Immune checkpoint inhibitors are successful immunotherapy modalities that enhance CD8+ T-cell responses. Although T cells are initially primed in draining lymph nodes, the mechanisms that underlie their reactivation inside the tumor microenvironment are less clear. Recent studies have found that not only is the cross-priming of conventional type 1 dendritic cells (cDC1) required to initiate CD8+ T-cell responses during tumor progression, but it also plays a central role in immunotherapy-mediated reactivation of tumor-specific CD8+ T cells for tumor regression. Moreover, many cancer treatment modalities trigger type I IFN responses, which play critical roles in boosting cDC1 cross-priming and CD8+ T-cell reactivation. Inducing type I IFNs within tumors can overcome innate immune resistance and activate antitumor adaptive immunity. Here, we review recent studies on how type I IFN-cDC1 cross-priming reactivates CD8+ T cells and contributes to tumor control by cancer immunotherapy.

Cancer immunotherapy modulates the host's immunity to fight cancer, with immune checkpoint inhibitors (ICI) as successful examples. ICIs use antibodies to block immune inhibitory signals in T cells, such as cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) and the programmed cell death protein 1 (PD-1) pathway, to reinvigorate antitumor T-cell responses (1). T cells, particularly CD8+ cytotoxic T cells, are important targets of cancer immunotherapy because they discriminate tumor cells from normal cells and, thus, kill tumor cells by recognizing tumor-derived mutant neoantigens (2). CD4+ T cells can help not only CD8+ T-cell activation, but also direct tumor killing in some models (3, 4). However, only a few patients completely respond to immunotherapy, which indicates either that directly modulating surface immune inhibitory signals alone does not sufficiently reactivate T cells within tumors, or that tumor-specific T cells are not being generated in the first place. Research is elucidating mechanisms through which tumor-specific CD8+ T-cell responses are reactivated after treatment as a crucial step toward developing new strategies to boost responses to immunotherapy.

Dendritic cells (DC) are specialized antigen-presenting cells with a key role in priming T cells. Specifically, conventional type 1 DCs (cDC1) excel in cross-presenting antigens to initiate CD8+ T-cell responses, while cDC2s preferentially prime CD4+ T-cell responses (5). DCs' recognition of foreign antigens by pattern recognition receptors (PRR) often triggers the secretion of type I IFNs, which are required for activating DCs to prime T cells (6). cDC1s that cross-prime CD8+ T-cell responses are involved in spontaneously rejecting tumor immunity (7). However, after a tumor has been treated, whether the reactivation of dysfunctional CD8+ T cells requires cDC1 cross-priming and how this occurs are still unclear. cDC1s lie within both peripheral lymphoid and tumor tissues; CD8+ T-cell reactivation can occur within both sites (8, 9). We have summarized the studies that reported immunotherapy's antitumor effects, focusing on cDC1 cross-priming that fully reactivates CD8+ T cells. Interestingly, type I IFNs triggered by anticancer treatments are required to stimulate cDC1s (Fig. 1). Combining immunotherapy with traditional therapy modalities that trigger type I IFN signaling and DC cross-priming may enhance CD8+ T-cell responses and synergistically improve treatment efficacy.

Figure 1.

Reactivation of CD8+ T cells through cDC1 cross-priming after tumor treatment. Tumor-targeting antibodies (Ab), radiotherapy (RT), or some chemotherapeutic (doxorubicin, DDR inhibitors, and 6-thio-dG) treatments induce tumor cell death. Apoptotic tumor cells release tumor-associated antigens (TAA) and some danger signals [double-stranded RNA (dsRNA), double-stranded DNA (dsDNA), or HMGB1]. TAAs are taken up and processed by cDC1s and presented on MHC I. Danger signals are sensed by pattern recognition receptors (TLR3, cGAS, and TLR4), which induce type I IFN expression in tumor cells or APCs. Paracrine or autocrine type I IFNs act on cDC1s and induce MHC I, CD40, CD80, and CD86 to help activate CD8+ T cells. cDC1s activate CD8+ T cells by providing MHC I peptides for T-cell receptor (TCR) stimulation, costimulatory signals, and cytokines for third signaling. Activated CD8+ T cells express IFNγ, TNFα, and granzyme and kill tumor cells. PD-L1 is highly expressed on DCs, and PD-L1 blockade also activates DC cross-priming to reactivate CD8+ T cells. Adapted from an image created with BioRender.com.

Figure 1.

Reactivation of CD8+ T cells through cDC1 cross-priming after tumor treatment. Tumor-targeting antibodies (Ab), radiotherapy (RT), or some chemotherapeutic (doxorubicin, DDR inhibitors, and 6-thio-dG) treatments induce tumor cell death. Apoptotic tumor cells release tumor-associated antigens (TAA) and some danger signals [double-stranded RNA (dsRNA), double-stranded DNA (dsDNA), or HMGB1]. TAAs are taken up and processed by cDC1s and presented on MHC I. Danger signals are sensed by pattern recognition receptors (TLR3, cGAS, and TLR4), which induce type I IFN expression in tumor cells or APCs. Paracrine or autocrine type I IFNs act on cDC1s and induce MHC I, CD40, CD80, and CD86 to help activate CD8+ T cells. cDC1s activate CD8+ T cells by providing MHC I peptides for T-cell receptor (TCR) stimulation, costimulatory signals, and cytokines for third signaling. Activated CD8+ T cells express IFNγ, TNFα, and granzyme and kill tumor cells. PD-L1 is highly expressed on DCs, and PD-L1 blockade also activates DC cross-priming to reactivate CD8+ T cells. Adapted from an image created with BioRender.com.

Close modal

Conventional or classical DCs (cDC) are subsets of DCs arising from a common progenitor in the bone marrow that are especially adept at presenting exogenous and endogenous antigens to activate T cells (10). cDCs comprise two main distinct subsets, cDC1 and cDC2 (10, 11). Mouse and human cDC1s are well known for their ability to cross-present antigens and activate CD8+ cytotoxic T cells, although recently, the early priming of CD4+ T cells by cDC1s was found in the setting of cell-associated antigens (12). cDC1s express CD11c, major histocompatibility class II (MHC II), CD8α, XCR1, CLEC9a, CD24, and CD103. Human cDC1s also express CD141. The transcription factors, IRF8, ID2 and Batf3 are required for the development of cDC1. cDC1s include both lymphoid tissue–resident CD8α+ CD11b cDCs and nonlymphoid tissue–resident CD103+CD11b cDCs; the latter can migrate to draining lymph nodes after maturation upon activation (5). cDC2s correspond to the CD11b+ cDCs and preferentially initiate CD4+ T-cell responses in vivo (13). However, both mouse and human cDC2s can also cross-present soluble antigens to CD8+ T cells (11, 12). In addition to CD11c, MHC II, and CD11b, cDC2s also express Sirpα and CD301b (4). CD1c is a marker of cDC2 in humans, but not in mice. The development of cDC2 requires the transcription factors IRF4, RBPJ, KLF4, and RELB (4).

During cross-priming, exogenous antigens are taken up by antigen-presenting cells (APC), processed, and presented on MHC I, which eventually activates CD8+ T-cell responses (14). cDC1s and cross-priming play critical roles in the initiation of antitumor CD8+ T-cell responses (15). The lack of cDC1s in Batf3−/− mice prevents the priming of tumor-specific CD8+ T cells or tumor control (16). The unique role of cDC1s in cross-presenting tumor antigens might be related to the reduced antigen degradation in the endosomal compartment and the unique processing of cell-associated antigens (17, 18). The molecule WDFY4 was identified as being involved in cDC1s' cross-presentation of cell-associated antigens, perhaps through the regulation of vesicular trafficking pathways, but finding the molecules involved in this process will require much more research (18).

In addition to priming and activating, cDC1s also contribute to antitumor CD8+ T-cell responses through recruiting and positioning CD8+ T cells within tumor tissues. In a melanoma tumor model, CD103+ cDC1s within tumor tissues secreted the chemokines CXCL9 and CXCL10, which recruited adoptive transferred CXCR3+ effector CD8+ T cells for tumor clearance (19). Meanwhile, tumor cDC1s express higher amounts of IL12 than other DC subsets or macrophages and may help enhance the cytotoxic function of CD8+ effector T cells within the tumor microenvironment (TME; ref. 20).

Human and mouse type I IFNs include class II α-helical cytokines, such as IFNα (12 subtypes in humans and 14 subtypes in mice), IFNβ, IFNϵ, IFNκ, and IFNω (21). The IFNAR receptor, shared by all type I IFNs, is a heterodimer composed of two subunits and expressed in most cells (21). In cancer, type I IFNs, predominantly IFNα and IFNβ, play critical roles in preventing tumorigenesis and promoting antitumor immunity by acting on multiple cells (22, 23). Type I IFNs can directly induce tumor cells' apoptosis, inhibit their proliferation, and upregulate the presentation of cancer antigens. Type I IFNs promote cross-presentation of cDC1s to activate tumor-specific CD8+ T cells (23). Furthermore, type I IFNs are one of three signals that directly promote T-cell activation, expansion, and differentiation. Type I IFNs' antitumor effects are also complemented by their functions of recruiting CD8+ T cells through chemokine CXCL10, increasing natural killer–cell cytotoxicity, and suppressing the activity of protumorigenic immune cells, like regulatory T cells (Treg) or myeloid-derived suppressor cells (23, 24).

Among these functions, type I IFNs are well-known for promoting the activation of and cross-presentation by DCs (especially cDC1s), which are crucial for initiating antitumor CD8+ T-cell responses. DCs are particularly sensitive to and closely regulated by type I IFNs. Type I IFNs can reduce the rate of endosomal-lysosomal acidification to promote cell-associated antigen persistence in RAB5+ and RAB11+ compartments (6). Upon activation by type I IFNs, CCR7 expression drives the migration of antigen-bearing DCs to the lymph nodes to interact with and prime T cells. Type I IFNs can increase the survival of antigen-bearing cDC1s by upregulating the antiapoptotic genes bcl-2 and bcl-xL. Most importantly, type I IFNs enhance cDC1s' ability to activate CD8+ T cells through upregulating costimulatory molecules, including MHC I, MHC II, CD40, CD80, and CD86 (6).

Using spontaneously rejected tumor graft models, endogenous type I IFNs have been shown to play a central role in cancer immune editing (25). After tumor implantation, type I IFNs were increased within draining lymph nodes, and they enhanced cDC1s' cross-priming ability. Activating IFNAR signaling in cDC1s promoted the priming of tumor antigen–specific CD8+ T cells and resulted in the spontaneous rejection of immunogenic tumors (26, 27). After tumors are established, CD8+ T cells are usually tolerated and dysfunctional. Interestingly, studies have found that dysfunctional CD8+ T cells are reactivated after efficient tumor treatment, which also requires cDC1s activated by type I IFNs. In the next part, we will summarize these studies with a focus on how type I IFNs are induced and how cDC1s' cross-priming is activated to fully reactivate CD8+ T cells during cancer treatment.

The first-generation of FDA-approved tumor-targeting antibodies, including trastuzumab, cetuximab, and rituximab, were designed to target surface oncogenic receptors and were found to inhibit tumor cells by interrupting oncogenic signals, by antibody-dependent cytotoxicity (ADCC), or by complement-dependent cytotoxicity (CDC; refs. 28–31). Studies also found that CD8+ T-cell responses play critical roles during antibody-mediated tumor regression. Trastuzumab binds to HER2, which is overexpressed in a large number of breast cancers. HER2 receptor signaling is connected to higher malignancy, tumor relapse, and mortality (32). The antitumor effect of anti-HER2/neu antibodies was found to be significantly impaired in Rag−/− mice or CD8+ T-cell–depleted wild-type mice. This suggests that anti-HER2/neu antibodies efficiently control tumor growth, depending on tumor-specific CD8+ T cells (33). Moreover, anti-HER2/neu–treated tumor cells released HMGB1 danger signals and activated the MyD88 pathway, both of which are critical for establishing antibody-mediated adaptive immunity. Interestingly, anti-HER2/neu therapy depends on the release of both type I and type II IFNs, suggesting that anti-HER2/neu–treated tumors release HMGB1 to trigger MyD88-dependent Toll-like receptor (TLR) signaling and stimulate type I IFNs, which then activate DCs to prime CD8+ T-cell responses (34). Similar results were found with lymphoma therapy when delivering anti-CD20. Early studies found that anti-CD20 could control tumors by direct killing or via ADCC/CDC. Recently, CD8+ T cells were found to contribute to effective anti-mouse CD20 therapy in a syngeneic B-cell lymphoma mouse model (30, 35, 36). Mechanistically, anti-CD20 treatment promoted DC-mediated cross-presentation, during which macrophage-secreted type IFN was involved (35). Overall, these studies show the essential contribution of DCs and CD8+ T-cell responses for tumor control by oncogenic receptor–targeting antibodies.

CD47 was found to be expressed in a broad range of malignant cells, including acute myeloid leukemia, non-Hodgkin lymphoma, bladder cancer, and breast cancer. CD47 is involved in tumor progression by interacting with receptor signal regulatory protein α (SIRPα) on phagocytes to prevent phagocytosis of tumor cells. Previous studies that used xenograft tumor models showed that antibodies that block human CD47 clear tumors by phagocytosis (37, 38). However, studies in immunocompetent mice revealed that anti-CD47 inhibits tumor progression by enhancing tumor-specific CD8+ T cells (39). Despite being major phagocytes regulated by anti-CD47 within tumors, macrophages are not involved in CD8+ T-cell cross-priming (39). Recent studies have shown that anti-CD47 boosts DCs' cross-priming for CD8+ T-cell activation. Anti-CD47 treatment induced DCs to express higher IFNα levels; tumor control depends on IFNAR signaling in DCs. Through type I IFN, CD47 blockade may enable the activation of NADPH oxidase NOX2 in DCs, thus inhibiting phagosomal acidification and reducing the degradation of tumor mitochondrial DNA (mtDNA) in DCs (40). Interestingly, the induction of type I IFNs in DCs depends on the cyclic GMP-AMP synthase (cGAS)–stimulator of IFN genes (STING) pathway, but not on the conventional MyD88 pathway. Anti-CD47 induced the release of mtDNA, which was recognized by cGAS in DCs' cytosol, thereby inducing type I IFN production (40). Overall, these studies suggest that tumor death induced by tumor-targeting antibody treatment can actually induce type I IFNs, which boost cDC1 cross-priming and CD8+ T-cell activation for complete tumor clearance.

ICIs may release immune inhibitory signaling to activate T cells, but recent studies have shown that CD8+ T cells reactivated by cDC1s are also involved. Preclinical studies showed that the therapeutic effect of anti-CTLA-4 (2) or anti-PD-L1 (8) was abrogated in cDC1-deficient Batf3−/− mice. The antitumor effect of combined anti-PD-1 and anti-CTLA-4, or anti-PD-1 and anti-CD137 also depends on cDC1 (16, 41). There is a concern that the impaired ICI therapy in Batf3−/− mice was due to a deficiency in CD8+ T-cell priming during tumor establishment or in exhausted CD8+ T-cell reactivation after antibody treatment. Recent studies on anti-PD-L1/PD-1 antibody treatment mechanisms have revealed the following: (i) PD-1 blockade induced exhausted CD8+ T-cell proliferation that depends on costimulatory CD28 signaling, which indicates the involvement of APC priming in reactivating exhausted CD8+ T cells (42, 43). (ii) Tumor-infiltrating myeloid cells express much higher PD-L1 than tumor cells and are critical for anti-PD-L1–mediated tumor control (44, 45). Another study showed that both PD-L1 and B7.1 were expressed in peripheral and tumor-associated DCs. Blocking PD-L1 on DCs relieves B7.1 sequestration in cis by PD-L1, which allows the B7.1/CD28 interaction to increase T-cell priming (46). (iii) cDC1s upregulate PD-L1's expression upon antigen uptake and are essential in PD-L1 blockade (47). T cells are primed in draining lymph nodes or reactivated inside tumor tissues, but which are essential for ICIs' function is unclear. Some studies clearly showed that, during treatment, PD-L1 blockade promotes cDC1-mediated reactivation of tumor-infiltrating T cells to control tumors (47, 48). PD-L1 on cDC1s might be essential for limiting T-cell reactivation. Another study found that anti-PD-L1–mediated tumor control depends on the cGAS-STING pathway, which suggests that cGAS may trigger type I IFNs to regulate DC cross-priming during ICI (49). Overall, these studies showed that CD8+ T-cell reactivation induced by ICI treatment requires cDC1 cross-priming and occurs within tumors in addition to the lymph nodes. However, how type I IFN is induced and involved inside the TME remains to be determined.

Conventional anticancer therapies, such as radiotherapy and chemotherapy, have been developed to interfere with the tumor cell cycle, such as the synthesis of DNA or RNA, mitotic spindle formation, and specific oncogenic signaling pathways for cancer survival (50–52). These therapies have been thought to clear tumors through direct killing. However, accumulating studies show that these conventional therapies depend on both innate and adaptive immunity. This treatment-induced tumor cell damage triggers innate immune sensing, including type I IFN production, which activates DCs and further CD8+ T-cell responses, which contribute to complete tumor control (Fig. 1).

Tumor cell DNA damage sensed by the cGAS-STING pathway is an important signal that triggers innate sensing during both tumor establishment and conventional therapies (53–56). Radiotherapy is a traditional tumor treatment modality that is widely used in the clinic because of its direct tumoricidal effect. Recent studies found that radiotherapy induces the leakage of DNA from the nucleus or mitochondria into the cytosol; the cGAS-STING pathway recognition of this DNA in the cytosol is one of the important innate sensing mechanisms. Upon encountering double-stranded DNA, cGAS catalyzes the synthesis of cyclic-di-GMP-AMP, which in turn binds the adapter protein STING on the endoplasmic reticulum and promotes TBK1-dependent IRF3 and NF-κB activation to further produce type I IFNs, proinflammatory cytokines, and chemokines to initiate adaptive antitumor responses (57, 58). Radiotherapy-induced type I IFNs further enhance DC cross-priming and CD8+ T-cell response, which are required for tumor clearance (59–61). Moreover, radiotherapy and anti-PD-L1 treatment synergistically control tumor growth in a CD8+ T-cell–dependent manner (62).

Some drugs might preferentially induce DNA fragments in a unique way. For example, 6-thio-2′-deoxyguanosine (6-thio-dG), a nucleoside analogue, can be incorporated into de novo–synthesized telomeres by telomerase to induce damage on telomeric DNA (63). This damage can activate cGAS-STING and type I IFN production and thus, synergize with PD-L1 blockade therapy (64). However, there are negative mechanisms within apoptotic tumor cells that allow their nucleic acids to escape innate immune sensing. The injured DNA could induce DNA damage response (DDR) factors to initiate DNA repair. Inhibiting the DDR pathway by DDR inhibitors, like drugs targeting PARP and checkpoint kinase I, impairs DNA repair, and inducing more damage can trigger the cGAS-STING pathway and type I IFN production within tumor cells. DDR inhibitors can synergize with PD-L1 blockade to enhance antitumor CD8+ T-cell responses (65). Another study showed that activated caspases, especially CASP9, can promote degradation of genomic DNA without triggering PRR recognition and type I IFNs, and can lead to nonimmunogenic tumor cell death (66). Irradiation can induce tumors to release mtDNA fragments, and pan-caspase inhibitor emricasan or knocking out Casp9 significantly increases tumor cell–derived type I IFNs via cGAS-STING pathway activation (66).

The involvement of RNA damage in triggering innate sensing and type I IFN production during chemotherapy is under debate. There are reports that treatment with anthracycline drugs, such as doxorubicin and mitoxantrone, can induce type I IFNs in tumors through the TLR3 pathway (24). However, the ligand here remains unknown; further research is needed to clarify this.

HMGB1 is another danger signal that has often been reported to be released after conventional therapies. Certain chemotherapeutics, including anthracycline and oxaliplatin, induce immunogenic cell death with the release of HMGB1. HMGB1 is recognized by TLR4 in DCs and activates downstream MyD88 signaling and type I IFN production (67).

IFNα has been used to treat tumors like melanoma and lymphoma. However, the clinical utilization of type I IFNs has been limited because of severe peripheral toxicity when it is delivered systemically (68). To specifically deliver type I IFNs into tumor tissues, some studies fused type I IFNs with tumor-targeting antibodies like anti-EGFR or anti-PD-L1 (69, 70). These fusion proteins increase DC cross-priming and tumor-specific CD8+ T-cell responses and synergize with anti-PD-L1 to control tumors. Various pattern recognition receptor agonists, which can induce type I IFNs, have also been put forward, including TLR agonists, STING agonists, and RIG-I agonists. Unfortunately, the therapeutic effect of many of these IFN inducers has not met expectations. Here, we summarize the progress of several type I IFN inducers in clinical trials (Table 1).

Table 1.

Type I IFN inducers in clinic.

Target PRRsIFN-1 inducerRelevant cancer typeCurrent phaseRemarksReferences
TLR7 Lipo-MERIT Melanoma Phase I It is RNA-lipoplexes and used as vaccine. Systemic IFNα and IP-10 were induced. (80, 81) 
RIG-1 Synthetic RNA oligonucleotide (MK-4621) Advanced solid tumors, leukemia Phase I Combined with pembrolizumab, modest antitumor activity. (82, 83) 
STING ADU-S100 Advanced solid tumors; lymphoma Phase I/II Combined with anti-CTLA4 or pembrolizumab. (84, 85) 
 MK-1454 HNSCC Phase I/II Single agent or combined with pembrolizumab. (86) 
 BMS-986301 Advanced solid tumors Phase I Single or combined with nivolumab and ipilimumab.  
TLR2/4 Bacillus Calmette–Guérin Superficial bladder carcinoma FDA approved (1997) Have clinical trials for other cancers. (87) 
TLR3/MDA5 Poly IC:LC Gliomas; breast, pancreatic, and ovarian cancer Phase I/II  (88, 89) 
TLR4 Monophosphoryl lipid A HPV-associated cervical cancer FDA approved (2009) Used as adjuvant to Cervarix. (87) 
TLR7 Imiquimod Basal cell carcinoma FDA approved (2004)  (87) 
TLR9 CpG Low-grade B-cell lymphoma, SCLC, NSCLC, CRC Phase I/II Limited antitumor effect. (90–92) 
Target PRRsIFN-1 inducerRelevant cancer typeCurrent phaseRemarksReferences
TLR7 Lipo-MERIT Melanoma Phase I It is RNA-lipoplexes and used as vaccine. Systemic IFNα and IP-10 were induced. (80, 81) 
RIG-1 Synthetic RNA oligonucleotide (MK-4621) Advanced solid tumors, leukemia Phase I Combined with pembrolizumab, modest antitumor activity. (82, 83) 
STING ADU-S100 Advanced solid tumors; lymphoma Phase I/II Combined with anti-CTLA4 or pembrolizumab. (84, 85) 
 MK-1454 HNSCC Phase I/II Single agent or combined with pembrolizumab. (86) 
 BMS-986301 Advanced solid tumors Phase I Single or combined with nivolumab and ipilimumab.  
TLR2/4 Bacillus Calmette–Guérin Superficial bladder carcinoma FDA approved (1997) Have clinical trials for other cancers. (87) 
TLR3/MDA5 Poly IC:LC Gliomas; breast, pancreatic, and ovarian cancer Phase I/II  (88, 89) 
TLR4 Monophosphoryl lipid A HPV-associated cervical cancer FDA approved (2009) Used as adjuvant to Cervarix. (87) 
TLR7 Imiquimod Basal cell carcinoma FDA approved (2004)  (87) 
TLR9 CpG Low-grade B-cell lymphoma, SCLC, NSCLC, CRC Phase I/II Limited antitumor effect. (90–92) 

Abbreviations: CRC, colorectal cancer; HNSCC, head and neck squamous cell carcinoma; HPV, human papillomavirus; SCLC, small cell lung cancer.

Better understanding the dual effects of type I IFNs in antitumor immunity may help in developing more effective therapies. Even though it is clear that type I IFNs can activate DCs and promote antitumor CD8+ T-cell responses, sustained exposure to type I IFNs can sometimes impair the host's immunity. First, in both ICI therapy and radiotherapy, the extended exposure of tumor cells to type I IFNs can induce a PD-L1–independent multigenic resistance program (71–73). Second, type I IFNs are strong agonists of inhibitory PD-L1 on DCs and other immune cells, which could induce T-cell exhaustion and even apoptosis (74–76). TMEs with type I IFN activation in plasmacytoid DCs exhibit an immunosuppressive signature, marked by PD-L1, IDO expression, and Treg recruitment, which also supports findings on the negative effects of type I IFNs on antitumor immunity (77). In the future, a potential strategy for exploiting type I IFNs may be to target or induce type I IFNs to specific DC subsets (cDC1s), but not cancer cells within the TME. The construction of an attenuated IFNα and antibody fusion protein is a smart design to fulfill this purpose (78, 79). In the meantime, it is better to combine type I IFNs and inducers with ICIs to overcome the resistance caused by PD-L1 induction.

cDC1 cross-priming is involved not only in initiating a CD8+ T-cell response in the host against tumor establishment, but also in reactivating CD8+ T cells inside the TME. To overcome ICI resistance, one needs to explore treatment modalities that can not only kill tumors, but also trigger innate sensing for more type I IFNs inside the TME. The next generation of IFN-based therapy must be designed to target tumor tissues or even specific cells (cDC1s) efficiently while avoiding a peripheral sink effect and side effects.

No disclosures were reported.

This work was supported, in part, by Cancer Prevention & Research Institute of Texas grant RP180725 to Y.-X. Fu and R. Hannan. We thank Drs. Damiana Chiavolini and Jonathan Feinberg for scientific editing.

1.
Sharma
P
,
Allison
JP
. 
Dissecting the mechanisms of immune checkpoint therapy
.
Nat Rev Immunol
2020
;
20
:
75
6
.
2.
Gubin
MM
,
Zhang
X
,
Schuster
H
,
Caron
E
,
Ward
JP
,
Noguchi
T
, et al
Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens
.
Nature
2014
;
515
:
577
81
.
3.
Tay
RE
,
Richardson
EK
,
Toh
HC
. 
Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms
.
Cancer Gene Ther
2020
;
28
:
5
17
.
4.
Garris
CS
,
Luke
JJ
. 
Dendritic cells, the T-cell-inflamed tumor microenvironment, and immunotherapy treatment response
.
Clin Cancer Res
2020
;
26
:
3901
7
.
5.
Merad
M
,
Sathe
P
,
Helft
J
,
Miller
J
,
Mortha
A
. 
The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting
.
Annu Rev Immunol
2013
;
31
:
563
604
.
6.
Lorenzi
S
,
Mattei
F
,
Sistigu
A
,
Bracci
L
,
Spadaro
F
,
Sanchez
M
, et al
Type I IFNs control antigen retention and survival of CD8α+ dendritic cells after uptake of tumor apoptotic cells leading to cross-priming
.
J Immunol
2011
;
186
:
5142
50
.
7.
Hildner
K
,
Edelson
BT
,
Purtha
WE
,
Diamond
M
,
Matsushita
H
,
Kohyama
M
, et al
Batf3 deficiency reveals a critical role for CD8 + dendritic cells in cytotoxic T cell immunity
.
Science
2008
;
322
:
1097
100
.
8.
Salmon
H
,
Idoyaga
J
,
Rahman
A
,
Leboeuf
M
,
Remark
R
,
Jordan
S
, et al
Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition
.
Immunity
2016
;
44
:
924
38
.
9.
Thompson
ED
,
Enriquez
HL
,
Fu
Y-X
,
Engelhard
VH
. 
Tumor masses support naive T cell infiltration, activation, and differentiation into effectors
.
J Exp Med
2010
;
207
:
1791
804
.
10.
Bottcher
JP
,
Reis e Sousa
C
. 
The role of type 1 conventional dendritic cells in cancer immunity
.
Trends Cancer
2018
;
4
:
784
92
.
11.
Wculek
SK
,
Cueto
FJ
,
Mujal
AM
,
Melero
I
,
Krummel
MF
,
Sancho
D
. 
Dendritic cells in cancer immunology and immunotherapy
.
Nat Rev Immunol
2020
;
20
:
7
24
.
12.
Ferris
ST
,
Durai
V
,
Wu
R
,
Theisen
DJ
,
Ward
JP
,
Bern
MD
, et al
cDC1 prime and are licensed by CD4(+)T cells to induce anti-tumour immunity
.
Nature
2020
;
584
:
624
9
.
13.
Gao
Y
,
Nish
SA
,
Jiang
R
,
Hou
L
,
Licona-Limon
P
,
Weinstein
JS
, et al
Control of T helper 2 responses by transcription factor IRF4-dependent dendritic cells
.
Immunity
2013
;
39
:
722
32
.
14.
Bevan
MJ
. 
Cross-priming
.
Nat Immunol
2006
;
7
:
363
5
.
15.
Sánchez-Paulete
AR
,
Teijeira
A
,
Cueto
FJ
,
Garasa
S
,
Pérez-Gracia
JL
,
Sánchez-Arráez
A
, et al
Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy
.
Ann Oncol
2017
;
28
:
xii44
55
.
16.
Sanchez-Paulete
AR
,
Cueto
FJ
,
Martinez-Lopez
M
,
Labiano
S
,
Morales-Kastresana
A
,
Rodriguez-Ruiz
ME
, et al
Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells
.
Cancer Discov
2016
;
6
:
71
9
.
17.
Savina
A
,
Jancic
C
,
Hugues
S
,
Guermonprez
P
,
Vargas
P
,
Moura
IC
, et al
NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells
.
Cell
2006
;
126
:
205
18
.
18.
Theisen
DJ
,
Davidson
JT
,
Briseño
CG
,
Gargaro
M
,
Lauron
EJ
,
Wang
Q
, et al
WDFY4 is required for cross-presentation in response to viral and tumor antigens
.
Science
2018
;
362
:
694
9
.
19.
Spranger
S
,
Dai
D
,
Horton
B
,
Gajewski
TF
. 
Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy
.
Cancer Cell
2017
;
31
:
711
23
.
20.
Broz
ML
,
Binnewies
M
,
Boldajipour
B
,
Nelson
AE
,
Pollack
JL
,
Erle
DJ
, et al
Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity
.
Cancer Cell
2014
;
26
:
938
.
21.
Pestka
S
,
Krause
CD
,
Walter
MR
. 
Interferons, interferon-like cytokines, and their receptors
.
Immunol Rev
2004
;
202
:
8
32
.
22.
Hervas-Stubbs
S
,
Perez-Gracia
JL
,
Rouzaut
A
,
Sanmamed
MF
,
Le Bon
A
,
Melero
I
. 
Direct effects of type I interferons on cells of the immune system
.
Clin Cancer Res
2011
;
17
:
2619
27
.
23.
Borden
EC
. 
Interferons α and β in cancer: therapeutic opportunities from new insights
.
Nat Rev Drug Discov
2019
;
18
:
219
34
.
24.
Sistigu
A
,
Yamazaki
T
,
Vacchelli
E
,
Chaba
K
,
Enot
DP
,
Adam
J
, et al
Cancer cell–autonomous contribution of type I interferon signaling to the efficacy of chemotherapy
.
Nat Med
2014
;
20
:
1301
9
.
25.
Dunn
GP
,
Bruce
AT
,
Sheehan
KCF
,
Shankaran
V
,
Uppaluri
R
,
Bui
JD
, et al
A critical function for type I interferons in cancer immunoediting
.
Nat Immunol
2005
;
6
:
722
9
.
26.
Fuertes
MB
,
Kacha
AK
,
Kline
J
,
Woo
S-R
,
Kranz
DM
,
Murphy
KM
, et al
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells
.
J Exp Med
2011
;
208
:
2005
16
.
27.
Diamond
MS
,
Kinder
M
,
Matsushita
H
,
Mashayekhi
M
,
Dunn
GP
,
Archambault
JM
, et al
Type I interferon is selectively required by dendritic cells for immune rejection of tumors
.
J Exp Med
2011
;
208
:
1989
2003
.
28.
Teeling
JL
,
French
RR
,
Cragg
MS
,
van den Brakel
J
,
Pluyter
M
,
Huang
H
, et al
Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas
.
Blood
2004
;
104
:
1793
800
.
29.
van Meerten
T
,
van Rijn
RS
,
Hol
S
,
Hagenbeek
A
,
Ebeling
SB
. 
Complement-induced cell death by rituximab depends on CD20 expression level and acts complementary to antibody-dependent cellular cytotoxicity
.
Clin Cancer Res
2006
;
12
:
4027
35
.
30.
Clynes
RA
,
Towers
TL
,
Presta
LG
,
Ravetch
JV
. 
Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets
.
Nat Med
2000
;
6
:
443
6
.
31.
Musolino
A
,
Naldi
N
,
Bortesi
B
,
Pezzuolo
D
,
Capelletti
M
,
Missale
G
, et al
Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer
.
J Clin Oncol
2008
;
26
:
1789
96
.
32.
Hudis
CA
. 
Trastuzumab–mechanism of action and use in clinical practice
.
N Engl J Med
2007
;
357
:
39
51
.
33.
Park
S
,
Jiang
Z
,
Mortenson
ED
,
Deng
L
,
Radkevich-Brown
O
,
Yang
X
, et al
The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity
.
Cancer Cell
2010
;
18
:
160
70
.
34.
Stagg
J
,
Loi
S
,
Divisekera
U
,
Ngiow
SF
,
Duret
H
,
Yagita
H
, et al
Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy
.
Proc Natl Acad Sci U S A
2011
;
108
:
7142
7
.
35.
Ren
Z
,
Guo
J
,
Liao
J
,
Luan
Y
,
Liu
Z
,
Sun
Z
, et al
CTLA-4 limits anti-CD20–mediated tumor regression
.
Clin Cancer Res
2017
;
23
:
193
203
.
36.
Maloney
DG
. 
Anti-CD20 antibody therapy for B-cell lymphomas
.
N Engl J Med
2012
;
366
:
2008
16
.
37.
Jaiswal
S
,
Jamieson
CH
,
Pang
WW
,
Park
CY
,
Chao
MP
,
Majeti
R
, et al
CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis
.
Cell
2009
;
138
:
271
85
.
38.
Chao
MP
,
Alizadeh
AA
,
Tang
C
,
Myklebust
JH
,
Varghese
B
,
Gill
S
, et al
Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma
.
Cell
2010
;
142
:
699
713
.
39.
Liu
X
,
Pu
Y
,
Cron
K
,
Deng
L
,
Kline
J
,
Frazier
WA
, et al
CD47 blockade triggers T cell-mediated destruction of immunogenic tumors
.
Nat Med
2015
;
21
:
1209
15
.
40.
Xu
MM
,
Pu
Y
,
Han
D
,
Shi
Y
,
Cao
X
,
Liang
H
, et al
Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein alpha signaling
.
Immunity
2017
;
47
:
363
73
.
41.
Beavis
PA
,
Henderson
MA
,
Giuffrida
L
,
Davenport
AJ
,
Petley
EV
,
House
IG
, et al
Dual PD-1 and CTLA-4 checkpoint blockade promotes antitumor immune responses through CD4(+)Foxp3(-) cell-mediated modulation of CD103(+) dendritic cells
.
Cancer Immunol Res
2018
;
6
:
1069
81
.
42.
Hui
E
,
Cheung
J
,
Zhu
J
,
Su
X
,
Taylor
MJ
,
Wallweber
HA
, et al
T cell costimulatory receptor CD28 is a primary target for PD-1–mediated inhibition
.
Science
2017
;
355
:
1428
33
.
43.
Kamphorst
AO
,
Wieland
A
,
Nasti
T
,
Yang
S
,
Zhang
R
,
Barber
DL
, et al
Rescue of exhausted CD8 T cells by PD-1–targeted therapies is CD28-dependent
.
Science
2017
;
355
:
1423
7
.
44.
Tang
H
,
Liang
Y
,
Anders
RA
,
Taube
JM
,
Qiu
X
,
Mulgaonkar
A
, et al
PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression
.
J Clin Invest
2018
;
128
:
580
8
.
45.
Lin
H
,
Wei
S
,
Hurt
EM
,
Green
MD
,
Zhao
L
,
Vatan
L
, et al
Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression
.
J Clin Invest
2018
;
128
:
805
15
.
46.
Mayoux
M
,
Roller
A
,
Pulko
V
,
Sammicheli
S
,
Chen
S
,
Sum
E
, et al
Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy
.
Sci Transl Med
2020
;
12
:
eaav7431
.
47.
Peng
Q
,
Qiu
X
,
Zhang
Z
,
Zhang
S
,
Zhang
Y
,
Liang
Y
, et al
PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade
.
Nat Commun
2020
;
11
:
4835
.
48.
Oh
SA
,
Wu
D-C
,
Cheung
J
,
Navarro
A
,
Xiong
H
,
Cubas
R
, et al
PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer
.
Nat Cancer
2020
;
1
:
681
91
.
49.
Wang
H
,
Hu
S
,
Chen
X
,
Shi
H
,
Chen
C
,
Sun
L
, et al
cGAS is essential for the antitumor effect of immune checkpoint blockade
.
Proc Natl Acad Sci U S A
2017
;
114
:
1637
42
.
50.
Bracci
L
,
Schiavoni
G
,
Sistigu
A
,
Belardelli
F
. 
Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer
.
Cell Death Differ
2014
;
21
:
15
25
.
51.
Deng
L
,
Liang
H
,
Fu
S
,
Weichselbaum
RR
,
Fu
YX
. 
From DNA damage to nucleic acid sensing: a strategy to enhance radiation therapy
.
Clin Cancer Res
2016
;
22
:
20
5
.
52.
Xu
MM
,
Pu
Y
,
Zhang
Y
,
Fu
YX
. 
The role of adaptive immunity in the efficacy of targeted cancer therapies
.
Trends Immunol
2016
;
37
:
141
53
.
53.
Marcus
A
,
Mao
AJ
,
Lensink-Vasan
M
,
Wang
L
,
Vance
RE
,
Raulet
DH
. 
Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response
.
Immunity
2018
;
49
:
754
63
.
54.
Harding
SM
,
Benci
JL
,
Irianto
J
,
Discher
DE
,
Minn
AJ
,
Reenberg
RAG
. 
Mitotic progression following DNA damage enables pattern recognition within micronuclei
.
Nature
2017
;
548
:
466
70
.
55.
Mackenzie
KJ
,
Carroll
P
,
Martin
CA
,
Murina
O
,
Fluteau
A
,
Simpson
DJ
, et al
cGAS surveillance of micronuclei links genome instability to innate immunity
.
Nature
2017
;
548
:
461
5
.
56.
Gluck
S
,
Guey
B
,
Gulen
MF
,
Wolter
K
,
Kang
TW
,
Schmacke
NA
, et al
Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence
.
Nat Cell Biol
2017
;
19
:
1061
70
.
57.
Ishikawa
H
,
Ma
Z
,
Barber
GN
. 
STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity
.
Nature
2009
;
461
:
788
92
.
58.
Wu
JX
,
Sun
LJ
,
Chen
X
,
Du
FH
,
Shi
HP
,
Chen
C
, et al
Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA
.
Science
2013
;
339
:
826
30
.
59.
Deng
L
,
Liang
H
,
Xu
M
,
Yang
X
,
Burnette
B
,
Arina
A
, et al
STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors
.
Immunity
2014
;
41
:
843
52
.
60.
Burnette
BC
,
Liang
H
,
Lee
Y
,
Chlewicki
L
,
Khodarev
NN
,
Weichselbaum
RR
, et al
The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity
.
Cancer Res
2011
;
71
:
2488
96
.
61.
Lee
Y
,
Auh
SL
,
Wang
Y
,
Burnette
B
,
Wang
Y
,
Meng
Y
, et al
Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment
.
Blood
2009
;
114
:
589
95
.
62.
Deng
L
,
Liang
H
,
Burnette
B
,
Beckett
M
,
Darga
T
,
Weichselbaum
RR
, et al
Irradiation and anti–PD-L1 treatment synergistically promote antitumor immunity in mice
.
J Clin Invest
2014
;
124
:
687
95
.
63.
Mender
I
,
Gryaznov
S
,
Dikmen
ZG
,
Wright
WE
,
Shay
JW
. 
Induction of telomere dysfunction mediated by the telomerase substrate precursor 6-thio-2'-deoxyguanosine
.
Cancer Discov
2015
;
5
:
82
95
.
64.
Mender
I
,
Zhang
A
,
Ren
Z
,
Han
C
,
Deng
Y
,
Siteni
S
, et al
Telomere stress potentiates STING-dependent anti-tumor immunity
.
Cancer Cell
2020
;
38
:
400
11
.
65.
Sen
T
,
Rodriguez
BL
,
Chen
LM
,
Della Corte
CM
,
Morikawa
N
,
Fujimoto
J
, et al
Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung cancer
.
Cancer Discov
2019
;
9
:
646
61
.
66.
Han
C
,
Liu
Z
,
Zhang
Y
,
Shen
A
,
Dong
C
,
Zhang
A
, et al
Tumor cells suppress radiation-induced immunity by hijacking caspase 9 signaling
.
Nat Immunol
2020
;
21
:
546
54
.
67.
Apetoh
L
,
Ghiringhelli
F
,
Tesniere
A
,
Obeid
M
,
Ortiz
C
,
Criollo
A
, et al
Toll-like receptor 4–dependent contribution of the immune system to anticancer chemotherapy and radiotherapy
.
Nat Med
2007
;
13
:
1050
9
.
68.
Sleijfer
S
,
Bannink
M
,
Gool
AR
,
Kruit
WHJ
,
Stoter
G
. 
Side effects of interferon-α therapy
.
Pharm World Sci
2005
;
27
:
423
31
.
69.
Yang
X
,
Zhang
X
,
Fu
ML
,
Weichselbaum
RR
,
Gajewski
TF
,
Guo
Y
, et al
Targeting the tumor microenvironment with interferon-β bridges innate and adaptive immune responses
.
Cancer Cell
2014
;
25
:
37
48
.
70.
Liang
Y
,
Tang
H
,
Guo
J
,
Qiu
X
,
Yang
Z
,
Ren
Z
, et al
Targeting IFNα to tumor by anti-PD-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance
.
Nat Commun
2018
;
9
:
4586
.
71.
Benci
JL
,
Xu
B
,
Qiu
Y
,
Wu
TJ
,
Dada
H
,
Twyman-Saint Victor
C
, et al
Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade
.
Cell
2016
;
167
:
1540
54
.
72.
Jacquelot
N
,
Yamazaki
T
,
Roberti
MP
,
Duong
CPM
,
Andrews
MC
,
Verlingue
L
, et al
Sustained type I interferon signaling as a mechanism of resistance to PD-1 blockade
.
Cell Res
2019
;
29
:
846
61
.
73.
Chen
J
,
Cao
Y
,
Markelc
B
,
Kaeppler
J
,
Vermeer
JAF
,
Muschel
RJ
. 
Type I IFN protects cancer cells from CD8+ T cell–mediated cytotoxicity after radiation
.
J Clin Invest
2019
;
129
:
4224
38
.
74.
Muhlbauer
M
,
Fleck
M
,
Schutz
C
,
Weiss
T
,
Froh
M
,
Blank
C
, et al
PD-L1 is induced in hepatocytes by viral infection and by interferon-alpha and -gamma and mediates T cell apoptosis
.
J Hepatol
2006
;
45
:
520
8
.
75.
Bazhin
AV
,
Von Ahn
K
,
Fritz
J
,
Werner
J
,
Karakhanova
S
. 
Interferon-α up-regulates the expression of PD-L1 molecules on immune cells through STAT3 and p38 signaling
.
Front Immunol
2018
;
9
:
2129
.
76.
Schreiner
B
,
Mitsdoerffer
M
,
Kieseier
BC
,
Chen
L
,
Hartung
H-P
,
Weller
M
, et al
Interferon-β enhances monocyte and dendritic cell expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: relevance for the immune modulatory effect in multiple sclerosis
.
J Neuroimmunol
2004
;
155
:
172
82
.
77.
Pinto
A
,
Rega
A
,
Crother
TR
,
Sorrentino
R
. 
Plasmacytoid dendritic cells and their therapeutic activity in cancer
.
Oncoimmunology
2012
;
1
:
726
34
.
78.
Cauwels
A
,
Van Lint
S
,
Paul
F
,
Garcin
G
,
De Koker
S
,
Van Parys
A
, et al
Delivering type I interferon to dendritic cells empowers tumor eradication and immune combination treatments
.
Cancer Res
2018
;
78
:
463
74
.
79.
Cauwels
A
,
Van Lint
S
,
Garcin
G
,
Bultinck
J
,
Paul
F
,
Gerlo
S
, et al
A safe and highly efficient tumor-targeted type I interferon immunotherapy depends on the tumor microenvironment
.
Oncoimmunology
2018
;
7
:
e1398876
.
80.
Kranz
LM
,
Diken
M
,
Haas
H
,
Kreiter
S
,
Loquai
C
,
Reuter
KC
, et al
Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy
.
Nature
2016
;
534
:
396
401
.
81.
Sahin
U
,
Oehm
P
,
Derhovanessian
E
,
Jabulowsky
RA
,
Vormehr
M
,
Gold
M
, et al
An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma
.
Nature
2020
;
585
:
107
12
.
82.
Barsoum
J
,
Renn
M
,
Schuberth
C
,
Jakobs
C
,
Schwickart
A
,
Schlee
M
, et al
Selective stimulation of RIG-I with a novel synthetic RNA induces strong anti-tumor immunity in mouse tumor models
.
Cancer Immunol Res
2017
;
5
(
3 Suppl
):
Abstract nr B44
.
83.
Moreno
V
,
Gaudy-Marqueste
C
,
Wermke
M
,
Italiano
A
,
Romano
E
,
Marabelle
A
, et al
794 Safety and efficacy results from a phase 1/1b study of intratumoral MK-4621, a retinoic acid-inducible gene I (RIG-I) agonist, plus intravenous pembrolizumab in patients with advanced solid tumors
.
J Immunother Cancer
2020
;
8
:
A842
.
84.
Zandberg
DP
,
Ferris
R
,
Laux
D
,
Mehra
R
,
Nabell
L
,
Kaczmar
J
, et al
A phase II study of ADU-S100 in combination with pembrolizumab in adult patients with PD-L1+recurrent or metastatic HNSCC: preliminary safety, efficacy and PK/PD results
.
Ann Oncol
2020
;
31
:
S1446
S7
.
85.
Meric-Bernstam
F
,
Sandhu
SK
,
Hamid
O
,
Spreafico
A
,
Kasper
S
,
Dummer
R
, et al
Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas
.
J Clin Oncol
2019
;
37
:
15s
(
suppl; abstr 2507
).
86.
Harrington
KJ
,
Brody
J
,
Ingham
M
,
Strauss
J
,
Cemerski
S
,
Wang
M
, et al
Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas
.
Ann Oncol
2018
;
29
:
712
.
87.
Vacchelli
E
,
Galluzzi
L
,
Eggermont
A
,
Fridman
WH
,
Galon
J
,
Sautès-Fridman
C
, et al
Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy
.
Oncoimmunology
2012
;
1
:
894
907
.
88.
Butowski
N
,
Chang
SM
,
Junck
L
,
DeAngelis
LM
,
Abrey
L
,
Fink
K
, et al
A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05)
.
J Neurooncol
2009
;
91
:
175
82
.
89.
Hartman
LL
,
Crawford
JR
,
Makale
MT
,
Milburn
M
,
Joshi
S
,
Salazar
AM
, et al
Pediatric phase II trials of poly-ICLC in the management of newly diagnosed and recurrent brain tumors
.
J Pediatr Hematol Oncol
2014
;
36
:
451
7
.
90.
Ursu
R
,
Carpentier
A
,
Metellus
P
,
Lubrano
V
,
Laigle-Donadey
F
,
Capelle
L
, et al
Intracerebral injection of CpG oligonucleotide for patients with de novo glioblastoma-A phase II multicentric, randomised study
.
Eur J Cancer
2017
;
73
:
30
7
.
91.
Weihrauch
MR
,
Richly
H
,
von Bergwelt-Baildon
MS
,
Becker
HJ
,
Schmidt
M
,
Hacker
UT
, et al
Phase I clinical study of the toll-like receptor 9 agonist MGN1703 in patients with metastatic solid tumours
.
Eur J Cancer
2015
;
51
:
146
56
.
92.
Thomas
M
,
Ponce-Aix
S
,
Navarro
A
,
Riera-Knorrenschild
J
,
Schmidt
M
,
Wiegert
E
, et al
Immunotherapeutic maintenance treatment with toll-like receptor 9 agonist lefitolimod in patients with extensive-stage small-cell lung cancer: results from the exploratory, controlled, randomized, international phase II IMPULSE study
.
Ann Oncol
2018
;
29
:
2076
84
.