Skip to main content

Evolution of cell therapy for renal cell carcinoma

Abstract

Treatment for renal cell carcinoma (RCC) has improved dramatically over the last decade, shifting from high-dose cytokine therapy in combination with surgical resection of tumors to targeted therapy, immunotherapy, and combination therapies. However, curative treatment, particularly for advanced-stage disease, remains rare. Cell therapy as a “living drug” has achieved hematological malignancy cures with a high response rate, and significant research efforts have been made to facilitate its translation to solid tumors. Herein, we overview the cellular therapies for RCC focusing on allogeneic hematopoietic stem cell transplantation, T cell receptor gene-modified T cells, chimeric antigen receptor (CAR) T cells, CAR natural killer (NK) cells, lymphokine-activated killer (LAK) cells, γδ T cells, and dendritic cell vaccination. We have also included perspectives for using other recent approaches, such as CAR macrophages, dendritic cell-cytokine induced killer cells and regulatory CAR-T cells to shed light on preclinical development of cell therapy and advancing cell therapy into clinic to achieve cures for RCC.

Introduction

Renal cell carcinoma (RCC) represents approximately 3% of adult cancers and has been widely recognized as a heterogeneous disease encompassing different subtypes [1]. About 70–80% of RCC cases have clear cell histology (ccRCC) [1, 2], which has a relatively poor prognosis, with 30% of patients developing metastatic ccRCC. ccRCC is characterized by inactivation of the von-Hippel-Lindau (VHL) gene. The dysfunction of VHL leads to hypoxia-inducible factor (HIF) hyperactivation, resulting in overexpression of many downstream genes involved in angiogenesis, metabolism, and cell-cycle regulation, which represent critical therapeutic targets for patients with ccRCC [3, 4] (Fig. 1). Papillary renal cell carcinoma (pRCC) is the second most common kidney cancer, accounting for 15% of kidney cancers [5]. pRCC has two major types, type 1 and type 2, categorized by the presence or absence of prominent nucleoli, respectively. 80% of type 1 pRCC have an alteration in MET proto-oncogene (MET) genetic sequence or copy number, making MET a potential target pathway [6].

Fig. 1
figure 1

Hypoxia-inducible factor (HIF) pathway. Under normoxia, von-Hippel-Lindau (VHL) binds to HIF1α, leading to HIF1α degradation. While under hypoxia or in clear cell renal cell carcinoma (ccRCC), the dysfunction of VHL results in HIF1α-HIF1β dimer formation and HIF hyperactivation, resulting in overexpression of many downstream genes involved in angiogenesis, metabolism, and cell-cycle regulation, including carbonic anhydrase IX (CAIX), platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF). Created with BioRender.com

Treatment of RCC has improved dramatically over the last decade, shifting from high-dose cytokine therapy in combination with surgical resection of tumors to extensive stage-dependent therapy regimens based on targeted therapies, highlighting the efficiency of antiangiogenic agents that targets the vascular endothelial growth factor (VEGF) pathway [7, 8] and immune checkpoint inhibitors (ICIs). In the European society for medical oncology (ESMO) clinical practice guideline, dual immunotherapy (ICI-ICI) or a combination of immunotherapy and antiangiogenic tyrosine kinase inhibitor (ICI-TKI) are recommended as the main first-line therapies for patients with advanced ccRCC. These therapies, their category, targets and comparative efficiency were summarized in Table 1 [9,10,11]. Despite all of these advances, curative treatment for advanced RCC remains rare [12] and the evolution of cell therapies, summarized in Fig. 2 and detailed in the text, represent a promising area for these cases. Moreover, Tables 2 and 3 summarize the results from the main pre-clinical and clinical adoptive cell therapies for RCC described below.

Table 1 Current targeted molecular agents recommended for the treatment of advanced/metastatic ccRCC
Fig. 2
figure 2

Cell therapies for renal cell carcinoma. A Allogeneic hematopoietic stem cell transplantation, presenting acute and chronic graft versus host disease (GvHD) with high transplant-related mortality; B Interleukin-2 (IL-2) and IL-2 receptor (IL-2R) variants, where mutants allowing only hIL-2Rβ activation on adoptive T cells but not the hIL-2Rα prevent T cells differentiation into Tregs and induce expansion of effector T cells against the tumor; C T cell receptor gene-modified T cells (TCR-T), which is consisted of a chimeric switch receptor (CSR) combining a ligand-binding domain (e.g., PD-1) with an alternative signaling domain (CD28) able to prevent T cell exhaustion and improve expansion; D Chimeric antigen receptor (CAR) T cells, and E CAR natural killer (NK) cells, both expressing engineered receptors designed against one or more antigens allowing immune cells activation against the tumor; F Lymphokine-activated killer (LAK) cells, that are T and NK cells, mainly expressing NK markers. Despite some efficiency against RCC metastasis, LAK cell therapy has been replaced by more specific cell-based immunotherapies; G γδ T cells, a subset of T cells with non-MHC‐restricted cytotoxic activity. These cells can be engineered for adoptive therapies, and the PD-1/PD-L1 axis does not abrogate their function; H Dendritic cell vaccination, where autologous DCs pulsed with peptides or tumor lysate-derived proteins can stimulate the generation of cytotoxic T cells in cancer patients. Created with BioRender.com

Table 2 Preclinical studies of cell therapies for RCC treatment
Table 3 Clinical studies of cell therapies for RCC treatment

Preclinical and clinical applications of adoptive cell therapies for RCC: acquired knowledge and perspectives

Allogeneic hematopoietic stem cell transplantation

Allogenic hematopoietic stem cell (HSC) transplantation, named allo-HSCT, has succeeded against primary and metastatic RCC due to an immune graft-versus-tumor (GvT) effect [49]; however, graft-versus-host disease (GvHD) challenges its application. Bregni et al. summarized the findings of 14 clinical studies on HSC transplantation for RCC patients, showing response rates ranging from 0 to 71% [49]. In half of the patients, acute and chronic GvHD were present, with transplant-related mortality observed in 0–33% of patients [49, 50]. These two post-transplant events, GvT and GvHD, are considered “two faces of the same coin” [51]. In the treatment of leukemia, administration of short-term immunosuppressive agents, camphorsulfonic acid (CsA) or methotrexate (MTX) at 10 mg/week, post allo-HSCT has been shown to significantly reduce GvHD without an appreciable impact on the GvT [52], which is promising to be translated to tame the immune overactivation in RCC treatment.

Alkylating agents alone or with total body irradiation (TBI) are the most used agents for myeloablative conditioning in clinical allo-HSCT. However, they are very cytotoxic and frequently induce and amplify GvHD. Some recent alternative approaches have been explored in a review paper by Saha and Blazar [53]. The use of radioimmunoconjugates in the conditioning has shown clinical benefits, with decreased relapse and no changes in transplant-related mortality. One interesting strategy allowed transplantation tolerance to fully major histocompatibility complex (MHC) mismatched donor marrow using nonmyeloablative preconditioning with busulfan or irradiation with lower systemic doses combined with costimulatory pathway blockade and/or immunosuppressive drugs. In preclinical setting, the use of antibodies and immunoconjugates as preconditioning methods for allo-HSCT have been tested, with the advantage of more specific targeting of hematopoietic stem and immune cells, reducing global toxicity. These antibodies can be used alone in high doses or in lower doses associated with traditional conditioning agents in reduced doses, inducing lower toxicity compared with the traditional methods and achieving comparable allo-engraftment [53].

Interleukin-2 (IL-2) and IL-2 receptor variants applied in adoptive cell therapies

Despite the importance of IL-2 against metastatic cancer [54], this cytokine has various limitations, including its dual action on regulatory T cells (Tregs) and effector T cells. IL-2 can induce CD4+ CD25+ Foxp3+ Treg expansion, especially in the immunosuppressive ICOS+ population [55]. In 2021, Motzer et al. engineered an orthogonal IL-2/IL-2R pair that do not cross react with their wild-type counterparts to specifically activate adoptively transferred T cells [56]. This method only activates the hIL-2Rβ on adoptive T cells but not the hIL-2Rα (CD25), preventing T cells differentiation into Tregs, thus ameliorating complications associated with conventional IL-2 therapy. Several IL-2 fusion proteins and variants are showing considerable promise in pre-clinical and clinical trials. PD-1-IL-2v, a new immunocytokine that binds PD-1 and IL-2Rβγ in cis, recovers the ability to differentiate PD-1+ T cell factor 1 (TCF-1)+ stem-like CD8+ T cells [57], which are critical for the success of PD-1 blockade based immunotherapies [58,59,60,61]. Another fusion protein, ALKS-4230, consisting of IL-2 and the extracellular domain of IL-2Rα, inhibiting interaction with IL-2Rα and preferentially binding to IL-2Rβγ [62, 63], is currently being investigated in Phase I/II clinical trial (NCT02799095). GI-102 has a CD80-ectodomain N terminal and IL-2Rβ targeted C terminal domain, is being tested in Phase I/II trials (NCT05824975) as a single agent. An IL-2 mutant (F42A, Y45A and L72G), fused to an anti-fibroblast activation protein mAb 4B9 [64], named FAP-IL-2v (simlukafusp alfa) is in Phase I/II clinical trials for RCC and other various carcinomas through combination therapies with trastuzumab, cetuximab, bevacizumab, and pembrolizumab (NCT02627274, NCT03193190, NCT03063762, NCT03386721, and NCT03875079). Cergutuzumab, sharing the same IL-2 mutein, fused with a carcinoembryonic antigen (CEA) targeted antibody [65], is also being tested in the clinic as a single therapeutic (NCT02004106), or in combination with atezolizumab (NCT02350673) for the treatment of metastatic solid tumors.

T cell receptor gene-modified T cells

T cell receptor-engineered T cells targeting 5T4 Tumor antigen

Trophoblast glycoprotein (TPBG, 5T4) is a heavily N-glycosylated antigen, a member of the family of proteins containing leucine-rich repeats (LRRs). The elevated prevalence of this protein is seen in human trophoblasts and across many primary and metastatic cancers, and its expression is restricted in adult systemic tissues [66]. The overexpression of 5T4 in ovarian, gastric, pancreatic, renal, and colorectal malignancies has been associated with poor prognosis and reduced OS of patients [22, 67,68,69,70]. Griffiths et al. have shown 5T4 positivity on three RCC cell lines (2220R, 2245R, and 2246R) and 20 RCC patient samples [22]. This study also proved that 5T4-targeted T cells could elicit cytotoxic activity on RCC tumor cells, paving the way for exploring T cell-based therapeutic strategies targeting 5T4-enriched tumors. A first line of 5T4 redirected CD8+ T cells selectively eliminated 5T4+ kidney, breast, and colorectal cancer cells in vitro [23].

Chimeric PD-1:28 receptor

One of the main obstacles to achieving a durable antitumor response with T-cell-mediated therapy is the exhaustion/inactivation of the T cells due to immunosuppressive factors in the tumor microenvironment (TME) [71] (Fig. 3). Multiple workarounds for this problem have been proposed and executed, including constructing CAR-T cells with various payloads to prevent or revert T cell exhaustion. A chimeric switch receptor (CSR) combines a ligand-binding domain with an alternative signaling domain. Several such constructs have been used for enhancing and altering signaling pathways in adoptive cell therapy (ACT) [72,73,74,75]. The PD-1:28 CSR is a fusion of the extracellular domain of PD-1 and the signaling domain of the costimulatory receptor CD28. In highly immunosuppressive TMEs, activation through the costimulatory domain has been shown to prevent T cell exhaustion and improve expansion [76]. At the same time, blocking immune checkpoint signals such as the PD-1/PD-L1 interaction is crucial to avoid T cell inactivation and restore effector function [77]. The PD-1:28 CSR restored effector functions essential for tumor cytotoxicity, preventing Th2 polarization and blocked PD-1 function in cell lines derived from various malignancies, including RCC, squamous cell carcinoma, melanoma, and glioblastoma [24]. CAR-T cells expressing this chimeric receptor reduced the susceptibility to tumor-induced hypofunction in various solid malignancies compared to CAR-T cells that do not carry the receptor [78]. The results from such studies and others are encouraging and point to the potential of chimeric receptors to increase the efficacy of immunotherapeutic strategies.

Fig. 3
figure 3

Immunosuppressive tumor microenvironment (TME). The TME is comprised of tumor cells, stroma, and exhausted immune cells, including dendritic cells (DCs), T cells, NK cells, B cells and macrophages. Created with BioRender.com

Chimeric antigen receptor (CAR) T cells

CARs are genetically engineered receptors designed against one or more antigens and expressed on immune cells (Fig. 4). Their extracellular domain is usually an scFv capable of specifically binding antigens overexpressed at the surface of tumor cells, linked to a hinge domain (e.g., CD8, CD28, IgG1, or IgG4) and a transmembrane domain (e.g., CD28, 4-1BB or CD8), fused to one or more variable intracellular costimulatory domains (e.g., CD28, 4-1BB, or OX40 – not present in first generation CARs) and a CD3ζ activation domain, leading to full T cell activation after contact with the target antigen [27]. CAR-T cell therapy has led to significant advances in cancer cell immunotherapy, resulting in great success in treating hematological malignancies [79], with recent advances for solid tumor treatments [80], including RCC [81].

Fig. 4
figure 4

Chimeric antigen receptor (CAR) structure. The CAR consists of an extracellular-hinge region, usually based on a single chain viable fragment (scFv), linked to a transmembrane region and intracellular costimulatory (e.g., 4-1BB, CD28) and stimulatory domains (CD3z). The CAR can be expressed in different immune cells (e.g., T cells, NK cells, Macrophages) and recognize specific tumor antigens independently of the major histocompatibility complex (MHC) presentation. This image summarizes all the molecules that are currently being evaluated as potential targets for CAR in RCC: carbonic anhydrase IX (CAIX), CD70, tyrosine-protein kinase Met (c-Met), mucin-1 (MUC1), receptor orphan tyrosine kinase receptor 2 (ROR2), epidermal growth factor receptor (EGFR). Created with BioRender.com

Lessons learned from the first CAR-T cell therapy (G250)

Carbonic anhydrases (CAs) are metalloenzymes that catalyze the reversible hydration of carbon dioxide to bicarbonate and hydrogen ions, controlling the pH of different body compartments. CAIX is a CA isoform overexpressed in hypoxic conditions and constitutively expressed in the majority of cases of the most common type of RCC, the ccRCC, due to mutations of the VHL gene, CAIX is being used as a ccRCC biomarker and has been shown to have prognostic implications [82, 83].

The first anti-CAIX CAR-T cells clinically tested for the treatment of metastatic ccRCC were CD4TM-γ expressing the first-generation CAR based on murine monoclonal antibody (mAb) G250 applied in high sequential doses in combination with IL-2. Patients showed disease progression, and liver toxicity was attributed to a specific scFv CAIX G250 attack against bile duct epithelial cells [41]. In a subsequent clinical protocol with the same CAR-T cells, the authors extended their observations to attenuate the on-target off-tumor cytotoxicity against bile duct epithelial cells, by testing the use of chimeric G250 mAb as a pre-treatment strategy. However, no effective antitumor response was obtained; but the results suggested that using a human scFv anti-CAIX and other generations of CAR vectors could enhance their efficiency [42].

Carbonic anhydrase IX-specific CAR-T cells alone or in association

In preclinical studies, Lo et al. tested two main constructs preclinically: a 1st generation humanized anti-CAIX scFv G36 CAR with only CD3ζ activation domain (G36-CD3ζ); and a 2nd generation G36 CAR containing the costimulatory cassette CD28 (G36-CD28ζ). The G36-CD28ζ CAR-T cells exhibited superiority compared to the first-generation equivalent CAR, with partial tumor regression observed in 67% of the cases [25]. To improve the efficacy of G36-CD28ζ against ccRCC, Suarez et al. tested armored anti-CAIX G36 CAR-T cells secreting anti-programmed cell death ligand-1 (PD-L1) IgG1 or IgG4 mAb, which target PD-L1 positive ccRCC cells and block the PD-1/PD-L1 pathway. These CAR-T cells, in a dose equivalent to 108 CAR CD8 T cells/kg, were able to reverse T cell exhaustion, decreasing 30–70% expression of exhaustion markers in tumor-infiltrating lymphocytes (TILs). There was an improvement in antitumor activity, with a robust decrease in tumor weight by 50–80% compared to a parental anti-CAIX CAR-T cell [26]. The anti-CAIX G36 CAR CD28 or BBζ peripheral blood mononuclear cells (PBMCs) secreting anti-PD-L1 IgG4 were recently tested in low doses in a similar orthotopic ccRCC model, equivalent to  107 CAR-T PBMCs/kg, and the CD28-based construction was superior to BBζ when immune checkpoint blockade via PD-L1 was used in combination. This treatment reduced tumor weight by 60%, avoided the occurrence of metastasis, and showed a 50% reduction in the co-expression of T cell exhaustion markers on viable TILs. The authors suggest a synergistic effect of CD28-based CARs with a PD-L1 blockade concerning the reversal of immunosuppression. No hepatotoxicity or nephrotoxicity was observed [84].

Recently, Wang et al. compared different anti-CAIX G36 CAR constructs (BBζ, 28ζ, 28BBζ) and CD4/CD8 cell compositions in an orthotopic mouse model bearing human ccRCC. The results showed that anti-CAIX G36 BBζ CAR-T cells with a CD4/CD8 ratio of 2:1 demonstrated complete tumor regression and exhibited decreased exhaustion genes revealed by single-cell RNA sequencing (scRNAseq) [28].

The combination of the TKI sunitinib with another anti-CAIX CAR-T containing a CD8α transmembrane domain and the intracellular domains of 4-1BB and CD3ζ in a subcutaneous mouse lung metastasis model of human RCC has led to the survival of all mice at the end of the experiment (day 60), with decreased tumor burden compared to anti-CAIX CAR-T cells or sunitinib alone. Sunitinib enhanced the proliferation and infiltration of CAIX-CAR-T cells, with decreased frequency of myeloid-derived suppressor cells in tumors [29]. Another study combined an oncolytic adenovirus (OAV) carrying decorin with CAIX-targeted CAR consisting of an scFv, a CD8α transmembrane domain, and 4-1BB/ CD3 zeta signaling intracellular domains. The CAIX CAR-T and OAV-Decorin (OAV-DEC) construct proved to have a significant specific killing effect on CAIX-positive RCC cells in vitro and displayed synergistic antitumor effects. In a subcutaneous xenograft model of human RCC, the combination OAV-DEC + CAIX-CAR-T reduced the tumor volume by 87%, while OAV + CAIX-CAR-T reduced the tumor volume by 54% [39].

CD70 targeted CAR-T cells

CD70 is a membrane protein that binds to the tumor necrosis factor receptor (TNFR) known as CD27. Hematologic malignancies and solid tumors, including about 40% of RCC cases, may constitutively express CD70 in high levels [85, 86]. Anti-CD70 CAR-T cells have shown an antitumor effect on RCC in preclinical studies leading to lysis of target cells and increased levels of IL-2, TNF-α, and IFN-γ released by CAR-T cells. Also, in the same study, the addition of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib (OLA) associated with CD70 CAR-T showed an increase in CD8 + infiltration and a better survival rate among tumor-bearing mice [30]. Another preclinical study evaluating allogeneic cells identified CD70 CAR-T binding epitopes that exhibited important antitumor activity against RCC cell lines and in a xenograft mouse model of RCC derived from patients [31]. Moreover, we developed dual-targeted anti-CAIX/CD70 CAR-T cells to enlarge the target cell population and mitigate tumor heterogeneity [87, 88].

Clinical trials are in progress to investigate the safety and efficacy of CD70-targeted CAR-T cells (NCT02830724) against several solid tumors including RCC. The food and drug administration (FDA) has granted fast-track designation to the Phase I (TRAVERSE) study to investigate the efficacy of an allogeneic CAR-T cell therapy that targets CD70 (NCT04696731). A Phase I multicenter trial (COBALT-RCC) of CRISPR-(CTX-130) in fourteen subjects with stage IV CD70 positive ccRCC, from which six presented refractory disease, has led to 8% durable CR (18+ months) and 69.2% SD. The treatment induced an acceptable safety profile, with most patients presenting low cytokine release syndrome (CRS) grades. No patients had GvHD, neurotoxicity, or hemophagocytic lymphohistiocytosis [40].

Other CAR-T cells for RCCs

In an orthotopic mouse model, CAR-T cells targeting tyrosine-protein kinase Met (c-Met) were presented as an option for treating pRCC. Administration of CAR-T cells induced an apparent suppression of tumor growth, and complete tumor regression was achieved in approximately 60% of the mice. In addition, the study verified the synergistic increase in therapeutic efficacy when in combination with axitinib [32]. However, the clinical study did not show an objective response to treatment (NCT01218867). Other clinical trials of c-Met CAR-T cells based on different vectors are advancing (NCT03638206).

An allogeneic CAR-T cell therapy designed to target cancer cells that express the cell surface-associated C-terminal antigen Mucin-1 (P-MUC1C-ALLO1) will be tested for safety, tolerability, and response to treatment in patients with solid cancers, including RCC (NCT05239143). Phase I/II clinical trial is investigating the therapeutic effects of CAR-T CCT 301-38 or CCT 301-59 cells in stage IV metastatic patients at different molecular targets, that RCC patients with receptor orphan tyrosine kinase receptor 2 (ROR2) receive CCT31-59 while AXL-positive patients undergo CCT 301 − 38 (NCT03393936).

CAR-NK cells or other genetically engineered NK cells

A wide range of sources can provide NK cells, such as peripheral blood, cord blood, induced pluripotent stem cells (iPSC), and an established NK cell line (such as NK92) can also be used [89]. Allogeneic NK cells can be infused back into patients regardless of donor-patient human leukocyte antigen (HLA) type matching, being an exciting alternative to reduce the cost of CAR-based cell therapy [90,91,92]. The positive rationale for using CAR-NK cell therapy includes that it is less likely to cause side effects like CRS and GvHD [89, 91] and this CAR-NK can overcome endogenous resistance mechanisms in tumor cells [93]. Nevertheless, some difficulties have been reported, such as low transduction efficiency compared to T cells and poor expansion when peripheral-blood-derived NK cells are used (Fig. 5) [94]. When the source of NK cells is from umbilical cord blood, these issues appear to be minimized; however, the relative immaturity of these cells constitutes a possible disadvantage. The use of NK-92 cell lines facilitates engineering and expansion, but it faces some challenges due to safety considerations, the necessity of special cell processing [33, 94], and poor long-term persistence [94, 95].

Fig. 5
figure 5

Natural killer (NK) cells for adoptive cell therapies. Description of advantages and drawbacks of NK cells used for adoptive cell therapies against RCC, showing the possible sources of these cells. Created with BioRender.com

Other challenges in CAR-NK cell therapy production include prolonging the cell survival post-infusion with significant persistence in the peripheral blood, optimizing cell cytotoxic capabilities [90], and increasing CAR-NK cell traffic to the tumor site [89]. A review of six male RCC patients between 50 and 70 years old and no previous treatment has shown that RCC can alter the classical characteristics of NK cells towards a decidua NK-like program, limiting their cytotoxic capacity and inducing angiogenesis [96], pointing out a possible new challenge to use CAR-NK therapy against RCC.

CAIX-specific NK92 cells

NK92 is an IL-2-dependent immortalized cell line derived from a patient with lymphoma. As such, despite the already established general safety of infusion, NK92 must be irradiated before its clinical use [34, 97]. The theoretical advantages of NK92 include its easy expansion and availability, resulting in reduced time to start treatment with lower costs [34].

CAIX-specific CAR-NK92 cells have been described as a potential killer of RCC cells in vitro and in vivo in a mouse model of human RCC. This 3rd generation CAIX-CAR includes an anti-CAIX scFv (LV5), CD8 hinge and transmembrane regions, and CD28, 4-1BB, and CD3ζ intracellular domains [34]. CAIX-specific CARNK92 at an effector: target (E:T) ratio of 30:1 induced specific cell lysis varying from 25-55% for different RCC cell lines, with an unobtrusive 10% increase when CAIX-specific CARNK92 was combined with bortezomib, a proteasome inhibitor [34]. The combination induced a very significant tumor volume decrease in a mouse model of subcutaneous human RCC [34].

EGFR-Specific CAR-NK92

An epidermal growth factor receptor (EGFR)-specific 3rd generation CAR was tested against EGFR+ RCC cells in vitro and in a subcutaneously human RCC-bearing mouse model. When these CAR-T cells are combined with low doses of cabozantinib – a tyrosine kinase inhibitor (TKI) that significantly decreases PD-L1 expression and increases EGFR expression in RCC cells in vitro 5-fold lower tumor volume were found when compared with CAR-NK92 cells or cabozantinib treatment alone [35].

CD70-directed CAR-NK cells

CD70 has also been explored as a target for therapies based on NK cells. A recently opened Phase I/II clinical trial evaluates cord blood-derived NK cells expressing a CD70-targeting CAR engineered to secrete IL-15. This treatment has been performed in association with lymphodepleting chemotherapy for the treatment of advanced RCC, mesothelioma, and osteosarcoma (NCT05703854), with no published results up to November 2023.

NK92 expressing CXCR2

Chemokines regulate immune cell migration by binding to their corresponding chemokine receptors. Many solid tumors, including RCC, release ligands for the C-X-C motif chemokine receptor 2 (CXCR2), but NK cells in peripheral blood seem to lose CXCR2 expression [36]. For this reason, patients with solid tumors that received adoptive NK cell infusion exhibited poor migration of NK cells to the tumor. Genetically modified NK cells re-expressing CXCR2 showed an increased ability to migrate toward CXCR2-ligand-expressing tumors, with better adhesion properties and more significant killing of target cells [36]. NK cells transduced with CXCR2 showed a 2-fold increase in their migration ability to CXCR2 ligands secreted by RCC cell lines compared to NK cells transduced with NGFR [36]. Elevated numbers of tumor infiltrating NK (TINK) cells are related to a better RCC prognosis [36].

Dendritic cell (DC) vaccination

DC vaccines work through induction and endorsement of an immune reaction to eradicate tumor cells. Autologous DCs pulsed with peptides or tumor lysate-derived proteins can stimulate the generation of cytotoxic T cells in cancer patients. Four main methods were applied to use DCs as cell-based vaccines against cancer: co-culture of DCs with isolated autologous tumor tissues, co-culture of DCs with synthetic peptides or recombinant proteins of a tumor antigen, transfection of DCs with a specific plasmid to express tumor antigens, or fusion of DCs with complete tumor cells via polyethylene glycol [98,99,100,101,102,103]. The result of a Phase I/II trial showed DCs pulsed with telomerase and surviving-derived HLA-A2 binding peptides in association with low dose IL-2 was able to promote stable disease for more than 8 weeks in 13 out of 27 patients with RCC [43].

Other adoptive cell therapies for RCC

Lymphokine-activated killer (LAK) cells (NK-like T cells)

Cappuzzello et al. reported the cytotoxic effector functions of NK-resistant tumor-killing cells, termed LAK cells [104]. LAK cells constitute T and NK cells, mainly expressing NK markers such as CD3-CD56 + and NKG2D for HLA-independent killing mechanisms [105, 106]. Autologous-activated LAK cells were infused in metastatic RCC (mRCC) patients with IL-2, and the response rate reported was relatively low with specific side effects [107]. A feasibility clinical trial was then conducted on 10 mRCC patients treated using lymphokine-activated natural killer (LANAK) cells associated with IL-2. The trial yielded 3/10 PR, 4/10 CR, and 1/10 SD with immunotherapy alone, and 2/10 CR after immunotherapy plus surgery, with IL-2 toxicities observed in all treated patients [44]. This better antitumor response allow us to conclude that using well-defined effector cells like NK cells rather than a heterogenous cell population is better for effective treatment [108] Tumor reduction and clinical toxicity were not correlated with LAK cell lytic activity or dose in RCC [109]. A Phase II trial recruited 94 patients with advanced RCC and treated them with LAK cells associated with IL-2 or bolus IL-2 injection with continuous infusion. The two groups achieved objective responses (OR) of 19% and 15%, respectively [46]. In another study for advanced RCC, LAK cells were systemically administered to patients between one to three times a week, followed by a bolus injection of 5000 IU IL-2 twice daily. These cells were localized in the lung but not in the tumor tissue and used to treat pulmonary RCC metastasis. In this study, 50% of the metastatic sites, such as bone, muscle, and lymph node metastases, showed regression in 9 patients treated by arterial LAK therapy, with no severe side effects [110]. LAK cell therapy has been replaced by more specific cell-based immunotherapies [111].

γδ T cells (from TIL and PBMCs)

γδ T cells are a distinct subset of T cells abundant in mucosal organs that constitute less than 5% of the peripheral blood lymphocytes [112, 113]. γδ T cells are non-HLA‐restricted cytotoxic cells and play an important immune role in innate and adaptive immunity by directly recognizing and killing pathogens and activating T and B lymphocytes by releasing certain cytokines [114]. Clinical trials have reported the safety and efficacy of activated γδ T cells in patients with non small cell lung cancer (NSCLC), RCC, melanoma, and breast cancer [45, 115,116,117]. Vγ9Vδ2 T cell subset is the most convenient to isolate and expand, especially from human PBMCs. Many studies have shown that these γδ T cells can be genetically engineered to have superior and specific cytotoxic effects against tumors, resulting in advanced adoptive immunotherapies. Chemotherapy-resistant γδ T cells have been developed by introducing O6-alkylguanine DNA alkyl transferase, a DNA repair enzyme, into Vγ9Vδ2 T cells by lentiviral transduction to confer resistance to the chemotherapeutic drug temozolomide (TMZ) to treat glioblastoma. Chemotherapy-resistant γδ T cells were reported to have a superior antitumor potential in the presence of TMZ [118]. CD19-specific CAR-T cells employing ex vivo expanded Vγ9Vδ2 T cells showed cytolytic effects against CD19+ cancer cells [119]. Different approaches have been studied and reviewed to engineer γδ T cells to overcome their clonal heterogeneity for optimal functionality. T cells engineered with defined γδ T cell repertoires are the autologous αβ T cells transduced with high-affinity Vγ9Vδ2 TCRs. These engineered cells have been subjected to a clinical trial for safety and efficacy assessment [120]. Another excellent feature of Vγ9Vδ2 T cells is that the PD-1/PD-L1 axis does not abrogate their function [121]. Based on all these properties, γδ T cells can be a good immunotherapy source for immunosuppressive solid tumors such as ccRCC. In a recent study, IL-15 activated γδ T cells showed an improved cytotoxic effect in RCC patient-derived xenograft (PDX) mouse model [37]. Lee et al. have characterized CD3low Vγ9Vδ1 T cells and explored their effector and cytotoxic function in 20 treatment naive RCC tumor samples from patients suggesting them as novel therapeutic candidates for RCC treatment in high-risk patients [38].

Cytokine induced killer (CIK) cells

CIK is a novel strategy of cancer cell immunotherapy based on modification, manipulation, and co-opting of autologous or allogeneic primary CD3+ CD56- T cells and CD3+ CD56+ NKT cells [122,123,124], in which NKT cells can recognize tumor cells in a HLA-unrestricted manner [122, 125, 126]. The results of clinical trials have shown all 40 enrolled patients treated with autologous CIK cells with significantly improved overall health conditions and OR [127]. Autologous CIK cells in combination with ICIs (pembrolizumab) [128], inflammatory cytokines (IL-2) [128], and TKIs (sorafenib) [129] have exhibited synergistic effects on RCC tumors in the clinic. Moreover, co-culturing DCs and CIK cells, termed DC-CIK, have improved anti-tumor activity and proliferation of CIK cells, might resulting from the capacity of DCs to decrease Tregs [130]. The combination therapies of DC-CIKs with ICIs (pembrolizumab) [48] (NCT03190811), and TKIs (sunitinib/sorafenib) [47] in RCC clinical trails further increase therapeutic efficacy compared to the monotherapy. Combining CIK cells with a DC vaccine also has displayed more robust antitumor activity and less severe side effects in RCC treatment [131] (NCT01924156).

Discussion

In this manuscript, we reviewed the history of cell therapy for RCC, presenting the advances and perspectives that describe a promising scenario for the use of cell therapies to treat RCC. However, several challenges must be overcome to enable safer and more effective performances of these treatments.

CAIX has recently been reborn as an exciting target for RCC cell therapy, mainly when CAR-T cells were used. Our in vitro and in vivo preclinical studies using humanized mice bearing human RCC [132] have been encouraging from the standpoint of antitumor efficacy and minimal on-target off-tumor side effects [28, 133,134,135]. Besides CAIX, oncofetal antigen 5T4 (with overexpression found in over 75% of RCC patient samples) [22] and the CD70 (overexpressed in about 40% of RCC patients) [86, 88, 136], are relevant targets to be further explored for the development of new cell immunotherapies for RCC management.

T cell exhaustion due to immunosuppressive factors in the TME of solid tumors, including RCC, is a substantial obstacle to improve its therapeutic efficiency. There are some prominent cell therapies in the preclinical development for RCC. Among them, we highlight the PD-1:28 CSR, which improved CAR-T cell efficiency and upgraded low-avidity T cells, blocking T cell inactivation via PD-1 to restore their effector functions and enhance tumor cytotoxicity [24]. Further development of the PD-1:28 CSR could create a first-line treatment strategy against solid malignancies that are refractory to conventional immunotherapeutic techniques. Anti-CAIX G36 CAR-T cells were also tested in a configuration capable of blocking immune checkpoint via releasing ICI in the TME.

Despite the undeniable potential of NK cells as an anti-cancer therapeutic tool and the known correlation between NK infiltration and improved survival of RCC patients [137], there are still challenges that must be surpassed to improve the efficiency of NK therapies against RCC, such as the tendency of NK cells to move to a decidua NK-like program in RCC, characterized by limited cytotoxicity and proangiogenic functions [96]. There are ongoing clinical trials with CAR-NK cell therapies for RCC treatment, such as CD70 targeted IL-15 CAR-NK cells. Also for other solid tumors, target MUC1, NKG2D ligands, and ROBO1 [89, 95]. DC-based vaccination in combination with low-dose IL-2 is a current regimen for advanced RCC.

Some recent approaches with exciting results for treating other solid tumors but underexplored in the context of RCC cellular therapies, such as CAR macrophages (CAR-M) and CAR-Treg [138]. Macrophages are abundant in solid tumors especially RCC [71, 139], and display superior capacities of tumor tissue homing compared to T cells [140]. CAR-M targeting CD19, CD22, HER-2, CD5, and carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) have been developed to fight against solid tumors and hematopoietic malignancies with improved tumor control and significant activation of TME [141,142,143,144]. Recently, it reported a Phase I first in human study of an anti-HER2 CAR-M in patients with HER-2 overexpressing solid tumors [145], shedding the light on translating CAR-M for RCC therapy. CAR regulatory T cells (CAR-Treg) could also be an interesting strategy to be applied in RCC treatment, especially in circumstance of immunosuppression, such as in GvHD. Anti-EGFR CAR-Treg with CD28 has shown antigen-specific infiltration as well as suppresses the function of T effector cells (Teffs) in vivo [146], providing a potential treatment for eliminating toxicities post CAR-T infusion [147].

Availability of data and materials

Not applicable, all information in this review can be found in the reference list.

Abbreviations

5T4:

Trophoblast glycoprotein

ACT:

Adoptive cell therapy

CAIX:

Carbonic anhydrase IX

CAR-T:

Chimeric antigen receptor T cell

CAR-M :

Chimeric antigen receptor Macrophage

CAR-NK:

Chimeric antigen receptor natural killer cell

CAR-Treg:

Chimeric antigen receptor regulatory T cell

CAs:

Carbonic anhydrases

ccRCC:

Clear cell renal cell carcinoma

CEA:

Carcinoembryonic antigen

CEACAM5:

Carcinoembryonic antigen-related cell adhesion molecule 5

CIK:

Cytokine induced killer cells

CRS:

Cytokine release syndrome

CsA:

Camphorsulfonic acid

CSR:

Chimeric switch receptor

CTLA-4:

Cytotoxic T-lymphocyte associated protein 4

CXCR2:

C-X-C motif chemokine receptor 2

DC:

Dendritic cell

E:T:

Effector: target

EGFR:

Epidermal growth factor receptor

ESMO:

European Society for Medical Oncology

FDA :

Food and Drug Administration

FGFR:

Fibroblast growth factor receptor

GvHD:

Graft-versus-host disease

GvT:

Graft vs tumor

HER-2:

Human epidermal growth factor receptor -2

HIF:

Hypoxia-inducible factor

HLA:

Human leukocyte antigen

HSC:

Hematopoietic stem cell

HSCT:

Hematopoietic stem cell transplantation

ICI-ICI:

Dual immunotherapy

ICIs:

Immune checkpoint inhibitors

ICI-TKI:

A combination of immunotherapy and antiangiogenic tyrosine kinase inhibitor

IL-2:

Interleukin-2

IL-2Rα:

Interleukin-2 receptor α

IV:

Intravenous

LAK:

Lymphokine-activated killer

LANAK:

Lymphokine activated natural killer cell

LRRs:

Leucine-rich repeats

mAb:

Monoclonal antibody

MET:

MET proto-oncogene

MHC:

Major histocompatibility complex

mTOR:

Mammalian target of rapamycin

MTX:

Methotrexate

NGFR:

Neural growth factor receptor

NK:

CAR natural killer

NSCLC :

Non-small-cell lung cancer

OAV:

Oncolytic adenovirus

OAV-DEC:

OAV-Decorin

OLA:

Olaparib

ORR:

Overall response rate

OS:

Overall survival

PARP:

Poly (ADP-ribose) polymerase

PBMCs :

Peripheral blood mononuclear cells

PD-1:

Programmed cell death receptor-1

PD-L1:

PD-1 ligand

PDX:

Patient-derived xenograft

PFS:

Progression free survival

pRCC:

Papillary renal cell carcinoma

RCC:

Renal cell carcinoma

rIL-2:

Recombinant interleukin-2

ROR2:

Orphan tyrosine kinase receptor 2

scRNAseq:

Single-cell RNA sequencing

TBI:

Total body irradiation

TCF-1:

 T cell factor 1

TCR:

T cell receptor

TILs:

Tumor-infiltrating lymphocytes

TINK:

Tumor infiltrating natural killer cell

TKI:

Tyrosine kinase inhibitor

TME:

Tumor microenvironment

TMZ:

Temozolomide

TNFR:

Tumor necrosis factor receptor

Tregs:

Regulatory T cells

VEGF:

Vascular endothelial growth factor

VEGFR:

Vascular endothelial growth factor receptor

VHL:

Von-Hippel-Lindau

References

  1. Cohen HT, McGovern FJ. Renal-cell carcinoma. N Engl J Med. 2005;353:2477–90. https://doi.org/10.1056/NEJMra043172.

    Article  CAS  PubMed  Google Scholar 

  2. Dutcher JP. Recent developments in the treatment of renal cell carcinoma. Ther Adv Urol. 2013;5:338–53. https://doi.org/10.1177/1756287213505672.PMC3825112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Shen C, Kaelin WG Jr. The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol. 2013;23:18–25. https://doi.org/10.1016/j.semcancer.2012.06.001.PMC3663044.

    Article  CAS  PubMed  Google Scholar 

  4. Ruf M, Mittmann C, Nowicka AM, Hartmann A, Hermanns T, Poyet C, et al. pVHL/HIF-regulated CD70 expression is associated with infiltration of CD27 + lymphocytes and increased serum levels of soluble CD27 in clear cell renal cell carcinoma. Clin Cancer Res. 2015;21:889–98. https://doi.org/10.1158/1078-0432.Ccr-14-1425.

    Article  CAS  PubMed  Google Scholar 

  5. Akhtar M, Al-Bozom IA, Al HT. Papillary renal cell carcinoma (PRCC): an update. Adv Anat Pathol. 2019;26:124–32. https://doi.org/10.1097/pap.0000000000000220.

    Article  PubMed  Google Scholar 

  6. Albiges L, Guegan J, Le Formal A, Verkarre V, Rioux-Leclercq N, Sibony M, et al. MET is a potential target across all papillary renal cell carcinomas: result from a large molecular study of pRCC with CGH array and matching gene expression array. Clin Cancer Res. 2014;20:3411–21. https://doi.org/10.1158/1078-0432.Ccr-13-2173.

    Article  CAS  PubMed  Google Scholar 

  7. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–24. https://doi.org/10.1056/NEJMoa065044.

    Article  CAS  PubMed  Google Scholar 

  8. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–8. https://doi.org/10.1200/jco.2009.23.9764.

    Article  CAS  PubMed  Google Scholar 

  9. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30:706–20. https://doi.org/10.1093/annonc/mdz056.

    Article  CAS  PubMed  Google Scholar 

  10. Powles T, Albiges L, Bex A, Grünwald V, Porta C, Procopio G, et al. ESMO clinical practice guideline update on the use of immunotherapy in early stage and advanced renal cell carcinoma. Ann Oncol. 2021;32:1511–9. https://doi.org/10.1016/j.annonc.2021.09.014.

    Article  CAS  PubMed  Google Scholar 

  11. Vano YA, Ladoire S, Elaidi R, Dermeche S, Eymard JC, Falkowski S, et al. First-line treatment of metastatic clear cell renal cell carcinoma: what are the most appropriate combination therapies? Cancers (Basel). 2021;13:5548. https://doi.org/10.3390/cancers13215548.PMC8583335.

    Article  CAS  PubMed  Google Scholar 

  12. Atkins MB, Tannir NM. Current and emerging therapies for first-line treatment of metastatic clear cell renal cell carcinoma. Cancer Treat Rev. 2018;70:127–37. https://doi.org/10.1016/j.ctrv.2018.07.009.

    Article  CAS  PubMed  Google Scholar 

  13. Santoni M, Rizzo A, Mollica V, Rosellini M, Marchetti A, Fragomeno B, et al. Pembrolizumab plus Lenvatinib or axitinib compared to nivolumab plus ipilimumab or cabozantinib in advanced renal cell carcinoma: a number needed to treat analysis. Expert Rev Pharmacoecon Outcomes Res. 2022;22:45–51. https://doi.org/10.1080/14737167.2021.1937130.

    Article  PubMed  Google Scholar 

  14. Matsumoto J, Iwata N, Watari S, Ushio S, Shiromizu S, Takeda T, et al. Adverse events of axitinib plus pembrolizumab versus lenvatinib plus pembrolizumab: a pharmacovigilance study in food and drug administration adverse event reporting system. Eur Urol Focus. 2023;9:141–4. https://doi.org/10.1016/j.euf.2022.07.003.

    Article  PubMed  Google Scholar 

  15. Jin H, D’Urso V, Neuteboom B, McKenna SD, Schweickhardt R, Gross AW, et al. Avelumab internalization by human circulating immune cells is mediated by both fc gamma receptor and PD-L1 binding. Oncoimmunology. 2021;10:1958590. https://doi.org/10.1080/2162402X.2021.1958590.PMC8409756.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Numakura K, Muto Y, Naito S, Hatakeyama S, Kato R, Koguchi T, et al. Outcomes of axitinib versus sunitinib as first-line therapy to patients with metastatic renal cell carcinoma in the immune-oncology era. Cancer Med. 2021;10:5839–46. https://doi.org/10.1002/cam4.4130.PMC8419787.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Choueiri TK, Hessel C, Halabi S, Sanford B, Michaelson MD, Hahn O, et al. Cabozantinib versus sunitinib as initial therapy for metastatic renal cell carcinoma of intermediate or poor risk (Alliance A031203 CABOSUN randomised trial): progression-free survival by Independent review and overall survival update. Eur J Cancer. 2018;94:115–25. https://doi.org/10.1016/j.ejca.2018.02.012.PMC6057479.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Liu XL, Xue HY, Chu Q, Liu JY, Li J. Comparative efficacy and safety of sunitinib vs sorafenib in renal cell carcinoma: A systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e19570. https://doi.org/10.1097/MD.0000000000019570.PMC7220148.

    Article  CAS  PubMed  Google Scholar 

  19. Carducci MA. Flashback foreword: pazopanib in renal cell carcinoma and overall survival with sunitinib versus interferon-α in metastatic renal cell carcinoma. J Clin Oncol. 2023;41:1955–6. https://doi.org/10.1200/jco.23.00153.

    Article  PubMed  Google Scholar 

  20. Rini BI, Pal SK, Escudier BJ, Atkins MB, Hutson TE, Porta C, et al. Tivozanib versus sorafenib in patients with advanced renal cell carcinoma (TIVO-3): a phase 3, multicentre, randomised, controlled, open-label study. Lancet Oncol. 2020;21:95–104. https://doi.org/10.1016/S1470-2045(19)30735-1.

    Article  CAS  PubMed  Google Scholar 

  21. Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, et al. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma. N Engl J Med. 2021;384:1289–300. https://doi.org/10.1056/NEJMoa2035716.

    Article  CAS  PubMed  Google Scholar 

  22. Griffiths RW, Gilham DE, Dangoor A, Ramani V, Clarke NW, Stern PL, et al. Expression of the 5T4 oncofoetal antigen in renal cell carcinoma: a potential target for T-cell-based immunotherapy. Br J Cancer. 2005;93:670–7. https://doi.org/10.1038/sj.bjc.6602776.PMC2361613.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xu Y, Morales AJ, Cargill MJ, Towlerton AMH, Coffey DG, Warren EH, et al. Preclinical development of T-cell receptor-engineered T-cell therapy targeting the 5T4 Tumor antigen on renal cell carcinoma. Cancer Immunol Immunother. 2019;68:1979–93. https://doi.org/10.1007/s00262-019-02419-4.PMC6877496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Schlenker R, Olguín-Contreras LF, Leisegang M, Schnappinger J, Disovic A, Rühland S, et al. Chimeric PD-1:28 receptor upgrades low-avidity T cells and restores effector function of Tumor-infiltrating lymphocytes for adoptive cell therapy. Cancer Res. 2017;77:3577–90. https://doi.org/10.1158/0008-5472.Can-16-1922.

    Article  CAS  PubMed  Google Scholar 

  25. Lo ASY, Xu C, Murakami A, Marasco WA. Regression of established renal cell carcinoma in nude mice using lentivirus-transduced human T cells expressing a human anti-CAIX chimeric antigen receptor. Mol Ther Oncolytics. 2014;1:14003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Suarez ER, Chang DK, Sun J, Sui J, Freeman GJ, Signoretti S, et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget. 2016;7:34341–55. https://doi.org/10.18632/oncotarget.9114.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Campos NSP, Souza BS, Silva G, Porto VA, Chalbatani GM, Lagreca G, et al. Carbonic anhydrase IX: a renewed target for cancer immunotherapy. Cancers (Basel). 2022;14. https://doi.org/10.3390/cancers14061392.PMC8946730.

  28. Wang Y, Buck A, Grimaud M, Culhane AC, Kodangattil S, Razimbaud C, et al. Anti-CAIX BBzeta CAR4/8 T cells exhibit superior efficacy in a ccRCC mouse model. Mol Ther Oncolytics. 2022;24:385–99. https://doi.org/10.1016/j.omto.2021.12.019.

  29. Li H, Ding J, Lu M, Liu H, Miao Y, Li L, et al. CAIX-specific CAR-T cells and sunitinib show synergistic effects against metastatic renal cancer models. J Immunother. 2020;43:16–28. https://doi.org/10.1097/CJI.0000000000000301.

    Article  CAS  PubMed  Google Scholar 

  30. Ji F, Zhang F, Zhang M, Long K, Xia M, Lu F, et al. Targeting the DNA damage response enhances CD70 CAR-T cell therapy for renal carcinoma by activating the cGAS-STING pathway. J Hematol Oncol. 2021;14:152. https://doi.org/10.1186/s13045-021-01168-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Panowski SH, Srinivasan S, Tan N, Tacheva-Grigorova SK, Smith B, Mak YSL, et al. Preclinical development and evaluation of allogeneic CAR T cells targeting CD70 for the treatment of renal cell carcinoma. Can Res. 2022;82:2610–24. https://doi.org/10.1158/0008-5472.CAN-21-2931.

    Article  CAS  Google Scholar 

  32. Mori JI, Adachi K, Sakoda Y, Sasaki T, Goto S, Matsumoto H, et al. Anti-tumor efficacy of human anti-c-met CAR-T cells against papillary renal cell carcinoma in an orthotopic model. Cancer Sci. 2021;112:1417–28. https://doi.org/10.1111/cas.14835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Schönfeld K, Sahm C, Zhang C, Naundorf S, Brendel C, Odendahl M, et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol Ther. 2015;23:330–8. https://doi.org/10.1038/mt.2014.219.PMC4445620.

    Article  PubMed  Google Scholar 

  34. Zhang Q, Xu J, Ding J, Liu H, Li H, Lu M, et al. Bortezomib improves adoptive carbonic anhydrase IX‑specific chimeric antigen receptor‑modified NK92 cell therapy in mouse models of human renal cell carcinoma. Oncol Rep. 2018;40:3714–24. https://doi.org/10.3892/or.2018.6731.

    Article  CAS  PubMed  Google Scholar 

  35. Zhang Q, Tian K, Xu J, Zhang H, Li L, Fu Q, et al. Synergistic effects of cabozantinib and EGFR-specific CAR-NK-92 cells in renal cell carcinoma. J Immunol Res. 2017;2017:6915912. https://doi.org/10.1155/2017/6915912.PMC5750507.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kremer V, Ligtenberg MA, Zendehdel R, Seitz C, Duivenvoorden A, Wennerberg E, et al. Genetic engineering of human NK cells to express CXCR2 improves migration to renal cell carcinoma. J Immunother Cancer. 2017;5:73. https://doi.org/10.1186/s40425-017-0275-9.PMC5604543.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Zhang B, Li H, Liu W, Tian H, Li L, Gao C, et al. Adoptive cell therapy of patient-derived renal cell carcinoma xenograft model with IL-15-induced γδT cells. Med Oncol. 2021;38:30. https://doi.org/10.1007/s12032-021-01474-1.

    Article  CAS  PubMed  Google Scholar 

  38. Lee HW, Park C, Joung JG, Kang M, Chung YS, Oh WJ, et al. Renal cell carcinoma-infiltrating CD3(low) Vγ9Vδ1 T cells represent potentially novel Anti-tumor Immune players. Curr Issues Mol Biol. 2021;43:226–39. https://doi.org/10.3390/cimb43010019.PMC8929056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang C, Fang L, Wang X, Yuan S, Li W, Tian W, et al. Oncolytic adenovirus-mediated expression of decorin facilitates CAIX-targeting CAR-T therapy against renal cell carcinoma. Mol Ther Oncolytics. 2022;24:14–25. https://doi.org/10.1016/j.omto.2021.11.018.

    Article  CAS  PubMed  Google Scholar 

  40. Sumanta P, Ben T, John H, Michael H, Adrian S, Neeraj A, et al. 558 CTX130 allogeneic CRISPR-Cas9–engineered chimeric antigen receptor (CAR) T cells in patients with advanced clear cell renal cell carcinoma: results from the phase 1 COBALT-RCC study. J Immuno Ther Cancer. 2022;10:A584. https://doi.org/10.1136/jitc-2022-SITC2022.0558.

    Article  Google Scholar 

  41. Lamers CHJ, Sleijfer S, Vulto AG, Kruit WHJ, Kliffen M, Debets R, et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. 2006;24:e20–22. https://doi.org/10.1200/JCO.2006.05.9964.

    Article  PubMed  Google Scholar 

  42. Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. 2013;21:904–12. https://doi.org/10.1038/mt.2013.17.PMC5189272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Berntsen A, Trepiakas R, Wenandy L, Geertsen PF, Straten T, Andersen P. Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: a clinical phase 1/2 trial. J Immunother. 2008;31:771–80. https://doi.org/10.1097/CJI.0b013e3181833818.

    Article  CAS  PubMed  Google Scholar 

  44. Escudier B, Farace F, Angevin E, Charpentier F, Nitenberg G, Triebel F, et al. Immunotherapy with interleukin-2 (IL2) and lymphokine-activated natural killer cells: improvement of clinical responses in metastatic renal cell carcinoma patients previously treated with IL2. Eur J Cancer. 1994;30a:1078–83. https://doi.org/10.1016/0959-8049(94)90460-x.

    Article  CAS  PubMed  Google Scholar 

  45. Bennouna J, Bompas E, Neidhardt EM, Rolland F, Philip I, Galéa C, et al. Phase-I study of Innacell Gammadelta, an autologous cell-therapy product highly enriched in gamma9delta2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother. 2008;57:1599–609. https://doi.org/10.1007/s00262-008-0491-8.

    Article  CAS  PubMed  Google Scholar 

  46. Weiss GR, Margolin KA, Aronson FR, Sznol M, Atkins MB, Dutcher JP, et al. A randomized phase II trial of continuous infusion interleukin-2 or bolus injection interleukin-2 plus lymphokine-activated killer cells for advanced renal cell carcinoma. J Clin Oncol. 1992;10:275–81. https://doi.org/10.1200/JCO.1992.10.2.275.

    Article  CAS  PubMed  Google Scholar 

  47. Mai H-X, Mei G-H, Zhao F-L, Li B-T, Tang Y-Y, Zhang B, et al. Retrospective analysis on the efficacy of sunitinib/sorafenib in combination with dendritic cells-cytokine-induced killer in Metastasis renal cell carcinoma after radical nephrectomy. J Cancer Res Ther. 2018;14:427–S432. https://doi.org/10.4103/0973-1482.180609.

    Article  CAS  Google Scholar 

  48. Chen CL, Pan QZ, Weng DS, Xie CM, Zhao JJ, Chen MS, et al. Safety and activity of PD-1 blockade-activated DC-CIK cells in patients with advanced solid tumors. OncoImmunology. 2018;7:e1417721. https://doi.org/10.1080/2162402X.2017.1417721.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Bregni M, Herr W, Blaise D. Allogeneic stem cell transplantation for renal cell carcinoma. Expert Rev Anticancer Ther. 2011;11:901–11. https://doi.org/10.1586/era.11.12.

    Article  CAS  PubMed  Google Scholar 

  50. Takahashi Y, Harashima N, Kajigaya S, Yokoyama H, Cherkasova E, McCoy JP, et al. Regression of human kidney cancer following allogeneic stem cell transplantation is associated with recognition of an HERV-E antigen by T cells. J Clin Invest. 2008;118:1099–109. https://doi.org/10.1172/jci34409.PMC2248804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bernasconi P, Borsani O. Immune escape after hematopoietic stem cell transplantation (HSCT): from mechanisms to novel therapies. Cancers (Basel). 2019;12:69. https://doi.org/10.3390/cancers12010069.PMC7016529.

    Article  PubMed  Google Scholar 

  52. Huang XJ, Wang Y, Liu DH, Xu LP, Liu KY, Chen H, et al. Administration of short-term immunosuppressive agents after DLI reduces the incidence of DLI-associated acute GVHD without influencing the GVL effect. Bone Marrow Transplant. 2009;44:309–16. https://doi.org/10.1038/bmt.2009.26.

    Article  CAS  PubMed  Google Scholar 

  53. Saha A, Blazar BR. Antibody based conditioning for allogeneic hematopoietic stem cell transplantation. Front Immunol. 2022;13:1031334. https://doi.org/10.3389/fimmu.2022.1031334.PMC9632731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Jiang T, Zhou C, Ren S. Role of IL-2 in cancer immunotherapy. Oncoimmunology. 2016;5:e1163462. https://doi.org/10.1080/2162402x.2016.1163462.PMC4938354.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hong M, Clubb JD, Chen YY. Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell. 2020;38:473–88. https://doi.org/10.1016/j.ccell.2020.07.005.

    Article  CAS  PubMed  Google Scholar 

  56. Sockolosky JT, Trotta E, Parisi G, Picton L, Su LL, Le AC, et al. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science. 2018;359:1037–42. https://doi.org/10.1126/science.aar3246.PMC5947856.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Codarri Deak L, Nicolini V, Hashimoto M, Karagianni M, Schwalie PC, Lauener L, et al. PD-1-cis IL-2R agonism yields better effectors from stem-like CD8 + T cells. Nature. 2022;610:161–72. https://doi.org/10.1038/s41586-022-05192-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, et al. Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity. 2019;50:195–211.e110. https://doi.org/10.1016/j.immuni.2018.12.021.

    Article  CAS  PubMed  Google Scholar 

  59. Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature. 2019;576:465–70. https://doi.org/10.1038/s41586-019-1836-5.PMC7108171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20:326–36. https://doi.org/10.1038/s41590-019-0312-6.PMC6673650.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hudson WH, Gensheimer J, Hashimoto M, Wieland A, Valanparambil RM, Li P, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1(+) stem-like CD8(+) T cells during chronic Infection. Immunity. 2019;51:1043–1058.e1044. https://doi.org/10.1016/j.immuni.2019.11.002.PMC6920571.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lopes JE, Fisher JL, Flick HL, Wang C, Sun L, Ernstoff MS, et al. ALKS 4230: a novel engineered IL-2 fusion protein with an improved cellular selectivity profile for cancer immunotherapy. J Immunother Cancer. 2020;8:e000673. https://doi.org/10.1136/jitc-2020-000673.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Overwijk WW, Tagliaferri MA, Zalevsky J. Engineering IL-2 to Give new life to T cell immunotherapy. Annu Rev Med. 2021;72(1):281–311.

  64. Waldhauer I, Gonzalez-Nicolini V, Freimoser-Grundschober A, Nayak TK, Fahrni L, Hosse RJ, et al. Simlukafusp alfa (FAP-IL2v) immunocytokine is a versatile combination partner for cancer immunotherapy. MAbs. 2021;13:1913791. https://doi.org/10.1080/19420862.2021.1913791.PMC8115765.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Klein C, Waldhauer I, Nicolini VG, Freimoser-Grundschober A, Nayak T, Vugts DJ, et al. Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based immunocytokine for combination cancer immunotherapy: Overcoming limitations of aldesleukin and conventional IL-2-based immunocytokines. Oncoimmunology. 2017;6:e1277306. https://doi.org/10.1080/2162402x.2016.1277306.PMC5384349.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Southall PJ, Boxer GM, Bagshawe KD, Hole N, Bromley M, Stern PL. Immunohistological distribution of 5T4 antigen in normal and malignant tissues. Br J Cancer. 1990;61:89–95. https://doi.org/10.1038/bjc.1990.20.PMC1971328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wrigley E, McGown AT, Rennison J, Swindell R, Crowther D, Starzynska T, et al. 5T4 oncofetal antigen expression in ovarian carcinoma. Int J Gynecol Cancer. 1995;5:269–74. https://doi.org/10.1046/j.1525-1438.1995.05040269.x.

    Article  PubMed  Google Scholar 

  68. Naganuma H, Kono K, Mori Y, Takayoshi S, Stern PL, Tasaka K, et al. Oncofetal antigen 5T4 expression as a prognostic factor in patients with gastric cancer. Anticancer Res. 2002;22:1033–8.

    CAS  PubMed  Google Scholar 

  69. Starzynska T, Marsh PJ, Schofield PF, Roberts SA, Myers KA, Stern PL. Prognostic significance of 5T4 oncofetal antigen expression in colorectal carcinoma. Br J Cancer. 1994;69:899–902. https://doi.org/10.1038/bjc.1994.173.PMC1968915.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. He P, Jiang S, Ma M, Wang Y, Li R, Fang F, et al. Trophoblast glycoprotein promotes pancreatic ductal adenocarcinoma cell Metastasis through Wnt/planar cell polarity signaling. Mol Med Rep. 2015;12:503–9. https://doi.org/10.3892/mmr.2015.3412.

    Article  CAS  PubMed  Google Scholar 

  71. Kastrunes G, Abbas R, Wang Y, Marasco WA. The updated tumor immune microenvironment TIME landscape of clear cell renal cell carcinoma ccRCC. Biomed J Sci Tech Res. 2022;44:35454–6.

    Google Scholar 

  72. Leen AM, Sukumaran S, Watanabe N, Mohammed S, Keirnan J, Yanagisawa R, et al. Reversal of tumor immune inhibition using a chimeric cytokine receptor. Mol Ther. 2014;22:1211–20. https://doi.org/10.1038/mt.2014.47.PMC4048899.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Bollard CM, Rössig C, Calonge MJ, Huls MH, Wagner HJ, Massague J, et al. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood. 2002;99:3179–87. https://doi.org/10.1182/blood.v99.9.3179.

    Article  CAS  PubMed  Google Scholar 

  74. Zhao S, Wang C, Lu P, Lou Y, Liu H, Wang T, et al. Switch receptor T3/28 improves long-term persistence and antitumor efficacy of CAR-T cells. J Immunother Cancer. 2021;9. https://doi.org/10.1136/jitc-2021-003176.PMC8638458.

  75. Hoogi S, Eisenberg V, Mayer S, Shamul A, Barliya T, Cohen CJ. A TIGIT-based chimeric co-stimulatory switch receptor improves T-cell anti-tumor function. J Immunother Cancer. 2019;7:243. https://doi.org/10.1186/s40425-019-0721-y.PMC6734436.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Maric M, Zheng P, Sarma S, Guo Y, Liu Y. Maturation of cytotoxic T lymphocytes against a B7-transfected nonmetastatic Tumor: a critical role for costimulation by B7 on both tumor and host antigen-presenting cells. Cancer Res. 1998;58:3376–84.

    CAS  PubMed  Google Scholar 

  77. Makuku R, Khalili N, Razi S, Keshavarz-Fathi M, Rezaei N. Current and future perspectives of PD-1/PDL-1 blockade in cancer immunotherapy. J Immunol Res. 2021;2021:6661406. https://doi.org/10.1155/2021/6661406.PMC7925068.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Liu X, Ranganathan R, Jiang S, Fang C, Sun J, Kim S, et al. A chimeric switch-receptor targeting PD1 augments the efficacy of second-generation CAR T cells in advanced solid tumors. Cancer Res. 2016;76:1578–90. https://doi.org/10.1158/0008-5472.Can-15-2524.PMC4800826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Xiao X, Wang Y, Zou Z, Yang Y, Wang X, Xin X, et al. Combination strategies to optimize the efficacy of chimeric antigen receptor T cell therapy in haematological malignancies. Front Immunol. 2022;13:954235. https://doi.org/10.3389/fimmu.2022.954235.PMC9460961.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through the solid-tumour microenvironment. Nat Rev Drug Discov. 2021;20:531–50. https://doi.org/10.1038/s41573-021-00189-2.

    Article  CAS  PubMed  Google Scholar 

  81. Rababah F, Alabduh T, Awawdeh A, Shatnawi T, Al-Shdaifat M, Ibdah E, et al. Chimeric antigen receptor T cells therapy in solid tumors. Clin Transl Oncol. 2023;25:2279–96. https://doi.org/10.1007/s12094-023-03122-8.

    Article  CAS  PubMed  Google Scholar 

  82. Pantuck AJ, Klatte T, Seligson D, Atkins M, Belldegrun A. Carbonic anhydrase IX as a predictive biomarker for clear cell renal cell carcinoma. J Clin Oncol. 2008;26:3105–7; author reply 3107-3109. https://doi.org/10.1200/JCO.2008.16.1935.

    Article  CAS  PubMed  Google Scholar 

  83. Clark PE. The role of VHL in clear-cell renal cell carcinoma and its relation to targeted therapy. Kidney Int. 2009;76:939–45. https://doi.org/10.1038/ki.2009.296.PMC2963106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. de Campos NS, de Oliveira Beserra A, Pereira PH, Chaves AS, Fonseca FL, da Silva Medina T, et al. Immune checkpoint blockade via PD-L1 potentiates more CD28-based than 4–1BB-based anti-carbonic anhydrase IX chimeric antigen receptor T cells. Int J Mol Sci. 2022;23(10):5448.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Yang M, Tang X, Zhang Z, Gu L, Wei H, Zhao S, et al. Tandem CAR-T cells targeting CD70 and B7–H3 exhibit potent preclinical activity against multiple solid tumors. Theranostics. 2020;10:7622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jilaveanu LB, Sznol J, Aziz SA, Duchen D, Kluger HM, Camp RL. CD70 expression patterns in renal cell carcinoma. Hum Pathol. 2012;43:1394–9. https://doi.org/10.1016/j.humpath.2011.10.014.PMC3374042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Wang Y, Suarez ER, Chang M, Jennings R, Signoretti S, Zhu Q, et al. Abstract 3179: design and activity of 2nd generation, dual-targeted CAR T cell factories against ccRCC. Can Res. 2019;79:3179–3179. https://doi.org/10.1158/1538-7445.Am2019-3179.

    Article  Google Scholar 

  88. Wang Y, Grimaud M, Buck A, Fayed A, Chang M, Jennings R, et al. Abstract 6606: develop dual-targeted CAR-T cells to achieve RCC cures. Can Res. 2020;80:6606–6606. https://doi.org/10.1158/1538-7445.Am2020-6606.

    Article  Google Scholar 

  89. Daher M, Rezvani K. Outlook for new CAR-based therapies with a focus on CAR NK cells: what lies beyond CAR-engineered T cells in the race against cancer. Cancer Discov. 2021;11:45–58. https://doi.org/10.1158/2159-8290.cd-20-0556.PMC8137521.

    Article  CAS  PubMed  Google Scholar 

  90. Wrona E, Borowiec M, Potemski P. CAR-NK cells in the treatment of solid tumors. Int J Mol Sci. 2021;22. https://doi.org/10.3390/ijms22115899.PMC8197981.

  91. Wang W, Jiang J, Wu C. CAR-NK for tumor immunotherapy: clinical transformation and future prospects. Cancer Lett. 2020;03(472):175–80. https://doi.org/10.1016/j.canlet.2019.11.033.

    Article  CAS  Google Scholar 

  92. Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, et al. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol. 2022;13:953849. https://doi.org/10.3389/fimmu.2022.953849.PMC9381932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, et al. ErbB2/HER2-Specific NK Cells for targeted therapy of glioblastoma. J Natl Cancer Inst. 2016;108:108. https://doi.org/10.1093/jnci/djv375.

    Article  CAS  Google Scholar 

  94. Schmidt P, Raftery MJ, Pecher G. Engineering NK cells for CAR therapy-recent advances in gene transfer methodology. Front Immunol. 2020;11:611163. https://doi.org/10.3389/fimmu.2020.611163.PMC7817882.

    Article  CAS  PubMed  Google Scholar 

  95. Khawar MB, Sun HCARNK. Cells: From natural basis to design for kill. Front Immunol. 2021;12:707542. https://doi.org/10.3389/fimmu.2021.707542.PMC8712563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Guan Y, Chambers CB, Tabatabai T, Hatley H, Delfino KR, Robinson K, et al. Renal cell tumors convert natural killer cells to a proangiogenic phenotype. Oncotarget. 2020;11:2571–85. https://doi.org/10.18632/oncotarget.27654.PMC7335666.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Siegler EL, Zhu Y, Wang P, Yang L. Off-the-shelf CAR-NK cells for cancer immunotherapy. Cell Stem Cell. 2018;23:160–1. https://doi.org/10.1016/j.stem.2018.07.007.

    Article  CAS  PubMed  Google Scholar 

  98. Daftarian P, Kaifer AE, Li W, Blomberg BB, Frasca D, Roth F, et al. Peptide-conjugated PAMAM dendrimer as a universal DNA vaccine platform to target antigen-presenting CellsPPD as a Universal platform for genetic vaccine. Can Res. 2011;71:7452–62.

    Article  CAS  Google Scholar 

  99. Koido S, Kashiwaba M, Chen D, Gendler S, Kufe D, Gong J. Induction of antitumor immunity by vaccination of dendritic cells transfected with MUC1 RNA. J Immunol. 2000;165:5713–9.

    Article  CAS  PubMed  Google Scholar 

  100. Butterfield LH, Ribas A, Potter DM, Economou JS. Spontaneous and vaccine induced AFP-specific T cell phenotypes in subjects with AFP-positive hepatocellular cancer. Cancer Immunol Immunother. 2007;56:1931–43.

    Article  CAS  PubMed  Google Scholar 

  101. Stober TZ, Reimann J, Schirmbeck R. Dendritic cells pulsed with exogenous Hepatitis B surface antigen particles efficiently present epitopes to MHC class I-restricted cytotoxic T cells. Eur J Immunol. 2002;32:1099–108.

    Article  CAS  PubMed  Google Scholar 

  102. Dashti A, Ebrahimi M, Hadjati J, Memarnejadian A, Moazzeni SM. Dendritic cell based immunotherapy using tumor stem cells mediates potent antitumor immune responses. Cancer Lett. 2016;374:175–85.

    Article  CAS  PubMed  Google Scholar 

  103. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu Y-J, et al. Immunobiol Dendritic Cells. Annu Rev Immunol. 2000;18:767–811.

    Article  CAS  PubMed  Google Scholar 

  104. Cappuzzello E, Sommaggio R, Zanovello P, Rosato A. Cytokines for the induction of antitumor effectors: the paradigm of Cytokine-Induced Killer (CIK) cells. Cytokine Growth Factor Rev. 2017;36:99–105. https://doi.org/10.1016/j.cytogfr.2017.06.003.

    Article  CAS  PubMed  Google Scholar 

  105. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid Tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med. 1982;155:1823–41. https://doi.org/10.1084/jem.155.6.1823.PMC2186695.

    Article  CAS  PubMed  Google Scholar 

  106. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 1999;285:727–9. https://doi.org/10.1126/science.285.5428.727.

    Article  CAS  PubMed  Google Scholar 

  107. Nakano E, Iwasaki A, Seguchi T, Sugao H, Tada Y, Matsuda M, et al. [Usefulness and limitation of immunotherapy of metastatic renal cell carcinoma with autologous lymphokine-activated killer cells and interleukin 2]. Nihon Hinyokika Gakkai Zasshi. 1991;82:395–404. https://doi.org/10.5980/jpnjurol1989.82.395.

    Article  CAS  PubMed  Google Scholar 

  108. Hercend T, Farace F, Baume D, Charpentier F, Droz JP, Triebel F, et al. Immunotherapy with lymphokine-activated natural killer cells and recombinant interleukin-2: a feasibility trial in metastatic renal cell carcinoma. J Biol Response Mod. 1990;9:546–55.

    CAS  PubMed  Google Scholar 

  109. Boldt DH, Mills BJ, Gemlo BT, Holden H, Mier J, Paietta E, et al. Laboratory correlates of adoptive immunotherapy with recombinant interleukin-2 and lymphokine-activated killer cells in humans. Cancer Res. 1988;48:4409–16.

    CAS  PubMed  Google Scholar 

  110. Hayakawa M. Lymphokine-activated killer (LAK) therapy for advanced renal cell carcinoma: clinical study on arterial LAK therapy and experimental study on LAK cell activity]. Hinyokika Kiyo. 1992;38:1311–8.

    CAS  PubMed  Google Scholar 

  111. Wendel P, Reindl LM, Bexte T, Künnemeyer L, Särchen V, Albinger N, et al. Arming Immune cells for battle: a brief journey through the advancements of T and NK Cell Immunotherapy. Cancers (Basel). 2021;13(6):1481. https://doi.org/10.3390/cancers13061481.PMC8004685.

    Article  CAS  PubMed  Google Scholar 

  112. Parker CM, Groh V, Band H, Porcelli SA, Morita C, Fabbi M, et al. Evidence for extrathymic changes in the T cell receptor gamma/delta repertoire. J Exp Med. 1990;171:1597–612. https://doi.org/10.1084/jem.171.5.1597.PMC2187908.

    Article  CAS  PubMed  Google Scholar 

  113. Itohara S, Farr AG, Lafaille JJ, Bonneville M, Takagaki Y, Haas W, et al. Homing of a gamma delta thymocyte subset with homogeneous T-cell receptors to mucosal epithelia. Nature. 1990;343:754–7. https://doi.org/10.1038/343754a0.

    Article  CAS  PubMed  Google Scholar 

  114. Bonneville M, O’Brien RL, Born WK. Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol. 2010;10:467–78. https://doi.org/10.1038/nri2781.

    Article  CAS  PubMed  Google Scholar 

  115. Nakajima J, Murakawa T, Fukami T, Goto S, Kaneko T, Yoshida Y, et al. A phase I study of adoptive immunotherapy for recurrent non-small-cell Lung cancer patients with autologous gammadelta T cells.Eur. J Cardiothorac Surg. 2010;37:1191–7. https://doi.org/10.1016/j.ejcts.2009.11.051.

    Article  Google Scholar 

  116. Abe Y, Muto M, Nieda M, Nakagawa Y, Nicol A, Kaneko T, et al. Clinical and immunological evaluation of zoledronate-activated Vgamma9gammadelta T-cell-based immunotherapy for patients with multiple myeloma. Exp Hematol. 2009;37:956–68. https://doi.org/10.1016/j.exphem.2009.04.008.

    Article  CAS  PubMed  Google Scholar 

  117. Meraviglia S, Eberl M, Vermijlen D, Todaro M, Buccheri S, Cicero G, et al. In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced Breast cancer patients. Clin Exp Immunol. 2010;161:290–7. https://doi.org/10.1111/j.1365-2249.2010.04167.x.PMC2909411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lamb LS Jr, Bowersock J, Dasgupta A, Gillespie GY, Su Y, Johnson A, et al. Engineered drug resistant γδ T cells kill glioblastoma cell lines during a chemotherapy challenge: a strategy for combining chemo- and immunotherapy. PLoS One. 2013;8:e51805. https://doi.org/10.1371/journal.pone.0051805.PMC3543433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Deniger DC, Switzer K, Mi T, Maiti S, Hurton L, Singh H, et al. Bispecific T-cells expressing polyclonal repertoire of endogenous γδ T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol Ther. 2013;21:638–47. https://doi.org/10.1038/mt.2012.267.PMC3589159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Straetemans T, Kierkels GJJ, Doorn R, Jansen K, Heijhuurs S, Dos Santos JM, et al. GMP-Grade Manufacturing of T Cells Engineered to Express a defined γδTCR. Front Immunol. 2018;9:1062. https://doi.org/10.3389/fimmu.2018.01062.PMC5988845.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Tomogane M, Sano Y, Shimizu D, Shimizu T, Miyashita M, Toda Y, et al. Human Vγ9Vδ2 T cells exert anti-tumor activity independently of PD-L1 expression in Tumor cells. Biochem Biophys Res Commun. 2021;573:132–9. https://doi.org/10.1016/j.bbrc.2021.08.005.

    Article  CAS  PubMed  Google Scholar 

  122. Schmidt TL, Negrin RS, Contag CH. A killer choice for cancer immunotherapy. Immunol Res. 2014;58:300–6.

    Article  CAS  PubMed  Google Scholar 

  123. Lee HK, Kim YG, Kim JS, Park EJ, Kim B, Park KH, et al. Cytokine-induced killer cells interact with Tumor lysate–pulsed dendritic cells via CCR1 signaling. Cancer Lett. 2016;378:142–9.

    Article  CAS  PubMed  Google Scholar 

  124. Linn Y, Lau L, Hui KM. Generation of cytokine-induced killer cells from leukaemic samples with in vitro cytotoxicity against autologous and allogeneic leukaemic blasts. Br J Haematol. 2002;116:78–86.

    Article  CAS  PubMed  Google Scholar 

  125. Pievani A, Belussi C, Klein C, Rambaldi A, Golay J, Introna M. Enhanced killing of human B-cell Lymphoma targets by combined use of cytokine-induced killer cell (CIK) cultures and anti-CD20 antibodies. Blood J Am Soc Hematol. 2011;117:510–8.

    CAS  Google Scholar 

  126. Verneris MR, Karami M, Baker J, Jayaswal A, Negrin RS. Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8 + T cells. Blood. 2004;103:3065–72.

    Article  CAS  PubMed  Google Scholar 

  127. Zhang J, Zhu L, Wei J, Liu L, Yin Y, Gu Y, et al. The effects of cytokine-induced killer cells for the treatment of patients with solid tumors: a clinical retrospective study. J Cancer Res Clin Oncol. 2012;138(6):1057–62. https://doi.org/10.1007/s00432-012-1179-1.

    Article  CAS  PubMed  Google Scholar 

  128. Wang Z, Liu X, Till B, Sun M, Li X, Gao Q. Combination of cytokine-induced killer cells and programmed cell death-1 blockade works synergistically to enhance therapeutic efficacy in metastatic renal cell carcinoma and Non-small cell lung cancer. Front Immunol. 2018;9:1513. https://doi.org/10.3389/fimmu.2018.01513.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Yang Y, Lin H, Zhao L, Song Y, Gao Q. Combination of sorafenib and cytokine-induced killer cells in metastatic renal cell carcinoma: a potential regimen. Immunotherapy. 2017;9:629–35. https://doi.org/10.2217/imt-2016-0133.

    Article  CAS  PubMed  Google Scholar 

  130. Pan Y, Tao Q, Wang H, Xiong S, Zhang R, Chen T, et al. Dendritic cells decreased the concomitant expanded Tregs and Tregs related IL-35 in cytokine-induced killer cells and increased their cytotoxicity against Leukemia cells. PloS One. 2014;9:e93591.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Cui Y, Yang X, Zhu W, Li J, Wu X, Pang Y. Immune response, clinical outcome and safety of dendritic cell vaccine in combination with cytokine–induced killer cell therapy in cancer patients. Oncol Lett. 2013;6(2):537–41. https://doi.org/10.3892/ol.2013.1376.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Wang Y, Shelton SE, Kastrunes G, Barbie DA, Freeman GJ, Marasco WA. Preclinical models for development of immune-oncology therapies. Immuno-oncol Insights. 2022;3(8):379–98. https://doi.org/10.18609/ioi.2022.41. PMC10150782.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Wang Y, Buck A, Grimaud M, Culhane A, Braun D, Kodangattil S, et al. Abstract 2814: Anti-CAIX Immune restoring (IR) CAR-T cells display superior antitumor activity and reverse immunosuppressive TME in a humanized ccRCC orthotopic mouse model. Can Res. 2022;82:2814–2814. https://doi.org/10.1158/1538-7445.Am2022-2814.

    Article  Google Scholar 

  134. Wang Y, Buck A, Lynch M, Kastrunes G, Cho J-W, Grimaud M, et al. 295 Affinity fine-tuning anti-CAIX CAR-T cells mitigate on-target off-tumor side effects. J Immunother Cancer. 2022;10:A310. https://doi.org/10.1136/jitc-2022-SITC2022.0295.

    Article  Google Scholar 

  135. Wang Y, Buck A, Kastrunes G, Abbas R, Lynch M, Zhong Z, et al. Abstract 886: fine-tuned CAIX targeted CAR-T cells exhibit superior efficacy and mitigate on-target off-tumor side effects. Can Res. 2023;83:886–886. https://doi.org/10.1158/1538-7445.Am2023-886.

    Article  Google Scholar 

  136. Wang Y, Buck A, Grimaud M, Kodangattil S, Razimbaud C, Fayed A, et al. Abstract 62: development of dual-targeted fine-tuned immune restoring (DFIR) CAR T cell therapy for clear cell renal cell carcinoma (ccRCC). Can Res. 2021;81:62–62. https://doi.org/10.1158/1538-7445.Am2021-62.

    Article  Google Scholar 

  137. Terren I, Orrantia A, Mikelez-Alonso I, Vitalle J, Zenarruzabeitia O, Borrego F. NK cell-based immunotherapy in renal cell carcinoma. Cancers (Basel). 2020;12(2):316. https://doi.org/10.3390/cancers12020316.PMC7072691.

    Article  CAS  PubMed  Google Scholar 

  138. Marofi F, Motavalli R, Safonov VA, Thangavelu L, Yumashev AV, Alexander M, et al. CAR T cells in solid tumors: challenges and opportunities. Stem Cell Res Ther. 2021;12:1–16.

    Article  Google Scholar 

  139. Braun DA, Street K, Burke KP, Cookmeyer DL, Denize T, Pedersen CB, et al. Progressive immune dysfunction with advancing Disease stage in renal cell carcinoma. Cancer Cell. 2021;39:632–648.e638. https://doi.org/10.1016/j.ccell.2021.02.013.PMC8138872.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhou X, Liu X, Huang L. Macrophage-mediated tumor cell phagocytosis: opportunity for nanomedicine intervention. Adv Funct Mater. 2021;31:2006220.

    Article  CAS  PubMed  Google Scholar 

  141. Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, et al. Chimeric antigen receptors that trigger phagocytosis. Elife. 2018;7:e36688.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Biglari A, Southgate TD, Fairbairn LJ, Gilham DE. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther. 2006;13:602–10.

    Article  CAS  PubMed  Google Scholar 

  143. Zhang W, Liu L, Su H, Liu Q, Shen J, Dai H, et al. Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix. Br J Cancer. 2019;121:837–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M. Hum chimeric antigen receptor macrophages cancer immunotherapy. Nat Biotechnol. 2020;38:947–53. https://doi.org/10.1038/s41587-020-0462-y.PMC7883632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Reiss K, Ueno N, Yuan Y, Johnson M, Dees EC, Chao J, et al. 633 a phase 1, first in human (FIH) study of autologous macrophages containing an anti-HER2 chimeric antigen receptor (CAR) in participants with HER2 overexpressing solid tumors. J Immunother Cancer. 2022;10:A664–4. https://doi.org/10.1136/jitc-2022-SITC2022.0633.

    Article  Google Scholar 

  146. Boroughs AC, Larson RC, Choi BD, Bouffard AA, Riley LS, Schiferle E, et al. Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. JCI Insight. 2019;5:e126194. https://doi.org/10.1172/jci.insight.126194.PMC6538349.

    Article  PubMed  Google Scholar 

  147. Good Z, Spiegel JY, Sahaf B, Malipatlolla MB, Ehlinger ZJ, Kurra S, et al. Post-infusion CAR TReg cells identify patients resistant to CD19-CAR therapy. Nat Med. 2022;28:1860–71. https://doi.org/10.1038/s41591-022-01960-7.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the Assistant Secretary of Defense for Health Affairs endorsed by the Department of Defense, through the FY21 Translational Research Partnership Award (W81XWH-21-1-0442) and FY21 Idea Development Award (W81XWH-21-1-0482) to Wayne A. Marasco. Opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endorsed by the Department of Defense. In addition, this work was supported by the Wong Family Award and KCA Trailblazer Award to Yufei Wang.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, YW, ERS; data accumulation, writing, and original draft preparation: YW, ERS, GK, NSPdC, RA, RSP, NM, GMC, VD, WAM; figure preparation: YW, ERS, NM, NSPdC; revision: YW, ERS, NM, GK; supervision: WAM. All authors have read and approved the submitted version of the manuscript.

Corresponding author

Correspondence to Wayne A. Marasco.

Ethics declarations

Ethics approval and consent to participate

Not applicable to this review.

Consent for publication

All authors agree with the content of the manuscript and consent to publication.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Suarez, E.R., Kastrunes, G. et al. Evolution of cell therapy for renal cell carcinoma. Mol Cancer 23, 8 (2024). https://doi.org/10.1186/s12943-023-01911-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12943-023-01911-x

Keywords