-line monotherapies for RCC, governed by stringent guidelines, with the selection of treatment and dosing regimens dictated by the clinical frameworks of individual patients. Unsurprisingly, TKI-ICI combination therapies were considered and developed from the inception of ICI clinical use, considering the added benefits of targeting RCC from both fronts (25). Nowadays, anti-VEGF-ICI combinations have proven more effective than TKI or ICI monotherapies in augmenting patient survival rates (26). By directing drugs against immune suppression and angiogenesis, two of the main survival mechanisms of RCC are engaged simultaneously (27). Clinical evidence also shows that TKI therapy initiated immediately after ICI treatment, in patient with poor response to immunotherapy, can be successful in stabilizing RCC progression (28, 29). Further, TKI with multi-kinase activity, in particular those targeting the MAPK, can also suppress pathways driving the secretion of immunosuppressive cytokines. In these combination therapies, TKI alter the dynamics of the TME, offering a line of sight for ICI-primed T-cells to engage and suppress RCC cells. The combination of two ICI – dual therapy – has also recently shown added benefit in the treatment of advanced RCC. This treatment doubles down on ICI activity, by targeting two separate checkpoint receptors in the T-cell using different ICI, hence amplifying the immune response towards the cancer cells (30).

The side effects of mono and combination ICI therapies are reasonably manageable throughout the treatment phases. However, dual ICI therapy can cause severe side effects in certain patients, with damage observed in the liver and lungs. These off-target toxic effects are comparable to an autoimmune reaction originating from a substantial up-regulation of cytotoxic immune responses (31). Therefore, understanding the patient-specific factor that leads to such adverse reactions will prove very beneficial in the selection of treatment. By improving mRCC treatment, TKI and ICI combination therapies can allegedly counteract certain aspects of the drug resistance experienced by monotherapies. Currently, the molecular inner works of these pharmacological interactions are not fully understood (22). Nonetheless, the development and implementation of TKI and ICI, and subsequent applications is a testimony to the advances made in tumor-immunology and our understanding of RCC pathophysiology. Current treatments under clinical development are mainly focusing on combinations of TKI and ICI to boost patient survival and potentially deliver curative effects (32); nonetheless, novel classes of drugs are being considered.

The Next Generation

As TKI and ICI have shown, drugs with a high target specificity are critical to enhancing efficacy and therefore, positive clinical outcomes. A significant proportion of the research portfolio of both established pharmaceutical enterprises as well as new ventures are now dedicated to the development of so-called new drug and therapy modalities (NDTM) (33-36). This non-official designation has acted as an umbrella term to describe mainly experimental molecules and therapies that have physiochemical, pharmacokinetics, and biological properties substantially different from traditional small molecules. Widely varying in their nature, NDTM are often large molecules, and even cells, that offer high target specificity with potential marginal toxicity (37). ICI fit under the designation of an NDTM, nevertheless, ICI are biologicals, a class of drugs approved for clinical use for over 20 years. NDTM is the term usually employed to describe classes of drugs and treatments considered experimental, also referred to as next-generation therapeutics.

The advent of multi-omics, automated workflows, and advanced in vitro models, to name a few tools now commonplace in biomedical research, has helped to fast-track the identification of new therapeutical targets as well the development of new drugs (38, 39). NDTM are being developed virtually across all therapeutic areas, some with a focus on conditions with no previous effective treatment (e.g., chronic kidney disease, Duchenne muscular dystrophy) (40). mRCC have also been the focus of much interest, with drugs aimed at further characteristic features of RCC.

Anti-cancer Vaccines

Cancer-based vaccine applications have been under development since the late XX century. These vaccines are designed to enhance the cancer-directed immune response by priming the immune-system to recognize neoantigens (41). The mutational turnover in developing tumors generates proteins unique to individual cancers – neoantigens – that are also expressed in cell surfaces. Neoantigens are identified as foreign elements by T-cells, contrary to MHC antigens, and can initiate an immune cascade. mRCC is a prominent cancer to employ a vaccine given its stable mutation rate and high proportion of neoantigens (42). Although pre-clinical studies have shown that vaccines encoding neoantigens can amplify T-cell mediated tumor cell death, poor tumor infiltration by immune cells, inhibition of T-cell by cytokines activity, and the turn-over of neoantigens are limiting factors that significantly hamper an efficacious response under normal circumstances (43).

To overcome these limitations, the combination of mRNA-vaccine and ICI is being considered. This approach aims at amplifying the activity of ICI by recruiting a larger population of T-cells to the TME. The potential benefits of this synergy include a prolonged ICI activity, where these CP blockers affect a high-density of T-cells, and overcoming poor ICI patient response by directing T-cells directly to the tumors, evading the immune-suppressive microenvironment. The deployment of neoantigen vaccines requires a personalized medicine strategy. The tumor-specific nature of antigens requires the vaccines to be patient-specific as well (44). An ongoing clinical trial (NCT02950766) is set to establish the effectiveness of a personalized vaccine – NeoVax – and Ipilimumab.

Despite the potential benefits, several challenges remain in the implementation of this type of co-therapy. Patient-specific vaccines will require a dedicated approach where tumor specimens are analyzed and vaccines designed to carry neoantigen epitopes, in an adequate timeframe. mRNA vaccines, similar to those brought into the spotlight during the Sars-CoV2 global pandemic, could also be employed to code neoantigens (45). Considering that neoantigen turnover can render vaccines ineffective, predictive tools can be used to maximize epitope identification. In the near future, clinical evidence will dictate the feasibility of such an approach, which can potentially represent another major step toward a curative treatment for mRCC.

Anti-sense Oligonucleotides

Anti-sense oligonucleotides (ASO) are naked, single-stranded, oligonucleotide sequences with chemical modifications to their backbones that confer these molecules high biological stability. Nowadays, tailored chemical modifications are used to imprint ASO with a large spectrum of properties. In general, ASO have limiting nuclease degradation, which together with their size, contributes to limited tissue penetration and distribution (46). On the other hand, ASO have slow elimination rates and tend to accumulate in the organs they reach, albeit to limited concentrations (47). ASO bind and promote the degradation of mRNA precluding protein translation or promoting alternative splicing, hence their ability to virtually target any protein of interest with very high specificity. It is believed that thanks to their accumulation and protracted degradation these molecules can exert their pharmacological effects for extended periods. Nowadays, a limited number of ASO have been approved for clinical use, namely neurodegenerative diseases (48). This is exemplified by the treatment of spinal muscular atrophy (SMA), a previously untreatable condition, with Nusinersen, an ASO that recovers the expression of a defective neuron motor protein by alternative splicing (49). Nonetheless, significant safety and efficacy considerations remain during ASO development, namely the long-term implications of tissue accumulation and potential off-target toxic effects.

Initial studies demonstrated that a phosphorothioate cross-linked ASO against VEGF mRNA was effective in reducing VEGF protein levels and limiting RCC cell proliferation in vitro. In RCC xenografts, the angiogenic activity and tumor size substantially diminished in nude mice treated with this ASO (50). Specifically targeting cellular proliferation via ASO has also been shown to halt proliferation in RCC xenografts. An anti- MKI67 (marker of proliferation Ki-67) ASO reduced the development of metastasis in mice, leaving tumor vessel density unchanged (51). Human trials of MG98, an anti-DNA-methyltransferase 1 (DNMT1) ASO had a good safety record. Hypermethylation is a characteristic of the RCC phenotype, with the expression of multiple genes truncated at the DNA by the activity of methylases. Hence, the rationale for targeting DNMT1. However, trial results proved inconclusive or fell short of the desired clinical endpoints (52, 53).

Recent advances in oligonucleotide technology and a far better understanding of the biological activity of ASO, their pharmacokinetics, dosing regimens, and routes of admiration still make ASO an appealing therapeutic approach, despite the limited clinical success of RCC therapy. Anti-angiogenic ASO have the potential to yield a sustained effect, given their low clearance and high stability, relative to a TKI. Currently, no significant study is seemingly ongoing to address RCC treatment. As an increasing number of ASO-based therapies are approved, this molecule is expected to achieve a foothold in several therapeutic areas once their applications become more commonplace (54). It would not be unsurprising to see ASO-RCC applications revisited, in particular those that could potentiate immune therapies. Combination therapies involving ICI and anti-proliferative/angiogenic ASO could prove beneficial. Sustained inhibition of tumor development may result in an enhanced immune response (55).

Anticalins and Bispecific Peptides

Anticalins are engineered low molecular weight proteins derived from Lipocalins, a family of small proteins, which consist of less than 200 amino acids (around 20 kDa). Lipocalins are extracellular soluble proteins with carrier functions, associated with cellular communication, and to date, a compressive picture of their regulatory functions is still elusive (56). Anticalins act as antibody mimetics and are taunted as alternatives to monoclonal antibodies. They display improved pharmacokinetic properties relative to antibodies, including efficient tissue penetration and renal clearance. These artificial proteins can also be designed to bind smaller molecules, such as steroids or metabolites, in addition to larger structural molecules (e.g., membrane receptor antigens). Moreover, the structure of anticalins is divergent from that of antibodies, with little to no homology. Therefore, immunogenic side-effects associated with antibody (i.e., biologicals) therapy could be mitigated (57).

The efficacy and safety of anticalins in the treatment of RCC have been shown in both pre-clinical and human trials. PRS-050 is a high-affinity anticalin that binds VEGF and antagonizes its interaction with VEGF receptors, effectively shutting any mitogenic-associated signaling. In humanized mice, PRS-050 reduced tumor microvascular density, with results comparable to bevacizumab, an anti-VEGF monoclonal antibody. Most interestingly, PRS-050 did not lead to platelet aggregation and thrombosis, which were observed after bevacizumab treatment (58). In RCC patients, this anticalin did not yield an immunogenic response and its administration resulted in undetectable levels of free VEGF in the subjects, with virtually all circulating VEGF bound to the anticalin (59).

Several anticalins are now in early to mid-stage development, with regulatory approval still in the future (60, 61). Anticalins have drawn much attention in the immune-therapy field, in particular with the development of bispecific peptides, which combine both an anticalin and an antibody. These conjugates harness the specificity of both classes of drugs to achieve an additive effect and boost the immune response. This is exemplified by PRS-352/S095025, which consists of an anticalin against OX40, a co-stimulatory immune checkpoint expressed in resting T-cells, and a monoclonal antibody targeting PD-L1, a suppressive immune checkpoint expressed by tumor cells. Results from recent pre-clinical studies show that this bispecific molecule triggers a strong PD-L1-dependant OX40 activation of T-cells, superior to that of PD-L1 antibodies (62).

With immune-therapy-based anticalins and bispecific peptides expected to reach late-stage development in the coming years, the true scope of the potential benefits relative to approved ICI, will remain to be seen. Nonetheless, they could represent a leap forward in RCC treatment if they can combine enhanced efficacy with limited side effects.

Proteolysis Targeting Chimeras

Proteolysis Targeting Chimeras (PROTAC) mediate the selective degradation of endogenous proteins by recruiting the activity of the ubiquitin-proteasome complex. PROTACS are heterobifunctional molecules, with two protein-binding domains conjugated by a molecular linker (63). A ligand domain that is designed to bind to a given protein of interest (POI) and a domain that engages with members of the E3 ubiquitin ligases family. By simultaneously binding to a POI and an E3 ligase, PROTACS trigger selective proteolysis by ubiquitination of the POI (64). These molecules are highly selective and highly effective in knocking-out their targets, in essence removing a protein rather than inhibiting its activity. This relatively new approach has gathered significant interest in cancer research. In particular, in the search for new therapies for cancer types with dated or ineffective treatments. PROTACS can virtually be designed to eliminate key proteins known to be over-expressed in cancer or intermediaries in tumor-specific processes. Moreover, these bifunctional molecules have physiochemical and pharmacokinetic properties more aligned with traditional small molecules, given their relatively reduced size (around 1,000 Da). This fact reduces the uncertainties surrounding drug distribution and elimination routes inherent to other new drug modality classes (65).

A promising example of an anti-cancer PROTAC is the development of ARV-110. This androgen receptor (AR) degrader is being tested in the treatment of another advanced urological cancer, metastatic castration-resistant prostate cancer (CRPC), in patients with progressive disease after conventional chemotherapy (NCT03888612) (66). AR activities govern prostate cancer growth and in CRPC its activity is believed to be dramatically deregulated. Pre-clinical data show that ARV-110 substantially reduces AR expression (i.e., >90%) and tumor growth in both wild-type and common AR genetic variants (63, 67).

To date, there are seemingly no PROTAC applications targeting RCC, either experimental or clinical. Nonetheless, as applications emerge, these molecules could find their way into the arsenal of drugs against RCC. A potential avenue is the impact of PROTAC on inhibiting the inflammatory response, by selectively excluding regulators of immune pathways (68, 69). This could prove beneficial in dampening the cytokine blockade that RCC relies on to keep immune cells at bay. Targeting abnormal hypoxia-induced factor (HIF) expression in RCC could also be addressed using PROTAC activity. HIF deregulation during the onset of RCC is derived from the loss of the von Hippel-Lindau protein (VHL), itself an E3 ligase. Bypassing VHL activity and promoting HIF degradation via an alternative ligase could help reduce the hypoxic nature of the TME and act concomitantly with other therapies (e.g., ICI).

Cellular Therapy

Innovative cellular therapies, which involve the administration of autologous or allogenic cells into a patient, have recently been brought to the spotlight including the approval of chimeric antigen receptor (CAR) T-cells. CAR T-cells are a type of immune therapy that involves the collection and isolation of T-cells from a patient, their expansion ex-vivo, the introduction of a CAR using genetic engineering tools, and the infusion of the transformed T-cell back into the patient (70). The CAR is at the heart of this therapy and consists of an artificial membrane receptor with the dual function of binding a specific cancer antigen and activating T-cells. CAR T-cells specifically recognize their target and trigger an immunogenic response directed at the intended tumor cells, which can include cytolytic activity and the secretion of immune factors that promote further CAR T-cell recruiting (71). Initially developed to treat blood cancers (e.g., acute lymphoblastic leukemia), CAR T-cells have shown dramatic efficacy, often leading to complete cancer remission, where preceding treatment failed (72). However, this therapy requires dedicated facilities, specialized personnel, and a tight manufacturing and delivery timeframe, far from an off-the-shelf solution. Preceding CAR T-cells therapy, patients undergo immunedepletion chemotherapy to facilitate the integration of the newly infused transformed T-cells.

The remarkable results of CAR T-cell therapy are derived from the potent and sustained immune response these cells can deliver. CAR-binding to their target cancer cells can also stimulate the proliferation of these transformed T-cells, hence amplifying their therapeutical range. A significant challenge with this type of therapy is, unsurprisingly, off-target immune effects. CAR-T cell can overreach their intended anti-tumor effect and induce severe toxicities derived from extensive cytokine release (73). Engineering CAR T-cells to avoid such side effects and strategies to mitigate them are focal areas in the development of new applications.

Several ongoing human trials are ongoing to determine the safety and potential efficacy of CAR T-cells therapies in the treatment of RCC (74). Despite their success in blood tumors, this cell therapy still has significant limitations while addressing solid tumors. Tumor trafficking and infiltration as well as the immunosuppressive TME are major hurdles in delivering CAR T-cells to their intended malignant targets (75). Antigen depletion and tumor off-target recognition are also important considerations in the CAR design. The heterogenicity of solid tumors could result in CAR T-cells targeting a specific sub-population of cancer cells. Nonetheless, this fact could be offset by an amplified immune response directed at the wider tumor. Solid tumors share antigens with healthy cells, albeit subjected to extensive variation, which can potentially lead to unintended tissue damage. CAR T-cells can be generated from distinct T-cell populations, which enables the deployment of transformed immune cells with different functions that can act in more than one front (76). Beyond direct cancer cell targeting, these cells can potentially alter the dynamics of the TME and suppress barriers to the recruitment of additional immune cells. Nephrotoxicity is a major concern with employing this approach to treat RCC. Toxic events derived from off-target immune activity could result in irreversible kidney damage leading to diminished renal function and ultimately renal failure. All these prospects are likely to remain unresolved in the foreseeable future. Nonetheless, the CAR T-cell field is fast progressing, with five therapies approved in the last five years (77). First-generation CAR T-cell therapies incorporate well-characterized immunestimulant antigens (e.g., CD19). A recent human trial (NCT03970382) has demonstrated the efficacy of this treatment using CAR designed to incorporate patient-specific mutated neoantigens. This approach relied on the analysis of samples from solid tumors and the generation of personalized libraries of predicted neoantigens of T-cell receptors (78). Several other important hurdles can be expected to be resolved in the coming years, opening CAR T-cell therapy to many different types of cancers.

Conclusion

Both current and future therapeutical approaches to mRCC are mainly centered around immune therapies. Harnessing the destructive capacities of immune cells has proven a winning strategy in the fight against many cancers. The introduction of new drugs or therapies for mRCC is still several years in the future, and in the short-term, the optimization of ICI treatments and the expansion of their combination therapies can be expected to further improve the clinical outcomes of RCC patients, by improving efficacies or mitigating drug resistance effects. Modulating the immune response can potentially become a frontier in the development of highly efficacious treatments. Coincidentally, an overreaching immune response is in a way opposite to drug resistance, where enhanced drug efficacy represents a problem. It is important to consider that the therapies discussed in this perspective have only been approved for clinical use for a few years or are in different stages of development. As our knowledge progresses, the framework for immune therapies will likely see major changes and breakthroughs by the end of this century. Personalized medicine approaches can effectively minimize both drug resistance phenomena and side effects by selecting the most optimal drug or therapy (or combination) given the nature of the tumor and other patient-specific considerations (such as the immune status).

With research spanning respiratory, immune, metabolic, and neurological diseases, among others, and beyond oncology, the next generation of medications can shift the current paradigms of cancer treatment and see the inception of curative mRCC therapies. NDTM allegedly hold the potential to solve many issues surrounding present mRCC drug resistance, but it has to be considered that different challenges will arise, as discussed in this perspective. Economic, social, and logistical factors will also greatly impact the introduction of treatments. Ultimately, strategic decisions weighing in all the factors, from efficacy to cost to availability, will eventually dictate which NDM and therapies will be implemented. With a remarkable number of new drugs and therapy modalities under development, that includes further classes of molecules (e.g., antibody-drug conjugates, bicyclic peptides, oligo-antibody conjugates) the prospects for a wide and effective choice of therapies available to patients is promising.

Footnotes

  • Conflicts of Interest

    The Author declares that there are no conflicts of interest in relation to this study.

  • Received January 5, 2023.
  • Revision received January 18, 2023.
  • Accepted January 19, 2023.
  • Copyright © 2023 The Author(s). Published by the International Institute of Anticancer Research.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Tran J and
    2. Ornstein MC
    : Clinical review on the management of metastatic renal cell carcinoma. JCO Oncol Pract 18(3): 187-196, 2022. PMID: 34529499. DOI: 10.1200/OP.21.00419
    OpenUrlCrossRefPubMed
  2. ↵
    1. Huang L,
    2. Jiang S and
    3. Shi Y
    : Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001-2020). J Hematol Oncol 13(1): 143, 2020. PMID: 33109256. DOI: 10.1186/s13045-020-00977-0
    OpenUrlCrossRefPubMed
  3. ↵
    1. Gotink KJ and
    2. Verheul HM
    : Anti-angiogenic tyrosine kinase inhibitors: what is their mechanism of action? Angiogenesis 13(1): 1-14, 2010. PMID: 20012482. DOI: 10.1007/s10456-009-9160-6
    OpenUrlCrossRefPubMed
  4. ↵
    1. Robert C
    : A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun 11(1): 3801, 2020. PMID: 32732879. DOI: 10.1038/s41467-020-17670-y
    OpenUrlCrossRefPubMed
  5. ↵
    1. Lavacchi D,
    2. Pellegrini E,
    3. Palmieri VE,
    4. Doni L,
    5. Mela MM,
    6. Di Maida F,
    7. Amedei A,
    8. Pillozzi S,
    9. Carini M and
    10. Antonuzzo L
    : Immune checkpoint inhibitors in the treatment of renal cancer: current state and future perspective. Int J Mol Sci 21(13): 4691, 2020. PMID: 32630154. DOI: 10.3390/ijms21134691
    OpenUrlCrossRefPubMed
  6. ↵
    1. Jiao Q,
    2. Bi L,
    3. Ren Y,
    4. Song S,
    5. Wang Q and
    6. Wang YS
    : Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol Cancer 17(1): 36, 2018. PMID: 29455664. DOI: 10.1186/s12943-018-0801-5
    OpenUrlCrossRefPubMed
  7. ↵
    1. Barrueto L,
    2. Caminero F,
    3. Cash L,
    4. Makris C,
    5. Lamichhane P and
    6. Deshmukh RR
    : Resistance to checkpoint inhibition in cancer immunotherapy. Transl Oncol 13(3): 100738, 2020. PMID: 32114384. DOI: 10.1016/j.tranon.2019.12.010
    OpenUrlCrossRefPubMed
  8. ↵
    1. Vasan N,
    2. Baselga J and
    3. Hyman DM
    : A view on drug resistance in cancer. Nature 575(7782): 299-309, 2019. PMID: 31723286. DOI: 10.1038/s41586-019-1730-1
    OpenUrlCrossRefPubMed
  9. ↵
    1. Emran TB,
    2. Shahriar A,
    3. Mahmud AR,
    4. Rahman T,
    5. Abir MH,
    6. Siddiquee MF,
    7. Ahmed H,
    8. Rahman N,
    9. Nainu F,
    10. Wahyudin E,
    11. Mitra S,
    12. Dhama K,
    13. Habiballah MM,
    14. Haque S,
    15. Islam A and
    16. Hassan MM
    : Multidrug resistance in cancer: Understanding molecular mechanisms, immunoprevention and therapeutic approaches. Front Oncol 12: 891652, 2022. PMID: 35814435. DOI: 10.3389/fonc.2022.891652
    OpenUrlCrossRefPubMed
  10. ↵
    1. Neul C,
    2. Schaeffeler E,
    3. Sparreboom A,
    4. Laufer S,
    5. Schwab M and
    6. Nies AT
    : Impact of membrane drug transporters on resistance to small-molecule tyrosine kinase inhibitors. Trends Pharmacol Sci 37(11): 904-932, 2016. PMID: 27659854. DOI: 10.1016/j.tips.2016.08.003
    OpenUrlCrossRefPubMed
  11. ↵
    1. Mao Q and
    2. Unadkat JD
    : Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport—an update. AAPS J 17(1): 65-82, 2015. PMID: 25236865. DOI: 10.1208/s12248-014-9668-6
    OpenUrlCrossRefPubMed
    1. Poller B,
    2. Iusuf D,
    3. Sparidans RW,
    4. Wagenaar E,
    5. Beijnen JH and
    6. Schinkel AH
    : Differential impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) on axitinib brain accumulation and oral plasma pharmacokinetics. Drug Metab Dispos 39(5): 729-735, 2011. PMID: 21282407. DOI: 10.1124/dmd.110.037317
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Lagas JS,
    2. van Waterschoot RA,
    3. Sparidans RW,
    4. Wagenaar E,
    5. Beijnen JH and
    6. Schinkel AH
    : Breast cancer resistance protein and P-glycoprotein limit sorafenib brain accumulation. Mol Cancer Ther 9(2): 319-326, 2010. PMID: 20103600. DOI: 10.1158/1535-7163.MCT-09-0663
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Kamli H,
    2. Li L and
    3. Gobe GC
    : Limitations to the therapeutic potential of tyrosine kinase inhibitors and alternative therapies for kidney cancer. Ochsner J 19(2): 138-151, 2019. PMID: 31258426. DOI: 10.31486/toj.18.0015
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Sharma R,
    2. Kadife E,
    3. Myers M,
    4. Kannourakis G,
    5. Prithviraj P and
    6. Ahmed N
    : Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma. J Exp Clin Cancer Res 40(1): 186, 2021. PMID: 34099013. DOI: 10.1186/s13046-021-01961-3
    OpenUrlCrossRefPubMed
  15. ↵
    1. Piva F,
    2. Giulietti M,
    3. Santoni M,
    4. Occhipinti G,
    5. Scarpelli M,
    6. Lopez-Beltran A,
    7. Cheng L,
    8. Principato G and
    9. Montironi R
    : Epithelial to mesenchymal transition in renal cell carcinoma: implications for cancer therapy. Mol Diagn Ther 20(2): 111-117, 2016. PMID: 26940073. DOI: 10.1007/s40291-016-0192-5
    OpenUrlCrossRefPubMed
  16. ↵
    1. Centanni M,
    2. Moes DJAR,
    3. Trocóniz IF,
    4. Ciccolini J and
    5. van Hasselt JGC
    : Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin Pharmacokinet 58(7): 835-857, 2019. PMID: 30815848. DOI: 10.1007/s40262-019-00748-2
    OpenUrlCrossRefPubMed
  17. ↵
    1. Yu EM,
    2. Linville L,
    3. Rosenthal M and
    4. Aragon-Ching JB
    : A contemporary review of immune checkpoint inhibitors in advanced clear cell renal cell carcinoma. Vaccines (Basel) 9(8): 919, 2021. PMID: 34452045. DOI: 10.3390/vaccines9080919
    OpenUrlCrossRefPubMed
  18. ↵
    1. Mier JW
    : The tumor microenvironment in renal cell cancer. Curr Opin Oncol 31(3): 194-199, 2019. PMID: 30985497. DOI: 10.1097/CCO.0000000000000512
    OpenUrlCrossRefPubMed
  19. ↵
    1. Guida M,
    2. Casamassima A,
    3. Monticelli G,
    4. Quaranta M and
    5. Colucci G
    : Basal cytokines profile in metastatic renal cell carcinoma patients treated with subcutaneous IL-2-based therapy compared with that of healthy donors. J Transl Med 5: 51, 2007. PMID: 17953739. DOI: 10.1186/1479-5876-5-51
    OpenUrlCrossRefPubMed
  20. ↵
    1. Karar J and
    2. Maity A
    : PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci 4: 51, 2011. PMID: 22144946. DOI: 10.3389/fnmol.2011.00051
    OpenUrlCrossRefPubMed
  21. ↵
    1. Ballesteros PÁ,
    2. Chamorro J,
    3. Román-Gil MS,
    4. Pozas J,
    5. Gómez Dos Santos V,
    6. Granados ÁR,
    7. Grande E,
    8. Alonso-Gordoa T and
    9. Molina-Cerrillo J
    : Molecular mechanisms of resistance to immunotherapy and antiangiogenic treatments in clear cell renal cell carcinoma. Cancers (Basel) 13(23): 5981, 2021. PMID: 34885091. DOI: 10.3390/cancers13235981
    OpenUrlCrossRefPubMed
  22. ↵
    1. Zhang Z,
    2. Li Q,
    3. Wang F,
    4. Ma B,
    5. Meng Y and
    6. Zhang Q
    : Identifying hypoxia characteristics to stratify prognosis and assess the tumor immune microenvironment in renal cell carcinoma. Front Genet 12: 606816, 2021. PMID: 34194463. DOI: 10.3389/fgene.2021.606816
    OpenUrlCrossRefPubMed
  23. ↵
    1. Pinto PC,
    2. Rönnau C,
    3. Burchardt M and
    4. Wolff I
    : Kidney cancer and chronic kidney disease: too close for comfort. Biomedicines 9(12): 1761, 2021. PMID: 34944574. DOI: 10.3390/biomedicines9121761
    OpenUrlCrossRefPubMed
  24. ↵
    1. Yang Y,
    2. Psutka SP,
    3. Parikh AB,
    4. Li M,
    5. Collier K,
    6. Miah A,
    7. Mori SV,
    8. Hinkley M,
    9. Tykodi SS,
    10. Hall E,
    11. Thompson JA and
    12. Yin M
    : Combining immune checkpoint inhibition plus tyrosine kinase inhibition as first and subsequent treatments for metastatic renal cell carcinoma. Cancer Med 11(16): 3106-3114, 2022. PMID: 35304832. DOI: 10.1002/cam4.4679
    OpenUrlCrossRefPubMed
  25. ↵
    1. Ueda K,
    2. Suekane S,
    3. Kurose H,
    4. Ito N,
    5. Ogasawara N,
    6. Hiroshige T,
    7. Chikui K,
    8. Ejima K,
    9. Uemura K,
    10. Nakiri M,
    11. Nishihara K,
    12. Matsuo M and
    13. Igawa T
    : Improved survival of real-world Japanese patients with advanced renal cell carcinoma treated with immuno-oncology combination therapy. Anticancer Res 42(9): 4573-4580, 2022. PMID: 36039432. DOI: 10.21873/anticanres.15960
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Jonasch E,
    2. Atkins MB,
    3. Chowdhury S and
    4. Mainwaring P
    : Combination of anti-angiogenics and checkpoint inhibitors for renal cell carcinoma: Is the whole greater than the sum of its parts? Cancers (Basel) 14(3): 644, 2022. PMID: 35158916. DOI: 10.3390/cancers14030644
    OpenUrlCrossRefPubMed
  27. ↵
    1. Kato T,
    2. Nagahara A,
    3. Kawamura N,
    4. Nakata W,
    5. Soda T,
    6. Matsuzaki K,
    7. Hatano K,
    8. Kawashima A,
    9. Ujike T,
    10. Imamura R,
    11. Nishimura K,
    12. Takada S,
    13. Tsujihata M,
    14. Yamaguchi S,
    15. Takao T,
    16. Nakai Y,
    17. Nakayama M,
    18. Nonomura N and
    19. Uemura M
    : Real-world outcomes of tyrosine kinase inhibitors immediately after immune checkpoint inhibitors in renal cell carcinoma. Anticancer Res 41(11): 5811-5816, 2021. PMID: 34732455. DOI: 10.21873/anticanres.15398
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Tachibana H,
    2. Inakawa T,
    3. Nemoto Y,
    4. Ishihara H,
    5. Fukuda H,
    6. Yoshida K,
    7. Iizuka J,
    8. Hashimoto Y,
    9. Tanabe K,
    10. Kondo T and
    11. Takagi T
    : Efficacy of cabozantinib for papillary compared with clear-cell renal cell carcinoma following immune checkpoint inhibitor treatment. Anticancer Res 42(6): 3151-3158, 2022. PMID: 35641290. DOI: 10.21873/anticanres.15804
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Schmidinger M,
    2. Shariat SF and
    3. Fajkovic H
    : Dual immune checkpoint inhibition in metastatic renal cell carcinoma: Editorial re.: Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced RCC: extended 4-year follow-up of the phase III CheckMate 214 trial. ESMO Open 6(1): 100035, 2021. PMID: 33421736. DOI: 10.1016/j.esmoop.2020.100035
    OpenUrlCrossRefPubMed
  30. ↵
    1. Martins F,
    2. Sofiya L,
    3. Sykiotis GP,
    4. Lamine F,
    5. Maillard M,
    6. Fraga M,
    7. Shabafrouz K,
    8. Ribi C,
    9. Cairoli A,
    10. Guex-Crosier Y,
    11. Kuntzer T,
    12. Michielin O,
    13. Peters S,
    14. Coukos G,
    15. Spertini F,
    16. Thompson JA and
    17. Obeid M
    : Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol 16(9): 563-580, 2019. PMID: 31092901. DOI: 10.1038/s41571-019-0218-0
    OpenUrlCrossRefPubMed
  31. ↵
    1. Braun DA,
    2. Bakouny Z,
    3. Hirsch L,
    4. Flippot R,
    5. Van Allen EM,
    6. Wu CJ and
    7. Choueiri TK
    : Beyond conventional immune-checkpoint inhibition - novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol 18(4): 199-214, 2021. PMID: 33437048. DOI: 10.1038/s41571-020-00455-z
    OpenUrlCrossRefPubMed
  32. ↵
    1. Blanco MJ and
    2. Gardinier KM
    : New chemical modalities and strategic thinking in early drug discovery. ACS Med Chem Lett 11(3): 228-231, 2020. PMID: 32184948. DOI: 10.1021/acsmedchemlett.9b00582
    OpenUrlCrossRefPubMed
    1. Kansy M and
    2. Caron G
    : New therapeutic modalities in drug discovery and development: Insights & opportunities. ADMET DMPK 9(4): 227-230, 2021. PMID: 35300374. DOI: 10.5599/admet.1209
    OpenUrlCrossRefPubMed
    1. Garg AD,
    2. Coulie PG,
    3. Van den Eynde BJ and
    4. Agostinis P
    : Integrating next-generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol 38(8): 577-593, 2017. PMID: 28610825. DOI: 10.1016/j.it.2017.05.006
    OpenUrlCrossRefPubMed
  33. ↵
    1. Sönmez MG and
    2. Sönmez LO
    : New treatment modalities with vaccine therapy in renal cell carcinoma. Urol Ann 11: 119-125, 2019. DOI: 10.4103/UA.UA_166_17
    OpenUrlCrossRefPubMed
  34. ↵
    1. Roth A
    : New drug modalities demand a refined preclinical safety assessment: a call for patient-relevant tissue models. Toxicol Sci 189(1): 5-6, 2022. PMID: 36017663. DOI: 10.1093/toxsci/kfac069
    OpenUrlCrossRefPubMed
  35. ↵
    1. Paul D,
    2. Sanap G,
    3. Shenoy S,
    4. Kalyane D,
    5. Kalia K and
    6. Tekade RK
    : Artificial intelligence in drug discovery and development. Drug Discov Today 26(1): 80-93, 2021. PMID: 33099022. DOI: 10.1016/j.drudis.2020.10.010
    OpenUrlCrossRefPubMed
  36. ↵
    1. Li S and
    2. Xia M
    : Review of high-content screening applications in toxicology. Arch Toxicol 93(12): 3387-3396, 2019. PMID: 31664499. DOI: 10.1007/s00204-019-02593-5
    OpenUrlCrossRefPubMed
  37. ↵
    1. Yamamura T,
    2. Horinouchi T,
    3. Adachi T,
    4. Terakawa M,
    5. Takaoka Y,
    6. Omachi K,
    7. Takasato M,
    8. Takaishi K,
    9. Shoji T,
    10. Onishi Y,
    11. Kanazawa Y,
    12. Koizumi M,
    13. Tomono Y,
    14. Sugano A,
    15. Shono A,
    16. Minamikawa S,
    17. Nagano C,
    18. Sakakibara N,
    19. Ishiko S,
    20. Aoto Y,
    21. Kamura M,
    22. Harita Y,
    23. Miura K,
    24. Kanda S,
    25. Morisada N,
    26. Rossanti R,
    27. Ye MJ,
    28. Nozu Y,
    29. Matsuo M,
    30. Kai H,
    31. Iijima K and
    32. Nozu K
    : Development of an exon skipping therapy for X-linked Alport syndrome with truncating variants in COL4A5. Nat Commun 11(1): 2777, 2020. PMID: 32488001. DOI: 10.1038/s41467-020-16605-x
    OpenUrlCrossRefPubMed
  38. ↵
    1. Donninger H,
    2. Li C,
    3. Eaton JW and
    4. Yaddanapudi K
    : Cancer vaccines: Promising therapeutics or an unattainable dream. Vaccines (Basel) 9(6): 668, 2021. PMID: 34207062. DOI: 10.3390/vaccines9060668
    OpenUrlCrossRefPubMed
  39. ↵
    1. Lin MJ,
    2. Svensson-Arvelund J,
    3. Lubitz GS,
    4. Marabelle A,
    5. Melero I,
    6. Brown BD and
    7. Brody JD
    : Cancer vaccines: the next immunotherapy frontier. Nat Cancer 3(8): 911-926, 2022. PMID: 35999309. DOI: 10.1038/s43018-022-00418-6
    OpenUrlCrossRefPubMed
  40. ↵
    1. Blass E and
    2. Ott PA
    : Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol 18(4): 215-229, 2021. PMID: 33473220. DOI: 10.1038/s41571-020-00460-2
    OpenUrlCrossRefPubMed
  41. ↵
    1. De Mattos-Arruda L,
    2. Vazquez M,
    3. Finotello F,
    4. Lepore R,
    5. Porta E,
    6. Hundal J,
    7. Amengual-Rigo P,
    8. Ng CKY,
    9. Valencia A,
    10. Carrillo J,
    11. Chan TA,
    12. Guallar V,
    13. McGranahan N,
    14. Blanco J and
    15. Griffith M
    : Neoantigen prediction and computational perspectives towards clinical benefit: recommendations from the ESMO Precision Medicine Working Group. Ann Oncol 31(8): 978-990, 2020. PMID: 32610166. DOI: 10.1016/j.annonc.2020.05.008
    OpenUrlCrossRefPubMed
  42. ↵
    1. Xu H,
    2. Zheng X,
    3. Zhang S,
    4. Yi X,
    5. Zhang T,
    6. Wei Q,
    7. Li H and
    8. Ai J
    : Tumor antigens and immune subtypes guided mRNA vaccine development for kidney renal clear cell carcinoma. Mol Cancer 20(1): 159, 2021. PMID: 34872567. DOI: 10.1186/s12943-021-01465-w
    OpenUrlCrossRefPubMed
  43. ↵
    1. Crooke ST
    : Molecular mechanisms of antisense oligonucleotides. Nucleic Acid Ther 27(2): 70-77, 2017. PMID: 28080221. DOI: 10.1089/nat.2016.0656
    OpenUrlCrossRefPubMed
  44. ↵
    1. Crooke ST,
    2. Wang S,
    3. Vickers TA,
    4. Shen W and
    5. Liang XH
    : Cellular uptake and trafficking of antisense oligonucleotides. Nat Biotechnol 35(3): 230-237, 2017. PMID: 28244996. DOI: 10.1038/nbt.3779
    OpenUrlCrossRefPubMed
  45. ↵
    1. Scharner J and
    2. Aznarez I
    : Clinical applications of single-stranded oligonucleotides: Current landscape of approved and in-development therapeutics. Mol Ther 29(2): 540-554, 2021. PMID: 33359792. DOI: 10.1016/j.ymthe.2020.12.022
    OpenUrlCrossRefPubMed
  46. ↵
    1. Chiriboga CA
    : Nusinersen for the treatment of spinal muscular atrophy. Expert Rev Neurother 17(10): 955-962, 2017. PMID: 28884620. DOI: 10.1080/14737175.2017.1364159
    OpenUrlCrossRefPubMed
  47. ↵
    1. Shi W and
    2. Siemann DW
    : Inhibition of renal cell carcinoma angiogenesis and growth by antisense oligonucleotides targeting vascular endothelial growth factor. Br J Cancer 87(1): 119-126, 2002. PMID: 12085267. DOI: 10.1038/sj.bjc.6600416
    OpenUrlCrossRefPubMed
  48. ↵
    1. Kausch I,
    2. Jiang H,
    3. Brocks C,
    4. Bruderek K,
    5. Krüger S,
    6. Sczakiel G,
    7. Jocham D and
    8. Böhle A
    : Ki-67-directed antisense therapy in an orthotopic renal cell carcinoma model. Eur Urol 46(1): 118-24; discussion 124-5, 2004. PMID: 15183557. DOI: 10.1016/j.eururo.2004.03.016
    OpenUrlCrossRefPubMed
  49. ↵
    1. Plummer R,
    2. Vidal L,
    3. Griffin M,
    4. Lesley M,
    5. de Bono J,
    6. Coulthard S,
    7. Sludden J,
    8. Siu LL,
    9. Chen EX,
    10. Oza AM,
    11. Reid GK,
    12. McLeod AR,
    13. Besterman JM,
    14. Lee C,
    15. Judson I,
    16. Calvert H and
    17. Boddy AV
    : Phase I study of MG98, an oligonucleotide antisense inhibitor of human DNA methyltransferase 1, given as a 7-day infusion in patients with advanced solid tumors. Clin Cancer Res 15(9): 3177-3183, 2009. PMID: 19383817. DOI: 10.1158/1078-0432.CCR-08-2859
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Winquist E,
    2. Knox J,
    3. Ayoub JP,
    4. Wood L,
    5. Wainman N,
    6. Reid GK,
    7. Pearce L,
    8. Shah A and
    9. Eisenhauer E
    : Phase II trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma: a National Cancer Institute of Canada Clinical Trials Group investigational new drug study. Invest New Drugs 24(2): 159-167, 2006. PMID: 16502349. DOI: 10.1007/s10637-006-5938-1
    OpenUrlCrossRefPubMed
  51. ↵
    1. Damase TR,
    2. Sukhovershin R,
    3. Boada C,
    4. Taraballi F,
    5. Pettigrew RI and
    6. Cooke JP
    : The limitless future of RNA therapeutics. Front Bioeng Biotechnol 9: 628137, 2021. PMID: 33816449. DOI: 10.3389/fbioe.2021.628137
    OpenUrlCrossRefPubMed
  52. ↵
    1. Xiong H,
    2. Veedu RN and
    3. Diermeier SD
    : Recent advances in oligonucleotide therapeutics in oncology. Int J Mol Sci 22(7): 3295, 2021. PMID: 33804856. DOI: 10.3390/ijms22073295
    OpenUrlCrossRefPubMed
  53. ↵
    1. Stopková R,
    2. Otčenášková T,
    3. Matějková T,
    4. Kuntová B and
    5. Stopka P
    : Biological roles of lipocalins in chemical communication, reproduction, and regulation of microbiota. Front Physiol 12: 740006, 2021. PMID: 34594242. DOI: 10.3389/fphys.2021.740006
    OpenUrlCrossRefPubMed
  54. ↵
    1. Schlehuber S and
    2. Skerra A
    : Anticalins as an alternative to antibody technology. Expert Opin Biol Ther 5(11): 1453-1462, 2005. PMID: 16255649. DOI: 10.1517/14712598.5.11.1453
    OpenUrlCrossRefPubMed
  55. ↵
    1. Gille H,
    2. Hülsmeyer M,
    3. Trentmann S,
    4. Matschiner G,
    5. Christian HJ,
    6. Meyer T,
    7. Amirkhosravi A,
    8. Audoly LP,
    9. Hohlbaum AM and
    10. Skerra A
    : Functional characterization of a VEGF-A-targeting Anticalin, prototype of a novel therapeutic human protein class. Angiogenesis 19(1): 79-94, 2016. PMID: 26650228. DOI: 10.1007/s10456-015-9490-5
    OpenUrlCrossRefPubMed
  56. ↵
    1. Mross K,
    2. Richly H,
    3. Fischer R,
    4. Scharr D,
    5. Büchert M,
    6. Stern A,
    7. Gille H,
    8. Audoly LP and
    9. Scheulen ME
    : First-in-human phase I study of PRS-050 (Angiocal), an Anticalin targeting and antagonizing VEGF-A, in patients with advanced solid tumors. PLoS One 8(12): e83232, 2013. PMID: 24349470. DOI: 10.1371/journal.pone.0083232
    OpenUrlCrossRefPubMed
  57. ↵
    1. Deuschle FC,
    2. Ilyukhina E and
    3. Skerra A
    : Anticalin® proteins: from bench to bedside. Expert Opin Biol Ther 21(4): 509-518, 2021. PMID: 33074019. DOI: 10.1080/14712598.2021.1839046
    OpenUrlCrossRefPubMed
  58. ↵
    1. Rothe C and
    2. Skerra A
    : Anticalin(®) proteins as therapeutic agents in human diseases. BioDrugs 32(3): 233-243, 2018. PMID: 29748739. DOI: 10.1007/s40259-018-0278-1
    OpenUrlCrossRefPubMed
  59. ↵
    1. Pattarini L,
    2. Pavlidou M,
    3. Morales A,
    4. Peper-gabriel J,
    5. Riviere M,
    6. Demarles D,
    7. Scholer-dahirel A,
    8. Blanc V and
    9. Olwill S
    : Abstract LB220: The anticalin-antibody bispecific PRS-352/S095025 strongly stimulates human CD4+ T cells in a PD-L1-dependent manner. Cancer Research 82(12_Suppl): LB220-LB220, 2022. DOI: 10.1158/1538-7445.am2022-lb220
    OpenUrlCrossRef
  60. ↵
    1. Sun X,
    2. Gao H,
    3. Yang Y,
    4. He M,
    5. Wu Y,
    6. Song Y,
    7. Tong Y and
    8. Rao Y
    : PROTACs: great opportunities for academia and industry. Signal Transduct Target Ther 4: 64, 2019. PMID: 31885879. DOI: 10.1038/s41392-019-0101-6
    OpenUrlCrossRefPubMed
  61. ↵
    1. Smith BE,
    2. Wang SL,
    3. Jaime-Figueroa S,
    4. Harbin A,
    5. Wang J,
    6. Hamman BD and
    7. Crews CM
    : Differential PROTAC substrate specificity dictated by orientation of recruited E3 ligase. Nat Commun 10(1): 131, 2019. PMID: 30631068. DOI: 10.1038/s41467-018-08027-7
    OpenUrlCrossRefPubMed
  62. ↵
    1. Neklesa TK,
    2. Winkler JD and
    3. Crews CM
    : Targeted protein degradation by PROTACs. Pharmacol Ther 174: 138-144, 2017. PMID: 28223226. DOI: 10.1016/j.pharmthera.2017.02.027
    OpenUrlCrossRefPubMed
  63. ↵
    1. Petrylak D,
    2. Gao X,
    3. Vogelzang N,
    4. Garfield M,
    5. Taylor I,
    6. Dougan moore M,
    7. Peck R and
    8. Burris H
    : First-in-human phase I study of ARV-110, an androgen receptor (AR) PROTAC degrader in patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC) following enzalutamide (ENZ) and/or abiraterone (ABI). Journal of Clinical Oncology 38(15_suppl): 3500-3500, 2020. DOI: 10.1200/jco.2020.38.15_suppl.3500
    OpenUrlCrossRef
  64. ↵
    1. Salami J,
    2. Alabi S,
    3. Willard RR,
    4. Vitale NJ,
    5. Wang J,
    6. Dong H,
    7. Jin M,
    8. McDonnell DP,
    9. Crew AP,
    10. Neklesa TK and
    11. Crews CM
    : Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance. Commun Biol 1: 100, 2018. PMID: 30271980. DOI: 10.1038/s42003-018-0105-8
    OpenUrlCrossRefPubMed
  65. ↵
    1. Bassi ZI,
    2. Fillmore MC,
    3. Miah AH,
    4. Chapman TD,
    5. Maller C,
    6. Roberts EJ,
    7. Davis LC,
    8. Lewis DE,
    9. Galwey NW,
    10. Waddington KE,
    11. Parravicini V,
    12. Macmillan-Jones AL,
    13. Gongora C,
    14. Humphreys PG,
    15. Churcher I,
    16. Prinjha RK and
    17. Tough DF
    : Modulating PCAF/GCN5 immune cell function through a PROTAC approach. ACS Chem Biol 13(10): 2862-2867, 2018. PMID: 30200762. DOI: 10.1021/acschembio.8b00705
    OpenUrlCrossRefPubMed
  66. ↵
    1. Komech E,
    2. Koltakova A,
    3. Shmidt E,
    4. Barinova A,
    5. Salnikova M,
    6. Minervina A,
    7. Korotaeva T,
    8. Lebedev Y and
    9. Zvyagin I
    : Pos0004 T-cell repertoire of synovial fluid in spondyloarthropathies exhibits hallmarks of hla-dependent clonal expansions and remains stable over 1.5 years. Ann Rheum Dis 80: 204, 2021. DOI: 10.1136/annrheumdis-2021-eular.3498
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Mehrabadi AZ,
    2. Ranjbar R,
    3. Farzanehpour M,
    4. Shahriary A,
    5. Dorostkar R,
    6. Hamidinejad MA and
    7. Ghaleh HEG
    : Therapeutic potential of CAR T cell in malignancies: A scoping review. Biomed Pharmacother 146: 112512, 2022. PMID: 34894519. DOI: 10.1016/j.biopha.2021.112512
    OpenUrlCrossRefPubMed
  68. ↵
    1. Alnefaie A,
    2. Albogami S,
    3. Asiri Y,
    4. Ahmad T,
    5. Alotaibi SS,
    6. Al-Sanea MM and
    7. Althobaiti H
    : Chimeric antigen receptor T-cells: an overview of concepts, applications, limitations, and proposed solutions. Front Bioeng Biotechnol 10: 797440, 2022. PMID: 35814023. DOI: 10.3389/fbioe.2022.797440
    OpenUrlCrossRefPubMed
  69. ↵
    1. Zhang X,
    2. Zhu L,
    3. Zhang H,
    4. Chen S and
    5. Xiao Y
    : CAR-T cell therapy in hematological malignancies: Current opportunities and challenges. Front Immunol 13: 927153, 2022. PMID: 35757715. DOI: 10.3389/fimmu.2022.927153
    OpenUrlCrossRefPubMed
  70. ↵
    1. Schubert ML,
    2. Schmitt M,
    3. Wang L,
    4. Ramos CA,
    5. Jordan K,
    6. Müller-Tidow C and
    7. Dreger P
    : Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol 32(1): 34-48, 2021. PMID: 33098993. DOI: 10.1016/j.annonc.2020.10.478
    OpenUrlCrossRefPubMed
  71. ↵
    1. Schepisi G,
    2. Conteduca V,
    3. Casadei C,
    4. Gurioli G,
    5. Rossi L,
    6. Gallà V,
    7. Cursano MC,
    8. Brighi N,
    9. Lolli C,
    10. Menna C,
    11. Farolfi A,
    12. Burgio SL,
    13. Altavilla A,
    14. Martinelli G and
    15. De Giorgi U
    : Potential application of chimeric antigen receptor (CAR)-T cell therapy in renal cell tumors. Front Oncol 10: 565857, 2020. PMID: 33072597. DOI: 10.3389/fonc.2020.565857
    OpenUrlCrossRefPubMed
  72. ↵
    1. Sterner RC and
    2. Sterner RM
    : CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J 11(4): 69, 2021. PMID: 33824268. DOI: 10.1038/s41408-021-00459-7
    OpenUrlCrossRefPubMed
  73. ↵
    1. Zhang H,
    2. Zhao P and
    3. Huang H
    : Engineering better chimeric antigen receptor T cells. Exp Hematol Oncol 9(1): 34, 2020. PMID: 33292660. DOI: 10.1186/s40164-020-00190-2
    OpenUrlCrossRefPubMed
  74. ↵
    1. Qu C,
    2. Zhang H,
    3. Cao H,
    4. Tang L,
    5. Mo H,
    6. Liu F,
    7. Zhang L,
    8. Yi Z,
    9. Long L,
    10. Yan L,
    11. Wang Z,
    12. Zhang N,
    13. Luo P,
    14. Zhang J,
    15. Liu Z,
    16. Ye W,
    17. Liu Z and
    18. Cheng Q
    : Tumor buster - where will the CAR-T cell therapy ‘missile’ go? Mol Cancer 21(1): 201, 2022. PMID: 36261831. DOI: 10.1186/s12943-022-01669-8
    OpenUrlCrossRefPubMed
  75. ↵
    1. Foy SP,
    2. Jacoby K,
    3. Bota DA,
    4. Hunter T,
    5. Pan Z,
    6. Stawiski E,
    7. Ma Y,
    8. Lu W,
    9. Peng S,
    10. Wang CL,
    11. Yuen B,
    12. Dalmas O,
    13. Heeringa K,
    14. Sennino B,
    15. Conroy A,
    16. Bethune MT,
    17. Mende I,
    18. White W,
    19. Kukreja M,
    20. Gunturu S,
    21. Humphrey E,
    22. Hussaini A,
    23. An D,
    24. Litterman AJ,
    25. Quach BB,
    26. Ng AHC,
    27. Lu Y,
    28. Smith C,
    29. Campbell KM,
    30. Anaya D,
    31. Skrdlant L,
    32. Huang EY,
    33. Mendoza V,
    34. Mathur J,
    35. Dengler L,
    36. Purandare B,
    37. Moot R,
    38. Yi MC,
    39. Funke R,
    40. Sibley A,
    41. Stallings-Schmitt T,
    42. Oh DY,
    43. Chmielowski B,
    44. Abedi M,
    45. Yuan Y,
    46. Sosman JA,
    47. Lee SM,
    48. Schoenfeld AJ,
    49. Baltimore D,
    50. Heath JR,
    51. Franzusoff A,
    52. Ribas A,
    53. Rao AV and
    54. Mandl SJ
    : Non-viral precision T cell receptor replacement for personalized cell therapy. Nature: 1-10, 2022. PMID: 36356599. DOI: 10.1038/s41586-022-05531-1
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 43 (3)
Anticancer Research
Vol. 43, Issue 3
March 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The Potential Impact of New Drug and Therapeutic Modalities on Drug Resistance to Renal Cell Carcinoma
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
The Potential Impact of New Drug and Therapeutic Modalities on Drug Resistance to Renal Cell Carcinoma
PEDRO CAETANO PINTO
Anticancer Research Mar 2023, 43 (3) 983-991; DOI: 10.21873/anticanres.16242

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
The Potential Impact of New Drug and Therapeutic Modalities on Drug Resistance to Renal Cell Carcinoma
PEDRO CAETANO PINTO
Anticancer Research Mar 2023, 43 (3) 983-991; DOI: 10.21873/anticanres.16242
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Resistance to Targeted and Immune Therapy
    • The Next Generation
    • Anti-cancer Vaccines
    • Anti-sense Oligonucleotides
    • Anticalins and Bispecific Peptides
    • Proteolysis Targeting Chimeras
    • Cellular Therapy
    • Conclusion
    • Footnotes
    • References
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Renal Cell Cancer - Insights in Drug Resistance Mechanisms
  • Google Scholar

More in this TOC Section

  • Cancer Stem Cells in Glioblastoma: The Role of the mTOR Pathway
  • Immunotherapy and Chemotherapy for Advanced or Recurrent Endometrial Carcinoma
  • Cannabinoids as Potential Therapeutic Agents in the Treatment of Pancreatic Cancer
Show more Review

Similar Articles

Keywords

  • Renal cell carcinoma
  • targeted therapy
  • Immune therapy
  • new drug modalities
  • drug-resistance
  • review
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire