Abstract
CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily of receptors expressed on a variety of cell types. The CD40-CD40L interaction gives rise to many immune events, including the licensing of dendritic cells to activate CD8+ effector T cells, as well as the facilitation of B cell activation, proliferation, and differentiation. In malignant cells, the expression of CD40 varies among cancer types, mediating cellular proliferation, apoptosis, survival and the secretion of cytokines and chemokines. Agonistic human anti-CD40 antibodies are emerging as an option for cancer treatment, and early-phase clinical trials explored its monotherapy or combination with radiotherapy, chemotherapy, immune checkpoint blockade, and other immunomodulatory approaches. In this review, we present the current understanding of the mechanism of action for CD40, along with results from the clinical development of agonistic human CD40 antibodies in cancer treatment (selicrelumab, CDX-1140, APX005M, mitazalimab, 2141-V11, SEA-CD40, LVGN7409, and bispecific antibodies). This review also examines the safety profile of CD40 agonists in both preclinical and clinical settings, highlighting optimized dosage levels, potential adverse effects, and strategies to mitigate them.
Keywords: CD40, molecular mechanism, agonistic antibodies, toxicity, cancer
Graphical Abstract

1. Introduction
Multiple signals are necessary for naïve T cells to differentiate into effector T cells in an adaptive immune response. Antigen-presenting cells (APCs), including dendritic cells (DCs), B cells, and macrophages, are essential for the delivery of primary signals to T cell receptors (TCRs) and secondary signals from costimulatory molecules. Additional tertiary signals include the secretion of cytokines, which modulate the activation and skew the differentiation of effector cells, including T cells [1].
One key coordinator in the activation process of effector cells is CD40, a costimulatory receptor molecule belonging to the tumor necrosis factor (TNF) receptor superfamily [1]. CD40 is mainly expressed in B lymphocytes, DCs, and monocytes [2]. The ligand of CD40, CD40L (also known as CD154), is primarily expressed by activated T cells. To maintain immune homeostasis, naïve T regulatory cells express low basal levels of CD40L, which can be upregulated upon activation [3]. In addition, ligation of CD40 by activated CD4+ T cells “license” DCs for CD8+ effector T cell activation [4]. The CD40 ligation on DCs also upregulates the cell surface expression of major histocompatibility complex (MHC), and the production of proinflammatory cytokines, such as IL-12 [5]. With viral infection, CD40L upregulation on DCs may directly prime CD8+ T cells independent of CD4+ T cells, promoting cytotoxic T lymphocyte (CTLs) responses [6]. For B cells, CD40 ligation upregulates costimulatory molecules, for example, B7-1/2, which reciprocally prime effector T cell activation [4]. CD40L is also expressed on platelets, and under inflammatory conditions, is induced in natural killer cells [7], mast cells, and basophils [8].
CD40 activation may modulate the tumor microenvironment (TME) independent of canonical innate immune sensors, such as Toll-like receptors (TLRs), inflammasomes, and stimulator of interferon genes (STING) [9]. CD40 knockout mice exhibited impaired T cell priming of B cells to class switching or to germinal center formation [10], and also exhibited significantly higher occurrence of spontaneous tumors [11]. In epithelial cancer and melanoma cells, CD40L ligation to CD40 mediates immunogenic cell death that leads to the activation of dendritic cells and CTLs in the TME [12, 13]. CD40 activation also extends efficiency to immune checkpoint blockade (ICB) in “cold” tumors that are typically resistant to checkpoint inhibitors, such as pancreatic cancer and melanoma tumors [13, 14]. Thus, CD40-CD40L interaction plays a critical role in activating the anti-tumor immune responses, which sparked CD40 agonism as a promising target for cancer immunotherapy. More than 23 years ago, agonistic CD40 antibodies were shown to mimic CD40L and initiate downstream anti-tumor immune signaling, particularly activation of CTLs [15]. Although toxicity issues hindered this agonist approach as a monotherapy, CD40 monoclonal antibodies have recently demonstrated their effectiveness in vivo for cancer therapy, especially when used in conjunction with other treatments [16]. Together, CD40-activating monoclonal antibody promotes CD40 agonism as a potentially significant yet unexplored means to broaden the scope of existing cancer immunotherapy.
2. CD40/CD40L downstream signaling mechanism
CD40 largely relies on the recruitment of the TNF receptor-associated factor (TRAF) family to transduce a wide range of downstream signals (Figure 1). TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6 have been reported to bind to CD40, among which TRAF2, TRAF3, and TRAF6 most significantly contribute to CD40 intracellular signaling [17]. The most well-documented CD40 signaling pathway is the non-canonical nuclear factor kappa-B (NF-κB) pathway. Induction of this pathway involves NF-κB-inducing kinase (NIK)-mediated activation of inhibitor of NF-κB kinase α (IKKα). Subsequently, IKKα triggers selective proteolysis of p100 to produce p52, which then promotes the transcription of target genes [18]. Another CD40 downstream signaling pathway is the canonical NF-κB pathway. With NF-κB activator 1 (Act1) acting as an adaptor, diverse immune receptors activate a trimeric IκB kinase (IKK) complex (IKKα/β/γ) through the kinase TGFβ-activated kinase 1 (TAK1) [19]. Subsequently, the IKK complex mediates NF-κB inhibitor (IκB) degradation, leading to nuclear translocations of p50/p65 and p65/p65 dimers [18]. In B cells, NF-κB signaling promotes activation, differentiation, and proliferation [17, 18]. While in DCs, activation of the non-canonical NF-κB pathway is critical for DC maturation [20] and NIK-mediated cross-priming of CTLs [21]. Other signal transduction pathways mediated by the CD40-TRAF complex include the three mitogen-activated protein kinase (MAPK) pathways - the extracellular signal-regulated kinases (ERKs) pathway, the c-Jun N-terminal kinases (JNKs) pathway, and the p38 pathway [19]. Differential activation of MAPKs leads to the activation of a unique set of transcription factors, resulting in distinct biological responses [4, 20]. In B cells, CD40-dependent activation of JNK and p38 can lead to proliferation, antibody production, and Ig switching [22]. In DCs, p38 MAPK signaling is crucial for IL-12 production that then promotes differentiation of T helper type 1 (Th1) lymphocytes [23, 24]. Another well-known downstream pathway of CD40-TRAF interaction is the phosphoinositide 3-kinase (PI3K) pathway. In B cells, CD40-PI3K regulates most cell fate decisions, including the development and differentiation of bone marrow precursors and B cell receptor (BCR)-mediated proliferation [25]. In monocytes, CD40-triggered PI3K-AKT signaling plays a role in regulating the production of TNF-α and Interleukin-10 (IL-10) [26]. In DCs, the CD40-TRAF6-mediated PI3K/AKT pathway promotes cell survival via suppressing apoptosis factors, such as BCL2-associated agonist of cell death (BAD), caspase 3, and caspase 9 [24].
Figure 1. Downstream signaling pathways of CD40/CD40L.
CD40 ligation initiates diverse downstream signaling pathways. Independent of TRAF mediation, CD40 activates the Janus kinase 3 (JAK3), phosphorylating and activating the signal transducer and activator of the transcription 5 (STAT5). Several pathways are TRAF-dependent. In the canonical nuclear factor kappa-B (NF-κB) pathway mediated by the NF-κB activator 1 (Act1) and kinase TGFβ-activated kinase 1 (TAK1), activation of the trimeric IκB kinase (IKK) complex (IKKα/β/γ) mediates NF-κB inhibitor (IκB) degradation, leading to nuclear translocations of the p50/p65 dimers. Induction of the non-canonical NF-κB pathway involves NF-κB-inducing kinase (NIK)-mediated activation of the inhibitor of NF-κB kinase α (IKKα), which triggers selective proteolysis of p100 to produce p52 and promotes transcription of p52/RelB. Three mitogen-activated protein kinase (MAPK) pathways are activated by CD40 ligation, including the extracellular signal-regulated kinases (ERKs) pathway, the c-Jun N-terminal kinases (JNKs) pathway, and the p38 pathway. The TRAF6-mediated phosphoinositide 3-kinase (PI3K)- protein kinase B (AKT) pathway also plays a crucial role via suppressing caspase-9/caspase3- associated apoptosis. CD40 ligation on cancer cells can lead to direct apoptosis via two distinct pathways downstream of JNK activation. In the intrinsic pathway, JNK activates the cytochrome c/caspase-9/caspase-3-associated intrinsic apoptosis in a caspase-8-dependent manner. In the extrinsic pathway, JNK triggers the intracellular TNF-related apoptosis-inducing ligand (TRAIL) induction, initiating caspase-10-associated apoptosis.
In addition to TRAF-mediated pathways, CD40 also activates the Janus kinase 3 (JAK3), which then phosphorylates and activates the signal transducer and activator of the transcription 5 (STAT5) [27]. In B cells, upon CD40 stimulation, JAK3 phosphorylation does not occur, indicating that JAK may not be crucial for CD40 signaling in B cells. However, monocytes induce JAK3 phosphorylation after CD40 engagement, inducing STAT5 dimerization and translocation to trigger targeted gene transcription [27]. In DCs, CD40-dependent JAK/STAT activation induces the Th1-polarizing cytokine IL-12, IFN-γ, and costimulatory molecule expressions, influencing the maturation of monocyte-derived dendritic cells [28].
In general, it is evident that the CD40/CD40L downstream signaling effects vary depending on cell types (Figure 1).
3. CD40/CD40L upstream signaling mechanism
Compared to CD40 downstream signaling pathways, less is known about the upstream signals regulating the CD40 expression [29] (Table 1). CD40 has a TATA-less promoter that contains four NF-κB sites and three IFN-γ activation sites (GAS) elements [30]. Mechanisms of induction of CD40 vary among cell types. In human macrophages and microglia, CD40 transcription is promoted through the canonical NF-κB pathway and STAT1 activity. LPS induces the production of IFNβ, which activates STAT-1α. Moreover, LPS induces the acetylation and phosphorylation of H3 and H4, facilitating recruitment of NF-κB, STAT-1α, and RNA polymerase II to the CD40 promoter in a time-dependent manner [30]. In human macrophages, IL-6 can induce CD40 expression through upregulating STAT3 and hypoxia-inducible factor (HIF)-1α [31]. In mouse macrophages, transcription factors such as STAT-1α and ETS (PU.1, SP1-B) have been shown to mediate CD40 expression after IFN-γ stimulation [32]. After LPS stimulation, CD40 expression is upregulated by NF-κB in a p65-dependent manner [33]. In human DCs, CD40 is transcriptionally regulated by RUNX1 and RUNX3 and by ribonucleotide binding proteins RPL26, RPL4, RPL8, and RPS9 [34, 35]. In contrast, arginine and serine-rich coiled-coil 2 (RSRC2) were shown to suppress CD40 expression [35]. In human B cells, AKNA, a human AT-hook protein, has been shown to bind to the A/T-rich promoter regions of CD40 and CD40 ligand (CD40L) to stimulate expression of both ligand and receptor [36]. Moreover, disruptions in the binding of EBF1 and STAT1 can significantly decrease CD40 promoter activity in human B cells, indicating their stimulatory roles in CD40 expression [37].
Table 1.
Transcription factors upregulating CD40
| Cell Type | Origin | Transcription Factor | Induced by | References |
|---|---|---|---|---|
| Macrophages and Microglia | Human | NF-κB STAT-1α RNA polymerase II |
LPS | [30] |
| Macrophages | Mouse | NF-κB PU.1, Spi-B STAT-1α Sp1 |
LPS IFNγ IFNγ or LPS-induced IFNβ N/A |
[33] [32] [30] [32] [33] |
| Dendritic Cells | Human | RUNX1 and RUNX3 RPL26, RPL4, RPL8, RPS9 |
LPS or TNFα or SAG N/A |
[34] [35] |
| B cell | Human | AKNA EBF1, STAT1 |
N/A | [36] [37] |
| Malignant Melanoma | Human | N/A | IFNγ, TNFα | [40] |
NF-κB: nuclear factor kappa-B; STAT-1α: signal transducers and activators of transcription 1α; RUNX1: RUNX family transcription factor 1; RUNX3: RUNX family transcription factor 3; RPL26: ribosomal protein L26; RPL4: ribosomal protein L4; RPL8: ribosomal protein L8; RPS9: ribosomal protein S9; AKNA: AT-Hook transcription factor; EBF1: EBF transcription factor 1; STAT-1: signal transducers and activators of transcription 1; LPS: bacterial lipopolysaccharides; IFNγ: interferon gamma; IFNβ: interferon beta; TNFα: tumor necrosis factor α; SAG: superantigen.
Apart from inducing signals, several signals were found to repress CD40 activation in mouse models. While IFN-γ is a potent inducer of CD40 expression in macrophages, IL-4, a Th2-derived cytokine that acts antagonistically to IFN-γ, was found to suppress IFNγ-induced CD40 gene expression in macrophages and microglia, in a STAT6-dependent manner [38]. Moreover, another immunosuppressive cytokine named transforming growth factor-β (TGF-β) was reported to inhibit IFN-γ-induced CD40 in microglia by enhancing the degradation of CD40 mRNA [39]. In mouse macrophages, SP1 was shown as a critical regulator by which phosphorylation can downregulate basal CD40 gene expression [33].
In human malignant melanoma cell lines, CD40 expression is enhanced by IFN-γ and TNF-α [40]. However, the transcription factors regulating CD40 expression in malignant cells remain poorly defined.
4. Role of CD40 activation in immune cells
On immature DCs, CD40 ligation stimulates the production of inflammatory cytokines, such as IL-1α, IL-1β, TNF-α, IL-6, and IL-8 [1, 41] (Figure 2). CD40 activation also upregulates the expression of costimulatory molecules such as CD80, CD86, and adhesion molecules such as CD54 (ICAM-1) and CD58 (LFA-30) [24, 41]. The costimulatory signals can “license” DCs to present exogenous antigens in the context of MHC-I molecules, and thus cross-present to CD8+ T cells[4]. Moreover, it has recently been shown that the CD40L-expressing CD8+ T cells may upregulate IL-12 production by DCs via CD40L-CD40 interactions, which forms a positive-feedback loop to promote their own proliferation and differentiation [42]. Mature DCs and macrophages secrete IL-12, which upon CD40L stimulation, results in sustained polarization to Th1 response and activation of NK-cell antitumor responses [23]. In macrophages, CD40 ligation stimulates a tumoristatic effect through increasing IFN-γ, TNF-α, T-cell dependent nitric oxide production, and antibody-dependent cellular cytotoxicity (ADCC). Also, CD40-activated macrophages can induce apoptosis in tumor cells in vitro, such as in mouse lymphoma cells (L5178Y) [43]. CD40 is constitutively expressed on platelets and its ligation is critical for platelet homeostasis and activation during inflammation [44]. In B cells, CD40 ligation promotes germinal center formation, where B cells undergo differentiation and proliferation through clonal expansion, somatic hypermutation, and immunoglobin class switching, resulting in high-affinity antibody production [17, 18].
Figure 2. The role of CD40-CD40L interaction on immune and malignant cells.
Schematic of the effect of CD40 ligation by membrane-bound (mCD40L) or soluble (sCD40L) form of CD40L on immune and cancer cells.
CD40 stimulation can also be generated by a soluble form of CD40L (sCD40L), which co-exists with the membrane-bound form of CD40L (mCD40L) [45]. The trimeric sCD40L can be generated by enzymatic cleavage of mCD40L by matrix metalloproteinases (MMPs) [45]. Activation by sCD40L triggers trimerization of the receptor, which can activate both the canonical and non-canonical NF-κB pathways [46]. The human native sCD40L is biologically active [47] and generates similar activities as mouse sCD40L, such as promoting B cell differentiation and proliferation [48]. In contrast, stimulation of endothelial cells by platelet-derived sCD40L promotes tumor angiogenesis, shedding light on potential negative effects with delivery of sCD40L as a therapeutic (discussed later) [49]. Early clinical testing of sCD40L did not produce positive results [50], which may due to the fact that sCD40L activity was dependent on FcγR-mediated cross-linking [51], and CD40-expressing immune cells were eliminated by ADCC [52]. Recently, hexavalent forms of sCD40L, such as the hexavalent receptor agonist (HERA), were developed to improve drug efficiency by circumventing the need for FcγR. HERA fusion proteins comprise three receptor-binding domains in a single chain arrangement linked to an Fc-silenced human IgG, resulting in a hexavalent molecule [52]. HERA-CD40L enhances activation of signal transduction in DCs via signaling through the NF-κB, AKT, p38, ERK1/2, JNK, and STAT1 pathways. This helps establish robust antitumor immune responses both in vitro and in vivo via increasing intratumor CD8+ T cell and promoting macrophage polarization from the pro-tumor M2 macrophages to antitumor M1 macrophages [52, 53].
5. Role of CD40 activation in malignant cells
CD40 is expressed in a variety of malignant cells, and based on tumor cell types, CD40-CD40L ligation differentially mediates cell survival, proliferation, and apoptosis [54, 55] (Figure 2). In malignant B cells, low-level constitutive CD40 engagement has been shown to promote sustained proliferation, such as in non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL) cells [54, 56]. Related mechanisms include upregulating the expression of anti-apoptotic factors, such as Bcl-XL and TNF-α-induced protein 3 (TNF-A IP3, or A20) [57, 58]. In fact, TNF-A IP3 can suppress inflammatory and autoimmune responses induced by the NF-kB pathway in B cells, and is critical in maintaining normal B cell differentiation of the marginal zone B and B1 cell subsets [59]. Moreover, CD40 activation induces cytokines with pro-survival activities in B cells, including IL-6, TNF-α, and GM-CSF, that can potentially affect malignant B cell survival, migration, and interaction with other cells in the TME [60]. Paradoxically, CD40-CD40L ligation has also been reported to induce apoptosis in various malignant B cells, for example, in Burkitt lymphoma (BL) [55]. The dual roles of apoptosis and proliferation can be explained by a balance determined by two factors during T cell-B cell interaction. Firstly, higher relative abundance of Th1 cells compared to Th2 cells lead to higher secretion of cytokines such as IL-2 and IFN-γ. This, in turn, triggers the expression of APO-1/FAS on B cells, such as the BL cell line Ramos, thereby promoting apoptosis [61]. Secondly, the timing and quality of BCR stimulation by antigen is crucial. Compared to B cells capturing antigen without BCRs or with desensitized receptors, BCRs stimulated by a sudden burst of new foreign antigen receive full intracellular signaling. The presence of prior BCR signaling, the binding of FasL to Fas on B cells does not eliminate the B cells but instead promotes cell division and proliferation [62].
Similarly, under CD40 stimulation, a variety of carcinoma cells can be induced to undergo apoptosis, including CD40 expressing human malignant melanoma [40], multiple myeloma [63], B-cell lymphoma [64], bladder cancer [15], and colorectal cancer (CRC) [65]. Studies in the epithelial context have unraveled several potential mechanisms of CD40-mediated intrinsic pathway of apoptosis of carcinoma cells. Using membrane-bound CD40L, it is shown that cell death is induced through the upregulation of TRAF3 protein, which activates the JNK pathway, initiating cytochrome c/caspase-9/caspase-3-associated intrinsic apoptosis [65, 66]. Moreover, the TRAF6-mediated activation of caspase-8 is essential for this process [66]. In addition, an extrinsic pathway downstream of JNK activation was reported to operate alongside with the intrinsic apoptosis pathway in CRC, in which JNK triggers the induction of the intracellular TNF-related apoptosis-inducing ligand (TRAIL), initiating caspase-10-asscociated apoptosis. This concurrent action results in a faster apoptosis process compared to other types of carcinoma cells [67].
6. CD40 expression in tumor cells and regulation of the TME
CD40 expression dynamically varies among cancers. Our group investigated the change of CD40 expression in a pan-cancer analysis. Of the twenty-three cancer types screened, CD40 downregulation occurred in eleven, while upregulation occurred in five [68]. Previous studies have also supported the downregulation of CD40 during tumor progression in several cancers, including malignant melanomas and gastric tumors, in which the latter associated with decreased apoptosis and increased proliferation of the immunosuppressive myeloid-derived suppressor cells (MDSC) [40, 69]. Activation of CD40 in cancer cells has frequently been associated with antitumor immunity. Hill et al. (2005) showed that CD40 activation in human cervical carcinoma cells can activate NF-κB and MAPK signaling pathways, upregulate cell surface markers and intracellular molecules to facilitate antigen processing and presentation, and enhance tumor recognition by CTLs [70]. CD40/CD40L expression in melanoma was shown to correlate with not only type-I antitumor T-cell and APC responses in the TME but also a better predicted response to ICB therapy [68]. While CD40 activation or upregulation was beneficial to tumor killing, contrary results were found in lung and oesophageal cancer, where upregulated CD40 expression on cancer cells correlated with poor prognosis [71, 72]. These contradictory results emphasize the potential of CD40 as a specific immunological target and that strategies of CD40 therapeutics need to be customized depending on cancer types [73].
While it is difficult to predict prognosis from CD40 expression levels, DNA methylation of CD40 has been discovered as an innovative noninvasive biomarker for monitoring the prognosis and diagnosis of several types of cancers, including gastric, breast, colorectal, and prostate cancers [74-76]. Co-expressed genes of CD40 have also been examined in three immunotherapy-responsive and/or resistant cancer types (clear cell renal cell carcinomas, cutaneous melanoma, pancreatic adenocarcinomas) (reported in [77]). Eight genes correlated with CD40 expression are shared between all three cancer types: KIAA1949 (a potential tumor suppressor) [78], IL-27 receptor alpha (IL-27RA) (immune regulation) [79], proteasome subunit beta type-8 (PSMB8) (antigen presentation-related proteasome), tripartite motif containing 21 (TRIM21) (antibody receptor involved in various immune responses) [80], vesicle-associated membrane protein 5 (VAMP5) (involved in vesicle activities) [81], caspase 4 (inflammasome activator) [82], thymidine phosphorylase (TP) (involved in tumor proliferation and apoptosis) [83], and syntaxin 4 (STX4) (involved in membrane fusion) [77, 84]. These results entail that CD40 signaling activities are closely related to immune modulation, proliferation, apoptosis, and vesicular trafficking, thus providing insights into co-targets with CD40 agonist-based therapy.
7. CD40 agonists in clinical trials
Several properties modulate the function of agonistic human anti-CD40 antibodies: fragment crystallizable (Fc)-mediated interactions, isotype, epitope, and specificity.
Cross-linking of the IgG1 monoclonal antibody Fc region through interacting with Fc gamma receptors (FcγR) is crucial for effective CD40 downstream signaling events. The clustering of CD40 receptors amplifies downstream signaling triggered by CD40 activation, thus optimizing immunostimulatory effects. Mice possess four distinct IgG Fc receptors, including activating receptors FcγRI, FcγRIII, FcγRIV, and the inhibitory receptor FcγRIIB [51]. Surprisingly, the inhibitory FcγRIIB, that generally plays a suppressive role in the immune system, is indispensable for in vivo antitumor activities. However, in vitro, the activating FcγRs retained effective cross-linking effects, of which the strengths do not correlate with binding affinity. Together, these findings likely reflect the importance of the bioavailability of FcγRIIB in vivo as other activating FcγRs in vitro to enable cross-linking [24]. While FcγR-mediated crosslinking can be achieved, limited evidence supports efficient in vivo crosslinking effects for therapeutic purposes [85]. To maximize therapeutic potential, Fc affinity for FcγRIIB has therefore been optimized in monoclonal antibodies [85-88]. As expected, these Fc-enhanced anti-CD40 agonist antibodies were shown to induce robust antitumor efficacy both in vitro [86] and in vivo [88] and are currently being explored in early-phase clinical trials.
Unlike IgG1 monoclonal antibodies, IgG2 monoclonal antibodies impart agonistic and antitumor activities independent of FcγR [89]. Structural analysis revealed that the IgG2 isotype contains a more rigid hinge region due to differential disulfide bonding, contributing to improved clustering of CD40 receptors [89, 90]. The agonistic potency of IgG2 monoclonal antibodies was further corroborated by isotype switching both in vitro and in vivo. A clinically relevant human CD40 IgG4 antagonist antibody (bleselumab) bearing antagonistic epitopes was transformed in two formats: IgG1 and IgG2. While the IgG1 format retained its antagonism, the IgG2 format was converted to a superagonist, eliciting >4-fold CD8+ T cell expansion via receptor clustering in vivo. Although the underlying mechanism remains unknown, the conversion from an antagonist to a superagonist demonstrated that sufficient FcγR engagement could overcome antagonistic epitopes and improve antitumor activity [90].
More recently, it has been demonstrated that receptor epitope location combines with isotype to determine the biological activity of human anti-CD40 antibodies. As a TNFR, CD40 consists of 4 cysteine-rich domain (CRD) subunits, CRD1-4, that antibodies can selectively target to enhance agonist activity. Regardless of isotype difference and FcγR dependency, antibodies targeting the membrane distal CRD1 domain were shown to induce the highest agonistic activity, while those binding CRD2-4 display antagonistic properties. This is because antibodies that target epitopes distal to the membrane enable access to FcγRs domains with less steric hindrance [91].
Lastly, bispecific antibodies are rapidly growing as a new therapeutic for their ability to directly modulate the TME via targeting both CD40 and tumor antigens [92]. There are currently nine agonistic human anti-CD40 antibodies in clinical development: 4 IgG1 antibodies (APX005M, mitazalimab, 2141-V11, SEA-CD40), 2 IgG2 antibodies (selicrelumab and CDX-1140), one IgG mutant antibody (LVGN7409), and two bispecific antibodies (RO7300490, ABBV-428). This review also discusses other CD40 agonistic therapies currently in clinical stages, including one multidomain protein (MP0317) and three oncolytic viruses (NG-350A, LOAd703, MEM-288).
7.1. Selicrelumab
Selicrelumab (also known as CP-870,893, RO7009789) is a fully human IgG2 antibody that does not depend on crosslinking via the Fc domain to activate CD40 [93]. To date, selicrelumab is the most extensively studied CD40 agonist in clinical trials. It has been tested as monotherapy and in combination with radiotherapy, chemotherapy, immune checkpoint inhibitors (ICI), and anti-angiogenic therapy (reported in [77]). More recently, a phase 1 study evaluated neoadjuvant selicrelumab with or without chemotherapy (nab-paclitaxel and gemcitabine) before surgery in patients with pancreatic ductal adenocarcinoma (PDAC) (NCT02588443). The 1-year OS rate (Median + SE) was 81.8% + 11.8% for patients receiving selicrelumab monotherapy and 100% for patients treated by selicrelumab with chemotherapy, respectively. Selicrelumab was well-tolerated as related adverse events (AE) were mostly mild. Evidence of selicrelumab-induced TME modulation, including T-cell infiltration, DC maturation, macrophage repolarization, and decreased tumor stroma density, was found [94].
7.2. CDX-1140
CDX-140, a human IgG2 monoclonal antibody, drives NF-kB activation. It binds a domain that is outside the CD40L binding site, and its activation mechanism is independent of FcR cross-linking. It is shown to synergize with the naturally expressed CD40L [95]. Data from a phase I trial evaluating CDX-1140 with or without anti-PD-1 therapy (pembrolizumab) have shown a good safety profile at the dosage of 1.5 mg/kg of CDX-1140 in combination with 200 mg of pembrolizumab. Clinical benefits were observed, especially in patients with non-small cell lung cancer (NSCLC), since 80% (4 out of 5) evaluable patients experienced stable disease (SD) , while all had progressive disease (PD) on prior anti-PD-1/L1 based therapies (NCT03329950) [96].
7.3. APX005M
APX005M is a humanized rabbit IgG1 monoclonal CD40 antibody. With a point mutation in the Fc domain, APX005M has enhanced binding affinity with FcγRIIb, facilitating crosslinking of CD40 and mediating ADCC [86]. Multiple early-phase clinical trials evaluated APX005M in combination with immunotherapies. One phase II study evaluated APX005M in combination with chemotherapy (nab-paclitaxel and gemcitabine) with or without anti-PD-1 (nivolumab) in patients with metastatic pancreatic cancer. It was shown that APX005 did not significantly improve OS: in 105 patients, the primary endpoint of 1-year overall survival (OS) was met for nivo/chemo but not for APX005M /chemo or APX005M/nivo/chemo. However, survival after APX005M/chemo correlates with more significant intratumoral CD4+ T cell infiltration, circulating differentiated CD4+ T cells, and APCs [97] (NCT03214250). The first-in-human phase I study evaluated APX005M and an inhibitor of macrophage recruitment, colony-stimulating factor 1 receptor (CSF1R) inhibitor (cabiralizumab), with or without nivolumab in patients with melanoma, kidney cancer, or non–small cell lung cancer resistant to anti-PD-1/PD-L1. In this study, Weiss et al. (2021) demonstrated the safety and pharmacodynamic activity of the dual macrophage-polarizing therapy with or without nivolumab (NCT03502330) [98]. Recently, in a subsequent phase II study, Weiss et al. (2023) further investigated APX005M in combination with nivolumab in patients with metastatic melanoma following disease progression on anti-PD-1. The objective response rate (ORR) was 15%, and the median duration of response was at least 26 months, rendering the drug response safe, durable, and prolonged [99].
7.4. Mitazalimab
Mitazalimab (also known as ADC-1013 and JNJ-64457107) is a human IgG1 monoclonal antibody targeting CD40. It is engineered to have high potency and affinity in binding to CD40 in an FcγR-dependent manner, which benefits intratumoral administration to reduce dosage and adverse immune-related events [87]. Systemic administration of mitazalimab is also beneficial, although this method has been associated with lower antitumor efficacy and higher toxicities than intratumoral delivery [100]. In a phase I study in patients with advanced solid tumors, intravenous administration of 0.075-2 mg/kg of mitazalimab demonstrated a manageable safety profile when administered once every two weeks, both with or without corticosteroid pre-infusion [101]. Although the MTD and RP2D were not determined, the principal DLT reported was drug-induced liver injury (grade-3 ALT/AST + grade-2 bilirubin increase). Mitazalimab exhibits a favorable pharmacokinetic (PK) profile, as demonstrated by target-mediated drug disposition and rapid decline in serum concentrations [102]. Clinical, PK, and pharmacodynamics (PD) assessments demonstrate broad physiologic activation of CD40 after intravenous administration. In fact, through analyzing cell-free RNA from the whole blood of pre- and post-mitazalimab-treated patients, significant CD40-related transcriptional activity was identified in patients without corticoid pre-treatment [103]. Gene signatures related to the activation of B cell and myeloid/DC were upregulated post-treatment, confirming the mitazalimab-triggered CD40 agonism [103] (NCT02829099).
7.5. 2141-V11
2141-V11 is a fully human IgG1 CD40 antibody, a Fc-optimized version of selicrelumab in which the human IgG1 Fc is engineered with 5-point mutations that enhance Fc binding to FcγRIIB [88]. A phase I study evaluated 2141-V11 in patients with solid tumors with metastasis to the skin and demonstrated signs of antitumor activity in both local and distant lesions (NCT04059588). Another phase I study has been investigating the safety and dosage of 2141-V11 monotherapy in patients with non-muscle invasive bladder cancer (NMIBC) who did not respond to standard treatment (NCT05126472). Local intravesical delivery is used to mitigate systemic toxicity [104]. Lastly, phase I study has been examining 2141-V11 in patients with recurrent glioblastoma. Interim data have demonstrated that intratumoral administration of 2141-V11 in combination with immunotoxin (D2C7-IT) via convection-enhanced delivery (CED) was safe and somewhat effective: among the 5 patients enrolled, early signs of tumor response were observed at one patient treated at 0.70 mg, and two patients treated at 2.0 mg (NCT04547777) [105].
7.6. SEA-CD40
SEA-CD40 is a fully human, non-fucosylated IgG1 monoclonal CD40 antibody derived from the normally fucosylated CD40 monoclonal antibody, dacetuzumab. Non-fucosylated antibodies have enhanced binding to the FcγRIIIa receptor, potentially improving the agonistic efficacy. A first-in-human phase I trial evaluated intravenous SEA-CD40 monotherapy in patients with advanced solid tumors and lymphoma. SEA-CD40 infusion demonstrated potent immune activation via inducing cytokines associated with immune activation and trafficking, including interferon (IFN)-γ-inducible protein-10 (IP-10), monocyte chemoattractant protein-1 (MCP-1), monokine induced by IFN-γ (MIG), and macrophage inflammatory protein-1b (MIP-1β). Moreover, trafficking of innate and adaptive immune cells was evident, specifically T cells (CD4+ and CD8+), NK cells, and monocytes. Optimal immune activation was reached at 10–30 μg/kg via intravenous delivery. However, minimal antitumor effect of SEA-CD40 was illustrated with 1.8% (1/56) partial response in a patient with basal cell carcinoma and 1.8% (1/56) complete response in a patient with follicular lymphoma [106]. The Coveler group examined SEA-CD40 in combination with chemotherapy (nab-paclitaxel and gemcitabine) and pembrolizumab in patients with metastatic PDAC. Objective responses were observed in 48% of patients at 10 μg/kg and 38% at 30 μg/kg. The median duration of response was 5.7 months for both dosage groups. This might indicate that the higher dosage of 30 μg/kg can lead to adverse events in patients who are less tolerated, counteracting the potentially higher antitumor outcomes. Overall, the combination of SEA-CD40 at 10 μg/kg with chemotherapy and pembrolizumab demonstrates an acceptable safety profile and evidence of antitumor activity in patients with PDAC (NCT02376699) [107].
7.7. LVGN7409
LVGN7409 is an IgG mutant recombinant monoclonal CD40 antibody with an engineered Fc segment to bind FcγRIIB selectively. LVGN7409 activates CD40 in an Fc-FcγRIIB cross-link dependent manner and thus operates optimally in CD40 and FcγRIIB enriched TME [108]. Currently, there are two phase I studies of LVGN7409 in dose-establishment phases in patients with locally advanced, metastatic, or recurrent/refractory malignancy. The first-in-human phase I study of LVGN7409 has been evaluating LVGN7409 with or without anti-PD-1 antibody (LVGN3616) and/or CD137 agonist antibody (LVGN6051). Intravenous LVGN7409 monotherapy has been well tolerated up to 2 mg/kg (NCT04635995).
7.8. Bispecific antibodies
Bispecific antibodies can target both CD40 and tumor-associated antigens, therefore minimizing off-target effects by selectively modifying the TME.
RO7300490 is a novel bispecific antibody targeting CD40 and fibroblast activating protein (FAP), the latter of which is often over-expressed in tumor tissues and low in healthy tissues. Thus, RO7300490 enables direct remodeling of the TME while potentially avoiding toxicities associated with systemic CD40 activation [85]. Currently, one phase I study is evaluating RO7300490 with or without atezolizumab in patients with advanced solid tumors (NCT04857138).
ABBV-428 is another bispecific antibody that is designed to achieve localized activation of CD40 by interacting with the tumor antigen mesothelin, a cell-surface molecule that in cancers is highly expressed on epithelial mesotheliomas and carcinomas of the pancreas, lung, and ovary [109]. ABBV-428 targets mesothelin via a C-terminal single-chain variable fragment flanking Fc-modified human IgG1 and CD40 via an N-terminal single-chain variable fragment. Simultaneous binding to mesothelin and CD40 is required to activate CD40, leading to potential APC activation and immunomodulation of the Treg and M2-like macrophage in the TME [109]. The first-in-human phase I study evaluated ABBV-428 with or without nivolumab in patients with advanced solid tumors (NCT02955251). From interim data of patients receiving ABBV-428 monotherapy, the best clinical response was SD in 36% of patients treated at the recommended phase II dose (3.6 mg/kg). Toxicities of CD40 agonism, such as cytokine release syndrome were not observed, demonstrating an acceptable safety profile. However, ABBV-428 produced minimal clinical activity as there were no consistent changes from baseline in intratumoral CD8+ T cells, programmed death ligand-1 (PD-L1+) cells, or immune-related gene expression detected post-ABBV-428 treatment [109]. This could be explained by the low mesothelin expression on tumor cells, which would limit tumor accessibility and crosslinking of CD40 by ABBV-428 [110].
7.9. Others
7.9.1. MP0317
A novel therapy, MP0317, is a multidomain-designed ankyrin repeat protein (DARPin®) agonistic to CD40. DARPin® drug candidates are created based on naturally occurring ankyrin repeat domains, which possess properties ideal for uses as scaffold drugs, such as high thermal-dynamic, storage stability, and high solubility. MP0317 is exclusively active in the presence of both FAP and CD40 and thus is expected to preferentially activate T cells in the TME and avoid systemic toxicities [111]. The first-in-human phase I study of MP0317 (NCT05098405) aimed to evaluate its safety/tolerability profile, PK/PD characteristics, and recommended dose for combination therapy in patients with advanced solid tumors. Interim clinical data have confirmed that MP0317 is safe and well-tolerated. However, the lack of proinflammatory circulating cytokines (TNFα, IL-2, IL-6, and IL-8) raised a concern for its antitumor efficacy. However, evidence of colocalization of MP0317 with FAP and CD40 from paired pre- and on-treatment tumor biopsies suggested tumor-localized CD40 interaction [112].
7.9.2. Oncolytic Viruses
Oncolytic virus immunotherapy (OVT) utilizes natural or genetically modified viruses to selectively propagate in cancer cells, leaving healthy cells unharmed. OVT acts by two principles: it directly infects and lyses tumor cells, subsequently triggering antitumor immunity by activating innate and adaptive immune responses. Oncolytic viruses were encoded to express costimulatory molecules that are needed for T cell activation to enhance their antitumor effects [113]. Oncolytic adenovirus (OAd) is one of the most frequently employed OVTs, which utilizes adenovirus, a non-enveloped, double-stranded linear DNA virus that binds and infects tumor cells via the adenoviral fiber knob [114]. This review focuses on one CD40 agonist OAd currently in clinical trials, NG-350A.
NG-350A is an adenoviral tumor-specific immunogene vector that expresses a potent, fully human agonistic anti-CD40 antibody. The tumor-specific immunogene vector bears transgenes selectively to tumors and was clinically validated for intravenous delivery. This platform enables NG-350A to selectively replicate in tumor cells [115]. NG-350A re-educates the immunosuppressive TME while avoiding systemic toxicity [116]. The first-in-human phase 1a study evaluated NG-350A with or without pembrolizumab in patients with metastatic/advanced epithelial tumors (NCT03852511). No objective responses were observed; however, SD was found in 50% of patients treated with NG-350A monotherapy. The treatments were well-tolerated, and elevated peak cytokine responses higher than those of systemic anti-CD40 Abs have demonstrated local immunological tumor impact without CD40-related systemic toxicity [117].
Challenges remain for the success of OAd-based cancer therapies. Scant evidence has shown that OAd could effectively eliminate the tumor stem cells, in which their self-renewal capacity and differentiation potential have contributed to the metastasis of tumor cells and also resistance to radiation or chemotherapy [118].
8. Toxicity
While promising in principle, CD40 agonists have been associated with different levels of toxicities both in the clinic and animal models. As shown in Table 2 of recent clinical trials, several CD40 agonistic antibodies with abundant clinical records – CDX-1140 [96], APX005M [119], mitazalimab [120] – were reported to be well tolerated, with most treatment-related adverse events (TRAE) being grade 1 or 2. In two phase I trials with intravenously delivered selicrelumab and LVGN7409, respectively, CRS and alterations in liver function (elevation in ALT and AST) were transient [94, 121].
Table 2.
Clinical studies of CD40 agonistic antibodies in cancer treatment, alone or in combination with other treatments.
| Therapy | Interventions | Route of Delivery |
Phase | Cancer Type |
Study/ NCT number |
Status | Dosage (mg /kg) |
Outcome | Side Effects |
|---|---|---|---|---|---|---|---|---|---|
| Selicrelumab | w/w.o nab-paclitaxel and + atezolizumab |
Intra venous,(neo | I | PDAC | NCT02588443 [94] | Completed | 0.2 | OS=23.4 mo, median DFS=1 | Neoadjuvant therapy (n=16): grade 1 CRS (56%). Adjuvant therapy (n=13): grade |
| Intratumoral | Ib | B cell lymphoma | NCT03892525 | Terminated | N/A | N/A | N/A | ||
| CDX-1140 | w/w.o pembrolizumab | Infusion | I | Advanced solid tumor | NCT03329950 [96] | Completed | 0.72 or 1.5 Q3 W | CR (4%) SD (36%) |
At 1.5 mg/kg dose level (n=21): arthralgia (62%), fatigue (62%), nausea (48%), diarrhea (48%), vomiting (43%), myalgia (43%), fever (38%), chills (38%), AST increase (38%), bilirubin increase (24%), ALT increase (19%), CRS (19%) |
| + pembrolizumab | Infusion | I/Ib | Epithelial cancer | NCT04520711 | Completed | N/A | N/A | N/A | |
| + TCRT + radiotherapy + CDX-301 + Poly-ICLC |
Intratumoral | I | Metastatic solid tumors | NCT04616248 | Completed | N/A | N/A | N/A | |
| + CDX-301 | Intravenous injection | I | TNBC | NCT05029999 | Recruiting | 1.5 Q4 W | N/A | N/A | |
| + odetiglucan | N/A | I | PDAC | NCT05484011 | Recruiting | 0.7 2 or 1.5 Q3 W | N/A | N/A | |
| + bevacizumab + pembrolizumab + capecitabine + oxaliplatin + pembrolizumab |
N/A | II | Ovarian cancer | NCT05231122 | Not yet recruiting | N/A | N/A | N/A | |
| Intravenous | I/II | Biliary tract cancer | NCT05849480 | Not yet recruiting | 0.36-1.5 Q3 W | N/A | N/A | ||
| APX 005 M | + nab-paclitaxel and gemcitabine w/w.o nivolumab | Intravenous | II | Pancreatic cance | NCT03214250 [97] | Completed | 0.1 and 0.3 Q4 W | Primary endpoint of 1yr OS met for nivo/chemo (57.7%) but not for sotiga/chemo (48.1%) or sotiga/nivo/chemo (41.3%) | n=105: CRS, infusion reactions, thrombocytopenia and elevated liver function (87% total). CRS in nivo/chemo (0%), sotiga/chemo (24%) and sotiga/nivo/chemo (34%) |
| + domvanalimab + zimberelimab |
Infusion | Ib/II | NCT05419479 | Recruiting | N/A | N/A | N/A | ||
| monotherapy | N/A | I | Pediatric braintumors | NCT03389802 | Ongoing | 0.1 Q3 W | N/A | N/A | |
| + cabirali zumab w/w.o nivolumab | Intravenous infusion | Metastatic melanoma, Kidney cancer, NSCLC | NCT03502330 [98] | Ongoing | 0.03 Q2 W | SD (8%) DP (62%) | n=26: asymptomatic elevations of lactate dehydrogenase (100%), creatine kinase (96%), AST (96%), and ALT (73%); periorbital edema (65%); fatigue (50%); DLT of acute respiratory distress syndrome (3.8%) | ||
| w/w.o radiotherapy | Infusion | II | NCT04337931 | Completed | N/A | N/A | N/A | ||
| + nivolumab | N/A | II | Metastastic melanoma | [99] | Ongoing | 0.3 Q3 W | ORR(15%) PR(13%) SD(36.8%) |
n=38: at least one AE (89%); grade 3 AE related to APX005M (13%): pyrexia, chills, nausea, fatigue, pruritus, elevated liver function, rash, vomiting, headache, arthralgia, asthenia, myalgia, and diarrhea | |
| Mitazalimab | w/w.o corticos teroid pre-infusion | Intravenous | I | Advanced solid tumor | NCT02829099 [102] | Completed | 0.075-2 Q2 W | Mitazalimab has shown to be safe and well tolerated at at doses up to 1.2 mg/kg | n=95: gatigue (44.2%), pyrexia (38.9%), pruritus (38.9%), chills (27.4%), and headache (26.3%). grade-3 or higher TEAEs (56.8%): gamma-glutamyl transferase increase (7.4%), AST increase (5.3%), and anemia (5.3%) |
| + mFOLFIRINOX | Intravenous | Ib/II | mPDAC | NCT04888312 [120] | Ongoing | 0.45 - 0.9 | ORR (52.2%) | n=5, grade 1 or 2 AEs (80%) | |
| + MesoPher + modified FOLFIRINOX |
Intravenous after chemo | I | Metastic pancreatic cancer | NCT05650918 | Rrecruiting | 0.075-0.15 - 0.3-0.6 or 1.2 | N/A | N/A | |
| 2141-V11 | monotherapy | Intratumoral | I | Solid tumor | NCT04059588 [128] | Completed | N/A | N/A | N/A |
| monotherapy | Intravesical instillation | I | Nonmuscle invasive bladder cancer | NCT05126472 [104] | Recruiting | 0.012-0.033-0.12 - 1.2 Q1 W | N/A | N/A | |
| + D2C7-IT | Intratumoral, convection enhanced delivery (CED) | I | Malignant glioma | NCT04547777 [105] | Recruiting | 0.012, 0.033, 0.12 0.35 | Early signs of tumor response (25%) | n=8: headache (grade 3=12.5%; grade 2=25%); paresthesia (grade 3=12.5%; grade 2=12.5%); dysphasia (grade 3 =12.5%); pyramidal tract disorder (grade 3=12.5%; grade 2=12.5%); and depressed level of consciousness (grade 2=12.5%) | |
| SEA-CD40 | monotherapy | Intravenous injection | I | Advanced solid tumors, Lymphoma | NCT02376699 [106] | Terminated | 0.0006, 0.003, 0.01, 0.03, 0.045, 0.06 | 0.03mg/kg was well tolerated | n=67: infusion/hypersensitivity reactions (73%) |
| + nab-paclitaxel and gemcitabine + pembrolizumab |
N/A | I | mPDAC | NCT02376699 [107] | 0.01 or 0.03 Q4 W | OR at 0.01 mg/kg (48%) OR at 0.03 mg/kg (38%) | n=61: fatigue (84%), nausea (74%), and neutropenia (67%); grade ≤3 TEAEs: neutropenia (61%), anemia (33%), and thrombocytopenia (20%); TEAE-caused discontinuation of treatment (10%) | ||
| + carboplatin + pemetrexed + pembrolizumab |
N/A | II | NSCLC Melanoma | NCT04993677 | Ongoing | N/A | N/A | N/A | |
| LVGN7409 | w/w.o LVGN3616 w/w.o LVGN3616 + LVGN6051 | Intravenous infusion | I | Locally advanced, metastatic or recurrent/refract orymalignancy | NCT04635995 [121] | Recruiting | 0.01-0.1, then conventional “3 + 3” design from 0.3 | SD(44%) | n=12: infusion related reaction (58%), amylase increased, lipase increase, ALT increase and AST increase (42% each), bilirubin increase, WBC or neutrophil decrease, nausea, and cytokine release syndrome (8.3% each); grade 3 TRAEs (17%): amylase/lipase increase and ALT/AST increase (8.3% each) |
| monotherapy | Intravenous infusion | I | NCT05152212 | Recruiting | N/A | N/A | N/A | ||
| RO7300490 | w/w.o atezolizumab | I | Intravenous | Advanced solid tumor | NCT04857138 | Recruiting | N/A | N/A | N/A |
| ABBV-428 | w/w.o atezolizumab | Intravenous infusion | I | Advanced solid tumor | NCT02955251 [109] | Completed | 0.01-3.6 | SD at 3.6 mg/kg (36%) | n=59: infusion-related reactions (12%) |
PDAC: pancreatic ductal adenocarcinoma; mPDAC: metastatic pancreatic ductal adenocarcinoma; TNBC: triple-negative breast cancer; NSCLC: non-small cell lung cancer; OS: overall survival; CR: complete response; PR: partial response; SD: stable disease; DFS: disease-free survival; AE: adverse events; TRAE: treatment-related adverse event; CRS: cytokine release syndrome; ALT: aminotransferase; AST: aspartate aminotransferase; WBC: white blood cells.
In contrast, several clinical trials have reported severe side effects, specifically grade 3 or higher treatment-emergent adverse events (TEAEs) and non-transient alterations in liver function. In a terminated phase I study of intravenous SEA-CD40 (0.01 or 0.03 mg/kg) in combination with chemotherapy and pembrolizumab in patients with PDAC, grade 3 or higher TEAEs occurred, including neutropenia (61%), anemia (33%), and thrombocytopenia (20%). TEAEs that lead to treatment discontinuation were reported in 10% of patients across two dosage groups [107]. In another phase I study of mitazalimab with or without corticosteroids in patients with advanced solid tumors, grade-3 or higher TEAEs were reported in 56.8% and 60% in cohorts with and without corticosteroids, respectively. Other grade-3 or higher TEAEs include gamma-glutamyl transferase increase (7.4%), AST increase (5.3%), and anemia (5.3%) [102].
To control toxicity, clinical methods have been modified. Firstly, local intratumoral delivery of CD40 agonist antibodies has been utilized to induce an abscopal effect [122], potentially reduce toxicity while retaining the antitumor effect [100, 123]. Recent studies using intratumoral delivery include treating malignant solid tumors with 2141-V11 (NCT04616248) and CDX-1140 (NCT04059588), respectively, as well as treating malignant glioma (MG) with 2141-V11 by convection-enhanced delivery (CED) (NCT04547777) [105].
Another possible approach to down-tuning treatment-induced toxicity is the sequence of treatments between CD40 agonism therapy, chemotherapy, and surgery. Using mouse models of PDAC, Byrne et al. showed that CD40 agonist antibody was lethal in a CSF1R-dependent manner when administered 48h before chemotherapy [9]. Moreover, delivering chemotherapy five days after CD40 antibody was shown to allow time for tumor stroma disruption, improving subsequent tumor killing [124]. Therefore, an alternative schedule between chemotherapy and CD40 agonists has been utilized in the clinic; for example, chemotherapy would be given on days 1, 8, and 15, and selicrelumab on day 3 [94]. This chemo-CD40 agonist antibody – chemo “sandwich” dosing schedule also aligned with previous preclinical [9] and clinical [125] studies, as well as current trials with SEA-CD40 [107], CDX1140 (NCT05849480), and APX005M [97]. Furthermore, the sequence with surgery was shown to mediate toxicity. In a phase 1 study in patients with PDAC, both neoadjuvant and adjuvant selicrelumab with or without chemotherapy were evaluated. In the neoadjuvant group, selicrelumab was given two days after chemotherapy and before surgery [94]. TRAEs were primarily mild at grade 1 or 2, with no CRS or serious adverse events observed. However, the adjuvant therapy was associated with grade 2 CRS, a series of grade 3 or higher AEs, and severe adverse events. The mechanism behind these AEs remains to be discovered.
While dosages selected for clinical trials vary from 0.01 mg/kg to 3.6 mg/kg (most frequently at 0.01-0.3 mg/kg) depending on the CD40 agonist used (Table 2), in vivo mouse studies using CD40 rat anti-mouse IgG2a mAb (clone FGK45) have commonly used 1.5-10 mg/kg (30-200μg/dose), with 5 mg/kg (100μg/dose) being the most frequently employed dosage across treatment groups (Table 3) [126-130]. The minimum dosage of 1.5 mg/kg in mouse studies converts to exactly 0.01 mg/kg in human after metabolic normalization, validating the safety of initial dosages used in current clinical studies. However, a lower dosage comparable to that in the clinic at 0.1 mg/kg occurred in one humanized mouse model, which carries human Fcγ receptors (FcγRs) and human CD40 (hFcγR/hCD40) instead of their mouse homologs. The CD40 agonist used was 2141-V11. As mentioned above, it is a fully human anti-CD40 agonist antibody with enhanced binding to FcγRIIB. Establishing the effect of FcγRIIB enhancement under physiological conditions, this mouse model was able to mirror and predict dose-limiting toxicities occurring in patients. While the MTD of the parental antibody 2141-IgG2 was found to be 0.2 mg/kg, doses higher than 0.1 mg/kg of 2141-V11 led to profound transaminitis and hepatotoxicity, and evidence of intravascular thrombi and hepatocyte necrosis was also found in mice treated with concentrations equal or above 0.25 mg/kg. This model also provides insights into the constraints of using anti-CD40 agonistic antibodies in humans – when administered systematically, they may struggle to attain an optimal therapeutic concentration due to dose-limiting toxicities [128].
Table 3.
Preclinical studies of CD40 agonistic antibodies in cancer treatment, alone or in combination with other treatments.
| Therapy | Interventions | Route of Delivery |
Cancer Type | Dosage (mg/kg ) |
Outcome | Side Effects | Reference |
|---|---|---|---|---|---|---|---|
| Anti-mouse CD40 mAb (FGK45) | w/w.o nab-nabpaclitaxel and gemcitabine (CD40/Gem or Gem/CD40) w/w.o anti-CSF-1R | IP | PDA | 5 or 15 | No reduction in tumor volume. aCD40 was lethal when administered before chemotherapy (40%). Lethal hepatotoxicity was abrogated when macrophage activation was blocked using anti-CSF-1R mAb | 42% mice treated with 0.3mg/kg followed by Gem died vs. 58.1% in the 0.1 mg/kg group. In all mice: multifocal granulomatous and neutrophilic infiltration; significant increase in ALT and AST. Higher hepatotoxicity in CD40/Gem group. | [9] |
| Diphtheria toxin (DT) (Treg depletion) w/w.o TNF neutralization | IP | Mammary carcinoma (4T1.2) and Colon adenocarcinoma (MC38) | 5 | Mice with Treg depletion had higher TNF and IL-6 production. Concomitant TNF neutralization may reduce irAEs but also antitumor efficacy | DT + aCD40: significant weight loss, increase in clinical score, ALT and the proinflammatory cytokines. Concomitant anti-TNF abrogated weight loss, liver damage and colitis, resulting in an improved clinical score. | [126] | |
| + Radiotherapy + PDL1 | NDES or IP | Mammary carcinoma (4T1) | 5 | Localized immunotherapy via NDES + radiotherapy demonstrated an abscopal effect, augmented CD8+ T cells | IP vs. NDES CD40/PDL1: significant weight loss, worsened pathologic liver inflammation. | [127] | |
| w/w.o anti-PD-1, anti-CTLA4 | IP | Colon adenocarcinoma (MC38) | 5 | IL-12 and IFN-γ interdependently drive tissue-damaging effects in the liver and other tissues in a neutrophil-dependent way | IrAEs at liver and the GI tract. Portal and prominent lobular hepatitis and broad, confluent areas of necrosis. Colon crypt hyperplasia. | [131] | |
| + IL-2 w/w.o T-cell adoptive transfer | IP | Melanoma (B16) and Colon adenocarcinom a (MC38) | 10 | CD40 agonist expanded transferred T cell in vivo and improved antitumor effects | N/A | [130] | |
| monotherapy | IP | Melanoma (B16-OVAMO4) | 1.5 | Antitumor activity was significantly higher for mice treated with CD40 antibody with enhanced FcγRIIB binding | N/A | [51] | |
| monotherapy | IP | Breast cancer (4T1) and Hepatocellular carcinoma (RIL-175) | 5 | CD40 agonist matured murine and human hepatic MDSCs, relieving immunosuppression. | Liver damage occurred within 24h: ALT and AST elevations, and ROS-mediated hepatotoxicity. | [129] | |
| monotherapy | subcutanous or peritumoral or intravenous | Bladder cancer (MB49) | 0.5, 1.5, 5 | local peritumoral at low dose (0.01 and 0.03 mg.kg) improved survival and reduced toxicity than the intravenous systemic treatment | [100] | ||
| Human anti-CD40 agonist antibody | monotherapy | Intratumoral injectionor IP | Colon adenocarcinoma (MC38) | 0.1 | Intratumoral delivery led to a significantly lower tumor burden. | Dosage > 0.1 mg/led to profound increase in AST, ALT, and hepatotoxicity. Increasing dose worsened thrombocytopenia. | [128] |
NDES: nanofluidic drug-eluting seed; IrAE: immune-related adverse events; ALT: aminotransferase; AST: aspartate aminotransferase
To improve antitumor efficacy while minimizing toxicities, in vivo studies have been exploring tumor-targeted CD40 agonism. Our group has shown that the inhibition of RAS/PI3K/RAF-mediated pathways directly induces CD40 expression in melanoma cells, triggering immunogenic cell death [13]. Recently, a novel bispecific CD40 agonistic antibody named CEA-CD40 was developed. It stimulates CD40 selectively in the presence of carcinoembryonic antigen (CEA), a glycoprotein exclusively expressed on tumor cells, thereby activating the delivery of tumor antigen derived from CEA+ tumor-derived extracellular vesicles by DCs to trigger tumor-specific T cell cross-priming [131]. To further reduce systemic toxicities, in vivo studies have shifted to evaluate the effect of local delivery on the toxicity and antitumor effect. Subcutaneous delivery [100], intratumoral injection [128], and nanofluidic drug-eluting seed (NDES) [127] were shown to allow lower toxicity and better efficacy than intraperitoneal injection (Table 3). Using mouse models, recent studies have unraveled the key players mediating CRS and liver toxicity: Kupffer cells, neutrophils, macrophages, and platelets. Mechanistically, tissue-resident Kupffer cells sense lymphocyte-derived IFN-γ and produce IL-12 [132]. IFN-γ and IL-12 subsequently stimulate neutrophils that in turn upregulate and secrete TNF [133], and cause granulocytic inflammation, which results in liver toxicity. It has been further shown that CD40-mediated hepatotoxicity is associated with an accumulation of triple-positive (MHCII+, CD14+, and CD11b+) macrophages in the liver in an IL-12p40-dependent manner [132]. Last but not least, a causal relationship between liver macrophages and platelets has been described in anti-CD40 treatment-related hepatotoxicity, whereby the FcγRIIB expression by Kupffer cells might lead to crosslinking of CD40 antibodies and subsequent platelet activation [134]. Therefore, to reduce toxicity, mechanism-based interventions have been employed, such as blocking macrophage activation [9] and neutralizing TNF [126]. However, antitumor efficacy could be compromised with this approach [126]. OVT “armed” with different adjuvants can be used to enhance antitumor efficacy while minimizing systemic toxicity. Arming oncolytic viruses with T cell costimulatory molecules, such as CD40, OX40, ICAM-1, and B7-1, can enhance APC function. In addition, a variety of cytokines that modulate antitumoral responses, such as GM-CSF, IL-2 and IFN-γ, have been armed with oncolytic viruses to promote proapoptotic lymphocyte functions in the TME, and have demonstrated higher antitumor efficacy with no or repressed systemic toxicity in the clinic [113].
9. Future directions
Several strategies can be incorporated with CD40 agonism therapy in cancer treatment, including cytokine therapies, targeted therapies of the RAS/RAF/PI3K pathway, and chimeric antigen receptor (CAR) T therapies.
9.1. Cytokine therapy
CD40 agonism can be combined with cytokine therapies targeting the MDSCs, macrophages, or NK cells, potentially mounting complementary and synergistic antitumor effects. For example, the C-X-C chemokine receptor 2 (CXCR2) signaling axis plays an essential role in the recruitment of neutrophils and immunosuppressive MDSCs into the TME [135]. The deletion of CXCR2 in a BrafV600E/PTEN−/− melanoma mouse model skewed the TME toward antitumor immunity via reducing MDSCs and promoting CD8+ T cell infiltration via B-cell derived C-X-C Motif Chemokine Ligand 11 (CXCL11) [136, 137]. A CXCR2 antagonist, such as SX-682, was reported to significantly decrease the expression of chemokines associated with MDSC recruitment and suppress progenitor cells via upregulating Tfcp2l1, a transcription factor regulating tumor suppression and stemness [137]. By combining CXCR2 antagonism with CD40 agonism, multiple aspects of the TME can potentially be complementarily targeted, with the former inhibiting the recruitment of immunosuppressive cells and the latter enhancing immunosurveillance.
Macrophage-based cytokine therapies can be further expanded to combine with CD40 agonism. Most TMEs are encompassed with the immunosuppressive tumor-associated macrophages (TAM) of the M2 type, which in several cancer types has been associated with the expression of the immunoregulatory IL-10 [138], promoting tumor growth and correlating with poor outcomes [139]. Therefore, the key to macrophage-based therapies has been modulating or reversing the M2 macrophages towards the M1 macrophages to overturn the immunosuppressive milieu [139]. One example that acts by such a mechanism is the clinically relevant CSF-1 inhibitor, as mentioned above [119]. Blockade of the immunosuppressive IL-10 or TGF-β may also modulate TAMs, decrease tumor stemness [140], and restore their antitumor effect, specifically by promoting IL-12 expression and activating type I antitumor immune responses [141]. Combining macrophage-modulating cytokine therapy with CD40 agonism may be beneficial, firstly because of their complementarily mechanisms of action, as reducing immunosuppressive TAMs while increasing immunosurveillance. Secondly, a perpetuating synergistic effect may arise since CD40 ligation on macrophages can activate their production of proinflammatory cytokines, including IFN-γ, which upregulate CD40 expression on the macrophages and lead to downstream autocrine tumoristatic effects [32].
CD40 agonism may also be combined with cytokine therapies targeting NK cells, which are expressed at low frequency in tumors compared to myeloid and major lymphoid cells [142]. Interleukin-2 (IL-2) and interleukin-5 (IL-5) are two stimulatory molecules for NK cells [142]. Clinical data have shown that the recombinant forms of IL-2 [143] and IL-5 [144] can induce robust activation of CD8+ T cells and NK cells within the TME in patients with solid tumors. Combining NK cell therapy that enhances the direct killing of tumor cells with CD40 agonism can activate both the innate and adaptive immune systems, thus mounting a more comprehensive antitumor response. In addition, a synergistic effect may result since activated NK cells produce IFN-γ and TNF, which in turn, upregulate CD40 expression on macrophages and promote type I antitumor immune responses [32].
The anti-apoptotic protein TNF-A IP3 (A20) was reported to protect malignant B cells against cell death under CD40 stimulation [59]. IL-17 signal is known to maintain the baseline production of the anti-inflammatory TNF-Α IP3 (A20) and restrain the aberrant activation of JNK and NF-kB pathways in melanoma and breast cancer cells [145]. In CRC, IL-17 receptor deletion correlates with tumor invasion, growth, metastasis, and poor clinical outcome [146]. The paradoxical role of IL-17 and IL-17-producing Th17 cells in cancers may interfere the therapeutic outcome of CD40 agonism [147], and thus, finetuning the IL-17/A20 signaling with CD40 agonism therapy is intriguing for future investigation.
9.2. Targeted therapy of the RAS/RAF/PI3K pathways
CD40 agonism can also be combined with targeted therapy in cancer patients exhibiting mutations in the RAS/RAF/PI3K pathways. RAS mutations occupy approximately 19% of all cancers, among which KRAS is the most frequently mutated isoform [148]. RAS-mutant tumors are thought to be highly aggressive and recurrent, correlating with specific clinicopathological features, such as the shifted location of metastasis, lower tumor-infiltrating lymphocyte grade, and shorter OS in certain types of cancers. Therefore, the inhibition of RAS – most frequently directly or through downstream proteins – has become a burgeoning field. A KRASG12C inhibitor named AMG510 exhibited encouraging results both in vivo [149] and in patients with NSCLC and colorectal cancer [150]. In breast cancer where the RAS-downstream PI3K/mTOR pathway is frequently altered, our group showed that PI3K/mTOR inhibition (gedatolisib) plus microtubule poison paclitaxel (PTX) halted the PyMT breast tumor growth and improved response to ICB [151]. In addition, dual inhibition of the PI3K/AKT/mTOR and the RAF-downstream MAPK/MEK/ERK pathway was mechanistically warranted: both being downstream of receptor tyrosine kinase (RTK) activation of RAS, such that the inhibition of only one pathway would cause compensatory enhancement of the other, offsetting ideal tumor suppression [152]. Therapeutic effects of the dual inhibition of PI3K and MAPK pathways were also confirmed by preclinical studies in breast cancer and melanoma models [152]. The combination of CD40 agonism with targeted therapies of the RAS/RAF/PI3K pathways, specifically of MEK, was reported to produce a synergistic effect without impeding T cell functions in murine melanoma (B16-OVA) and colon cancer (MC-38). Mechanistically, the MEK inhibitor (GDC-0973) produced pro-immunogenic effects by inhibiting Tregs, M2 macrophages, and MDSCs, while the CD40 agonist complementarily promoted antigen presentations of DC, CD8+ T cells and M1 macrophages [153]. Such combinative therapies shed light on the personalization of treatment tailored to cancer patients harboring RAS/RAF/PI3K mutations.
9.3. CAR T therapy
Engineered CD40 agonists with selective binding to tumor cells can be combined with CAR T therapies. Adoptive transfer of CAR T cells with constitutive expression of CD40L was reported with robust antitumor efficacy by licensing APCs, which then mobilized endogenous tumor-recognizing T cells and enhanced cytokine productions [154]. The addition of the tumor-specific CD40 agonists may complement the proinflammatory responses by CAR T cells by inducing direct apoptosis of tumor cells, creating a synergistic antitumor effect. It would be intriguing to explore whether CD40 agonists lacking selective affinity for tumor cells might bind and augment the recruitment of CAR T cells. Alternatively, there's the question of whether CD40 agonists could saturate CD40L-expressing CAR T cells, potentially diminishing their effectiveness.
9.4. Advanced methodologies and models
CD40 agonism can be explored in various preclinical models with quantitative analysis to improve the prediction of drug responses in patients. The patient-derived xenograft (PDX) established in humanized mice was recently optimized for evaluating both targeted therapy and immune therapy: human tumor tissues were transplanted and expanded in the humanized mice that were established by the engraftment of CD34+ hematopoietic stem cells. The translational utility was maximized in this model since human tumor tissues were grown in humanized mice competent for mounting antitumor responses [155]. Moreover, high-throughput screening containing an extensive collection of PDXs with diverse mutations enabled insights into the association between genotypes and drug efficacy and facilitated the exploration of predictive biomarkers [156]. In addition, the prediction of clinical trial responses from preclinical models can be further improved by multiplex immunohistochemistry/immunofluorescence and spatial imaging analysis of cells in the TME, which visualize desired biomarkers in the TME of the tumor tissue section and perform quantitative assessment of their spatial distribution [157].
Since the successful generation of organoids about a decade ago, patient-derived tumor organoids have become a favorable preclinical model. Organoids are 3D-cultured in vitro from patient-derived tumor tissues in an extracellular matrix, enabling mimicking of the genetic, phenotypic, and histopathologic characteristics of the original tumor, such as in colon, lung and breast cancer [158]. However, the variation in the physical structures of organoids poses challenges to reproducibility, which is alleviated by organoid-on-a-chip models. These models culture organoids in specific scaffolds that mimic the physiological aspects of the target organ, and allow coculture with immune cells via microfluidics technology, thereby enabling a uniform, monitorable interplay between the organoid and the human-like physiological environment [158]. Bridging the gap between patient-derived tissues, animal, and cell models by simulating human biological conditions, these preclinical models combined with quantitative analysis would enable faster, more cost-effective testing of CD40 agonists and generate more reliable and personalized prediction of drug responses.
Conclusions
The intricate role of CD40-CD40L signaling in a variety of immune cells and cancer types underscore its significance as a therapeutic target in cancer treatment. While the clinical development of CD40 agonists shows remarkable potential, continued research and careful consideration of dosage optimization and adverse effects management are crucial for harnessing the full therapeutic potential of CD40 agonism in the fight against cancer.
Highlights.
CD40L-CD40 signaling plays an intricate role in a variety of immune and malignant cells and the regulation of the tumor microenvironment.
CD40 is linked to various biological and immunological functions.
Preclinical studies and clinical development of CD40 agonism therapy underscore its significance in cancer treatment.
Careful consideration of dosage optimization and adverse effects management is crucial for the future design of CD40 agonism cancer therapy.
Several cancer treatment strategies, including cytokine therapies, targeted therapies of the RAS/RAF/PI3K pathway, and chimeric antigen receptor (CAR) T therapies, can potentially be incorporated with CD40 agonism therapy.
Acknowledgement
This work was supported by grants from the Lloyd Foundation Melanoma Research Grant (CY), NCI R01-CA116021 (AR, CY), NCI-R01-CA243326 (AR, CY), the Department of Veterans Affairs SRCS Award IK6BX005225 (AR) and the Department of Veterans Affairs MERIT Award 101BX002301 (AR).
Abbreviations
- Act1
activator 1
- ADCC
antibody-dependent cellular cytotoxicity
- AE
adverse event
- AKNA
AT-Hook transcription factor
- ALT
alanine aminotransferase
- AST
aspartate aminotransferase
- BAD
BCL2-associated agonist of cell death
- CR
complete response
- CRS
cytokine release syndrome
- DFS
disease-free survival
- EBF1
EBF transcription factor 1
- ERK
extracellular signal-regulated kinase
- IFNβ
interferon beta
- IFNγ
interferon gamma
- IKK
IκB kinase
- IKKα
inhibitor of NF-κB kinase α
- IrAE
immune-related adverse event
- IκB
NF-κB inhibitor
- JAK3
Janus kinase 3
- JNK
c-Jun N-terminal kinase
- LPS
lipopolysaccharides
- MAPK
mitogen-activated protein kinase
- mPDAC
metastatic pancreatic ductal adenocarcinoma
- NDES
nanofluidic drug-eluting seed
- NF-κB
nuclear factor kappa-B
- NSCLC
non-small cell lung cancer
- OS
overall survival
- PDAC
pancreatic ductal adenocarcinoma
- PI3K
phosphoinositide 3-kinase
- PR
partial response
- RPL26
ribosomal protein L26
- RPL4
ribosomal protein L4
- RPL8
ribosomal protein L8
- RPS9
ribosomal protein S9
- RUNX1
RUNX family transcription factor 1
- RUNX3
RUNX family transcription factor 3
- SAG
superantigen
- SD
stable disease
- STAT-1
signal transducers and activators of transcription 1
- STAT-1α
signal transducers and activators of transcription 1α
- STAT5
signal transducer and activator of the transcription 5
- TAK1
TGFβ-activated kinase 1
- TNBC
triple-negative breast cancer
- TNFα
tumor necrosis factor α
- TRAE
treatment-related adverse event
- TRAF
TNF receptor-associated factor
- WBC
white blood cells
Biographies

Yang Zhou
Yang Zhou is a senior undergraduate at Vanderbilt University, majoring in Molecular & Cellular Biology. Her research in the Lab at Vanderbilt's Department of Pharmacology focuses on investigating CD40 overexpression and targeted therapy in melanoma. She is a fellow of the SyBBURE Searle Undergraduate Research Program. She is also a Student Member of the American Association for Cancer Research. She serves as an Editorial Board Member of Vanderbilt's largest student-run STEM newspaper, Vanguard. She was also a News Editor Intern for the Chinese platform of Scientific American. With a deep passion for immunology, she aspires to pursue graduate studies in this field, aiming to contribute to bench-to-bedside translation.

Ann Richmond, Ph.D.
Ann Richmond is an Ingram Professor of Cancer Research, Director for the Program in Cancer Biology, Professor of Pharmacology and Dermatology at Vanderbilt University School of Medicine and at the TVHS Department of Veterans Affairs. Early in her career her lab identified and characterized one of the first chemokines, now known as CXCL1 and went on to characterize the role of this and other chemokines and their receptors in wound healing, inflammation, tumor growth and metastasis. Her research goals are to develop and translate new therapeutic approaches for treatment of malignant melanoma and breast cancer. With her research team she has developed new strategies for treatment of tumors that respond poorly to immune checkpoint inhibitors (ICI). They have shown that combining inhibitors of the PI3K/AKT pathway in breast cancer, or inhibitors of the RAS/PI3K/MEK pathways can significantly increase response to ICI therapy and in some instances result in tumor regression. She has been continuously funded by the NIH and the Department of Veterans Affairs throughout her career. Recent work from her group lead to an investigator initiated clinical trial to determine whether the small molecule inhibitor rigosertib will restore sensitivity to immune checkpoint inhibitors of tumors that have developed resistance to these therapies. She is a fellow of the American Association for Advancement of Science, was awarded the William S Middleton Award in 2016 for outstanding achievement in biomedical research by the Department of Veterans Affairs and she received the Society of Leukocyte Biology Legacy Award in 2019. Her research has been continuously supported by grants from the NIH and the Department of Veteran’s Affairs throughout her career. She has published over 200 peer-reviewed papers and trained over 50 graduate students and postdoctoral fellows over her career. She is currently the Director for the Program in Cancer Biology at Vanderbilt University School of Medicine. She serves on numerous editorial boards, NIH advisory boards, and grant review boards.

Chi Yan, Ph.D.
Dr. Chi Yan is an Assistant Professor in the Department of Pharmacology at Vanderbilt University School of Medicine, Nashville, TN, USA. He obtained his Ph.D. in Cancer Immunology from Dalhousie University, Canada, and his M.Sc. in Molecular Biology and Bioinformatics from Saint Mary’s University, Canada. Dr. Yan is a recipient of the prestigious Graduate Fellowship from CRTP-BHCRI in Cancer Research at CIHR, Canada, and the Graduate Fellowship from IWK Health Center, Canada. The goal of Dr. Yan’s research program is to better understand the interaction between tumor cells and immune cells so that one can improve current therapeutic strategies for the treatment of melanoma and breast cancer patients. Dr. Yan’s ongoing research at Vanderbilt University aims to enhance antitumor immune responses by combining personalized targeted and immune therapies. As an accomplished young scientist, Dr. Yan’s work has resulted in investigator-initiated clinical trial NCT05764395 and over 30 publications, including 24 first- or senior-authored articles, in highly-ranked journals that are considered among the top in the field, e.g., Cytokine & Growth Factor Reviews (2024), Molecular Cancer (June 2021, Nov 2021, and 2023), Cancers (2023), npj Precision Oncology (2022), Cancer Immunology Research (2021), and Journal of Clinical Investigation (2020) etc. In addition, Dr. Yan has peer-reviewed over 60 manuscripts for biomedical science journals, including but not limited to the prestigious Nature, Molecular Cancer, Journal of Clinical Investigation, Clinical Cancer Research, and Cancer Research etc. Dr. Yan has also served as Review Editor for Frontiers in Immunology, Frontiers in Oncology, Frontiers in Cell and Developmental Biology, and Frontiers in Pharmacology. Dr. Yan was invited to present his research work at local, national, and international conferences, including Innovation in Breast Cancer Symposium 2022 (Madrid, Spain & Virtual) and the AAI Annual Meeting - IMMUNOLOGY2022 with the awarded AAI Early Career Faculty Travel Grant, etc. Dr. Yan has recently been selected as the Vanderbilt University Early-Career Scientist nominee for the global 2024 Takeda Innovators in Science Award - Cancer Immunology. Dr. Yan received a Henry J Lloyd Foundation Melanoma Research Career Development Grant (PI), Vanderbilt-Ingram Cancer Center Support Grant (Co-PI), and is actively managing and serving as the Co-PI of two ongoing NIH R01 grants (CA116021 and CA243326). Dr. Yan served as the direct mentor of Yang Zhou (first author), and is the corresponding author of the published review in Cytokine & Growth Factor Reviews (2024).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest: The authors declare no potential conflicts of interest.
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Reference
- [1].Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ, Molecular mechanism and function of CD40/CD40L engagement in the immune system, Immunol Rev 229(1) (2009) 152–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Vonderheide RH, Prospect of targeting the CD40 pathway for cancer therapy, Clin Cancer Res 13(4) (2007) 1083–8. [DOI] [PubMed] [Google Scholar]
- [3].Guiducci C, Valzasina B, Dislich H, Colombo MP, CD40/CD40L interaction regulates CD4+CD25+ T reg homeostasis through dendritic cell-produced IL-2, Eur J Immunol 35(2) (2005) 557–67. [DOI] [PubMed] [Google Scholar]
- [4].Tang T, Cheng X, Truong B, Sun L, Yang X, Wang H, Molecular basis and therapeutic implications of CD40/CD40L immune checkpoint, Pharmacol Ther 219 (2021) 107709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].O'Sullivan B, Thomas R, CD40 and dendritic cell function, Crit Rev Immunol 23(1-2) (2003) 83–107. [DOI] [PubMed] [Google Scholar]
- [6].Johnson S, Zhan Y, Sutherland RM, Mount AM, Bedoui S, Brady JL, Carrington EM, Brown LE, Belz GT, Heath WR, Lew AM, Selected Toll-like receptor ligands and viruses promote helper-independent cytotoxic T cell priming by upregulating CD40L on dendritic cells, Immunity 30(2) (2009) 218–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Carbone E, Ruggiero G, Terrazzano G, Palomba C, Manzo C, Fontana S, Spits H, Karre K, Zappacosta S, A new mechanism of NK cell cytotoxicity activation: the CD40-CD40 ligand interaction, J Exp Med 185(12) (1997) 2053–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Gauchat JF, Henchoz S, Mazzei G, Aubry JP, Brunner T, Blasey H, Life P, Talabot D, Flores-Romo L, Thompson J, et al. , Induction of human IgE synthesis in B cells by mast cells and basophils, Nature 365(6444) (1993) 340–3. [DOI] [PubMed] [Google Scholar]
- [9].Byrne KT, Vonderheide RH, CD40 stimulation obviates innate sensors and drives T cell immunity in cancer, Cell Rep 15(12) (2016) 2719–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].van Essen D, Kikutani H, Gray D, CD40 ligand-transduced co-stimulation of T cells in the development of helper function, Nature 378(6557) (1995) 620–3. [DOI] [PubMed] [Google Scholar]
- [11].Mackey MF, Gunn JR, Ting PP, Kikutani H, Dranoff G, Noelle RJ, Barth RJ Jr., Protective immunity induced by tumor vaccines requires interaction between CD40 and its ligand, CD154, Cancer Res 57(13) (1997) 2569–74. [PubMed] [Google Scholar]
- [12].Tong AW, Stone MJ, Prospects for CD40-directed experimental therapy of human cancer, Cancer Gene Ther 10(1) (2003) 1–13. [DOI] [PubMed] [Google Scholar]
- [13].Yan C, Saleh N, Yang J, Nebhan CA, Vilgelm AE, Reddy EP, Roland JT, Johnson DB, Chen SC, Shattuck-Brandt RL, Ayers GD, Richmond A, Novel induction of CD40 expression by tumor cells with RAS/RAF/PI3K pathway inhibition augments response to checkpoint blockade, Mol Cancer 20(1) (2021) 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Morrison AH, Diamond MS, Hay CA, Byrne KT, Vonderheide RH, Sufficiency of CD40 activation and immune checkpoint blockade for T cell priming and tumor immunity, Proc Natl Acad Sci U S A 117(14) (2020) 8022–8031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Alexandroff AB, Jackson AM, Paterson T, Haley JL, Ross JA, Longo DL, Murphy WJ, James K, Taub DD, Role for CD40-CD40 ligand interactions in the immune response to solid tumours, Mol Immunol 37(9) (2000) 515–26. [DOI] [PubMed] [Google Scholar]
- [16].Richards DM, Sefrin JP, Gieffers C, Hill O, Merz C, Concepts for agonistic targeting of CD40 in immuno-oncology, Hum Vaccin Immunother 16(2) (2020) 377–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Hostager BS, Bishop GA, CD40-mediated activation of the NF-kappaB2 pathway, Front Immunol 4 (2013) 376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Sun SC, The non-canonical NF-kappaB pathway in immunity and inflammation, Nat Rev Immunol 17(9) (2017) 545–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Qian Y, Zhao Z, Jiang Z, Li X, Role of NF kappa B activator Act1 in CD40-mediated signaling in epithelial cells, Proc Natl Acad Sci U S A 99(14) (2002) 9386–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Sutherland CL, Heath AW, Pelech SL, Young PR, Gold MR, Differential activation of the ERK, JNK, and p38 mitogen-activated protein kinases by CD40 and the B cell antigen receptor, J Immunol 157(8) (1996) 3381–90. [PubMed] [Google Scholar]
- [21].Lind EF, Ahonen CL, Wasiuk A, Kosaka Y, Becher B, Bennett KA, Noelle RJ, Dendritic cells require the NF-kappaB2 pathway for cross-presentation of soluble antigens, J Immunol 181(1) (2008) 354–63. [DOI] [PubMed] [Google Scholar]
- [22].Gallagher E, Enzler T, Matsuzawa A, Anzelon-Mills A, Otero D, Holzer R, Janssen E, Gao M, Karin M, Kinase MEKK1 is required for CD40-dependent activation of the kinases Jnk and p38, germinal center formation, B cell proliferation and antibody production, Nat Immunol 8(1) (2007) 57–63. [DOI] [PubMed] [Google Scholar]
- [23].Macatonia SE, Hosken NA, Litton M, Vieira P, Hsieh CS, Culpepper JA, Wysocka M, Trinchieri G, Murphy KM, O'Garra A, Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells, J Immunol 154(10) (1995) 5071–9. [PubMed] [Google Scholar]
- [24].Yu Q, Kovacs C, Yue FY, Ostrowski MA, The role of the p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and phosphoinositide-3-OH kinase signal transduction pathways in CD40 ligand-induced dendritic cell activation and expansion of virus-specific CD8+ T cell memory responses, J Immunol 172(10) (2004) 6047–56. [DOI] [PubMed] [Google Scholar]
- [25].Donahue AC, Fruman DA, PI3K signaling controls cell fate at many points in B lymphocyte development and activation, Semin Cell Dev Biol 15(2) (2004) 183–97. [DOI] [PubMed] [Google Scholar]
- [26].Foey AD, Feldmann M, Brennan FM, CD40 ligation induces macrophage IL-10 and TNF-alpha production: differential use of the PI3K and p42/44 MAPK-pathways, Cytokine 16(4) (2001) 131–42. [DOI] [PubMed] [Google Scholar]
- [27].Hanissian SH, Geha RS, Jak3 is associated with CD40 and is critical for CD40 induction of gene expression in B cells, Immunity 6(4) (1997) 379–87. [DOI] [PubMed] [Google Scholar]
- [28].Marzaioli V, Canavan M, Floudas A, Wade SC, Low C, Veale DJ, Fearon U, Monocyte-derived dendritic cell differentiation in inflammatory arthritis is regulated by the JAK/STAT axis via NADPH oxidase regulation, Front Immunol 11 (2020) 1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Wu W, Alexis NE, Bromberg PA, Jaspers I, Peden DB, Mechanisms of LPS-induced CD40 expression in human peripheral blood monocytic cells, Biochem Biophys Res Commun 379(2) (2009) 573–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Qin H, Wilson CA, Lee SJ, Zhao X, Benveniste EN, LPS induces CD40 gene expression through the activation of NF-kappaB and STAT-1alpha in macrophages and microglia, Blood 106(9) (2005) 3114–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Yang F, He Z, Duan H, Zhang D, Li J, Yang H, Dorsey JF, Zou W, Nabavizadeh SA, Bagley SJ, Abdullah K, Brem S, Zhang L, Xu X, Byrne KT, Vonderheide RH, Gong Y, Fan Y, Synergistic immunotherapy of glioblastoma by dual targeting of IL-6 and CD40, Nat Commun 12(1) (2021) 3424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Nguyen VT, Benveniste EN, Involvement of STAT-1 and ets family members in interferon-gamma induction of CD40 transcription in microglia/macrophages, J Biol Chem 275(31) (2000) 23674–84. [DOI] [PubMed] [Google Scholar]
- [33].Tone M, Tone Y, Babik JM, Lin CY, Waldmann H, The role of Sp1 and NF-kappa B in regulating CD40 gene expression, J Biol Chem 277(11) (2002) 8890–7. [DOI] [PubMed] [Google Scholar]
- [34].Akhtar MN, Mishra M, Yadav V, Yadav M, Gujar R, Lal S, Kumar R, Khatri N, Sen P, Runx proteins mediate protective immunity against Leishmania donovani infection by promoting CD40 expression on dendritic cells, PLoS Pathog 16(12) (2020) e1009136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Zou M, Zhang X, Jiang D, Zhao Y, Wu T, Gong Q, Su H, Wu D, Moreland L, Li G, Transcriptional regulation of CD40 expression by 4 ribosomal proteins via a functional SNP on a disease-associated CD40 locus, Genes (Basel) 11(12) (2020) 1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Siddiqa A, Sims-Mourtada JC, Guzman-Rojas L, Rangel R, Guret C, Madrid-Marina V, Sun Y, Martinez-Valdez H, Regulation of CD40 and CD40 ligand by the AT-hook transcription factor AKNA, Nature 410(6826) (2001) 383–7. [DOI] [PubMed] [Google Scholar]
- [37].Putlyaeva LV, Demin DE, Korneev KV, Kasyanov AS, Tatosyan KA, Kulakovskiy IV, Kuprash DV, Schwartz AM, Potential Markers of Autoimmune Diseases, Alleles rs115662534(T) and rs548231435(C), Disrupt the binding of transcription factors STAT1 and EBF1 to the regulatory elements of human CD40 gene, Biochemistry (Mosc) 83(12) (2018) 1534–1542. [DOI] [PubMed] [Google Scholar]
- [38].Nguyen VT, Benveniste EN, IL-4-activated STAT-6 inhibits IFN-gamma-induced CD40 gene expression in macrophages/microglia, J Immunol 165(11) (2000) 6235–43. [DOI] [PubMed] [Google Scholar]
- [39].Nguyen VT, Walker WS, Benveniste EN, Post-transcriptional inhibition of CD40 gene expression in microglia by transforming growth factor-beta, Eur J Immunol 28(8) (1998) 2537–48. [DOI] [PubMed] [Google Scholar]
- [40].von Leoprechting A, van der Bruggen P, Pahl HL, Aruffo A, Simon JC, Stimulation of CD40 on immunogenic human malignant melanomas augments their cytotoxic T lymphocyte-mediated lysis and induces apoptosis, Cancer Res 59(6) (1999) 1287–94. [PubMed] [Google Scholar]
- [41].Chand Dakal T, Dhabhai B, Agarwal D, Gupta R, Nagda G, Meena AR, Dhakar R, Menon A, Mathur R, Mona V Yadav A Sharma, Mechanistic basis of co-stimulatory CD40-CD40L ligation mediated regulation of immune responses in cancer and autoimmune disorders, Immunobiology 225(2) (2020) 151899. [DOI] [PubMed] [Google Scholar]
- [42].Tay NQ, Lee DCP, Chua YL, Prabhu N, Gascoigne NRJ, Kemeny DM, CD40L expression allows CD8(+) T cells to promote their own expansion and differentiation through dendritic cells, Front Immunol 8 (2017) 1484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Buhtoiarov IN, Lum H, Berke G, Paulnock DM, Sondel PM, Rakhmilevich AL, CD40 ligation activates murine macrophages via an IFN-gamma-dependent mechanism resulting in tumor cell destruction in vitro, J Immunol 174(10) (2005) 6013–22. [DOI] [PubMed] [Google Scholar]
- [44].Inwald DP, McDowall A, Peters MJ, Callard RE, Klein NJ, CD40 is constitutively expressed on platelets and provides a novel mechanism for platelet activation, Circ Res 92(9) (2003) 1041–8. [DOI] [PubMed] [Google Scholar]
- [45].Yacoub D, Benslimane N, Al-Zoobi L, Hassan G, Nadiri A, Mourad W, CD154 is released from T-cells by a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and ADAM17 in a CD40 protein-dependent manner, J Biol Chem 288(50) (2013) 36083–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Seigner J, Basilio J, Resch U, de Martin R, CD40L and TNF both activate the classical NF-kappaB pathway, which is not required for the CD40L induced alternative pathway in endothelial cells, Biochem Biophys Res Commun 495(1) (2018) 1389–1394. [DOI] [PubMed] [Google Scholar]
- [47].Pietravalle F, Lecoanet-Henchoz S, Blasey H, Aubry JP, Elson G, Edgerton MD, Bonnefoy JY, Gauchat JF, Human native soluble CD40L is a biologically active trimer, processed inside microsomes, J Biol Chem 271(11) (1996) 5965–7. [DOI] [PubMed] [Google Scholar]
- [48].Mazzei GJ, Edgerton MD, Losberger C, Lecoanet-Henchoz S, Graber P, Durandy A, Gauchat JF, Bernard A, Allet B, Bonnefoy JY, Recombinant soluble trimeric CD40 ligand is biologically active, J Biol Chem 270(13) (1995) 7025–8. [DOI] [PubMed] [Google Scholar]
- [49].Bergmann S, Pandolfi PP, Giving blood: a new role for CD40 in tumorigenesis, J Exp Med 203(11) (2006) 2409–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Vonderheide RH, Dutcher JP, Anderson JE, Eckhardt SG, Stephans KF, Razvillas B, Garl S, Butine MD, Perry VP, Armitage RJ, Ghalie R, Caron DA, Gribben JG, Phase I study of recombinant human CD40 ligand in cancer patients, J Clin Oncol 19(13) (2001) 3280–7. [DOI] [PubMed] [Google Scholar]
- [51].Li F, Ravetch JV, Inhibitory Fcgamma receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies, Science 333(6045) (2011) 1030–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Merz C, Sykora J, Marschall V, Richards DM, Heinonen K, Redondo Muller M, Thiemann M, Schnyder T, Fricke H, Hill O, Gieffers C, The hexavalent CD40 agonist HERA-CD40L induces T-cell-mediated antitumor immune response through activation of antigen-presenting cells, J Immunother 41(9) (2018) 385–398. [DOI] [PubMed] [Google Scholar]
- [53].Frankish J, Mukherjee D, Romano E, Billian-Frey K, Schroder M, Heinonen K, Merz C, Redondo Muller M, Gieffers C, Hill O, Thiemann M, Honeychurch J, Illidge T, Sykora J, The CD40 agonist HERA-CD40L results in enhanced activation of antigen presenting cells, promoting an anti-tumor effect alone and in combination with radiotherapy, Front Immunol 14 (2023) 1160116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Pham LV, Tamayo AT, Yoshimura LC, Lo P, Terry N, Reid PS, Ford RJ, A CD40 Signalosome anchored in lipid rafts leads to constitutive activation of NF-kappaB and autonomous cell growth in B cell lymphomas, Immunity 16(1) (2002) 37–50. [DOI] [PubMed] [Google Scholar]
- [55].Funakoshi S, Longo DL, Beckwith M, Conley DK, Tsarfaty G, Tsarfaty I, Armitage RJ, Fanslow WC, Spriggs MK, Murphy WJ, Inhibition of human B-cell lymphoma growth by CD40 stimulation, Blood 83(10) (1994) 2787–94. [PubMed] [Google Scholar]
- [56].Furman RR, Asgary Z, Mascarenhas JO, Liou HC, Schattner EJ, Modulation of NF-kappa B activity and apoptosis in chronic lymphocytic leukemia B cells, J Immunol 164(4) (2000) 2200–6. [DOI] [PubMed] [Google Scholar]
- [57].Choi MS, Boise LH, Gottschalk AR, Quintans J, Thompson CB, Klaus GG, The role of bcl-XL in CD40-mediated rescue from anti-mu-induced apoptosis in WEHI-231 B lymphoma cells, Eur J Immunol 25(5) (1995) 1352–7. [DOI] [PubMed] [Google Scholar]
- [58].Sarma V, Lin Z, Clark L, Rust BM, Tewari M, Noelle RJ, Dixit VM, Activation of the B-cell surface receptor CD40 induces A20, a novel zinc finger protein that inhibits apoptosis, J Biol Chem 270(21) (1995) 12343–6. [DOI] [PubMed] [Google Scholar]
- [59].Chu Y, Vahl JC, Kumar D, Heger K, Bertossi A, Wojtowicz E, Soberon V, Schenten D, Mack B, Reutelshofer M, Beyaert R, Amann K, van Loo G, Schmidt-Supprian M, B cells lacking the tumor suppressor TNF-A IP3/A20 display impaired differentiation and hyperactivation and cause inflammation and autoimmunity in aged mice, Blood 117(7) (2011) 2227–36. [DOI] [PubMed] [Google Scholar]
- [60].Luqman M, Klabunde S, Lin K, Georgakis GV, Cherukuri A, Holash J, Goldbeck C, Xu X, Kadel EE 3rd, Lee SH, Aukerman SL, Jallal B, Aziz N, Weng WK, Wierda W, O'Brien S, Younes A, The antileukemia activity of a human anti-CD40 antagonist antibody, HCD122, on human chronic lymphocytic leukemia cells, Blood 112(3) (2008) 711–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Schattner EJ, Elkon KB, Yoo DH, Tumang J, Krammer PH, Crow MK, Friedman SM, CD40 ligation induces Apo-1/Fas expression on human B lymphocytes and facilitates apoptosis through the Apo-1/Fas pathway, J Exp Med 182(5) (1995) 1557–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Rathmell JC, Townsend SE, Xu JC, Flavell RA, Goodnow CC, Expansion or elimination of B cells in vivo: dual roles for CD40- and Fas (CD95)-ligands modulated by the B cell antigen receptor, Cell 87(2) (1996) 319–29. [DOI] [PubMed] [Google Scholar]
- [63].Dotti G, Savoldo B, Takahashi S, Goltsova T, Brown M, Rill D, Rooney C, Brenner M, Adenovector-induced expression of human-CD40-ligand (hCD40L) by multiple myeloma cells. A model for immunotherapy, Exp Hematol 29(8) (2001) 952–61. [DOI] [PubMed] [Google Scholar]
- [64].Heath AW, Chang R, Harada N, Santos-Argumedo L, Gordon J, Hannum C, Campbell D, Shanafelt AB, Clark EA, Torres R, et al. , Antibodies to murine CD40 stimulate normal B lymphocytes but inhibit proliferation of B lymphoma cells, Cell Immunol 152(2) (1993) 468–80. [DOI] [PubMed] [Google Scholar]
- [65].Georgopoulos NT, Merrick A, Scott N, Selby PJ, Melcher A, Trejdosiewicz LK, CD40-mediated death and cytokine secretion in colorectal cancer: a potential target for inflammatory tumour cell killing, Int J Cancer 121(6) (2007) 1373–81. [DOI] [PubMed] [Google Scholar]
- [66].Elmetwali T, Young LS, Palmer DH, CD40 ligand-induced carcinoma cell death: a balance between activation of TNFR-associated factor (TRAF) 3-dependent death signals and suppression of TRAF6-dependent survival signals, J Immunol 184(2) (2010) 1111–20. [DOI] [PubMed] [Google Scholar]
- [67].Ibraheem K, Yhmed AMA, Nasef MM, Georgopoulos NT, TRAF3/p38-JNK signalling crosstalk with intracellular-TRAIL/caspase-10-induced apoptosis accelerates ROS-driven cancer cell-specific death by CD40, Cells 11(20) (2022) 3274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Yan C, Richmond A, Hiding in the dark: pan-cancer characterization of expression and clinical relevance of CD40 to immune checkpoint blockade therapy, Mol Cancer 20(1) (2021) 146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Shen J, Chen X, Wang Z, Zhang G, Chen W, Downregulation of CD40 expression contributes to the accumulation of myeloid-derived suppressor cells in gastric tumors, Oncol Lett 8(2) (2014) 775–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Hill SC, Youde SJ, Man S, Teale GR, Baxendale AJ, Hislop A, Davies CC, Luesley DM, Blom AM, Rickinson AB, Young LS, Eliopoulos AG, Activation of CD40 in cervical carcinoma cells facilitates CTL responses and augments chemotherapy-induced apoptosis, J Immunol 174(1) (2005) 41–50. [DOI] [PubMed] [Google Scholar]
- [71].Ishikawa K, Miyamoto M, Yoshioka T, Kato T, Kaji M, Ohbuchi T, Hirano S, Itoh T, Dosaka-Akita H, Kondo S, Up-regulation of CD40 with juxtacrine activity in human nonsmall lung cancer cells correlates with poor prognosis, Cancer 113(3) (2008) 530–41. [DOI] [PubMed] [Google Scholar]
- [72].Matsumura Y, Hiraoka K, Ishikawa K, Shoji Y, Noji T, Hontani K, Itoh T, Nakamura T, Tsuchikawa T, Shichinohe T, Hirano S, CD40 expression in human esophageal squamous cell carcinoma is associated with tumor progression and lymph node metastasis, Anticancer Res 36(9) (2016) 4467–75. [DOI] [PubMed] [Google Scholar]
- [73].Piechutta M, Berghoff AS, New emerging targets in cancer immunotherapy: the role of Cluster of Differentiation 40 (CD40/TNFR5), ESMO Open 4(Suppl 3) (2019) e000510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Weiss JM, Gregory Alvord W, Quinones OA, Stauffer JK, Wiltrout RH, CD40 expression in renal cell carcinoma is associated with tumor apoptosis, CD8(+) T cell frequency and patient survival, Hum Immunol 75(7) (2014) 614–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Luo C, Lei T, Zhao M, Meng Q, Zhang M, CD40 is positively correlated with the expression of nucleophosmin in cisplatin-resistant bladder cancer, J Oncol 2020 (2020) 3676751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Amini M, Ghorban K, Mokhtarzadeh A, Dadmanesh M, Baradaran B, CD40 DNA hypermethylation in primary gastric tumors; as a novel diagnostic biomarker, Life Sci 254 (2020) 117774. [DOI] [PubMed] [Google Scholar]
- [77].Djureinovic D, Wang M, Kluger HM, Agonistic CD40 antibodies in cancer treatment, Cancers (Basel) 13(6) (2021) 1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Su YA, Yang J, Tao L, Nguyen H, He P, Undetectable and decreased expression of KIAA1949 (phostensin) encoded on chromosome 6p21.33 in human breast cancers revealed by transcriptome analysis, J Cancer 1 (2010) 38–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Bosmann M, Ward PA, Modulation of inflammation by interleukin-27, J Leukoc Biol 94(6) (2013) 1159–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Foss S, Bottermann M, Jonsson A, Sandlie I, James LC, Andersen JT, TRIM21-from intracellular immunity to therapy, Front Immunol 10 (2019) 2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Tajika Y, Takahashi M, Ueno H, Murakami T, Yorifuji H, Organization of organelles and VAMP-associated vesicular transport systems in differentiating skeletal muscle cells, Anat Sci Int 90(1) (2015) 33–9. [DOI] [PubMed] [Google Scholar]
- [82].Matikainen S, Nyman TA, Cypryk W, Function and regulation of noncanonical caspase-4/5/11 inflammasome, J Immunol 204(12) (2020) 3063–3069. [DOI] [PubMed] [Google Scholar]
- [83].Bronckaers A, Gago F, Balzarini J, Liekens S, The dual role of thymidine phosphorylase in cancer development and chemotherapy, Med Res Rev 29(6) (2009) 903–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Kennedy MJ, Davison IG, Robinson CG, Ehlers MD, Syntaxin-4 defines a domain for activity-dependent exocytosis in dendritic spines, Cell 141(3) (2010) 524–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Jhajj HS, Lwo TS, Yao EL, Tessier PM, Unlocking the potential of agonist antibodies for treating cancer using antibody engineering, Trends Mol Med 29(1) (2023) 48–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Filbert EL, Bjorck PK, Srivastava MK, Bahjat FR, Yang X, APX005M, a CD40 agonist antibody with unique epitope specificity and Fc receptor binding profile for optimal therapeutic application, Cancer Immunol Immunother 70(7) (2021) 1853–1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Mangsbo SM, Broos S, Fletcher E, Veitonmaki N, Furebring C, Dahlen E, Norlen P, Lindstedt M, Totterman TH, Ellmark P, The human agonistic CD40 antibody ADC-1013 eradicates bladder tumors and generates T-cell-dependent tumor immunity, Clin Cancer Res 21(5) (2015) 1115–26. [DOI] [PubMed] [Google Scholar]
- [88].Garris CS, Wong JL, Ravetch JV, Knorr DA, Dendritic cell targeting with Fc-enhanced CD40 antibody agonists induces durable antitumor immunity in humanized mouse models of bladder cancer, Sci Transl Med 13(594) (2021) eabd1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].White AL, Chan HT, French RR, Willoughby J, Mockridge CI, Roghanian A, Penfold CA, Booth SG, Dodhy A, Polak ME, Potter EA, Ardern-Jones MR, Verbeek JS, Johnson PW, Al-Shamkhani A, Cragg MS, Beers SA, Glennie MJ, Conformation of the human immunoglobulin G2 hinge imparts superagonistic properties to immunostimulatory anticancer antibodies, Cancer Cell 27(1) (2015) 138–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Yu X, Chan HTC, Fisher H, Penfold CA, Kim J, Inzhelevskaya T, Mockridge CI, French RR, Duriez PJ, Douglas LR, English V, Verbeek JS, White AL, Tews I, Glennie MJ, Cragg MS, Isotype switching converts anti-CD40 antagonism to agonism to elicit potent antitumor activity, Cancer Cell 37(6) (2020) 850–866 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Yu X, Chan HTC, Orr CM, Dadas O, Booth SG, Dahal LN, Penfold CA, O'Brien L, Mockridge CI, French RR, Duriez P, Douglas LR, Pearson AR, Cragg MS, Tews I, Glennie MJ, White AL, Complex interplay between epitope specificity and isotype dictates the biological activity of anti-human CD40 antibodies, Cancer Cell 33(4) (2018) 664–675 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Huang S, van Duijnhoven SMJ, Sijts A, van Elsas A, Bispecific antibodies targeting dual tumor-associated antigens in cancer therapy, J Cancer Res Clin Oncol 146(12) (2020) 3111–3122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Richman LP, Vonderheide RH, Role of crosslinking for agonistic CD40 monoclonal antibodies as immune therapy of cancer, Cancer Immunol Res 2(1) (2014) 19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Byrne KT, Betts CB, Mick R, Sivagnanam S, Bajor DL, Laheru DA, Chiorean EG, O'Hara MH, Liudahl SM, Newcomb C, Alanio C, Ferreira AP, Park BS, Ohtani T, Huffman AP, Vayrynen SA, Dias Costa A, Kaiser JC, Lacroix AM, Redlinger C, Stern M, Nowak JA, Wherry EJ, Cheever MA, Wolpin BM, Furth EE, Jaffee EM, Coussens LM, Vonderheide RH, Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer, Clin Cancer Res 27(16) (2021) 4574–4586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Vitale LA, Thomas LJ, He LZ, O'Neill T, Widger J, Crocker A, Sundarapandiyan K, Storey JR, Forsberg EM, Weidlick J, Baronas AR, Gergel LE, Boyer JM, Sisson C, Goldstein J, Marsh HC Jr., Keler T, Development of CDX-1140, an agonist CD40 antibody for cancer immunotherapy, Cancer Immunol Immunother 68(2) (2019) 233–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Sanborn R, Nashat G, Benedito C, Mark OH, 596 Results from a phase 1 study of CDX-1140, a fully human anti-CD40 agonist monoclonal antibody (mAb), in combination with pembrolizumab, J Immunother Cancer 10 (2022) A624–A624. [Google Scholar]
- [97].Padron LJ, Maurer DM, O'Hara MH, O'Reilly EM, Wolff RA, Wainberg ZA, Ko AH, Fisher G, Rahma O, Lyman JP, Cabanski CR, Yu JX, Pfeiffer SM, Spasic M, Xu J, Gherardini PF, Karakunnel J, Mick R, Alanio C, Byrne KT, Hollmann TJ, Moore JS, Jones DD, Tognetti M, Chen RO, Yang X, Salvador L, Wherry EJ, Dugan U, O'Donnell-Tormey J, Butterfield LH, Hubbard-Lucey VM, Ibrahim R, Fairchild J, Bucktrout S, LaVallee TM, Vonderheide RH, Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: clinical and immunologic analyses from the randomized phase 2 PRINCE trial, Nat Med 28(6) (2022) 1167–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Weiss SA, Djureinovic D, Jessel S, Krykbaeva I, A phase I study of APX005M and cabiralizumab with or without nivolumab in patients with melanoma, kidney cancer, or non-small cell lung cancer resistant to anti-PD-1/PD-L1, Clin Cancer Res 27(17) (2021) 4757–4767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Weiss SA, Sznol M, Shaheen M, Berciano-Guerrero MA, Munoz-Couselo E, Rodriguez-Abreu D, Boni V, Schuchter LM, Gonzalez Cao M, Arance Fernandez A, Wei W, Ganti AK, Hauke RJ, Berrocal A, Iannotti NO, Hsu FJ, Kluger HM, A phase II trial of the Cd40 agonist sotigalimab (APX005M) in combination with nivolumab in subjects with metastatic melanoma with disease progression on anti-PD-1, Clin Cancer Res (2023) CCR-23–0475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Sandin LC, Orlova A, Gustafsson E, Ellmark P, Tolmachev V, Totterman TH, Mangsbo SM, Locally delivered CD40 agonist antibody accumulates in secondary lymphoid organs and eradicates experimental disseminated bladder cancer, Cancer Immunol Res 2(1) (2014) 80–90. [DOI] [PubMed] [Google Scholar]
- [101].Calvo E, Moreno V, Perets R, Fourneau N, A phase I study to assess safety, pharmacokinetics (PK), and pharmacodynamics (PD) of JNJ-64457107, a CD40 agonistic monoclonal antibody, in patients (pts) with advanced solid tumors., Journal of Clinical Oncology 37(15_suppl) (2019) 2527–2527. [Google Scholar]
- [102].Moreno V, Perets R, Peretz-Yablonski T, Fourneau N, Girgis S, Guo Y, Hellemans P, Verona R, Pendas N, Xia Q, Geva R, Calvo E, A phase 1 study of intravenous mitazalimab, a CD40 agonistic monoclonal antibody, in patients with advanced solid tumors, Invest New Drugs 41(1) (2023) 93–104. [DOI] [PubMed] [Google Scholar]
- [103].Lindstedt M, Ellmark P, Sobti A, Smith KE, 35 Early pharmacodynamic changes measured by RNA sequencing in peripheral blood from patients in a phase 1 study with mitazalimab, a potent CD40 agonistic IgG1 monoclonal antibody, J Immunother Cancer 10 (2022) Issue Suppl 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Wong JL, Knorr D, Whiting K, Seshan VE, Phase I study of intravesical anti-CD40 agonist antibody 2141-V11 for non-muscle invasive bladder cancer unresponsive to Bacillus Calmette-Guerin (BCG) therapy., JCO 40 (2022) TPS4616–TPS4616. [Google Scholar]
- [105].Desjardins A, Chandramohan V, Landi DB, Johnson MO, A phase 1 trial of D2C7-it in combination with an Fc-engineered anti-CD40 monoclonal antibody (2141-V11) administered intratumorally via convection-enhanced delivery for adult patients with recurrent malignant glioma (MG). JCO 40 (2022) e14015–e14015. [Google Scholar]
- [106].Coveler AL, Smith DC, Phillips T, Curti BD, Goel S, Mehta AN, Kuzel TM, Markovic SN, Rixe O, Bajor DL, Gajewski TF, Gutierrez M, Lee HJ, Gopal AK, Caimi P, Heath EI, Thompson JA, Ansari S, Jacquemont C, Topletz-Erickson A, Zhou P, Schmitt MW, Grilley-Olson JE, Phase 1 dose-escalation study of SEA-CD40: a non-fucosylated CD40 agonist, in advanced solid tumors and lymphomas, J Immunother Cancer 11(6) (2023) e005584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Coveler AL, Cutierrez M, Vaccaro GM, Brown-Glaberman U, Updated results of a phase 1 study of SEA-CD40, gemcitabine, nab-paclitaxel, and pembrolizumab in patients with metastatic pancreatic ductal adenocarcinoma (PDAC; SGNS40-001). JCO 41 (2023) 708–708. [Google Scholar]
- [108].Liu L, Wu Y, Ye K, Cai M, Zhuang G, Wang J, Antibody-Targeted TNFRSF Activation for Cancer Immunotherapy: The role of FcgammaRIIB cross-linking, Front Pharmacol 13 (2022) 924197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Luke JJ, Barlesi F, Chung K, Tolcher AW, Kelly K, Hollebecque A, Le Tourneau C, Subbiah V, Tsai F, Kao S, Cassier PA, Khasraw M, Kindler HL, Fang H, Fan F, Allaire K, Patel M, Ye S, Chao DT, Henner WR, Hayflick JS, McDevitt MA, Fong L, Phase I study of ABBV-428, a mesothelin-CD40 bispecific, in patients with advanced solid tumors, J Immunother Cancer 9(2) (2021) e002015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Del Bano J, Flores-Flores R, Josselin E, Goubard A, Ganier L, Castellano R, Chames P, Baty D, Kerfelec B, A bispecific antibody-based approach for targeting mesothelin in triple negative breast cancer, Front Immunol 10 (2019) 1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Stumpp MT, Dawson KM, Binz HK, Beyond antibodies: the DARPin((R)) drug platform, BioDrugs 34(4) (2020) 423–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Winter HD, Fernandez E, Stavropoulou V, Stojcheva N, 1475 A phase 1 study to characterize the safety and tolerability of MP0317, a tumor targeting FAP dependent CD40 agonist DARPin, in patients with relapsed/refractory solid tumors, J Immunother Cancer 10 (2022) Issue Suppl 2. [Google Scholar]
- [113].Tian Y, Xie D, Yang L, Engineering strategies to enhance oncolytic viruses in cancer immunotherapy, Signal Transduct Target Ther 7(1) (2022) 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Zhao Y, Liu Z, Li L, Wu J, Zhang H, Zhang H, Lei T, Xu B, Oncolytic adenovirus: prospects for cancer immunotherapy, Front Microbiol 12 (2021) 707290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Bergin R, Pelletier A, Zonca M, Muntzer L, Abstract 2740: Safe and effective delivery of antibody fragments to the solid tumor microenvironment, Cancer Research 83 (2023) 2740. [Google Scholar]
- [116].Naing A, Rosen L, Camidge RD, Khalil D, 1011P FORTITUDE phase I study of NG-350A, a novel tumour-selective adenoviral vector expressing an anti-CD40 agonist antibody: Monotherapy dose escalation results, Annals of Oncology 32 (2021) S853–S854. [Google Scholar]
- [117].Rosen LS, Camidge DR, Khalil D, Lillie T, FORTITUDE: Results of a phase 1a study of the novel transgene-armed and tumor-selective vector NG-350A with and without pembrolizumab (pembro). JCO 40 (2022) 2559–2559. [Google Scholar]
- [118].Tan EW, Abd-Aziz N, Poh CL, Tan KO, Engineered oncolytic adenoviruses: an emerging approach for cancer therapy, Pathogens 11(10) (2022) 1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Weiss SA, Djureinovic D, Jessel S, Krykbaeva I, A phase I study of APX005M and cabiralizumab with or without nivolumab in patients with melanoma, kidney cancer, or non-small cell lung cancer resistant to anti-PD-1/PD-L1, Clin Cancer Res 27 (2021) 4757–4767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Laethem J-LV, Borbath I, Prenen H, Coaña Y.P.d., Mitazalimab in combination with mFOLFIRINOX in patients with metastatic pancreatic ductal adenocarcinoma (PDAC): safety data from part of the OPTIMIZE-1 study., JCO 40 (2022) e16237–e16237. [Google Scholar]
- [121].Fu S, Vandross AL, Hsu YH, Shi N, Early safety and efficacy from a phase I open-label clinical study of LVGN7409 (CD40 agonist antibody) in patients with advanced or metastatic malignancies., JCO 40 (2022) e14501–e14501. [Google Scholar]
- [122].Enell Smith K, Deronic A, Hagerbrand K, Norlen P, Ellmark P, Rationale and clinical development of CD40 agonistic antibodies for cancer immunotherapy, Expert Opin Biol Ther 21(12) (2021) 1635–1646. [DOI] [PubMed] [Google Scholar]
- [123].Fransen MF, Sluijter M, Morreau H, Arens R, Melief CJ, Local activation of CD8 T cells and systemic tumor eradication without toxicity via slow release and local delivery of agonistic CD40 antibody, Clin Cancer Res 17(8) (2011) 2270–80. [DOI] [PubMed] [Google Scholar]
- [124].Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL, IFNgamma and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma, Cancer Discov 6(4) (2016) 400–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Vonderheide RH, Burg JM, Mick R, Trosko JA, Li D, Shaik MN, Tolcher AW, Hamid O, Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors, Oncoimmunology 2(1) (2013) e23033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Jacoberger-Foissac C, Blake SJ, Liu J, McDonald E, Triscott H, Nakamura K, Smyth MJ, Teng MW, Concomitant or delayed anti-TNF differentially impact on immune-related adverse events and antitumor efficacy after anti-CD40 therapy, J Immunother Cancer 8(2) (2020) e001687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Liu HC, Viswanath DI, Pesaresi F, Xu Y, Zhang L, Di Trani N, Paez-Mayorga J, Hernandez N, Wang Y, Erm DR, Ho J, Susnjar A, Liu X, Demaria S, Chen SH, Teh BS, Butler EB, Xuan Chua CY, Grattoni A, Potentiating antitumor efficacy through radiation and sustained intratumoral delivery of anti-CD40 and anti-PDL1, Int J Radiat Oncol Biol Phys 110(2) (2021) 492–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Knorr DA, Dahan R, Ravetch JV, Toxicity of an Fc-engineered anti-CD40 antibody is abrogated by intratumoral injection and results in durable antitumor immunity, Proc Natl Acad Sci U S A 115(43) (2018) 11048–11053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Medina-Echeverz J, Ma C, Duffy AG, Eggert T, Hawk N, Kleiner DE, Korangy F, Greten TF, Systemic agonistic anti-CD40 treatment of tumor-bearing mice modulates hepatic myeloid-suppressive cells and causes immune-mediated liver damage, Cancer Immunol Res 3(5) (2015) 557–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Liu C, Lewis CM, Lou Y, Xu C, Peng W, Yang Y, Gelbard AH, Lizee G, Zhou D, Overwijk WW, Hwu P, Agonistic antibody to CD40 boosts the antitumor activity of adoptively transferred T cells in vivo, J Immunother 35(3) (2012) 276–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Sum E, Rapp M, Durr H, Mazumdar A, Romero PJ, Trumpfheller C, Umana P, The tumor-targeted CD40 agonist CEA-CD40 promotes T cell priming via a dual mode of action by increasing antigen delivery to dendritic cells and enhancing their activation, J Immunother Cancer 10(3) (2022) e003264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Bonnans C, Thomas G, He W, Jung B, Chen W, Liao M, Heyen J, Buetow B, Pillai S, Matsumoto D, Chaparro-Riggers J, Salek-Ardakani S, Qu Y, CD40 agonist-induced IL-12p40 potentiates hepatotoxicity, J Immunother Cancer 8(1) (2020) e000624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Siwicki M, Gort-Freitas NA, Messemaker M, Bill R, Gungabeesoon J, Engblom C, Zilionis R, Garris C, Gerhard GM, Kohl A, Lin Y, Zou AE, Cianciaruso C, Bolli E, Pfirschke C, Lin YJ, Piot C, Mindur JE, Talele N, Kohler RH, Iwamoto Y, Mino-Kenudson M, Pai SI, deVito C, Koessler T, Merkler D, Coukos A, Wicky A, Fraga M, Sempoux C, Jain RK, Dietrich PY, Michielin O, Weissleder R, Klein AM, Pittet MJ, Resident Kupffer cells and neutrophils drive liver toxicity in cancer immunotherapy, Sci Immunol 6(61) (2021) eabi7083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Salomon R, Rotem H, Katzenelenbogen Y, Weiner A, Cohen Saban N, Feferman T, Amit I, Dahan R, Bispecific antibodies increase the therapeutic window of CD40 agonists through selective dendritic cell targeting, Nat Cancer 3(3) (2022) 287–302. [DOI] [PubMed] [Google Scholar]
- [135].Bullock K, Richmond A, Suppressing MDSC recruitment to the tumor microenvironment by antagonizing CXCR2 to enhance the efficacy of immunotherapy, Cancers (Basel) 13(24) (2021) 6293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Yang J, Yan C, Vilgelm AE, Chen SC, Ayers GD, Johnson CA, Richmond A, Targeted deletion of CXCR2 in myeloid cells alters the tumor immune environment to improve antitumor immunity, Cancer Immunol Res 9(2) (2021) 200–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Yang J, Bergdorf K, Yan C, Luo W, Chen SC, Ayers GD, Liu Q, Liu X, Boothby M, Weiss VL, Groves SM, Oleskie AN, Zhang X, Maeda DY, Zebala JA, Quaranta V, Richmond A, CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth, Mol Cancer 22(1) (2023) 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho CM, Pryer N, Daniel D, Hwang ES, Rugo HS, Coussens LM, Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells, Cancer Cell 26(5) (2014) 623–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Tian L, Lei A, Tan T, Zhu M, Zhang L, Mou H, Zhang J, Macrophage-based combination therapies as a new strategy for cancer immunotherapy, Kidney Dis (Basel) 8(1) (2022) 26–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Yang L, Dong Y, Li Y, Wang D, Liu S, Wang D, Gao Q, Ji S, Chen X, Lei Q, Jiang W, Wang L, Zhang B, Yu JJ, Zhang Y, IL-10 derived from M2 macrophage promotes cancer stemness via JAK1/STAT1/NF-kappaB/Notch1 pathway in non-small cell lung cancer, Int J Cancer 145(4) (2019) 1099–1110. [DOI] [PubMed] [Google Scholar]
- [141].Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, van Damme J, Mantovani A, Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages, J Immunol 164(2) (2000) 762–7. [DOI] [PubMed] [Google Scholar]
- [142].Bald T, Krummel MF, Smyth MJ, Barry KC, The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapies, Nat Immunol 21(8) (2020) 835–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Bentebibel SE, Hurwitz ME, Bernatchez C, Haymaker C, Hudgens CW, Kluger HM, Tetzlaff MT, Tagliaferri MA, Zalevsky J, Hoch U, Fanton C, Aung S, Hwu P, Curti BD, Tannir NM, Sznol M, Diab A, A first-in-human study and biomarker analysis of NKTR-214, a novel IL2Rbetagamma-Biased cytokine, in patients with advanced or metastatic solid tumors, Cancer Discov 9(6) (2019) 711–721. [DOI] [PubMed] [Google Scholar]
- [144].Margolin K, Morishima C, Velcheti V, Miller JS, Lee SM, Silk AW, Holtan SG, Lacroix AM, Fling SP, Kaiser JC, Egan JO, Jones M, Rhode PR, Rock AD, Cheever MA, Wong HC, Ernstoff MS, Phase I Trial of ALT-803, A novel recombinant IL15 complex, in patients with advanced solid tumors, Clin Cancer Res 24(22) (2018) 5552–5561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Yan C, Lei Y, Lin TJ, Hoskin DW, Ma A, Wang J, IL-17RC is critically required to maintain baseline A20 production to repress JNK isoform-dependent tumor-specific proliferation, Oncotarget 8(26) (2017) 43153–43168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Yan C, Huang WY, Boudreau J, Mayavannan A, Cheng Z, Wang J, IL-17R deletion predicts high-grade colorectal cancer and poor clinical outcomes, Int J Cancer 145(2) (2019) 548–558. [DOI] [PubMed] [Google Scholar]
- [147].Yan C, Richmond A, Th9 and Th17 cells: the controversial twins in cancer immunity, J Clin Invest 130(7) (2020) 3409–3411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Chen K, Zhang Y, Qian L, Wang P, Emerging strategies to target RAS signaling in human cancer therapy, J Hematol Oncol 14(1) (2021) 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Lanman BA, Allen JR, Allen JG, Amegadzie AK, Ashton KS, Booker SK, Chen JJ, Chen N, Frohn MJ, Goodman G, Kopecky DJ, Liu L, Lopez P, Low JD, Ma V, Minatti AE, Nguyen TT, Nishimura N, Pickrell AJ, Reed AB, Shin Y, Siegmund AC, Tamayo NA, Tegley CM, Walton MC, Wang HL, Wurz RP, Xue M, Yang KC, Achanta P, Bartberger MD, Canon J, Hollis LS, McCarter JD, Mohr C, Rex K, Saiki AY, San Miguel T, Volak LP, Wang KH, Whittington DA, Zech SG, Lipford JR, Cee VJ, Discovery of a covalent inhibitor of KRAS(G12C) (AMG 510) for the treatment of solid tumors, J Med Chem 63(1) (2020) 52–65. [DOI] [PubMed] [Google Scholar]
- [150].Fakih M, O’Neil B, Price TJ, Falchook GS, Phase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors., JCO 37 (2019) 3003–3003. [Google Scholar]
- [151].Yan C, Yang J, Saleh N, Chen SC, Ayers GD, Abramson VG, Mayer IA, Richmond A, Inhibition of the PI3K/mTOR pathway in breast cancer to enhance response to immune checkpoint inhibitors in breast cancer, Int J Mol Sci 22(10) (2021) 5207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Zhang Z, Richmond A, Yan C, Immunomodulatory properties of PI3K/AKT/mTOR and MAPK/MEK/ERK inhibition augment response to immune checkpoint blockade in melanoma and triple-negative breast cancer, Int J Mol Sci 23(13) (2022) 7353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Baumann D, Hagele T, Mochayedi J, Drebant J, Vent C, Blobner S, Noll JH, Nickel I, Schumacher C, Boos SL, Daniel AS, Wendler S, Volkmar M, Strobel O, Offringa R, Proimmunogenic impact of MEK inhibition synergizes with agonist anti-CD40 immunostimulatory antibodies in tumor therapy, NatCommun 11(1) (2020) 2176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Kuhn NF, Purdon TJ, van Leeuwen DG, Lopez AV, Curran KJ, Daniyan AF, Brentjens RJ, CD40 ligand-modified chimeric antigen receptor T cells enhance antitumor function by eliciting an endogenous antitumor response, Cancer Cell 35(3) (2019) 473–488 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Yan C, Nebhan CA, Saleh N, Shattuck-Brandt R, Chen SC, Ayers GD, Weiss V, Richmond A, Vilgelm AE, Generation of orthotopic patient-derived xenografts in humanized mice for evaluation of emerging targeted therapies and immunotherapy combinations for melanoma, Cancers (Basel) 15(14) (2023) 3695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, Zhang C, Schnell C, Yang G, Zhang Y, Balbin OA, Barbe S, Cai H, Casey F, Chatterjee S, Chiang DY, Chuai S, Cogan SM, Collins SD, Dammassa E, Ebel N, Embry M, Green J, Kauffmann A, Kowal C, Leary RJ, Lehar J, Liang Y, Loo A, Lorenzana E, Robert McDonald E 3rd, McLaughlin ME, Merkin J, Meyer R, Naylor TL, Patawaran M, Reddy A, Roelli C, Ruddy DA, Salangsang F, Santacroce F, Singh AP, Tang Y, Tinetto W, Tobler S, Velazquez R, Venkatesan K, Von Arx F, Wang HQ, Wang Z, Wiesmann M, Wyss D, Xu F, Bitter H, Atadja P, Lees E, Hofmann F, Li E, Keen N, Cozens R, Jensen MR, Pryer NK, Williams JA, Sellers WR, High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response, Nat Med 21(11) (2015) 1318–25. [DOI] [PubMed] [Google Scholar]
- [157].Yan C, Chen SC, Ayers GD, Nebhan CA, Roland JT, Weiss VL, Johnson DB, Richmond A, Proximity of immune and tumor cells underlies response to BRAF/MEK-targeted therapies in metastatic melanoma patients, NPJ Precis Oncol 6(1) (2022) 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Gronholm M, Feodoroff M, Antignani G, Martins B, Hamdan F, Cerullo V, Patient-derived organoids for precision cancer immunotherapy, Cancer Res 81(12) (2021) 3149–3155. [DOI] [PMC free article] [PubMed] [Google Scholar]


