Introduction
In the evolving landscape of bladder cancer (BCa) immunotherapy, this commentary offers a critical and timely reflection on a recent review that we published previously on targeting innate lymphoid cells (ILCs) in BCa. Even though BCa immunotherapy has traditionally focused on enhancing T-cell responses, the review explores the promising but underappreciated role of ILCs in BCa. From natural killer cells to the diverse ILC subtypes, these cells offer a dualistic impact on tumor progression and immune surveillance. By dissecting the review's findings and integrating recent discoveries, this commentary emphasizes the potential of targeting ILCs to boost therapeutic efficacy and provide new avenues for BCa treatment. This analysis not only reaffirms the importance of integrating ILCs into immunotherapy strategies but also advocates for the application of these insights into novel immunotherapeutic strategies.
The Twists and Turns of ILC Ontogeny
ILCs are a diverse family of immune cells with functions similar to T lymphocytes, including ILC1, ILC2, ILC3, natural killer (NK) cells, and lymphoid tissue inducer (LTi) cells [1-3]. Unlike T lymphocytes, ILCs do not possess antigen-specific receptors or undergo clonal selection and expansion. Instead, they respond to signals in their environment and regulate immune responses by specific cytokine secretion [2]. While the exact mechanisms of ILC differentiation are still not fully understood, it is widely accepted that ILCs originate from the common lymphoid progenitor (CLP) [4]. In mice, the CLP is characterized as Ly6D-, with the α4β7+ subset proposed as the earliest ILC progenitor, termed the α lymphoid progenitor (αLP) [5,6]. Additionally, the CLP gives rise to the early innate lymphoid progenitor (EILP), which differs from the αLP by lacking the interleukin-7 receptor (IL-7Rα, CD127) and the inhibitor of DNA binding 2 (Id2) [4,6]. Differentiation of both αLP and EILP is regulated by the transcription factor nuclear factor interleukin 3 regulated (NFIL3), which modulates Id2 expression [7,8]. Subsequent differentiation driven by transcription factor 7 (TCF-1) results in the formation of NK progenitors (NKP) and common helper-innate lymphoid cell progenitors (CHILP). This heterogeneous population includes both PLZF+ (zinc finger and BTB domain containing 16) and PLZF- cells, which can differentiate into NK cells, ILC1, ILC2, ILC3, and LTis, respectively [9]. The terminal differentiation of these ILC subsets relies on specific transcription factors: T-bet and Eomesodermin (Eomes) for NK cells, T-bet for ILC1s, RORα and GATA3 for ILC2s and RORγt for ILC3s and LTis [4].
To thoroughly address ILC ontogeny, it is essential to consider the distribution of CLPs during embryonic development, particularly in the differentiation of LTis. LTis were initially classified as a subset of ILC3s due to their similar transcriptomic profiles but have since been recognized as a distinct subset [4]. The differentiation of LTis in utero is believed to start in the aorta-gonad-mesonephros (AGM) region, where endothelial cells produce hematopoietic stem cells (HSCs) that subsequently migrate to the fetal liver (Figure 1) [10]. At embryonic day E16.5, fetal liver HSCs migrate to the bone marrow, where the majority of lymphoid differentiation occurs [10]. In contrast, the differentiation of LTis begins earlier in the fetal liver, where HSCs differentiate into CLP and LTi progenitor (LTiP) around E11.5. The LTiP then migrates to the periphery, a crucial step in LTi differentiation and the formation of lymph nodes (LNs) [10]. As the LTiP is exposed to retinoic acid in the periphery, the transcription of the retinoic acid-receptor-related orphan nuclear receptor gamma (RORγt) is induced [11]. The clustering of LTis and mesenchymal lymphoid tissue organizer (LTo) cells, essential for LN formation, occurs at E12.5. This clustering activates the lymphotoxin signaling pathway, leading to the maturation of LNs around E14.5. [10]. While most LTi differentiation in utero occurs in the periphery, a similar subset of LTis is also found in the fetal liver, where some acquire antigen-presenting capabilities through the expression of major histocompatibility complex (MHC)-II. This raises significant questions, particularly regarding whether the LTis observed in adults are true LTis or merely LTi-like cells that retain lymphotoxin signaling capacity. These differentiation divergences are not limited to LTis but extend to all ILC subsets. For instance, NK cells, which differentiate in the bone marrow, are unique in that they lack CD127 and thus develop in an IL-7-independent manner [12-14]. However, CD127+ NK cells have been identified in the LNs of mice, of which a percentage differentiate in the thymus [15]. Other tissue-resident (tr)NK cell subsets have also been identified in the liver, lung, and uterus, all of which demonstrate unique functions and phenotypes [16]. Similarly, cytotoxic intraepithelial ILCs are characterized by their absence of CD127, reliance on Eomes, and expression of CD103, an integrin that binds E-cadherin [17]. Distinct ILC2 and ILC3 subsets have also been described, such as the IL-10+ ILC2s, and the natural cytotoxicity receptor (NCR)+/NCR- ILC3 subsets [18,19]. The variability in transcription factor activity and the environmental conditions during differentiation significantly contribute to the uncertainty surrounding ILC lineage commitment and the progenitors involved [20].
Figure 1. Ontogeny of embryonic ILC differentiation. LTi differentiation in utero occurs prior to the other ILC subsets and is required for the formation of lymph nodes. Following the migration to the fetal liver, HSCs differentiate into CLP and LTiP, then travel to the periphery. Exposure to retinoic acid allows for the transcription of RORγt, and subsequent maturation of LTi. LTis and LTos then cluster, which precedes the LTβR. HSC: Hematopoetic Stem Cell; CLP: Common Lymphoid Progenitor; LTi: Lymphoid Tissue Inducer; LTiP: Lymphoid Tissue Inducer Progenitor; LTo: Lymphoid Tissue Organizer. Figure created in BioRender.
ILCs play distinct roles in modulating the immune response through their cytokine production. NK cells, known as the cytotoxic ILC subset and the innate counterparts to CD8+ T cells, produce inflammatory cytokines such as TNFα and IFNγ. They also mediate direct cytotoxicity by degrangulating, inducing target cell apoptosis, and performing antibody-dependent cell cytotoxicity [4]. ILC1s, generally thought to be the non-cytotoxic Th1 CD4+ counterpart, modulate inflammatory responses through their production of IFNγ [4]. However, a cytotoxic subset of ILC1s, termed “tumor-associated” ILC1s, has been found to express granzyme B and mediate NK cell-like cytotoxicity [21] ILC2s, the innate counterpart to Th2 CD4+ cells, modulate anti-inflammatory responses through their production of IL-5 and IL-13 [4,22]. ILC3s, the innate counterpart to Th17 CD4+ T cells, produce IL-17 and IL-22 and mediate gut immune responses [23].
Defining LTi cells and their functions is more complex due to their intricate ontogeny. LTi cells that form lymph nodes and differentiate in the periphery during embryogenesis are present for only a few weeks post-partum before being replaced by HSC-derived cells [10]. LTis that differentiate ex utero, known as LTi-like cells, develop in the bone marrow and express OX40L, CD30L, and LTα1β2 [24,25]. CXCL13 produced by both stromal cells and lymphocytes recruit LTis through CXCR5 signaling, inducing the formation of lymphoid structures, including the Peyer’s patches in the intestines [23,26]. During chronic inflammation and cancer, however, the recruitment of LTis to inflamed tissue can lead to the formation of ectopic tertiary lymphoid structures [27].
The Dynamic and Multifaceted Roles of ILCs in Bladder Cancer
BCa carries one of the highest tumor mutational burdens, and the resulting immunogenicity makes it a great target for immunotherapies [28]. Current immunotherapies approved for use in BCa are immune checkpoint inhibitors (ICI), which aim to prevent T cell exhaustion [29]. The activation of cytotoxic CD8+ T cells requires antigen presentation by the target via the MHC-I, which is often downregulated by cancer cells as a mechanism of immune evasion, rendering T cell mediated cytotoxicity ineffective [30]. In contrast, innate immune cells like ILCs modulate immune activity independent of antigen specificity or presentation [4]. NK cells, for instance, mediate direct cell lysis by recognizing markers upregulated during cellular stress and detecting the downregulation of MHC-I [31]. Additionally, ILC1 and ILC3 cells produce pro-inflammatory cytokines, amplifying inflammatory responses [4].
In BCa, studies show that ILCs modulate both an inflammatory and immunosuppressive tumor microenvironment (TME) [4]. As we have previously discussed, NK cells compose a significant portion of bladder tumor-infiltrating lymphocytes and are crucial for anti-tumor activity [4]. In the BCa TME, however, NK cells are found to express various markers of exhaustion, including Tim-3 and PD-1, reducing their cytotoxic capacity [4]. ILC1s, which canonically contribute to modulating a pro-inflammatory immune environment, display a unique Th17-like phenotype in BCa, characterized by the production of IL-17, playing an overall pro-tumorigenic role [4,32].
ILC2s are known for their overall immunosuppressive nature, owing to their production of anti-inflammatory cytokines and chemokines such as IL-13 and CXCL2 [4,33,34]. Specifically, in the hypoxic TME, ILC2s have been found to produce IL-10, an immunosuppressive cytokine and negative regulator of inflammation [35-37]. In BCa, ILC2s have been correlated with increased infiltration of myeloid-derived suppressive cells (MDSCs), specifically M2-polarized macrophages [34]. The ILC2-modulated shift in the TME has also been correlated with the recurrence and progression of BCa. Recently, however, an in vivo BCa model demonstrated that inhibiting or depleting ILC2s does not affect mouse survival or tumor development. Nonetheless, their exact contribution to BCa progression warrants further investigation [33].
The role of the ILC3 subset in BCa remains underexplored, but their highly plastic nature suggests they could have both beneficial and detrimental effects. As previously discussed, ILC1s in BCa have exhibited an ILC3-like phenotype and were found to accumulate in higher-stage tumors [32]. Additionally, the co-expression of CD69 and CD103, along with elevated levels of CCR6, on ILC3s contribute to increased recruitment to lymphoid tissue and tissue residency in BCa [4,38]. These LTi-like ILC3s are particularly interesting due to their association with tertiary lymphoid structure (TLS) formation [39].
LTis are responsible for the formation of TLS (Figure 2), ectopic aggregates of immune cells resembling secondary lymphoid organs (SLO) [26,40]. The binding of the lymphotoxin (LT)α1β1 expressed on LTi to the LTβR on stromal cells induces the release of chemokines including CXCL13, CXCL12, CCL21, and CCL19, which further recruit CD4+ T cells, follicular dendritic cells (FDC), and B cells [39]. Mature TLSs are structured similarly to SLO, with CD3+ T cell-rich areas as well as FDC and T follicular helper cells (Tfh) capable of forming germinal centers [39]. TLSs are becoming increasingly relevant in cancer due to their role in enhancing lymphocyte recruitment and maturation. However, in some malignancies, they have also been found to play an immunosuppressive role [41]. TLSs in renal cell carcinoma increased the population of tumor-specific IgG-producing plasma cells due to their in situ hypermutation and clonal expansion capabilities [42]. In BCa, mature TLSs correlated with increased immune infiltration and improved patient prognosis [42]. Additionally, TLSs expressing higher levels of CXCL13 were associated with better BCa outcomes and an improved response to ICI therapy, suggesting that CXCL13 is a possible biomarker for disease progression and ICI therapy effectiveness [42]. Due to the nature of BCa, biomarkers relating to ILC infiltration are difficult to quantify without invasive procedures, such as biopsies. For this reason, it is pertinent to define additional urinary biomarkers which can be used to screen for ILC infiltration.
Figure 2. LTi-like cells interact with stromal cells and induce the formation of TLSs in BCa. LTi-like cells respond to signaling chemokines produced by stromal cells in BCa via the CXCL13/CXCR5 axis. The stromal cells expressing LTβR and TNFR1 then bind and interact with LTα1β2 and TNFα expressed by the LTi, which produce cytokines that induce the formation of high endothelial venules and recruit B cells, T follicular helper cells, and follicular dendritic cells, which form germinal centers in the TLS. LTi: Lymphoid Tissue Inducer; TLS: Tertiary Lymphoid Structure; LTβR: Lymphotoxin Β Receptor; TNFα: Tumor Necrosis Factor α; LTα1β2: Lymphotoxin α1β2; TNFR1: Tumor Necrosis Factor Receptor. Figure created in BioRender.
It is evident that ILCs play a vital role in BCa prognosis, but the extent of their impact remains unclear due to limited research. While targeting cytotoxic ILC subsets appears to be a promising strategy for enhancing anti-tumor immunity, the plasticity of non-cytotoxic ILCs may also play a crucial role [43]. Evidence of ILC2 plasticity has emerged from studies in other malignancies. For instance, in hepatocellular carcinoma, a higher ILC2-to-ILC1 ratio with a unique KLRG1+ phenotype correlated with a better prognosis compared to KLRG-CXCL8hi ILC2s, which are known to promote the recruitment of immunosuppressive neutrophils [18]. Interestingly, an inflammatory ILC2 subset emerged following the reconstitution of allogeneic HSC transplantation, in response to inflammatory cytokines. [44]. Incubation of ILC2s with inflammatory cytokines in vitro also induced an inflammatory phenotype, as evidenced by changes in the transcriptome and chromatin modifications of STAT1 and IFNγ [44].
ILCs in Bladder Cancer Prognosis and Treatment–Past, Present, and Future
The downregulation of MHC-I as an immune evasion mechanism has been correlated with immune therapy failure and recurrence of BCa [45]. Due to the inhibitory mechanism of NK cells, the downregulation of MHC-I increases BCa susceptibility to NK cytotoxicity [4]. The adoptive transfer of NK cells is a promising therapeutic strategy in BCa due to their ability to mediate tumor lysis independently of antigen presentation [46]. However, its clinical efficacy is limited by several factors, including the exhaustion of transferred NK cells, which diminishes their cytotoxic function, and insufficient tumor infiltration. Additionally, the immunosuppressive tumor microenvironment, characterized by inhibitory cytokines, metabolic constraints, and physical barriers, further impairs NK cell persistence, infiltration, and antitumor activity, ultimately contributing to the suboptimal outcomes of adoptive NK cell therapy [47]. Current treatments targeting NK cells include immunomodulatory drugs, NK receptor agonists, interleukins, and chimeric antigen receptor (CAR)-expressing NK cells (Table 1) [48]. Lenalidomide, vactosertib, and durvalumab are three immunomodulatory drugs currently undergoing clinical trials for BCa. Lenalidomide (NCT01373294, NCT01342172, NCT01352962) functions as an NK cell activator by stimulating T cell production of IL-2 and IFNγ while vactosertib and durvalumab (NCT04064190) inhibit the TGFβ type 1 receptor kinase, inhibiting TGFβ-induced NK cell downregulation [48-52]. NK receptor agonists currently being tested target the co-receptors CD27, OX40, GITR, and 4-1BB [48,53,54]. Both OX40 and 4-1BB are also expressed in T cells, allowing these agonists to target both the innate and adaptive immune populations [48,55]. Other combination therapies targetting adaptive and innate immunity are also being tested in BCa, such as the combination of the anti-PD-1 drug, nivolumab, and anti-KIR2DL lirilumab in cisplatin-refractive muscle-invasive BCa (NCT0353245) [56]. Interleukin administration is another treatment that targets NK cells as well as other inflammatory immune cells [48]. For example, IL-2, an inflammatory cytokine required for NK cell proliferation, cytotoxicity, and survival, immunotherapy was approved nearly 20 years ago and remains a treatment choice as both a monotherapy and combination therapy in multiple metastatic cancers [57,58].
Alternative drug approaches targeting NK cells in BCa remain in the preclinical phase but show promise [59]. Recently, the anti-androgen drug enzalutamide demonstrated increased efficacy in mediating NK cell cytotoxicity both in vitro and in vivo [60]. Liu et al. demonstrated that signaling through the androgen receptor modulated the adenosine deaminase acting on RNA (ADAR) 2 enzyme, leading to decreased miR-200-3p via circ_0001005 sponging and resulting in increased PD-L1 expression in BCa [60]. Treatment of BCa cells with androgen-targeting drugs, including the FDA-approved prostate cancer treatment Enzalutamide, enhanced NK cell cytotoxicity by downregulating PD-L1 expression [60].
In an orthotopic murine model of BCa, treatment with a CD39 inhibitor significantly reduced tumor burden and increased infiltration of cytotoxic T lymphocytes (CTLs), NK cells, and conventional dendritic cells (cDCs), leading to enhanced CTL proliferation [61]. However, this anti-tumor effect was lost following NK cell depletion, highlighting the crucial role of NK cells in recruiting both cDCs and CTLs [61]. Additionally, Huyan et al. recently showed the impact of tumor-derived exosomes on NK cells. Exosomes released by the BCa cell line T24 induced NK cell apoptosis and downregulated the expression of DAP10, perforin, and CD96 through the delivery of microRNAs miR-221-5p and miR-186-5p [62].
Drug therapies targeting ILCs beyond NK cells remain limited due to sparse research on these subsets. However, existing studies are promising and warrant further investigation. For example, in an orthotopic model of pancreatic ductal adenocarcinoma, deleting IL-33 led to increased tumor burden due to reduced recruitment of ILC2s [50]. Conversely, treatment with recombinant IL-33 and anti-PD-1 decreased tumor burden, with IL-33 stimulation enhancing the sensitivity of ILC2s to anti-PD-1 therapy [63]. Additionally, stimulating NCR+ILC3s with IL-12 can activate the inflammatory cascade, suggesting that recombinant IL-12 treatment could be a viable therapeutic strategy [64]. These findings draw valuable lessons from other solid tumors and highlight the potential for targeting ILCs to enhance BCa treatment. However, special consideration is required when targeting ILCs due to their tissue-specific behavior. For instance, while recombinant IL-33 treatment reduces tumor burden in pancreatic ductal adenocarcinoma, in BCa, IL-33 may exacerbate the ILC2-mediated recruitment and polarization of MDSCs.
As research on ILCs advances, the heterogeneity within ILC subsets becomes increasingly evident. We have highlighted instances of ILC plasticity in BCa, such as the ILC3-like phenotype observed in ILC1s [32]. However, it remains unclear whether these phenotypic shifts are driven by the TME or are specific to bladder tissue-resident ILCs. To effectively target ILC subsets in BCa, it is essential to fully characterize the unique phenotype and function of tissue-resident ILCs. Additionally, the role of the bladder TME in shaping ILC differentiation and recruitment remains poorly understood and warrants further investigation. Identifying the signals that drive ILC differentiation may shed light on the mechanisms behind their skewed functionality in BCa. For example, in chronic obstructive pulmonary disease and asthma, upregulation of inflammatory cytokines IL-1β and IL-12 promotes the differentiation of ILC2s into ILC1s, leading to chronic inflammation and tissue damage [65]. Understanding whether similar mechanisms occur in BCa could inform therapeutic strategies, such as leveraging the ILC2 axis in recombinant IL-12 treatment.
Despite the established importance of ILCs, the ontogeny, residency, and phenotypic expression of ILCs in BCa remain largely unexplored. To develop effective treatments targeting ILCs, a thorough understanding of their functions and roles within BCa is imperative. Single-cell RNA sequencing technologies allow for precise phenotypic characterization of tissue-specific ILCs across various tissues. When combined with in vivo infection and cancer models, this approach can elucidate the plasticity of tissue-resident ILCs in disease. Additionally, chromatin immunoprecipitation sequencing (ChIP-seq) may offer a comprehensive view of ILCs in cancer, paving the way for the development of novel ILC-targeting therapies tailored to the site of malignancy. Lastly, advancing BCa treatment requires integrating ILC-targeted strategies into clinical research. This involves incorporating ILC profiling into clinical trials to correlate ILC subsets with treatment outcomes and exploring ILC modulation or depletion as potential adjuvant therapies. By embedding ILC-focused approaches into both preclinical and clinical studies, we can uncover strategies to overcome immunotherapy resistance and fully realize the potential of ILC-targeted therapies. To fully realize these benefits, identifying specific biomarkers to guide therapy choice is also essential. Embracing precision medicine will allow for the customization of treatments to individual patients, thereby enhancing their effectiveness and advancing overall treatment strategies in BCa.
Therapy |
Functional Target |
Approval Status |
Ref |
ALT-803/N-803 |
IL-15 agonist |
Awaiting FDA approval for use in high risk NMIBC |
[66,67] |
CG0070 |
GM-CSF expressing oncolytic adenovirus |
Phase III |
[68,69] |
Durvalumab |
TGFβ inhibitor |
FDA approved for biliary malignancy, lung cancer (small cell and non-small cell), and hepatocellular carcinoma. Phase II trial for efficacy in bBCa as an adjuvant |
[70,71] |
Lenalidomide |
Stimulates production of IFNγ and IL-2 by T cells |
Approved for multiple myeloma, specific lymphomas. Phase Ib/II trials for metastatic BCa in combination with gemcitabine and cisplatin. |
[50-52,72] |
MEDI6469 MOXR0916 INBRIX-106 |
Anti-OX40 antibody; OX40 agonist |
Terminated Terminated Phase 1 / 2 Trials |
[73-76] |
NK-CD24-CAR cell therapy |
Targets highly expressed CD24 in BCa to activate CAR-NK cells |
Pre-Clinical |
[77] |
Proleukin |
Synthetic IL-2 treatment |
FDA approved in melanoma and renal carcinoma |
[78] |
TRX518 |
Glucocorticoid-induced TNF receptor-related protein agonist |
Phase 1b for use in solid tumors |
[79] |
Urelumab Utomilumab |
CD137 (4-1BB) Agonist |
Awaiting FDA Approval; Phase II/dose escalation |
[80,81]; [82] |
Vactosertib |
TGFβ receptor inhibitor |
Phase 1b for use in multiple myeloma, as well as in combination with durvalumab in urothelial carcinoma |
[49,83,84] |
Varlilumab |
CD27 targetting monoclonal antibody |
Phase II for various solid tumors, as well as in combination with anti-PD-1 therapy |
[53,54,85] |
Nivolumab and Lirilumab |
Anti-PD-1 and anti-KIR2DL combination therapy |
Phase 1b for MIBC ineligible for cisplatin-based therapy |
[56] |
Funding
1) The Mays Family Cancer Center at the University of Texas Health San Antonio (2) Bladder Cancer Advocacy Network (BCAN), (3) Research Training Award (RP170345) from the Cancer Prevention & Research Institute of Texas, (4) Long School of Medicine at UTHSCSA and the Institute for the Integration of Medicine and Science (5) Mike Hogg Fund (6) American Cancer Society Research Scholar Grant (7) NCI R01 1R01CA281726-01A1 (8) ) the Max & Minnie Tomerlin Voelcker Fund.
References
2. Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015 Jan 15; 517(7534):293-301.
3. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, Powrie F, Spits H. Innate Lymphoid Cells: 10 Years On. Cell. 2018 Aug 23;174(5):1054-66.
4. Noel ODV, Hassouneh Z, Svatek RS, Mukherjee N. Innate Lymphoid Cells in Bladder Cancer: From Mechanisms of Action to Immune Therapies. Cancer Immunol Res. 2024 Feb 2;12(2):149-60.
5. Harly C, Cam M, Kaye J, Bhandoola A. Development and differentiation of early innate lymphoid progenitors. J Exp Med. 2018 Jan 2; 215(1):249-62.
6. Nagasawa M, Spits H, Ros XR. Innate Lymphoid Cells (ILCs): Cytokine Hubs Regulating Immunity and Tissue Homeostasis. Cold Spring Harb Perspect Biol. 2018 Dec 3;10(12):a030304.
7. Seillet C, Mielke LA, Amann-Zalcenstein DB, Su S, Gao J, Almeida FF, Shi W, Ritchie ME, Naik SH, Huntington ND, Carotta S, Belz GT. Deciphering the Innate Lymphoid Cell Transcriptional Program. Cell Rep. 2016 Oct 4; 17(2):436-47.
8. Xu W, Domingues RG, Fonseca-Pereira D, Ferreira M, Ribeiro H, Lopez-Lastra S, et al. NFIL3 orchestrates the emergence of common helper innate lymphoid cell precursors. Cell Rep. 2015 Mar 31; 10(12):2043-54.
9. Yang Q, Li F, Harly C, Xing S, Ye L, Xia X, et al. TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow. Nat Immunol. 2015 Oct; 16(10):1044-50.
10. Simic M, Manosalva I, Spinelli L, Gentek R, Shayan RR, Siret C, et al. Distinct Waves from the Hemogenic Endothelium Give Rise to Layered Lymphoid Tissue Inducer Cell Ontogeny. Cell Rep. 2020 Aug 11; 32(6):108004.
11. van de Pavert SA, Ferreira M, Domingues RG, Ribeiro H, Molenaar R, Moreira-Santos L, et al. Maternal retinoids control type 3 innate lymphoid cells and set the offspring immunity. Nature. 2014 Apr 3; 508(7494):123-7.
12. Walker JA, Clark PA, Crisp A, Barlow JL, Szeto A, Ferreira ACF, et al. Polychromic Reporter Mice Reveal Unappreciated Innate Lymphoid Cell Progenitor Heterogeneity and Elusive ILC3 Progenitors in Bone Marrow. Immunity. 2019 Jul 16;51(1):104-118.e7
13. Sheikh A, Abraham N. Interleukin-7 Receptor Alpha in Innate Lymphoid Cells: More Than a Marker. Front Immunol. 2019 Dec 11; 10:2897.
14. Possot C, Schmutz S, Chea S, Boucontet L, Louise A, Cumano A, et al. Notch signalling is necessary for adult, but not fetal, development of RORγt(+) innate lymphoid cells. Nat Immunol. 2011 Sep 11; 12(10):949-58.
15. Luther C, Warner K, Takei F. Unique progenitors in mouse lymph node develop into CD127+ NK cells: thymus-dependent and thymus-independent pathways. Blood. 2011 Apr 14; 117(15):4012-21.
16. Hashemi E, Malarkannan S. Tissue-Resident NK Cells: Development, Maturation, and Clinical Relevance. Cancers (Basel). 2020 Jun 12; 12(6):1553.
17. Fuchs A, Vermi W, Lee JS, Lonardi S, Gilfillan S, Newberry RD, et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity. 2013 Apr 18; 38(4):769-81.
18. Spits H, Mjösberg J. Heterogeneity of type 2 innate lymphoid cells. Nat Rev Immunol. 2022 Nov; 22(11):701-12.
19. Melo-Gonzalez F, Hepworth MR. Functional and phenotypic heterogeneity of group 3 innate lymphoid cells. Immunology. 2017 Mar; 150(3):265-75.
20. D'Aversa E, Salvatori F, Vaccarezza M, Antonica B, Grisafi M, Singh AV, et al. circRNAs as Epigenetic Regulators of Integrity in Blood-Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis. Cells. 2024 Aug 6; 13(16):1316.
21. Dadi S, Chhangawala S, Whitlock BM, Franklin RA, Luo CT, Oh SA, et al. Cancer Immunosurveillance by Tissue-Resident Innate Lymphoid Cells and Innate-like T Cells. Cell. 2016 Jan 28; 164(3):365-77.
22. Chung DC, Jacquelot N, Ghaedi M, Warner K, Ohashi PS. Innate Lymphoid Cells: Role in Immune Regulation and Cancer. Cancers (Basel). 2022 Apr 21; 14(9):2071.
23. Strober W. The LTi cell, an immunologic chameleon. Immunity. 2010 Nov 24;33(5):650-2.
24. Zhong C, Zheng M, Zhu J. Lymphoid tissue inducer-A divergent member of the ILC family. Cytokine Growth Factor Rev. 2018 Aug;42:5-12.
25. Withers DR. Lymphoid tissue inducer cells. Curr Biol. 2011 May 24;21(10):R381-2.
26. Groeneveld CS, Fontugne J, Cabel L, Bernard-Pierrot I, Radvanyi F, Allory Y, et al. Tertiary lymphoid structures marker CXCL13 is associated with better survival for patients with advanced-stage bladder cancer treated with immunotherapy. Eur J Cancer. 2021 May;148:181-9.
27. Gago da Graça C, van Baarsen LGM, Mebius RE. Tertiary Lymphoid Structures: Diversity in Their Development, Composition, and Role. J Immunol. 2021 Jan 15;206(2):273-81.
28. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017 Apr 19;9(1):34.
29. Wołącewicz M, Hrynkiewicz R, Grywalska E, Suchojad T, Leksowski T, Roliński J, et al. Immunotherapy in Bladder Cancer: Current Methods and Future Perspectives. Cancers (Basel). 2020 May 7;12(5):1181.
30. Mukherjee N, Ji N, Hurez V, Curiel TJ, Montgomery MO, Braun AJ, et al. Intratumoral CD56bright natural killer cells are associated with improved survival in bladder cancer. Oncotarget. 2018 Nov 23;9(92):36492-502.
31. Sivori S, Vacca P, Del Zotto G, Munari E, Mingari MC, Moretta L. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol. 2019 May;16(5):430-41.
32. Mukherjee N, Ji N, Tan X, Lin CL, Rios E, Chen CL, et al. Bladder tumor ILC1s undergo Th17-like differentiation in human bladder cancer. Cancer Med. 2021 Oct;10(20):7101-10.
33. Schneider AK, Domingos-Pereira S, Cesson V, Polak L, Fallon PG, Zhu J, et al. Type 2 innate lymphoid cells are not involved in mouse bladder tumor development. Front Immunol. 2024 Jan 12;14:1335326.
34. Chevalier MF, Trabanelli S, Racle J, Salomé B, Cesson V, Gharbi D, et al. ILC2-modulated T cell-to-MDSC balance is associated with bladder cancer recurrence. J Clin Invest. 2017 Aug 1;127(8):2916-29.
35. Seehus CR, Kadavallore A, Torre B, Yeckes AR, Wang Y, Tang J, et al. Alternative activation generates IL-10 producing type 2 innate lymphoid cells. Nat Commun. 2017 Dec 1;8(1):1900.
36. Ye L, Jin K, Liao Z, Xiao Z, Xu H, Lin X, et al. Hypoxia-reprogrammed regulatory group 2 innate lymphoid cells promote immunosuppression in pancreatic cancer. EBioMedicine. 2022 May;79:104016.
37. Saraiva M, O'Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol. 2010 Mar;10(3):170-81.
38. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009 Jan 16;206(1):35-41.
39. Kang W, Feng Z, Luo J, He Z, Liu J, Wu J, et al. Tertiary Lymphoid Structures in Cancer: The Double-Edged Sword Role in Antitumor Immunity and Potential Therapeutic Induction Strategies. Front Immunol. 2021 Jul 29;12:689270.
40. Chen J, Chen J, Wang L. Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes. J Zhejiang Univ Sci B. 2022 Oct 15;23(10):812-22.
41. Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008 Sep 20;26(27):4410-7.
42. Meylan M, Petitprez F, Becht E, Bougoüin A, Pupier G, Calvez A, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity. 2022 Mar 8;55(3):527-41.e5.
43. Singh AV. Commentary on "Peptide-Conjugated Nanoparticles as Targeted Anti-angiogenesis Therapeutic and Diagnostic in Cancer" by Shaker A. Mousa, Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States - Peptide-Conjugated Nanoparticles for Multimodal Nanomedicine. Curr Med Chem. 2020;27(17):2927-8.
44. Laurie SJ, Foster JP 2nd, Bruce DW, Bommiasamy H, Kolupaev OV, Yazdimamaghani M, et al. Type II innate lymphoid cell plasticity contributes to impaired reconstitution after allogeneic hematopoietic stem cell transplantation. Nat Commun. 2024 Jul 17;15(1):6000.
45. Kitamura H, Torigoe T, Honma I, Sato E, Asanuma H, Hirohashi Y, et al. Effect of human leukocyte antigen class I expression of tumor cells on outcome of intravesical instillation of bacillus calmette-guerin immunotherapy for bladder cancer. Clin Cancer Res. 2006 Aug 1;12(15):4641-4.
46. Ferreira-Teixeira M, Paiva-Oliveira D, Parada B, Alves V, Sousa V, et al. Natural killer cell-based adoptive immunotherapy eradicates and drives differentiation of chemoresistant bladder cancer stem-like cells. BMC Med. 2016 Oct 21;14(1):163.
47. Lian G, Mak TS, Yu X, Lan HY. Challenges and Recent Advances in NK Cell-Targeted Immunotherapies in Solid Tumors. Int J Mol Sci. 2021 Dec 23;23(1):164.
48. Wang YA, Ranti D, Bieber C, Galsky M, Bhardwaj N, Sfakianos JP, et al. NK Cell-Targeted Immunotherapies in Bladder Cancer: Beyond Checkpoint Inhibitors. Bladder Cancer. 2023 Jun 27;9(2):125-39.
49. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2021 Sep 24. Identifier NCT04064190, Vactosertib With Durvalumab in Urothelial Carcinoma Failing Checkpoint Inhibition. Available from: https://clinicaltrials.gov/study/NCT04064190.
50. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2024 Sep 26. Identifier NCT01352962, Gemcitabine, Carboplatin, and Lenalidomide for Treatment of Advanced/Metastatic Urothelial Cancer and Other Solid Tumors. Available from: https://clinicaltrials.gov/study/NCT01352962.
51. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2019 Aug 06. Identifier NCT01373294, Oral Lenalidomide and Intravesical BCG for Therapy of Bladder Cancer. Available from: https://clinicaltrials.gov/study/NCT01373294.
52. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2019 Apr 23. Identifier NCT01342172, Gemcitabine, Cisplatin, Plus Lenalidomide as First-line Therapy for Patients With Metastatic Urothelial Carcinoma. Available from: https://clinicaltrials.gov/study/NCT01342172.
53. Sanborn RE, Pishvaian MJ, Callahan MK, Weise A, Sikic BI, Rahma O, et al. Safety, tolerability and efficacy of agonist anti-CD27 antibody (varlilumab) administered in combination with anti-PD-1 (nivolumab) in advanced solid tumors. J Immunother Cancer. 2022 Aug; 10(8):e005147.
54. Burris HA, Infante JR, Ansell SM, Nemunaitis JJ, Weiss GR, Villalobos VM, et al. Safety and Activity of Varlilumab, a Novel and First-in-Class Agonist Anti-CD27 Antibody, in Patients With Advanced Solid Tumors. J Clin Oncol. 2017 Jun 20; 35(18):2028-36.
55. Sanmamed MF, Pastor F, Rodriguez A, Perez-Gracia JL, Rodriguez-Ruiz ME, Jure-Kunkel M, et al. Agonists of Co-stimulation in Cancer Immunotherapy Directed Against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol. 2015 Aug; 42(4):640-55.
56. Grivas P, Koshkin VS, Chu X, Cole S, Jain RK, Dreicer R, et al. PrECOG PrE0807: A Phase 1b Feasibility Trial of Neoadjuvant Nivolumab Without and with Lirilumab in Patients with Muscle-invasive Bladder Cancer Ineligible for or Refusing Cisplatin-based Neoadjuvant Chemotherapy. Eur Urol Oncol. 2024 Aug; 7(4):914-22.
57. Jiang T, Zhou C, Ren S. Role of IL-2 in cancer immunotherapy. Oncoimmunology. 2016 Apr 25; 5(6):e1163462.
58. Wang KS, Frank DA, Ritz J. Interleukin-2 enhances the response of natural killer cells to interleukin-12 through up-regulation of the interleukin-12 receptor and STAT4. Blood. 2000 May 15;95(10):3183-90.
59. Chandrasekar V, Ansari MY, Singh AV, Uddin S, Prabhu KS, Dash S, et al. Investigating the use of machine learning models to understand the drugs permeability across placenta. IEEE Access. 2023 May 4; 11:52726-39.
60. Liu Q, You B, Meng J, Huang CP, Dong G, Wang R, et al. Targeting the androgen receptor to enhance NK cell killing efficacy in bladder cancer by modulating ADAR2/circ_0001005/PD-L1 signaling. Cancer Gene Ther. 2022 Dec;29(12):1988-2000.
61. Liu L, Hou Y, Deng C, Tao Z, Chen Z, Hu J, et al. Single cell sequencing reveals that CD39 inhibition mediates changes to the tumor microenvironment. Nat Commun. 2022 Nov 8;13(1):6740.
62. Huyan T, Gao L, Gao N, Wang C, Guo W, Zhou X, et al. miR-221-5p and miR-186-5p Are the Critical Bladder Cancer Derived Exosomal miRNAs in Natural Killer Cell Dysfunction. Int J Mol Sci. 2022 Dec 2; 23(23):15177.
63. Moral JA, Leung J, Rojas LA, Ruan J, Zhao J, Sethna Z, et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature. 2020 Mar; 579(7797):130-5.
64. Castillo-González R, Valle-Noguera A, Gomez-Sánchez MJ, Xia P, Cruz-Adalia A. Innate lymphoid cells type 3 in cancer. Front Immunol. 2022 Oct 13; 13:1033252.
65. Bal Bernink JH, Nagasawa M, Groot J, Shikhagaie MM, Golebski K, et al. IL-1β, IL-4 and IL-12 control the fate of group 2 innate lymphoid cells in human airway inflammation in the lungs. Nat Immunol. 2016 Jun; 17(6):636-45.
66. FDA accepts ImmunityBio’s BLA resubmission as complete and sets new PDUFA date. ImmunityBio.
67. Chen W, Liu N, Yuan Y, Zhu M, Hu X, Hu W, et al. ALT-803 in the treatment of non-muscle-invasive bladder cancer: Preclinical and clinical evidence and translational potential. Front Immunol. 2022 Nov 10; 13:1040669.
68. Ramesh N, Ge Y, Ennist DL, Zhu M, Mina M, Ganesh S, et al. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor--armed oncolytic adenovirus for the treatment of bladder cancer. Clin Cancer Res. 2006 Jan 1; 12(1):305-13.
69. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2024 Aug 15. Identifier NCT04452591, Study of Cretostimogene Given in Patients With Non-Muscle Invasive Bladder Cancer, Unresponsive to Bacillus-Calmette-Guerin (BOND-003). Available from: https://clinicaltrials.gov/study/NCT04452591.
70. National Cancer Institute (U.S.). Durvalumab. 2017.
71. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2021 Feb 08. Identifier NCT02891161, Durvalumab And Radiation Therapy Followed by Adjuvant Durvalumab in Patients With Urothelial Cancer (T2-4 N0-2 M0) of the Bladder (DUART). Available from: https://clinicaltrials.gov/study/NCT02891161.
72. Agarwal N, Apolo AB, Tsao CK, Lee KM, Godbold JH, Soto R, et al. Phase Ib/II trial of gemcitabine, cisplatin, and lenalidomide as first-line therapy in patients with metastatic urothelial carcinoma. Oncologist. 2014 Sep; 19(9):915-6.
73. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2024 Jul 19. Identifier NCT04198766, Study of INBRX-106 and INBRX-106 in Combination With Pembrolizumab (Keytruda®) in Subjects With Locally Advanced or Metastatic Solid Tumors (Hexavalent OX40 Agonist). Available from: https://clinicaltrials.gov/study/NCT04198766.
74. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2022 Feb 14. Identifier NCT02559024, Anti-OX40 Antibody (MEDI6469) in Patients With Metastatic Colorectal Cancer. Available from: https://clinicaltrials.gov/study/NCT02559024.
75. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2019 May 31. Identifier NCT03029832, A Study of MOXR0916 in Combination With Atezolizumab Versus Atezolizumab Alone in Participants With Untreated Locally Advanced or Metastatic Urothelial Carcinoma Who Are Ineligible for Cisplatin-Based Therapy. Available from: https://clinicaltrials.gov/study/NCT03029832.
76. Hamid O, Chiappori AA, Thompson JA, Doi T, Hu-Lieskovan S, Eskens FALM, et al. First-in-human study of an OX40 (ivuxolimab) and 4-1BB (utomilumab) agonistic antibody combination in patients with advanced solid tumors. J Immunother Cancer. 2022 Oct; 10(10):e005471.
77. Söhngen C, Thomas DJ, Skowron MA, Bremmer F, Eckstein M, Stefanski A, et al. CD24 targeting with NK-CAR immunotherapy in testis, prostate, renal and (luminal-type) bladder cancer and identification of direct CD24 interaction partners. FEBS J. 2023 Oct; 290(20):4864-6.
78. National Cancer Institute (U.S.). Aldesleukin.
79. Davar D, Zappasodi R, Wang H, Naik GS, Sato T, Bauer T, et al. Phase IB Study of GITR Agonist Antibody TRX518 Singly and in Combination with Gemcitabine, Pembrolizumab, or Nivolumab in Patients with Advanced Solid Tumors. Clin Cancer Res. 2022 Sep 15; 28(18):3990-4002.
80. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2020 Oct 05. Identifier NCT02253992, An Investigational Immuno-therapy Study to Determine the Safety of Urelumab Given in Combination With Nivolumab in Solid Tumors and B-cell Non-Hodgkin's Lymphoma. Available from: https://clinicaltrials.gov/study/NCT02253992.
81. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2017 Apr 19. Identifier NCT02110082, Combination Study of Urelumab and Cetuximab in Patients With Advanced/Metastatic Colorectal Cancer or Advanced/Metastatic Head and Neck Cancer. Available from: https://clinicaltrials.gov/study/NCT02110082.
82. Hong DS, Gopal AK, Shoushtari AN, Patel SP, He AR, Doi T, et al. Utomilumab in Patients With Immune Checkpoint Inhibitor-Refractory Melanoma and Non-Small-Cell Lung Cancer. Front Immunol. 2022 Aug 2; 13:897991.
83. Malek E, Rana PS, Swamydas M, Daunov M, Miyagi M, Murphy E, et al. Vactosertib, a novel TGF-β1 type I receptor kinase inhibitor, improves T-cell fitness: a single-arm, phase 1b trial in relapsed/refractory multiple myeloma. Res Sq [Preprint]. 2023 Jul 17:rs.3.rs-3112163.
84. Metropulos AE, Munshi HG, Principe DR. The difficulty in translating the preclinical success of combined TGFβ and immune checkpoint inhibition to clinical trial. EBioMedicine. 2022 Dec;86:104380.
85. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2019 Dec 09. Identifier NCT02335918, A Dose Escalation and Cohort Expansion Study of Anti-CD27 (Varlilumab) and Anti-PD-1 (Nivolumab) in Advanced Refractory Solid Tumors. Available from: https://clinicaltrials.gov/study/NCT02335918.