The ability of conventional type-1 dendritic cells (cDC1) to cross-present tumor antigens to CD8+ T cells is critical for the induction of antitumor CTLs. Mice that are constitutively deficient in cDC1 cells have been reported to fail to respond to immunotherapy strategies based on checkpoint inhibitors. However, further work is needed to clarify the precise time during immunotherapy treatment that cDC1 cells are required for the beneficial effect of treatment. Here, we used a refined XCR1-DTR-Venus transgenic mouse model to acutely deplete cDC1 cells and trace their behavior using intravital microscopy. Diphtheria toxin–mediated cDC1 depletion prior to immunotherapy treatment with anti–PD-1 and/or anti-CD137 immunostimulatory mAbs completely ablated antitumor efficacy. The efficacy of adoptive T-cell therapy was also hampered by prior cDC1 depletion. After the onset of immunotherapy treatment, depletion of cDC1s only moderately reduced the therapeutic efficacy of anti–PD-1 and anti-CD137 mAbs. Intravital microscopy of liver-engrafted tumors revealed changes in the intratumoral behavior of cDC1 cells in mice receiving immunotherapy, and treatment with diphtheria toxin to deplete cDC1s impaired tumor T-cell infiltration and function. These results reveal that the functional integrity of the cDC1 compartment is required at the onset of various immunotherapies to successfully treat established tumors.

Significance:

These findings reveal the intratumoral behavior of cDC1 dendritic cells in transgenic mouse models and demonstrate that the efficacy of immunotherapy regimens is precluded by elimination of these cells.

Cross-priming is a key function that controls the induction of antigen-specific CTLs. The ability to uptake cellular remains and process the engulfed antigens for MHC class I presentation was discovered to be mainly performed by a minority subset of dendritic cells (DC; ref. 1). Such myeloid DCs are known as conventional-type 1 dendritic cells (cDC1; ref. 2). They were first identified in the spleen of mice as the CD11c+ DEC205+ CD8α+ cells whose involvement in CD8 T-cell responses was shown (3–5).

Detailed study of the transcripts and proteins expressed by DCs better profiled this cross-presenting subset that is characterized by the surface coexpression of CD11c, XCR1, and DNGR-1 (2). In mice, two subsets of cDC1 cells have been identified depending on expression of the CD103 integrin, which defines the CD103+ migratory cDC1s and a CD103 resident subtype constitutively located in secondary lymphoid organs (6).

Much of the research pertaining to cDC1 function comes from the fact that this subset of DC needs the transcription factors BATF3 and IRF8 for their ontogeny in the bone marrow (7). Hence, Batf3 knockout mice are selectively devoid of cDC1 cells (8, 9). Interestingly, Batf3−/− mice are severely deficient when it comes to mounting antiviral CTL responses as well as rejecting immunogenic spontaneously regressing tumor cell lines (8). Similarly, other mouse strains lacking cDC1 cells (e.g., XCR-1 DTA mice) show impaired antitumor immune responses (10). The main mechanisms carried out by cDC1 cells are MHC class I presentation of exogenous antigens (cross-presentation) and responsiveness to viral double-stranded RNA via endosomal TLR3 (11), giving rise to high levels of IL12 production (12). The function of these DCs is critically upregulated by type-I IFNs (13, 14) and activated helper CD4 T cells expressing CD40 L (13). The exact molecular mechanism that redirects endosomal engulfed material to MHC I for antigen presentation is as yet incompletely understood, although there is genetic evidence for the involvement of proteins related to vesicular trafficking such as WDFY4 (15) and Sec22b (16).

Two groups simultaneously reported that tumor immunotherapy with anti–PD-1 and anti-CD137 mAbs was ineffective in cDC1-deficient Batf3−/− mice (17, 18). The mechanisms could be traced to dysfunctional cross-priming of surrogate tumor antigens (18). Interestingly, adoptive T-cell therapy is also less efficient in Batf3 knockout mice showing a complete lack of antigen spreading that was needed to prevent tumor relapse (19–21). IL12 production by cDC1 cells has also been shown to play a key role in PD-1 blockade antitumor efficacy (22).

In humans, cDC1 cells can be identified because they share with their mouse counterparts the expression of XCR1 and DNGR-1 (23–25). In peripheral blood, they are best identified by the expression of CD141 (BDCA3; ref. 26). Numbers of cDC1 cells in mice and humans are greatly increased by treatment with a soluble form of Flt3L, which promotes their ontogeny in the bone marrow (27–29).

The involvement of cDC1 cells in cancer immunology and immunotherapy was underscored by observations according to which tumors failing to produce the CCL4 or CCL5 chemokines that attract cDC1 cells behave as poorly immunogenic (30, 31). In human tumors, a transcriptional cDC1 signature and the expression of Batf3 are strongly associated with the levels of CD8+ T-cell infiltration (32–34). Reportedly, migratory cDC1s take up tumor antigen in mouse models (35) and may transfer it to secondary lymphoid organ-resident cDC1 cells (36). cDC1 cells seem to have a close functional interaction with natural killer (NK) cells (32, 37), which is counter regulated by prostaglandins (38).

One important question that remains unanswered is whether cDC1 cells are required once immunotherapy has been instigated to mediate and sustain the beneficial effect of treatment. With the help of XCR1-DTR-Venus transgenic mice (39), we have demonstrated that diphtheria toxin (DT)–mediated depletion of cDC1 cells upon treatment hampers response to various immunotherapy agents. Moreover, microscopy imaging indicates that cDC1 cells actively interact with tumor antigen-specific T cells in the tumor microenvironment and are involved in tumor T-cell infiltration.

Mice and cell lines

Mice were housed at the animal facility of the CIMA Universidad de Navarra (Pamplona, Spain). XCR1-DTR-VENUS (Xcr1tm2(HBEGF/Venus)Ksho) mice were obtained from the RIKEN Bioresource Research Center and have been described elsewhere (39). Mice were bred homozygous and crossed with C57BL/6 mice (Envigo) to obtain F1 heterozygous XCR1-DTR-Venus mice. hCD2RFP (C57BL/6 hCD2-DsRed) transgenic mice were a kind gift from Mark Coles from University of York (York, United Kingdom; ref. 40) and were bred to OT-I mice (Jackson, C57BL/6-Tg(TcraTcrb)1100Mjb/J, RRID:IMSR_JAX:003831) to obtain F1 hCD2RFPxOT-I mice. Batf3−/− mice (B6.129S(C)-Batf3tm1Kmm/J, RRID:IMSR_JAX:013755) were kindly provided by Kenneth M. Murphy (Washington University, St. Louis, MO; ref. 8). OT-I and CD45.1 (Jackson) were bred to homocygosity to allow in vivo cell tracking. All animal procedures were approved by the Institutional Animal Experimentation Ethics Committee and the regional government of Navarra (protocol 036-18, 089-18, and 087-21). Ages of mice included in experiments ranged from 8 to 12 weeks.

B16.OVA mouse melanoma cells and MC38 mouse colon carcinoma cell lines were kindly gifted by Dr. Lieping Chen (Yale University, New Haven, CT) and Dr. Karl E Hellström (University of Washington, Seattle, WA), respectively. E0771 tumor cells were obtained from ATCC. B16.OVAiRFP670 tumor cells were generated by lipofectamine 2000 (Thermo Fisher Scientific) transfection of B16.OVA cells with piRFP670-N1 (a gift from Vladislav Verkhusha; Addgene plasmid #45457; http://n2t.net/addgene:45457; RRID:Addgene_45457; ref. 41), an expression plasmid and selected on the basis of FACS in a MoFloAstrios EQ cell sorter (Beckman Coulter). Cultures were tested for Mycoplasma monthly using the Mycoalert kit (Promega). These cell lines were authenticated by Idexx Radil (Case 6592-2012). Cell lines were subjected to two or three passages before using in the experiments. Activated OT-I cells for therapy and imaging experiments were prepared as described previously (20). Briefly, total splenocytes of OT-I or OT-I x hCD2RFP mice were cultured at 106 cells/mL with 100 ng/mL of SIINFEKL peptide and 30 U/mL of hIL2 (Proleukin, Novartis) for 48 hours, then the medium was removed and cells were cultured at the same cell density for an additional 48 hours with 30 U/mL hIL2 in the absence of SIINFEKL peptide. All cells were grown in RPMI1640 media supplemented with GlutaMAX (Gibco), 10% heat-inactivated FBS, 50 μmol/L of 2-mercaptoethanol, 100 U/mL of penicillin, and 100 μg/mL of streptomycin at 37°C with 5% CO2 (complete media).

Mouse tumor studies

A total of 5 × 105 MC38, B16.OVA, or E0771 tumor cells were subcutaneously injected into the right flank of XCR1-DTR-Venus or Batf3−/− mice. Mice were treated at the indicated time points with DT (Sigma) with a single intraperitoneal dose of 1 μg/mL plus two additional doses of 0.5 μg/mL every 3 days to maintain full cDC1 depletion (when indicated in the figures). Tumors were measured every 3 days with a caliper and the volume was calculated (length × width2/2). In addition, mice were monitored for survival and euthanized when any tumor size reached a diameter of 15 mm or mice showed signs of distress. In some experiments, C57BL/6 mice deficient for BATF3 were used. Mice were treated on the indicated days with 200 μg of anti–PD-1 (clone RPM1-14, Bio X cell, RRID:AB_1107747), anti-CD137 (clone 3H3, Bio X cell, RRID:AB_2687721), anti-CTLA4 (9D9, Bio X cell, RRID:AB_10949609), and Rat IgG control (Bio X cell, RRID:AB_2687813) when used in single treatment or with 100 μg/mL of each antibody when used in combination with other immunostimulatory antibodies. For adoptive transfer experiments, mice were intravenously injected with 2 × 106 activated OT-I cells that were isolated and cultured as explained above.

Flow cytometry

For the analysis of cDC1 depletion of XCR1-DTR-Venus in tumors, draining lymph nodes and spleen, MC38-bearing mice were treated with a single dose of 1 μg of DT and were sacrificed at the indicated time points. For the analysis of CD8 T cells, mice were injected with 0.5 × 106 B16.OVA cells. At day 6, mice were treated with 1 μg of DT, at day 7 mice were transferred with 2 × 106 activated OT-I CD8+ T cells, and at days 9 and 12 mice were treated with additional 0.5 μg of DT. Mice were treated with anti PD-1 and anti CD137 antibodies at day 10 and tumors and tumor-draining lymph nodes (TDLN) were harvested at day 14 after tumor inoculation. In the MC38 model, DT was injected at days 6 and 9 and antibodies at days 7 and 10 after tumor cell injection. Tumors and TDLN from euthanized mice were harvested at day 11. Organs were collected and single-cell suspensions were stained with the following antibodies: CD8α (BV510,RRID:AB_2561389; APC, RRID:AB_312750; BUV395, RRID:AB_2732919), F4/80 (PeCy7, RRID:AB_893490; BV421, RRID:AB_10901171), TCRβ BV605 (RRID:AB_2687544), CXCR6 PeCy7 (RRID:AB_2721669), PD-1 PercpCy5.5 (RRID:AB_10550092), Tox PE (RRID:AB_10853657), Ki67 AF488 (RRID:AB_10900418), TCF7 AF647 (RRID:AB_2797631), CD11c (APC, RRID:AB_313778; BV510, RRID:AB_2562010), IA/IE (APC Fluor 780, RRID:AB_1548783; Percp-Cy5.5, RRID:AB_2191072), CD103 PE (RRID:AB_535948), CD11b (BUV395, RRID:AB_2740936; PeCy7, RRID:AB_312798),CD45.2 (PB, RRID:AB_492873; BUV496, RRID:AB_2870691) CD45.1 BV786 (RRID:AB_2740538; all from BioLegend or BD, except TCF1 that was from Cell Signaling Technology and Tox from BD; ref. 42). Immunostained cells were analyzed with a Cytoflex LX or XS (Beckmann Coulter) or a FACScanto flow cytometer (BD). Fluorescence minus one or biological comparison controls were used for cell analysis. FlowJo software (TreeStar) was used for data analysis.

Multiplexed immunofluorescence staining

A six-color multiplex immunofluorescence panel based on tyramide signal amplification was used for simultaneous detection of CD3 (T cells, RRID:AB_443425), CD8 (CTLs, RRID:AB_2756376), Foxp3 (regulatory T cells, RRID:AB_2797979), CD4 (Th cells, RRID:AB_2798898), CD11b (myeloid cells, RRID:AB_1216361), and Ki67 (proliferating cells, RRID:AB_443209) and diamidino-2-phenylindole on tumor sections from formalin-fixed paraffin-embedded (FFPE) samples as described previously (43). Briefly, 4-μm-thick sections obtained from FFPE tissue blocks were deparaffinized and rehydrated from ethanol to water. Antigen retrieval with citrate (pH6, PerkinElmer) or ethylenediaminetetraacetic acid (EDTA; pH9, Dako) target retrieval solution was performed at the beginning of each sequential round of antibody staining. Each round consisted of heat-induced antigen retrieval followed by protein blocking (Antibody Diluent/Block, Akoya Bioscience), incubation with primary antibody, anti-rabbit secondary antibody (Opal Polymer anti-rabbit horseradish peroxidase Kit, Perkin Elmer) finishing with Opal fluorophore incubation diluted in 1XPlus Amplification Diluent (Akoya Bioscience). The panel primary antibody description is provided in Supplementary Materials and Methods and sections were mounted with Faramount Aqueous Mounting Medium (Dako).

Whole tissue sections were scanned on a Vectra-Polaris Automated Quantitative Pathology Imaging System (Akoya Biosciences). Akoya Biosciences’ Inform software (V.2.4.8) was used to remove the autofluorescence as determined by an unstained slide and to perform the spectral unmixing of the images. To analyze the invasive margin, six rectangular regions were randomly selected for each tumor containing the tumor margins, analyzed and the mean infiltration denoted by each marker for each tumor section was plotted.

Intravital microscopy experiments

For intravital microscopy of B16.OVA tumors engrafted in the liver, the spleens of XCR1-DTR-Venus mice were surgically exposed under isoflurane anesthesia and 106 B16.OVA.iRFP670 cells were injected in 50 μL of PBS. Then, spleens were surgically removed, and mice were allowed to recover. Four days later, mice were intravenously injected with activated 5 × 106 OT-IxhCD2RFP CTLs. Mice were intraperitoneally injected with anti–PD-1 plus anti-CD137 or Rat IgG control mAbs at day 7 or 9 after tumor cell injection and DT was given 24 hours before such Ab treatments as indicated in the figures. At days 9 and 11 after tumor cell injection, intravital confocal microscopy of the liver was performed as described previously (44, 45). Mice were injected with 5 μg of CD31-AF647 antibody (RRID:AB_2161030). Briefly, the liver was surgically exposed and isolated from respiratory movements by separating the rib cage by pulling with a thread and keeping the liver tissue adhered to the coverslip with the help of wet pieces of paper. The temperature of the mice was maintained with a rectal probe connected to a heating blanket (Kemp) and mice were kept asleep under Isoflurane anesthesia (2%). Mice were placed on a custom-built stage and imaged with an LSM880 inverted microscope (Zeiss) equipped with a 25X water immersion objective (NA, 0.8). Imaging sessions took from 2 to 4 hours per mice and time-lapse acquisitions lasted from 30 minutes to 2 hours with frames taken every 2 minutes. Several tumor foci were imaged per session and mice. Time-lapse videos were analyzed using the IMARIS (Bitplane) software. Both Venus-positive (Venus+) and red fluorescent protein–positive (RFP+) cells were segmented using the spots tool and tracked manually overtime. Individual cell motility parameters were generated from such tracks. Speed (track length/time), chemotactic index or straightness (track displacement length/track length), and motility index (track displacement length2/4*track duration) were calculated. Interactions of T cells with Venus+ cells were quantified selecting time points in which Venus+ cells were closer than 12 μm from an RFP+ CD8+ T cell.

Time-lapse videos were generated in IMARIS software and edited with Final Cut Pro (Apple software).

Quantification and statistical analysis

Data were processed using GraphPad Prism 6.0. Means and SEM are presented as averages and error bars unless otherwise indicated in the figure legends. All experimental repetitions and numbers of specimens and mice are indicated in the figure legends. For the statistical comparison of tumor growth, individual AUC were calculated for each mouse and compared using ANOVA tests, which were used to analyze statistical differences between independent groups unless otherwise indicated. P values are shown for any relevant statistical difference in the figures.

Data availability

The data generated in this study are available upon request from the corresponding authors.

Depletion of cDC1 cells in the tumor tissue microenvironment and TDLNs in XCR1-DTR-Venus transgenic mice

To investigate the need for the presence of cDC1 cells during immunotherapy, we made use of XCR1-DTR-Venus transgenic mice (39). cDC1 cells can be identified upon flow cytometry analysis of CD11c+ CD11b cells because of selective expression of the Venus fluorescent protein. We engrafted MC38-derived tumors under the skin of XCR1-DTR-Venus mice and intraperitoneally injected DT (Fig. 1A).

Figure 1.

XCR1-Venus-DTR transgenic mice permit tracing and depletion of cDC1 cells in lymphoid organs and engrafted tumor tissue. A, Schematic view of XCR1-DTR-Venus and a representative FACS dot plot of cells in the spleen showing CD11c on the x-axis and Venus fluorescent protein on the y-axis. A population of Venus+ cells can be observed only in the CD11c+ population. Representative microphotograph of fluorescent Venus+ cells within tumors is shown to highlight that cDC1 can also be tracked by fluorescence microscopy. B, Representative flow cytometry dot plots showing cDC1 depletion as the percentage of Venus+ cells among dendritic cells (CD45+, F4/80, CD11c+, MHC II+) or based on cDC1 surface phenotype in the spleen (CD45+, F4/80, CD11c+, MHC II+, CD8α+, CD11b), lymph nodes (CD45+, F4/80, CD11c+, MHC II+, CD103α+, CD11b), and in the leukocyte infiltrate of MC38-derived tumors (CD45+, F4/80, CD11c+, MHC II+, CD103α+, CD11b) 24 hours after DT intraperitoneal injection. C and D, Summary data from experiments as in B (n = 4 mice). Each dot represents a single mouse.

Figure 1.

XCR1-Venus-DTR transgenic mice permit tracing and depletion of cDC1 cells in lymphoid organs and engrafted tumor tissue. A, Schematic view of XCR1-DTR-Venus and a representative FACS dot plot of cells in the spleen showing CD11c on the x-axis and Venus fluorescent protein on the y-axis. A population of Venus+ cells can be observed only in the CD11c+ population. Representative microphotograph of fluorescent Venus+ cells within tumors is shown to highlight that cDC1 can also be tracked by fluorescence microscopy. B, Representative flow cytometry dot plots showing cDC1 depletion as the percentage of Venus+ cells among dendritic cells (CD45+, F4/80, CD11c+, MHC II+) or based on cDC1 surface phenotype in the spleen (CD45+, F4/80, CD11c+, MHC II+, CD8α+, CD11b), lymph nodes (CD45+, F4/80, CD11c+, MHC II+, CD103α+, CD11b), and in the leukocyte infiltrate of MC38-derived tumors (CD45+, F4/80, CD11c+, MHC II+, CD103α+, CD11b) 24 hours after DT intraperitoneal injection. C and D, Summary data from experiments as in B (n = 4 mice). Each dot represents a single mouse.

Close modal

Twenty-four hours after DT treatment, splenic CD11c+ CD11b CD8α+ cells almost disappeared, a phenomenon that was also observed in terms of the disappearance of MHC-II+ Venus+ cells (Fig. 1BD; Supplementary Fig. S1A). TDLNs experienced a similar outcome following DT treatment (Fig. 1BD; Supplementary Fig. S1A). In excised tumors, all Venus+ DCs and most CD103+ CD11c+ MHCII+ F4/80 DCs were absent as a result of DT treatment (Fig. 1BD; Supplementary Fig. S1A). In contrast, cDC2s were respected upon DT treatment (Supplementary Fig. S1B). Depletion of cDC1 cells following a single DT injection lasted for at least 5 days (Supplementary Fig. S1C). cDC1 cell numbers remained low for 5 days and eventually recovered when mice were analyzed at day 7 after DT treatment (Supplementary Fig. S1D).

cDC1 cells are required for the successful treatment of established tumors with various immunotherapy agents

We used the MC38 tumor model to characterize the need for cDC1 cells during treatment with efficacious immunotherapies. MC38-derived subcutaneous tumors partially respond to anti-CD137, or to anti–PD-1 mAbs and intensely respond to a combination of anti-CD137 plus anti–PD-1 mAbs (18). Experiments of DT depletion and immunostimulatory mAb treatments were performed in MC38 tumor-bearing XCR1-DTR-Venus transgenic mice and in Batf3 knockout mice (Fig. 2A). DT treatment started at day 4 after tumor inoculation, while the first immunostimulatory mAb was given at day 5. At day 4, the expansion of anti-gp70 tumor-reactive CD8 T cells were readily detectable in such tumor-bearing mice (Supplementary Fig. S2A). The partial efficacy of treatment of anti–PD-1 or anti-CD137 mAbs was lost in the groups of mice pretreated with DT (Fig. 2B). The synergistic immunotherapy combination of anti–PD-1 plus anti-CD137 mAbs was very efficacious, eradicating 14 of 20 tumors, and again such efficacy was completely lost in DT-treated mice and in cDC1 constitutively deficient Batf3−/− mice (Fig. 2C). In sentry mice from the experiments in Fig. 2B and C, cDC1 cells were verified as having been efficiently depleted (Supplementary Fig. S2B). Of note, DT treatment did not impair the efficacy of immunotherapy in wild-type (WT) mice (Supplementary Fig. S2C).

Figure 2.

Abrupt depletion of cDC1 dendritic cells during immunotherapy with immunostimulatory mAbs impairs its efficacy. A, Schematic timeline of subcutaneous tumor growth experiments in XCR1-DTR-Venus mice treated with the indicated immunostimulatory antibodies and/or DT. B, Tumor size follow-up studies in XCR1-DTR-Venus mice depleted or not from cDC1 and treated with 200 μg of anti–PD-1, anti-CD137, or Rat IgG as a control. Mean ± SEM is shown; n = 6 mice per group. C, Tumor size follow-up studies in XCR1-DTR-Venus mice or Batf3/ mice depleted or not of cDC1 and treated with 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG as a control. Mean ± SEM is shown. The proportions of cured mice per group are shown in the adjacent legends. D, Schematic timeline and tumor size follow-up studies in XCR1-DTR-Venus mice predepleted or not with intraperitoneal DT 2 days prior to MC38 tumor cell inoculation and treated with 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG (as shown in the scheme). E, Schematic timeline and tumor size follow-up studies in XCR1-DTR-Venus mice depleted or not with intraperitoneal DT starting one day after immunotherapy treatment with 200 μg of anti–PD-1 or anti-CD137 mAbs or 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG (as shown in the scheme). The proportions of cured mice per group are shown in the adjacent legend. Mean ± SEM is shown; n = 5, 6 mice per group. ANOVA tests of AUC were performed for statistical analyses; P values are shown for statistical comparisons.

Figure 2.

Abrupt depletion of cDC1 dendritic cells during immunotherapy with immunostimulatory mAbs impairs its efficacy. A, Schematic timeline of subcutaneous tumor growth experiments in XCR1-DTR-Venus mice treated with the indicated immunostimulatory antibodies and/or DT. B, Tumor size follow-up studies in XCR1-DTR-Venus mice depleted or not from cDC1 and treated with 200 μg of anti–PD-1, anti-CD137, or Rat IgG as a control. Mean ± SEM is shown; n = 6 mice per group. C, Tumor size follow-up studies in XCR1-DTR-Venus mice or Batf3/ mice depleted or not of cDC1 and treated with 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG as a control. Mean ± SEM is shown. The proportions of cured mice per group are shown in the adjacent legends. D, Schematic timeline and tumor size follow-up studies in XCR1-DTR-Venus mice predepleted or not with intraperitoneal DT 2 days prior to MC38 tumor cell inoculation and treated with 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG (as shown in the scheme). E, Schematic timeline and tumor size follow-up studies in XCR1-DTR-Venus mice depleted or not with intraperitoneal DT starting one day after immunotherapy treatment with 200 μg of anti–PD-1 or anti-CD137 mAbs or 100 μg of anti–PD-1 plus anti-CD137 or Rat IgG (as shown in the scheme). The proportions of cured mice per group are shown in the adjacent legend. Mean ± SEM is shown; n = 5, 6 mice per group. ANOVA tests of AUC were performed for statistical analyses; P values are shown for statistical comparisons.

Close modal

To study when cDC1 cells were needed for immunotherapy efficacy, we performed DT-mediated depletion at different time points. Mice that recovered from preengraftment transient depletion attained similar efficacy upon anti-CD137 plus anti–PD-1 treatment (Fig. 2D). However, tumor growth prior to immunotherapy onset was hastened in predepleted groups, suggesting an immunotherapy-independent cDC1-mediated control of tumor progression at least in this tumor model (Fig. 2D). When DT treatment was performed 1 day after the first immunotherapeutic mAb injections (Fig. 2E), anti–PD-1 and anti-CD137 efficacy were reduced upon cDC1 depletion but the efficacy of the combination of both antibodies was retained irrespective of cDC1 cell depletion at that timepoint.

In the clinic, anti–PD-1 plus anti-CTLA4 (nivolumab plus ipilimumab) constitutes the approved treatment of choice in a number of malignancies (46–48). Figure 3A shows that treatment with a mouse surrogate of such a combination lost most of its efficacy upon DT treatment against the transplanted E0771 breast cancer model. Interestingly, when testing cDC1 depletion in anti–PD-1– and anti-CTLA4–based immunotherapies, cDC1 cells were only required for the efficacy of PD-1 blockade but not for the efficacy of anti-CTLA4 mAbs (Fig. 3B).

Figure 3.

cDC1 depletion impairs the efficacy of PD-1 blockade and adoptive T-cell therapy. A, Schematic timeline (top), summary of tumor size follow-ups (left), and individual tumor growth of E0771 tumors (right) implanted in XCR1-DTR-Venus mice depleted or not of cDC1 cells and treated with 100 μg of anti–PD-1 plus anti–CTLA4 or Rat IgG. Mean ± SEM is shown; n = 6 mice per group. B, Experiments as in A but using 200 μg of anti–PD-1 or 200 μg of anti–CTLA4 as single immunotherapy treatments. C, Schematic timeline (top), summary of tumor size follow-ups (left), and individual tumor growth of B16.OVA tumors (right) implanted in XCR1-DTR-Venus mice depleted or not of cDC1 cells and treated with two intravenous infusions of 2 × 106 activated OT-I cells (black arrows). Mean ± SEM is shown; n = 8–12 mice per group. ANOVA tests of AUC were performed for statistical analyses; P values are shown.

Figure 3.

cDC1 depletion impairs the efficacy of PD-1 blockade and adoptive T-cell therapy. A, Schematic timeline (top), summary of tumor size follow-ups (left), and individual tumor growth of E0771 tumors (right) implanted in XCR1-DTR-Venus mice depleted or not of cDC1 cells and treated with 100 μg of anti–PD-1 plus anti–CTLA4 or Rat IgG. Mean ± SEM is shown; n = 6 mice per group. B, Experiments as in A but using 200 μg of anti–PD-1 or 200 μg of anti–CTLA4 as single immunotherapy treatments. C, Schematic timeline (top), summary of tumor size follow-ups (left), and individual tumor growth of B16.OVA tumors (right) implanted in XCR1-DTR-Venus mice depleted or not of cDC1 cells and treated with two intravenous infusions of 2 × 106 activated OT-I cells (black arrows). Mean ± SEM is shown; n = 8–12 mice per group. ANOVA tests of AUC were performed for statistical analyses; P values are shown.

Close modal

Adoptive T-cell therapy is also gaining clinical application (49, 50). To model it, we used B16.OVA engrafted tumors and adoptive transfer of activated OT-I TCR-transgenic T cells recognizing ovalbumin (OVA). Results in Fig. 3C indicate that the control of tumors by adoptively transferred OT-I cells was partially lost upon cDC1 depletion. These results are consistent with previous observations in Batf3−/− tumor-bearing mice treated with adoptive T-cell therapy (19), but highlight the relevance of cDC1s specifically during the administration of T cells.

In all previous experiments, we used homozygous XCR1-DTR-Venus knock-in mice, that happen to have a disrupted XCR1 locus due to the fact that depletions were more efficient in such DTR homozygous mice. However, this created a doubt as to the potential importance of XCR1 for the efficacy of immunotherapy. This was ruled out as shown in Supplementary Fig. S3, because therapies with anti-CD137+ anti–PD-1 (Supplementary Fig. S3A), anti–PD-1+anti-CTLA4 (Supplementary Fig. S3B) and OT-I adoptive T-cell transfer (Supplementary Fig. S3C) were as efficacious in homozygous as in heterozygous transgenic mice, which have a functional XCR1 gene in the latter case.

cDC1 depletion reduces T-cell infiltration into the tumor microenvironment under immunotherapy and alters CD8 T-cell stemness and exhaustion in tumor and TDLNs

Focusing on the efficacy of anti–PD-1+anti-CD137 antibodies, we used multiplex tissue immunofluorescence to study the lymphocyte infiltrates of MC38 tumors under therapy with and without cDC1 depletion.

As can be seen in Fig. 4A at two different magnifications, DT depletion caused a dramatic reduction of T cells infiltrating the tumor on day 6 after treatment onset, when mice had received two doses of immunostimulatory mAbs. Interestingly, upon cDC1 depletion, T cells remained on the rim of the invasive tumor margins without penetrating to the center of the engrafted tumor nodules (Fig. 4A). Quantitative analysis and statistical comparisons of the images are provided in Fig. 4B and C, showing that the observed effects regarding T-cell density were less prominent at the tumor margins.

Figure 4.

Depletion of cDC1 cells during immunotherapy with immunostimulatory mAbs reduces tumor infiltration by T cells. A, Representative multiplex immunofluorescence images of MC38 tumors treated with two doses of anti–PD-1 plus anti-CD137 ± DT (tumors were harvested on day 9 after tumor cell inoculation). Widefield views are shown on the left. Bars, 200 μm. Zoomed-in images of the areas indicated by white rectangles on the left are shown images on the right. Bars, 50 μm. Images are from a representative tumor section from each condition. B, Quantification of infiltrated immune cells in the MC38 tumors in images as in A. C, Quantification of T-cell infiltrates only in the tumor margins. n = 5 mice per group. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Figure 4.

Depletion of cDC1 cells during immunotherapy with immunostimulatory mAbs reduces tumor infiltration by T cells. A, Representative multiplex immunofluorescence images of MC38 tumors treated with two doses of anti–PD-1 plus anti-CD137 ± DT (tumors were harvested on day 9 after tumor cell inoculation). Widefield views are shown on the left. Bars, 200 μm. Zoomed-in images of the areas indicated by white rectangles on the left are shown images on the right. Bars, 50 μm. Images are from a representative tumor section from each condition. B, Quantification of infiltrated immune cells in the MC38 tumors in images as in A. C, Quantification of T-cell infiltrates only in the tumor margins. n = 5 mice per group. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Close modal

It has been recently reported that during chronic lymphocytic choriomeningitis (LCMV) infection cDC1 are able to support TCF1+ CD8+ precursor cells (51). The absence of cDC1 cells during the use of PD-1 blockade in the viral model induces a loss of CD8+ T cells, with precursor features (TCF1+) enforcing their excessive proliferation and leading faster to exhaustion. To test the functional effects of cDC1 absence during immunotherapy in the cancer model setting, we implanted B16.OVA tumors in XCR-1 DTR mice and adoptively transferred activated OVA-specific CD8+ CD45.1+ OT-I T cells to such mice to henceforth be able to trace antigen-specific CD8 T-cell responses (Fig. 5A). cDC1 depletion did not have any impact on the proportion of transferred OT-I T cells recovered from the tumor draining lymph nodes at the evaluated timepoint (5 days after starting cDC1 depletion, 4 days after immunotherapy onset). However, the recovered OT-I CD8+ T cells showed markedly less TCF1 expression, particularly in the absence of cDC1 during immunotherapy and showed upregulated CXCR6 expression as recently described in the LCMV chronic infection model (Fig. 5B; ref. 51). Immunotherapy enhanced the overall number of proliferating OT-I cells, but proliferating cells were TCF1low, mainly when cDC1 cells were absent. This was in contrast with the nondepleted mice in which proliferating cells were mainly TCF1hi (Fig. 5C). We also analyzed the endogenous CD8+ PD1+ cells in the TDLNs (Fig. 5D) and observed very similar results regarding TCF1 expression reduction (Fig. 5E) and higher ki67-denoted proliferation in such TCF1low CD8+ T cell (Fig. 5F). In the tumor compartment, we confirmed the impaired infiltration of transferred CD8+ T cells (Fig. 5G) as a result of cDC1 depletion. Moreover, we observed increased expression of the exhaustion marker TOX2 in the absence of cDC1 cells during immunotherapy in OT-I CD8+ tumor-infiltrating lymphocytes (Fig. 5H). However, in this intratumoral location, the effects of cDC1 depletion on TCF1 and proliferation were not as evident as in the TDLNs (Fig. 5,I and J). In the endogenous CD8+ tumor-infiltrating lymphocytes less tumor infiltration, less exhaustion marker expression levels and similar TCF1 levels were also observed (Fig. 5,KM).

Figure 5.

In the absence of cDC1 cells, antitumor CD8 T cells show reduced stemness and increased expression of exhaustion markers. Mice bearing subcutaneous B16.OVA tumors were transferred with 2 × 106 preactivated OT-I T cells and were treated or not with DT and anti–PD-1 +anti-CD137 antibodies. Cell suspensions derived from tumors and TDLNs were then analyzed by flow cytometry. OT-I cells were gated based on their CD45.1 expression and endogenous CD8 T cells were gated as CD45.2. AF show analyses performed in T cells from the TDLNs and GM show analyses performed in tumor-infiltrating CD8+ T cells. n = 6–7 mice per group. In the Rat IgG DT group, some marker analyses could not be performed in samples from a single mouse due to the very low numbers of recovered OT-I cells in the TDLN. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Figure 5.

In the absence of cDC1 cells, antitumor CD8 T cells show reduced stemness and increased expression of exhaustion markers. Mice bearing subcutaneous B16.OVA tumors were transferred with 2 × 106 preactivated OT-I T cells and were treated or not with DT and anti–PD-1 +anti-CD137 antibodies. Cell suspensions derived from tumors and TDLNs were then analyzed by flow cytometry. OT-I cells were gated based on their CD45.1 expression and endogenous CD8 T cells were gated as CD45.2. AF show analyses performed in T cells from the TDLNs and GM show analyses performed in tumor-infiltrating CD8+ T cells. n = 6–7 mice per group. In the Rat IgG DT group, some marker analyses could not be performed in samples from a single mouse due to the very low numbers of recovered OT-I cells in the TDLN. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Close modal

To test whether these changes in CD8 T cells were observable in another tumor model, we analyzed endogenous CD8+ T cells infiltrating MC38 tumors and from TDLNs in the same experimental conditions. The effects observed in the TDLN of such mice (Supplementary Figs. S4A–S4C) were similar to the ones observed in B16.OVA TDLNs, but even more clear, with almost complete loss of TCF1 expression upon cDC1 depletion. In MC38 tumors, infiltrating CD8+ T cells showed similar changes following cDC1 depletion, that included upregulation of exhaustion markers, loss of TCF1 expression and enhanced proliferation of the TCF1low population (Supplementary Figs. S4D–S4F).

To further understand the effects of cDC1 to sustain already activated T cells, we transferred T cells from MC38-bearing mice previously cured with immunotherapy (anti–PD-1+anti-CD137 treated) into Batf3−/− recipient mice having no cDC1 cells or control WT mice. Transferred T cells were able to more efficaciously control tumors in WT mice than in cDC1-deficient Batf3−/− mice (Supplementary Fig. S4G).

Intravital microscopy evidence for cDC1 sustainment of CD8+ T-cell responses in the tumor microenvironment under immunotherapy

The selective expression of Venus in XCR1+ cDC1 cells in XCR1-DTR-Venus mice offered an opportunity for in vivo time-lapse confocal microscopy to trace their presence and behavior. We chose a model in which mice bear metastatic melanoma lesions in the liver (Fig. 6A) to allow tumor visualization. In this setting, tumor cells are injected into the spleen to disseminate through the portal vein 10 days before imaging. Of note, spleens are removed to avoid unwanted tumor cell engraftment in this organ. Multiple tumor nodules appeared in the liver when a B16.OVA melanoma variant transfected to express the fluorescent protein iRFP670 was intrasplenically injected. Hence, fluorescent tumor cells expressing the surrogate antigen OVA can be visualized. Moreover, XCR1-DTR-Venus mice were adoptively transferred with preactivated TCR-transgenic OT-1 T cells that are cotransgenic for RFP to simultaneously permit visualization and tracking of anti-OVA tumor-specific CD8+ T cells (Fig. 6B). The liver central lobe can be surgically exposed and microscopy imaging can be performed for up to 4 or 5 hours. Figure 6B and Supplementary Movie S1 show that cDC1 cells (bright green), OT-I cells (red), and B16.OVA melanoma cells (blue) can be covisualized overtime in this experimental setting. OT-I cells were activated before adoptive transfer to bypass possible effects of cDC1 cells on priming in these experiments.

Figure 6.

cDC1 dendritic cells infiltrate the tumor microenvironment and better perform their functions under anti–PD-1 plus anti-CD137 immunotherapy. A, Scheme showing the setup for intravital microscopy of melanoma lesions spread to the liver of XCR1-DTR-Venus mice transferred with antitumor OT-I CD8+ T cells. B, Representative image of a metastatic melanoma lesion in the liver showing B16.OVAiRFP670 tumor cells (blue), infiltrating Venus+ cDC1 cells (intense green), and anti–OVA-specific red fluorescent OT-I cells (red). Images correspond to Supplementary Movie S1. C, Representative snapshots of a time-lapse sequence of a tumor visualized as in A, focusing on the behavior of cDC1 Venus+ cells in the tumor. The sequence shows DCs actively migrating (3 and 4) or not moving (1 and 2); interacting (1) or not (2) with intratumor OVA-specific OT-I CTLs. Individual tracks of moving cDC1 cells are overlaid as yellow lines. The sequence corresponds to Supplementary Movie S2. D, Representative images showing differential cDC1 infiltration in tumor lesions (circles) in Rat IgG- or anti–PD-1 + anti–CD137–treated mice following the experimental scheme shown in A and quantification of cDC1 density per tumor lesion. n = 3–4 mice per condition and each dot represents an individual tumor lesion. EG, Single-cell cDC1 behavior was tracked in time-lapse videos such as Supplementary Movies S1 and S2. cDC1 interaction time with CTLs (E), speed (F), and meandering index (directionality; G) is shown. n = 3–4 mice per condition; 2–3 lesions per mice; each dot represents a single tracked cell. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Figure 6.

cDC1 dendritic cells infiltrate the tumor microenvironment and better perform their functions under anti–PD-1 plus anti-CD137 immunotherapy. A, Scheme showing the setup for intravital microscopy of melanoma lesions spread to the liver of XCR1-DTR-Venus mice transferred with antitumor OT-I CD8+ T cells. B, Representative image of a metastatic melanoma lesion in the liver showing B16.OVAiRFP670 tumor cells (blue), infiltrating Venus+ cDC1 cells (intense green), and anti–OVA-specific red fluorescent OT-I cells (red). Images correspond to Supplementary Movie S1. C, Representative snapshots of a time-lapse sequence of a tumor visualized as in A, focusing on the behavior of cDC1 Venus+ cells in the tumor. The sequence shows DCs actively migrating (3 and 4) or not moving (1 and 2); interacting (1) or not (2) with intratumor OVA-specific OT-I CTLs. Individual tracks of moving cDC1 cells are overlaid as yellow lines. The sequence corresponds to Supplementary Movie S2. D, Representative images showing differential cDC1 infiltration in tumor lesions (circles) in Rat IgG- or anti–PD-1 + anti–CD137–treated mice following the experimental scheme shown in A and quantification of cDC1 density per tumor lesion. n = 3–4 mice per condition and each dot represents an individual tumor lesion. EG, Single-cell cDC1 behavior was tracked in time-lapse videos such as Supplementary Movies S1 and S2. cDC1 interaction time with CTLs (E), speed (F), and meandering index (directionality; G) is shown. n = 3–4 mice per condition; 2–3 lesions per mice; each dot represents a single tracked cell. Mann–Whitney U tests were performed for statistical analyses; P values are shown.

Close modal

Focusing on cDC1 cells (Fig. 6C), we were able to trace their behavior and their interactions with OT-I cells. As can be seen in Fig. 6C and Supplementary Movie S2, some cDC1 cells actively moved (termed migrating cells in Fig. 6C) and interacted with T cells (cell #2 in Fig. 6C). First, when these experiments were performed in mice receiving anti–PD-1 plus anti-CD137 mAbs, given 48 hours earlier, more cDC1 cells could be found in the metastatic foci (Fig. 6D). We observed a nonstatistically significant upregulation of CCL5 mRNA within MC38 tumors treated with immunotherapy but no major changes in other DC chemoattracting chemokines as XCL1 or CCL4 were found (Supplementary Fig. S5A) Monitoring cDC1 behavior overtime, we detected that a fraction of cDC1 cells established more durable interactions with OT-I cells in mice receiving immunotherapy as compared with cDC1 cells in mice receiving IgG control antibodies (Fig. 6E). However, the cDC1 cells also tended to move at higher speeds (Fig. 6F) in a direction that seemed purposeful, because the directionality of migration was also significantly higher under immunotherapy with the immunostimulatory antibodies (Fig. 6G). When such experiments were performed in the absence of OT-I (Supplementary Fig. S5B), we observed a similar effect regarding enhanced speed of cDC1 cells upon immunotherapy treatment but without increased directional movement, therefore implying that adoptively transferred OT-I cells may underlie enhanced DC activation in these experiments.

Next, we focused on T-cell behavior comparing conditions in which mice had been depleted of cDC1 cells 72 hours earlier with DT and/or given immunotherapy with anti–PD-1 plus anti-CD137 mAbs. Most importantly, under conditions of immunotherapy, OT-I cells readily infiltrated the tumor, but such an increase was impaired by DT-mediated depletion of cDC1 cells (Fig. 7A and B; Supplementary Movie S3).

Figure 7.

Impact of DT-mediated depletion of cDC1 cells on the behavior of antitumor CD8 T cells upon immunotherapy treatment. A, Representative snapshots of liver tumor lesions imaged as in Fig. 5 showing individual tracks (30-minute follow-up) of antitumor specific OT-I cells (red) to highlight enhanced focus of CTLs interacting with tumor cells. Tumors of XCR1-DTR-Venus mice treated with control IgG or anti–PD-1 + anti-CD137 mAbs and treated or not with DT are shown. B, Anti–OVA-specific CD8+ T-cell density in tumor lesions in each of the three conditions. n = 2–3 mice per condition. Dots, individual tumor lesions. C and D, Single-cell behavior of antitumor fluorescent OT-I cells was tracked in time-lapse videos as those shown in Supplementary Movie S3. Speed (C) and motility coefficient (D) are shown for T cells within the tumor lesions or in the surrounding liver tissue in XCR1-Venus mice treated with Rat IgG (control), anti–PD-1+anti-CD137 mAb ± DT depletion as indicated. n = 2–3 mice per condition (1–3 tumor lesions per mice). Dots, single cells. ANOVA tests were performed for statistical analyses; P values are shown. n.s., not significant.

Figure 7.

Impact of DT-mediated depletion of cDC1 cells on the behavior of antitumor CD8 T cells upon immunotherapy treatment. A, Representative snapshots of liver tumor lesions imaged as in Fig. 5 showing individual tracks (30-minute follow-up) of antitumor specific OT-I cells (red) to highlight enhanced focus of CTLs interacting with tumor cells. Tumors of XCR1-DTR-Venus mice treated with control IgG or anti–PD-1 + anti-CD137 mAbs and treated or not with DT are shown. B, Anti–OVA-specific CD8+ T-cell density in tumor lesions in each of the three conditions. n = 2–3 mice per condition. Dots, individual tumor lesions. C and D, Single-cell behavior of antitumor fluorescent OT-I cells was tracked in time-lapse videos as those shown in Supplementary Movie S3. Speed (C) and motility coefficient (D) are shown for T cells within the tumor lesions or in the surrounding liver tissue in XCR1-Venus mice treated with Rat IgG (control), anti–PD-1+anti-CD137 mAb ± DT depletion as indicated. n = 2–3 mice per condition (1–3 tumor lesions per mice). Dots, single cells. ANOVA tests were performed for statistical analyses; P values are shown. n.s., not significant.

Close modal

We analyzed the behavior of T cells under immunotherapy with and without cDC1 depletion. Previous results using anti-CD137 treatment had shown that OT-I cells upon such immunotherapy stayed focused on targets and showed less overall motility (52). In keeping with these previous findings, OT-I cells under anti–PD-1 plus anti-CD137 immunotherapy showed reduced speed and motility (distance away from the initial location; Fig. 7C and D) and were more focused on the tumor target cells (Supplementary Movie S3). Notably, this focusing effect was lost when cDC1 cells were depleted by DT. Interestingly, CD8+ T cells showed no differences in speed or motility in the surrounding nontumor liver tissue (Fig. 7C and D).

Overall, this intravital microscopy experimentation indicates that cDC1 cells are not only important to mediate infiltration of T cells recognizing tumor antigens, but also are able to change the functional T-cell behavior in the tumor tissue microenvironment.

The requirement for cDC1 cells to be present and functional immediately before immunotherapy treatments with immunostimulatory mAbs is clearly demonstrated here. We took advantage of XCR1-DTR-Venus transgenic mice, which had been used before to reveal the role of cDC1 cells in the cross-priming of listeria antigens as well as in LCMV chronic infection (39, 51). DT depletions in the tumor microenvironment and TDLNs were almost complete and lasted for at least 5 days. Nonetheless, our DT dosing scheme was more frequent to ensure complete cDC1 absence. These results are compatible with the lack of efficacy of various immunotherapies in Batf3−/− mice (17, 18, 53) and make the point that the presence of cDC1 cells at the onset of treatment is a must for immunotherapy to be efficacious. Our experiments do not yet address whether migratory (CD103+) or secondary lymphoid organ-resident (CD103) cDC1 cells are those that are necessary and it may well be the case that both dendritic cell subsets play key roles (36).

Our previous experiments in Batf3−/− mice had shown that these cells were critically needed for antigen-specific CD8 T-cell priming in tumor-bearing mice (18). Importantly, these cDC1 cells are also key to prime against antigen material derived from cytotoxicity as mediated by NK, CTL, and chimeric antigen receptor T cells (21, 54). Therefore, a virtuous circle is postulated according to which the cytotoxic effects of immunotherapy provide more raw material for tumor antigen cross presentation by cDC1 cells. The common feature is that cDC1 cells are continuously needed for immunotherapy agents such as anti–PD-1, anti-CD137, intratumoral poly I:C (53), radioimmunotherapy (55), and adoptive T-cell therapy (19–21). These facts speak of the key importance of the functions mediated by cDC1 cells in oncoimmunology approaches, either with approved or with promising agents under clinical trial development (56).

In keeping with previous observations (30), we observe that cDC1 cells are critical to allow intratumoral T-cell infiltration. Our multiplex immunofluorescence images and time-lapse confocal intravital imaging are conclusive in that regard. The underlying mechanisms remain under investigation and probably involve chemokine loops under the IL12-IFNγ axis as reported previously (30, 31). Observations following OT-I CD8+ T-cell transfer are also in agreement with this conclusion. Furthermore, cDC1 seem to be necessary to avoid the expression of exhaustion markers and maintain the markers related to stemness and self-renewal in such TCR transgenic OT-I cells (51).

Intravital imaging of tumor metastases nested in the liver permitted time-lapse imaging of the tumors and the surrounding nontumor liver parenchyma. Three facts became clear in these intravital microsocopy experiments: (i) cDC1 cells change their behavior under immunotherapy conditions within the tumor microenvironment; (ii) CD8+ T cells recognizing tumor antigens are attracted and stimulated by cDC1 cells in the baseline tumor microenvironment and more intensely once immunotherapy has been instigated; and (iii) cDC1 cells are required for optimal dynamics and function of CD8+ T cells within the tumor microenvironment during immunotherapy. The quantitative parameters of motility, number and duration of cellular interactions in the liver nested tumors are indirect measurements that estimate profound functional changes, even if the molecular mechanisms in action are probably multiple and remain to be fully elucidated. Complex bidirectional functional consequences of cDC1 interactions with cognate T cells are likely to ultimately underlie the efficacy of immunotherapy treatments.

Given the absolute requirement for the presence of cDC1 cells for immunotherapy of established tumors to be efficacious, strategies that enhance their presence and activity in tumors make a great deal of sense in clinical translation. For instance, sFLT3L (28, 29) or local delivery of chemokines attracting cDC1s (42) are strong partner candidates to enhance efficacy of various immunotherapy approaches.

I. Melero reports grants and personal fees from BMS, AstraZeneca, Genmab, Roche-Genentech, Alligator, Bioncotech; personal fees from F-Star, pharmamar, F-Star, Numab, Biolinerx, Dompe, Boston Therapeutics, Third Rock, Servier, and Pierre Fabre outside the submitted work; in addition, I. Melero has a patent for Bioncotech issued and a patent for Genmab pending. No disclosures were reported by the other authors.

A. Teijeira: Conceptualization, resources, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, writing–review and editing. S. Garasa: Data curation, formal analysis, validation, investigation, methodology. C. Luri-Rey: Investigation, methodology, writing–review and editing. C. de Andrea: Data curation, formal analysis, validation, methodology. M. Gato: Formal analysis, investigation, methodology, writing–review and editing. C. Molina: Investigation, methodology, writing–review and editing. T. Kaisho: Resources, writing–review and editing. A. Cirella: Investigation, methodology, writing–review and editing. A. Azpilikueta: Data curation, investigation, methodology, writing–review and editing. S.K. Wculek: Resources, methodology, writing–review and editing. J. Egea: Investigation, methodology, writing–review and editing. I. Olivera: Data curation, investigation, methodology, writing–review and editing. I. Rodriguez: Investigation, writing–review and editing. A. Rouzaut: Resources, formal analysis, writing–review and editing. V. Verkhusha: Resources, writing–review and editing. K. Valencia: Validation, investigation, writing–review and editing. D. Sancho: Conceptualization, resources, writing–review and editing. P. Berraondo: Resources, data curation, methodology, writing–review and editing. I. Melero: Conceptualization, resources, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing.

This work was supported by Spanish Ministry of Economy and Competitiveness and Spanish Ministry of Research [MINECO SAF2014-52361-R and SAF 2017-83267-C2-1R and PID2020-112892RB-100, PID2020-113174-RA-100 (AEI/FEDER,UE), financed by MCIN/AEI/10.13039/501100011033], Cancer Research Institute under the CRI-CLIP, Asociación Española Contra el Cancer (AECC) Foundation under Grant GCB15152947MELE, Joint Translational Call for Proposals 2015 (JTC 2015) TRANSCAN-2 (code: TRS-2016-00000371), projects PI14/01686, PI13/00207, PI16/00668, PI19/01128, funded by Instituto de Salud Carlos III and co-funded by European Union (ERDF, “A way to make Europe”), European Commission within the Horizon 2020 Programme (PROCROP - 635122), Gobierno de Navarra Proyecto LINTERNA Ref: 0011-1411, Mark Foundation, Fundación BBVA and Fundación Olga Torres. A. Teijeira is supported by the Ramon y Cajal program from the Spanish Ministry of Science (RYC2019-026406-I financiada por MCIN/AEI /10.13039/501100011033 y por El FSE invierte en tu futuro).

Esther Guirado and Dr. Belen Palencia are acknowledged for project managing, Dr. Diego Alignani for excellent flow cytometry assistance, the light microscopy unit and Carlos Ortiz de Solórzano for their support in microscopy experiments, and Dr. Paul Miller for English editing.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Joffre
OP
,
Segura
E
,
Savina
A
,
Amigorena
S
.
Cross-presentation by dendritic cells
.
Nat Rev Immunol
2012
;
12
:
557
69
.
2.
Cabeza-Cabrerizo
M
,
Cardoso
A
,
Minutti
CM
,
Pereira da Costa
M
,
Reis e Sousa
C
.
Dendritic cells revisited
.
Annu Rev Immunol
2021
;
39
:
131
66
.
3.
den Haan
JM
,
Lehar
SM
,
Bevan
MJ
.
CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo
.
J Exp Med
2000
;
192
:
1685
96
.
4.
Vremec
D
,
Zorbas
M
,
Scollay
R
,
Saunders
DJ
,
Ardavin
CF
,
Wu
L
, et al
.
The surface phenotype of dendritic cells purified from mouse thymus and spleen: investigation of the CD8 expression by a subpopulation of dendritic cells
.
J Exp Med
1992
;
176
:
47
58
.
5.
Steinman
RM
.
Decisions about dendritic cells: past, present, and future
.
Annu Rev Immunol
2012
;
30
:
1
22
.
6.
Edelson
BT
,
Kc
W
,
Juang
R
,
Kohyama
M
,
Benoit
LA
,
Klekotka
PA
, et al
.
Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells
.
J Exp Med
2010
;
207
:
823
36
.
7.
Murphy
TL
,
Grajales-Reyes
GE
,
Wu
X
,
Tussiwand
R
,
Briseno
CG
,
Iwata
A
, et al
.
Transcriptional control of dendritic cell development
.
Annu Rev Immunol
2016
;
34
:
93
119
.
8.
Hildner
K
,
Edelson
BT
,
Purtha
WE
,
Diamond
M
,
Matsushita
H
,
Kohyama
M
, et al
.
Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity
.
Science
2008
;
322
:
1097
100
.
9.
Ataide
MA
,
Komander
K
,
Knopper
K
,
Peters
AE
,
Wu
H
,
Eickhoff
S
, et al
.
BATF3 programs CD8(+) T cell memory
.
Nat Immunol
2020
;
21
:
1397
407
.
10.
Mattiuz
R
,
Brousse
C
,
Ambrosini
M
,
Cancel
JC
,
Bessou
G
,
Mussard
J
, et al
.
Type 1 conventional dendritic cells and interferons are required for spontaneous CD4(+) and CD8(+) T-cell protective responses to breast cancer
.
Clin Transl Immunology
2021
;
10
:
e1305
.
11.
Schulz
O
,
Diebold
SS
,
Chen
M
,
Naslund
TI
,
Nolte
MA
,
Alexopoulou
L
, et al
.
Toll-like receptor 3 promotes cross-priming to virus-infected cells
.
Nature
2005
;
433
:
887
92
.
12.
Gautier
G
,
Humbert
M
,
Deauvieau
F
,
Scuiller
M
,
Hiscott
J
,
Bates
EE
, et al
.
A type I interferon autocrine-paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells
.
J Exp Med
2005
;
201
:
1435
46
.
13.
Snijders
A
,
Kalinski
P
,
Hilkens
CM
,
Kapsenberg
ML
.
High-level IL-12 production by human dendritic cells requires two signals
.
Int Immunol
1998
;
10
:
1593
8
.
14.
Le Bon
A
,
Etchart
N
,
Rossmann
C
,
Ashton
M
,
Hou
S
,
Gewert
D
, et al
.
Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon
.
Nat Immunol
2003
;
4
:
1009
15
.
15.
Theisen
DJ
,
D
JTt
,
Briseno
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
.
16.
Cebrian
I
,
Visentin
G
,
Blanchard
N
,
Jouve
M
,
Bobard
A
,
Moita
C
, et al
.
Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells
.
Cell
2011
;
147
:
1355
68
.
17.
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
.
18.
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
.
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.
Etxeberria
I
,
Bolanos
E
,
Quetglas
JI
,
Gros
A
,
Villanueva
A
,
Palomero
J
, et al
.
Intratumor adoptive transfer of IL-12 mRNA transiently engineered antitumor CD8(+) T cells
.
Cancer Cell
2019
;
36
:
613
29
.
21.
Conde
E
,
Vercher
E
,
Soria-Castellano
M
,
Suarez-Olmos
J
,
Mancheno
U
,
Elizalde
E
, et al
.
Epitope spreading driven by the joint action of CART cells and pharmacological STING stimulation counteracts tumor escape via antigen-loss variants
.
J Immunother Cancer
2021
;
9
:
e003351
.
22.
Garris
CS
,
Arlauckas
SP
,
Kohler
RH
,
Trefny
MP
,
Garren
S
,
Piot
C
, et al
.
Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-gamma and IL-12
.
Immunity
2018
;
49
:
1148
61
.
23.
Poulin
LF
,
Reyal
Y
,
Uronen-Hansson
H
,
Schraml
BU
,
Sancho
D
,
Murphy
KM
, et al
.
DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues
.
Blood
2012
;
119
:
6052
62
.
24.
Bachem
A
,
Guttler
S
,
Hartung
E
,
Ebstein
F
,
Schaefer
M
,
Tannert
A
, et al
.
Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells
.
J Exp Med
2010
;
207
:
1273
81
.
25.
Sancho
D
,
Mourao-Sa
D
,
Joffre
OP
,
Schulz
O
,
Rogers
NC
,
Pennington
DJ
, et al
.
Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin
.
J Clin Invest
2008
;
118
:
2098
110
.
26.
Poulin
LF
,
Salio
M
,
Griessinger
E
,
Anjos-Afonso
F
,
Craciun
L
,
Chen
JL
, et al
.
Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells
.
J Exp Med
2010
;
207
:
1261
71
.
27.
Morse
MA
,
Nair
S
,
Fernandez-Casal
M
,
Deng
Y
,
St Peter
M
,
Williams
R
, et al
.
Preoperative mobilization of circulating dendritic cells by Flt3 ligand administration to patients with metastatic colon cancer
.
J Clin Oncol
2000
;
18
:
3883
93
.
28.
Hammerich
L
,
Marron
TU
,
Upadhyay
R
,
Svensson-Arvelund
J
,
Dhainaut
M
,
Hussein
S
, et al
.
Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination
.
Nat Med
2019
;
25
:
814
24
.
29.
Breton
G
,
Zheng
S
,
Valieris
R
,
Tojal da Silva
I
,
Satija
R
,
Nussenzweig
MC
.
Human dendritic cells (DCs) are derived from distinct circulating precursors that are precommitted to become CD1c+ or CD141+ DCs
.
J Exp Med
2016
;
213
:
2861
70
.
30.
Spranger
S
,
Bao
R
,
Gajewski
TF
.
Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
31.
Ruiz de Galarreta
M
,
Bresnahan
E
,
Molina-Sanchez
P
,
Lindblad
KE
,
Maier
B
,
Sia
D
, et al
.
β-Catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma
.
Cancer Discov
2019
;
9
:
1124
41
.
32.
Barry
KC
,
Hsu
J
,
Broz
ML
,
Cueto
FJ
,
Binnewies
M
,
Combes
AJ
, et al
.
A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments
.
Nat Med
2018
;
24
:
1178
91
.
33.
Garris
CS
,
Luke
JJ
.
Dendritic cells, the T-cell-inflamed tumor microenvironment, and immunotherapy treatment response
.
Clin Cancer Res
2020
;
26
:
3901
7
.
34.
Spranger
S
,
Luke
JJ
,
Bao
R
,
Zha
Y
,
Hernandez
KM
,
Li
Y
, et al
.
Density of immunogenic antigens does not explain the presence or absence of the T-cell-inflamed tumor microenvironment in melanoma
.
Proc Natl Acad Sci U S A
2016
;
113
:
E7759
E68
.
35.
Roberts
EW
,
Broz
ML
,
Binnewies
M
,
Headley
MB
,
Nelson
AE
,
Wolf
DM
, et al
.
Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma
.
Cancer Cell
2016
;
30
:
324
36
.
36.
Ruhland
MK
,
Roberts
EW
,
Cai
E
,
Mujal
AM
,
Marchuk
K
,
Beppler
C
, et al
.
Visualizing synaptic transfer of tumor antigens among dendritic cells
.
Cancer Cell
2020
;
37
:
786
99
.
37.
Bottcher
JP
,
Bonavita
E
,
Chakravarty
P
,
Blees
H
,
Cabeza-Cabrerizo
M
,
Sammicheli
S
, et al
.
NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control
.
Cell
2018
;
172
:
1022
37
.
38.
Bonavita
E
,
Bromley
CP
,
Jonsson
G
,
Pelly
VS
,
Sahoo
S
,
Walwyn-Brown
K
, et al
.
Antagonistic inflammatory phenotypes dictate tumor fate and response to immune checkpoint blockade
.
Immunity
2020
;
53
:
1215
29
.
39.
Yamazaki
C
,
Sugiyama
M
,
Ohta
T
,
Hemmi
H
,
Hamada
E
,
Sasaki
I
, et al
.
Critical roles of a dendritic cell subset expressing a chemokine receptor, XCR1
.
J Immunol
2013
;
190
:
6071
82
.
40.
Veiga-Fernandes
H
,
Coles
MC
,
Foster
KE
,
Patel
A
,
Williams
A
,
Natarajan
D
, et al
.
Tyrosine kinase receptor RET is a key regulator of Peyer's patch organogenesis
.
Nature
2007
;
446
:
547
51
.
41.
Shcherbakova
DM
,
Verkhusha
VV
.
Near-infrared fluorescent proteins for multicolor in vivo imaging
.
Nat Methods
2013
;
10
:
751
4
.
42.
Sanchez-Paulete
AR
,
Teijeira
A
,
Quetglas
JI
,
Rodriguez-Ruiz
ME
,
Sanchez-Arraez
A
,
Labiano
S
, et al
.
Intratumoral immunotherapy with XCL1 and sFlt3L encoded in recombinant semliki forest virus-derived vectors fosters dendritic cell-mediated T-cell cross-priming
.
Cancer Res
2018
;
78
:
6643
54
.
43.
Alvarez
M
,
Molina
C
,
De Andrea
CE
,
Fernandez-Sendin
M
,
Villalba
M
,
Gonzalez-Gomariz
J
, et al
.
Intratumoral co-injection of the poly I:C-derivative BO-112 and a STING agonist synergize to achieve local and distant anti-tumor efficacy
.
J Immunother Cancer
2021
;
9
:
e002953
.
44.
Marques
PE
,
Antunes
MM
,
David
BA
,
Pereira
RV
,
Teixeira
MM
,
Menezes
GB
.
Imaging liver biology in vivo using conventional confocal microscopy
.
Nat Protoc
2015
;
10
:
258
68
.
45.
Teijeira
A
,
Garasa
S
,
Gato
M
,
Alfaro
C
,
Migueliz
I
,
Cirella
A
, et al
.
CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity
.
Immunity
2020
;
52
:
856
71
.
46.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Rutkowski
P
,
Lao
CD
, et al
.
Five-year survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2019
;
381
:
1535
46
.
47.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Aren Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
.
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
90
.
48.
Yau
T
,
Kang
YK
,
Kim
TY
,
El-Khoueiry
AB
,
Santoro
A
,
Sangro
B
, et al
.
Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: The CheckMate 040 randomized clinical trial
.
JAMA Oncol
2020
;
6
:
e204564
.
49.
Morotti
M
,
Albukhari
A
,
Alsaadi
A
,
Artibani
M
,
Brenton
JD
,
Curbishley
SM
, et al
.
Promises and challenges of adoptive T-cell therapies for solid tumours
.
Br J Cancer
2021
;
124
:
1759
76
.
50.
Yang
JC
,
Rosenberg
SA
.
Adoptive T-cell therapy for cancer
.
Adv Immunol
2016
;
130
:
279
94
.
51.
Dahling
S
,
Mansilla
AM
,
Knopper
K
,
Grafen
A
,
Utzschneider
DT
,
Ugur
M
, et al
.
Type 1 conventional dendritic cells maintain and guide the differentiation of precursors of exhausted T cells in distinct cellular niches
.
Immunity
2022
;
55
:
656
70
.
52.
Weigelin
B
,
Bolanos
E
,
Teijeira
A
,
Martinez-Forero
I
,
Labiano
S
,
Azpilikueta
A
, et al
.
Focusing and sustaining the antitumor CTL effector killer response by agonist anti-CD137 mAb
.
Proc Natl Acad Sci U S A
2015
;
112
:
7551
6
.
53.
Aznar
MA
,
Planelles
L
,
Perez-Olivares
M
,
Molina
C
,
Garasa
S
,
Etxeberria
I
, et al
.
Immunotherapeutic effects of intratumoral nanoplexed poly I:C
.
J Immunother Cancer
2019
;
7
:
116
.
54.
Minute
L
,
Teijeira
A
,
Sanchez-Paulete
AR
,
Ochoa
MC
,
Alvarez
M
,
Otano
I
, et al
.
Cellular cytotoxicity is a form of immunogenic cell death
.
J Immunother Cancer
2020
;
8
:
e000325
.
55.
Rodriguez-Ruiz
ME
,
Rodriguez
I
,
Garasa
S
,
Barbes
B
,
Solorzano
JL
,
Perez-Gracia
JL
, et al
.
Abscopal effects of radiotherapy are enhanced by combined immunostimulatory mAbs and are dependent on CD8 T cells and crosspriming
.
Cancer Res
2016
;
76
:
5994
6005
.
56.
Etxeberria
I
,
Glez-Vaz
J
,
Teijeira
A
,
Melero
I
.
New emerging targets in cancer immunotherapy: CD137/4-1BB costimulatory axis
.
ESMO Open
2020
;
4
:
e000733
.