Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance

Similar documents
% of live splenocytes. STAT5 deletion. (open shapes) % ROSA + % floxed

well for 2 h at rt. Each dot represents an individual mouse and bar is the mean ±

L1 on PyMT tumor cells but Py117 cells are more responsive to IFN-γ. (A) Flow

SUPPLEMENTARY INFORMATION

Bezzi et al., Supplementary Figure 1 *** Nature Medicine: doi: /nm Pten pc-/- ;Zbtb7a pc-/- Pten pc-/- ;Pml pc-/- Pten pc-/- ;Trp53 pc-/-

Supplemental Table 1. Primer sequences for transcript analysis

Supplemental Figure 1. Signature gene expression in in vitro differentiated Th0, Th1, Th2, Th17 and Treg cells. (A) Naïve CD4 + T cells were cultured

Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis

Supplemental Figure 1

SUPPLEMENTARY INFORMATION

Nature Immunology: doi: /ni.3412

Supplemental Information. Granulocyte-Monocyte Progenitors and. Monocyte-Dendritic Cell Progenitors Independently

Combined Rho-kinase inhibition and immunogenic cell death triggers and propagates immunity against cancer

Nature Medicine doi: /nm.3957

Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Supplementary Figure 1. Deletion of Smad3 prevents B16F10 melanoma invasion and metastasis in a mouse s.c. tumor model.

Chronic variable stress activates hematopoietic stem cells

Supplemental Information. Aryl Hydrocarbon Receptor Controls. Monocyte Differentiation. into Dendritic Cells versus Macrophages

TITLE: MODULATION OF T CELL TOLERANCE IN A MURINE MODEL FOR IMMUNOTHERAPY OF PROSTATIC ADENOCARCINOMA

Supplementary Materials for

Supplementary Figure 1. Characterization of basophils after reconstitution of SCID mice

Supporting Information

Supplementary Figure 1 Chemokine and chemokine receptor expression during muscle regeneration (a) Analysis of CR3CR1 mrna expression by real time-pcr

Nature Immunology: doi: /ni Supplementary Figure 1. Examples of staining for each antibody used for the mass cytometry analysis.

sequences of a styx mutant reveals a T to A transversion in the donor splice site of intron 5

Supplementary Figure 1. mrna expression of chitinase and chitinase-like protein in splenic immune cells. Each splenic immune cell population was

pplementary Figur Supplementary Figure 1. a.

Targeting tumour associated macrophages in anti-cancer therapies. Annamaria Gal Seminar Series on Drug Discovery Budapest 5 January 2018

Appendix Figure S1 A B C D E F G H

IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia

NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies

Intracellular MHC class II molecules promote TLR-triggered innate. immune responses by maintaining Btk activation

Supplemental Table I.

Darwinian selection and Newtonian physics wrapped up in systems biology

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human)

Supplementary figure 1. Systemic delivery of anti-cd47 antibody controls tumor growth in

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Natural Killer Cells: Development, Diversity, and Applications to Human Disease Dr. Michael A. Caligiuri

SUPPLEMENTARY METHODS

Supplementary Figure 1: STAT3 suppresses Kras-induced lung tumorigenesis

Leukocyte Complexity Predicts Breast Cancer Survival and Functionally Regulates Response to Chemotherapy

Supplemental Information. Gut Microbiota Promotes Hematopoiesis to Control Bacterial Infection. Cell Host & Microbe, Volume 15

Supplemental Information. Tissue-Resident Macrophages in Pancreatic. Ductal Adenocarcinoma Originate from Embryonic

Supplementary Figure 1. NAFL enhanced immunity of other vaccines (a) An over-the-counter, hand-held non-ablative fractional laser (NAFL).

Dendritic cell subsets and CD4 T cell immunity in Melanoma. Ben Wylie 1 st year PhD Candidate

Supplementary Figure 1. Example of gating strategy

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

SUPPLEMENTARY FIGURES

Supplementary Figure 1 IL-27 IL

A Versatile Tool to Study Immune Checkpoint Therapeutics: Syngeneic Tumor Mouse Models in vivo

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow

Immune Checkpoints. PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich

Haematopoietic stem cells

Nature Immunology: doi: /ni Supplementary Figure 1. Transcriptional program of the TE and MP CD8 + T cell subsets.

Manipulating the Tumor Environment

Supplementary Fig. 1 p38 MAPK negatively regulates DC differentiation. (a) Western blot analysis of p38 isoform expression in BM cells, immature DCs

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

Dendritic cells in cancer immunotherapy Aimin Jiang

Supplementary Figures

Fluorochrome Panel 1 Panel 2 Panel 3 Panel 4 Panel 5 CTLA-4 CTLA-4 CD15 CD3 FITC. Bio) PD-1 (MIH4, BD) ICOS (C398.4A, Biolegend) PD-L1 (MIH1, BD)

Tumor Associated Macrophages as a Novel Target for Cancer Therapy

Supplementary Figure 1: TSLP receptor skin expression in dcssc. A: Healthy control (HC) skin with TSLP receptor expression in brown (10x

D CD8 T cell number (x10 6 )

Supplementary Figures

NKTR-255: Accessing IL-15 Therapeutic Potential through Robust and Sustained Engagement of Innate and Adaptive Immunity

Supplement Material. Spleen weight (mg) LN cells (X106) Acat1-/- Acat1-/- Mouse weight (g)

NK cell flow cytometric assay In vivo DC viability and migration assay

Frontiers of Science Foundation Scholar. Toby Bothwell. Freshman, University of Arkansas. final research manuscripts

ECM1 controls T H 2 cell egress from lymph nodes through re-expression of S1P 1

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

Interferon-dependent IL-10 production by Tregs limits tumor Th17 inflammation

Supplemental Information. Genomic Characterization of Murine. Monocytes Reveals C/EBPb Transcription. Factor Dependence of Ly6C Cells

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION

Generation of ST2-GFP reporter mice and characterization of ILC1 cells following infection

Supplemental Materials. Stromal Modulation Reverses Primary Resistance to Immune Checkpoint Blockade in. Pancreatic Cancer.

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

activation with anti-cd3/cd28 beads and 3d following transduction. Supplemental Figure 2 shows

BCR-ABL - LSK BCR-ABL + LKS - (%)

REPROGRAMING IMMUNITY IN RENAL CELL CARCINOMA

Supplementary Information. Tissue-wide immunity against Leishmania. through collective production of nitric oxide

Supplementary Figure S1. PTPN2 levels are not altered in proliferating CD8+ T cells. Lymph node (LN) CD8+ T cells from C57BL/6 mice were stained with

SUPPLEMENTARY INFORMATION

Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression

W/T Itgam -/- F4/80 CD115. F4/80 hi CD115 + F4/80 + CD115 +

Nature Immunology: doi: /ni Supplementary Figure 1. Cellularity of leukocytes and their progenitors in naive wild-type and Spp1 / mice.

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

National Surgical Adjuvant Breast and Bowel Project (NSABP) Foundation Annual Progress Report: 2009 Formula Grant

SUPPLEMENTARY INFORMATION

Supplementary Information:

Nature Genetics: doi: /ng Supplementary Figure 1

Supplementary Materials. for Garmy-Susini, et al, Integrin 4 1 signaling is required for lymphangiogenesis and tumor metastasis

Adaptive immune responses: T cell-mediated immunity

Emerging Concepts of Cancer Immunotherapy

SUPPLEMENTARY INFORMATION doi: /nature12026

Understanding the T cell response to tumors using transnuclear mouse models

Suppl Video: Tumor cells (green) and monocytes (white) are seeded on a confluent endothelial

Transcription:

Washington University School of Medicine Digital Commons@Becker Open Access Publications Breast and pancreatic cancer interrupt IRF-dependent dendritic cell development to overcome immune surveillance Melissa A. Meyer Washington University School of Medicine in St. Louis John M. Baer Washington University School of Medicine in St. Louis Brett L. Knolhoff Washington University School of Medicine in St. Louis Timothy M. Nywening Washington University School of Medicine in St. Louis Roheena Z. Panni Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Meyer, Melissa A.; Baer, John M.; Knolhoff, Brett L.; Nywening, Timothy M.; Panni, Roheena Z.; Su, Xinming; Weilbaecher, Katherine N.; Hawkins, William G.; Ma, Cynthia; Fields, Ryan C.; Linehan, David C.; Challen, Grant A.; Faccio, Roberta; Aft, Rebecca L.; and DeNardo, David G.,,"Breast and pancreatic cancer interrupt IRF-dependent dendritic cell development to overcome immune surveillance." Nature Communications.9,.. (). https://digitalcommons.wustl.edu/open_access_pubs/97 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact engeszer@wustl.edu.

Authors Melissa A. Meyer, John M. Baer, Brett L. Knolhoff, Timothy M. Nywening, Roheena Z. Panni, Xinming Su, Katherine N. Weilbaecher, William G. Hawkins, Cynthia Ma, Ryan C. Fields, David C. Linehan, Grant A. Challen, Roberta Faccio, Rebecca L. Aft, and David G. DeNardo This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/97

ARTICLE DOI:./s7--- OPEN Breast and pancreatic cancer interrupt IRF-dependent dendritic cell development to overcome immune surveillance 79():,; Melissa A. Meyer, John M. Baer, Brett L. Knolhoff, Timothy M. Nywening, Roheena Z. Panni,, Xinming Su, Katherine N. Weilbaecher,, William G. Hawkins,, Cynthia Ma,, Ryan C. Fields,, David C. Linehan, Grant A. Challen,,, Roberta Faccio, Rebecca L. Aft,,7 & David G. DeNardo,, Tumors employ multiple mechanisms to evade immune surveillance. One mechanism is tumor-induced myelopoiesis, whereby the expansion of immunosuppressive myeloid cells can impair tumor immunity. As myeloid cells and conventional dendritic cells (cdcs) are derived from the same progenitors, we postulated that myelopoiesis might impact cdc development. The cdc subset, cdc, which includes human CD + DCs and mouse CD + DCs, supports anti-tumor immunity by stimulating CD + T-cell responses. Here, to understand how cdc development changes during tumor progression, we investigated cdc bone marrow progenitors. We found localized breast and pancreatic cancers induce systemic decreases in cdcs and their progenitors. Mechanistically, tumor-produced granulocytestimulating factor downregulates interferon regulatory factor- in cdc progenitors, and thus results in reduced cdc development. Tumor-induced reductions in cdc development impair anti-tumor CD + T-cell responses and correlate with poor patient outcomes. These data suggest immune surveillance can be impaired by tumor-induced alterations in cdc development. Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA. Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA. Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA. Section of Stem Cell Biology, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA. Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA. 7 John Cochran St. Louis Veterans Administration Hospital, St. Louis, MO, USA. Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA. Correspondence and requests for materials should be addressed to D.G.D. (email: ddenardo@wustl.edu) NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

ARTICLE NATURE COMMUNICATIONS DOI:./s7--- To subvert immune surveillance, solid tumors disrupt tumor-targeted immune responses. Conventional dendritic cells (cdcs) support anti-tumor adaptive immunity by stimulating T cells, but cdcs often fail to accumulate in the tumor microenvironment,. Furthermore, those cdcs found in the tumor can be immature and are therefore less effective in antigen presentation and T-cell stimulation. Solid tumors also interfere with anti-tumor immune responses by stimulating immature granulocyte and monocyte production from bone marrow (BM) progenitors. Expanded myeloid cells are recruited to tumors where they can maintain an immature phenotype or differentiate into tumor-associated macrophages. All of these populations can suppress anti-tumor CD + T cells as well as promote tumor progression through support of angiogenesis and metastasis 7. Interestingly, cdcs are produced from the same BM progenitors as the expanding populations of granulocytes and monocytes,. Although granulocyte and monocyte differentiation is known to be dysregulated in cancer 7, we do not fully understand how tumors affect cdc differentiation. Because of their common origin, we hypothesized that tumor-induced expansion of immature granulocytes and monocytes occur at the expense of cdc differentiation. cdcs play an important role in initiating and maintaining adaptive immune responses. cdcs are split into two subsets: cdcs, which specialize in CD + T-cell activation, and cdcs, which specialize in CD + T-cell activation. The cdcs are marked by CD in humans and encompass both migratory CD + cdcs and lymphoid-resident CDα + cdcs in mice. cdcs are also found in both lymphoid and peripheral tissues and are marked by CDc in humans and CDb and Sirpα in mice. The development of cdcs is driven by the transcription factors interferon regulatory factor- (IRF), basic leucine zipper transcription factor ATF-like (Batf), and inhibitor of DNA binding (Id),7 9. The development of cdcs is driven by a different set of transcription factors including interferon regulatory factor (IRF). Because of their role in activating CD + T cells, cdcs have been implicated in supporting the T-cell response against solid tumors. CD + cdcs are known to cross-present antigen to activate CD + T cells and secrete factors that attract T cells into the tumor,,. Furthermore, CD + cdcs are important for transporting antigen into the draining lymph nodes (LNs), supporting T-cell activation and expansion,,. Given these functions, it is understandable that CD + cdcs have been implicated in initiation and maintenance of CD + T-cell responses against tumors. CD + cdcs are required to restrain tumor growth and support response to CD + T cellmediated chemo- and immune-therapies in multiple mouse models of solid tumors. In patients, intratumoral CD + cdc numbers correlate with better outcomes in many types of solid tumors, including breast cancer (BC),,. Thus, cdcs are important mediators of the anti-tumor CD + T-cell response and can function to control tumor progression in mice and humans. Given their important role in supporting anti-tumor immunity, we inquired whether tumor progression affects the generation of cdcs. Recent work has shown that after committing to the granulocyte, monocyte, or cdc lineage, cdc precursors can commit to the cdc subset during differentiation before leaving the BM 7. Given that differentiation choice can be made outside the tumor microenvironment, we hypothesized that systemic changes induced by tumors might impair cdc, and further cdc, commitment in the BM, and subsequently influence cdcs in the periphery and anti-tumor immunity. In this study, we show that tumors interrupt cdc, and specifically cdc, differentiation in BC and pancreatic ductal adenocarcinoma (PDAC) mouse models and patients. This interruption reduces the systemic cdc pool, negatively impacting CD + T-cell immunity and correlating with poor patient outcome. Our data illustrate a new mechanism by which tumors subvert anti-tumor immunity via dysregulation of cdc differentiation. Results BC and PDAC patients have reduced BM cdc differentiation. cdcs are important for initiating and sustaining anti-tumor T- cell responses,,. To understand the impact of tumors on development of cdcs, we profiled BM samples from human BC and PDAC patients with localized disease and no prior therapy. We analyzed the following cdc progenitors: macrophagedendritic cell progenitors (MDPs), which retain monocyte/macrophage potential; common dendritic cell progenitors (CDPs), which retain plasmacytoid DC (pdc) potential; and pre-dcs, which are committed to the cdc lineage. Relative to BM from healthy controls, BM from both BC and PDAC patients had decreased numbers of CDPs and pre-dcs. MDPs were reduced in BC patients but not PDAC patients (Fig. a, b, Supplementary Fig. a, Supplementary Table ). These data suggest the defect in BM progenitors is strongest and most consistent after progenitors start to commit to the cdc lineage. We also analyzed cells of the cdc subsets, cdcs and cdcs, found in the BM. Patient populations showed a decrease in CD + cdcs and CDc + cdcs (Fig. a, b). As illustrated in Fig. a, CD + cdcs are rare in the BM but are dramatically reduced in BC and PDAC patients. We validated these findings using a second cohort of BC patients (Supplementary Fig. b). In contrast to the cdc lineage, an expansion of BM immature granulocytes (CDb + CD Hi CD - CD + ), potentially a subset of myeloid-derived suppressor cells, was observed in both cancer types (Fig. b). This result is in agreement with published observations in several other solid tumor types,. To determine if the reduction in cdcs extended into the periphery, where cdcs are thought to function,, we analyzed BC and PDAC patient blood samples and found reduced pre-dc numbers and increased immature granulocyte numbers relative to healthy controls (Fig. c, Supplementary Fig. c). We validated these findings in a second cohort of PDAC patients (Supplementary Fig. d). Together, these data suggest that cancer results in decreased cdc lineage cells in the BM and decreases in the systemic pool of pre-dcs. Previous studies have shown that CD + cdc numbers and functions in the tumor are predictive of patient outcome,,, prompting us to assess whether changes in CD + cdcs in the BM prior to treatment or resection were also predictive of response to therapy. We observed higher numbers of CD + cdcs and lower numbers of granulocytes (CD + CDb + CSFR CD + CD ) in the BM of BC patients who achieved a pathological complete response (pcr) to neoadjuvant chemotherapy (Fig. d, Supplementary Table ). Notably, CDc + cdc numbers were not predictive of pcr, suggesting the predictive nature is specific to the CD + cdc subset (Supplementary Fig. e). These findings demonstrate that tumor-induced decreases in CD + cdc development in the BM could be an important indicator of patient immune competency and response to therapy. Local breast and pancreatic tumors systemically reduce cdcs. To more carefully study changes in cdc development during cancer progression, we evaluated three distinct genetic mouse models and four syngeneic orthotopic mouse models of BC and PDAC. Similar to the human cancer patients, we found six of these seven models had reductions in absolute numbers of BM CD + cdcs and most had reductions in the absolute number of BM pre-dcs (Fig. a c, Supplementary Fig. a d). To further NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

cdcs/granulocytes NATURE COMMUNICATIONS DOI:./s7--- ARTICLE investigate the cdc lineage in tumor-bearing mice, we employed PyMT-B, a cell line derived from the genetic MMTV-Polyoma Middle T (PyMT) mammary tumor mouse model on the C7BL/ background. Consistent with the patient data, mice bearing end-stage orthotopic PyMT-B mammary tumors showed no change in MDPs but a modest reduction in CDPs (Fig. b, Supplementary Fig. a). This finding suggests that tumors impact components of the cdc lineage. In contrast to cdcs, LyG + LyC + CSFR + immature granulocytes and LyG + CSFR mature granulocytes expand in the BM (Fig. b, Supplementary Fig. a). In addition, the changes in cdc progenitors and granulocytic populations were consistent in the genetic models of BC and PDAC: MMTV-PyMT FvB/N and KPC (p-cre;lsl- Kras GD ;Trp flox/+ C7BL/), respectively (Supplementary a Patient bone marrow Singlet-live CD + cdcs K K K K CDc + cdcs K K K K Healthy control K K K K K K K K K K K K CDc.9% CD CD/CD/CD9/CDb Flt HLA-DR K K K K K K K K K K K K K K K c-kit GM-CSFR Flt CDRA M-CSFR CDc CD Breast cancer.% b Patient bone marrow MDPs n.s.......... CDPs....... CD + cdcs CDc + cdcs CD Immature granulocytes.... Healthy controls Breast cancer Pancreatic cancer c Patient blood.... Immature granulocytes d Patient bone marrow CD + cdcs.. Granulocytes............ Healthy controls Breast cancer Pancreatic cancer.. pcr + + + Fig. Human breast and pancreatic cancers reduce dendritic cell progenitors and CD + cdcs. a Representative flow cytometry gating strategy for human BM pre-dcs, CD + cdcs, and CDc + cdcs, including representative final plots from a BC patient and from a healthy control. b Frequency of BM MDPs, CDPs, pre-dcs, CD + cdcs, CDc + cdcs, and immature granulocytes (CDb + CD High CD - CD + ) in baseline BC and PDAC patients relative to healthy controls. Data from BC cohort and PDAC cohort. Healthy controls n = ; BC n = ; PDAC n = 9. c Frequency of circulating blood pre-dcs and immature granulocytes in baseline BC and PDAC patients relative to healthy controls. Data are from BC cohort and PDAC cohort. Healthy controls n = ; BC n = ; PDAC n = 7. d Frequency of BM CD + cdcs and granulocytes (CD + CDb + CSFR CD + CD ), and the ratio of cdcs/granulocytes in BC patients prior to treatment or surgical intervention comparing those who achieved pathological complete response relative to patients who did not achieve pathological complete response in BC cohort ; n =. Additional cohorts illustrated in Supplementary Figure. Error bars represent mean +/ s.e.m. or box plot; p <., p <., p <., n.s., not significant by unpaired two-sided Student s t test NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

ARTICLE NATURE COMMUNICATIONS DOI:./s7--- Fig. e, f). Unlike cdcs, cdcs were not consistently decreased in the BM of tumor-bearing mice across several models (Fig. a c, Supplementary Fig. a d). Together, these data suggest that solid tumors reduce the number of cells that make up the cdc lineage, including cdc. Specific reductions in the cdc progenitors and cdcs suggest a selective interruption of cdc development. Recent data show that pre-dcs can commit to the cdc subset before leaving the BM, rather than receiving influence from factors at a peripheral site 7,9,. To evaluate this phenomenon, we assessed the number a Bone marrow Singlet-live K K K K K K K K MHCII CD MHCII Sirpα K Lineage K Flt CD + cdc Sirpα + cdc CDc Sirpα CDc Flt b d PyMT-B bone marrow Number per bone (x ) Number per bone (x ) K K K K K MDPs CDPs Immature granulocytes PyMT-B bone marrow Singlet-live-Lin-CDc + PyMT-B lymph node Number per LN (x ) K K K K CD + cdcs Number per bone (x ) Number per bone (x ) Granulocytes Number per bone (x ) K Blood Singlet-live n.s. Flt Lineage MHCII PyMT-B c-kit Number per bone (x ) CDc i Sirpα Sirpα + cdcs n.s. Pre-cDCs Primary tumor Number per bone (x ) c-kit Flt PyMT-B Number per plug (x ) Number per ml (x ) GEMM bone marrow MMTV-PyMT KPC PyMT-B blood Number per bone (x ) Number per bone (x ) % % Zbtb-GFP Zbtb-GFP Poly I:C OVA-TxRd plug CD + cdcs Plug CD + cdcs Pre-cDCs PyMT-B PyMT-B Number per LN (x ) Number per bone (x ) Number per ml (x ) Number per bone (x ) Number per bone (x ) CD + cdcs Pre-cDCs Granulocytes CD + cdcs e f g h c PyMT-B Draining lymph node CD + cdcs Number per ml (x ) Number per plug (x ) Number per bone (x ) Number per bone (x ) GEMM blood Sirpα + cdcs Sirpα + cdcs PyMT-B MMTV-PyMT Number per ml (x ) Plug OVA-specific CD + T cells n.s. KPC NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

NATURE COMMUNICATIONS DOI:./s7--- ARTICLE of DC-committed pre-dcs (pre-cdcs) in the BM using the gating strategy defined by Grajales-Reyes et al. 9 employing the Zbtb-GFP mouse. We found that pre-cdcs were decreased by % in the BM of tumor-bearing mice, which is a more substantial decrease than the reduction in the broader pre-dc population (Fig. d). These data suggest that tumors not only reduce the number of cdc progenitors, but also reduce the commitment of pre-dcs to the cdc lineage in the BM. To determine if changes in cdc progenitors result in systemic alterations in the pool of available cdcs, we analyzed circulating pre-dcs. We found fewer pre-dcs and more granulocytes in the blood of tumor-bearing mice relative to controls (Fig. e, f, Supplementary Fig. b). Similar observations were seen in the genetic BC model, MMTV-PyMT FVB/N, and genetic PDAC model, KPC (Fig. g), as well as two additional orthotopic BC and PDAC models (Supplementary Fig. b c). These changes in the pool of circulating pre-dcs correlated with reduced numbers of CD + cdcs in the uninvolved LNs of tumor-bearing mice (Fig. h, Supplementary Fig. c). Congruent with observations in BC and PDAC patients, these data show that mouse models of BC and PDAC, independent of genetic driver and strain, have systemically reduced numbers of cdc precursors and cdcs. Interestingly, pdcs were decreased in the BM, but circulating pdcs were not decreased (Supplementary Fig. g), suggesting they may be regulated differently. Research has shown that cdcs turn over at a high rate in tissues and require BM progenitors to provide new cdc 7.To determine if the decreased pool of circulating pre-dcs would result in altered recruitment and antigen presentation at new sites of inflammation, we measured the recruitment of CD + cdcs in the context of a tumor. PyMT-B mammary tumors were established in the th lower mammary fat pads. Then, a matrigel plug containing polyinosinic:polycytidylic acid (poly I:C) and ovalbumin (OVA)-Texas Red (TxRd) conjugate was implanted in the contralateral nd upper mammary fats pads of tumor-bearing and control mice. We found that the presence of the primary tumors resulted in decreased numbers of CD + cdcs recruited into the matrigel plug and decreased numbers of migratory CD + cdcs in the draining LN (Fig. i, Supplementary Fig. d). These changes in cdc recruitment correlated with reduced numbers of OVA-specific CD + T cells (CD + CD + Dextamer + ) in the matrigel plug (Fig. i). To verify that the reduced number of cdcs in the poly I:C plug was due to a decreased pool of circulating pre-dcs rather than changes in recruitment cytokines, we measured recruitment of CD + cdcs to a matrigel plug containing the recruitment factor C-C motif chemokine ligand (Ccl), which is known to be downregulated in some tumor types. Again, we saw a defect in CD + cdcs recruited to the Ccl-containing matrigel plug in tumor-bearing mice (Supplementary Fig. h). Similarly, we found there was no change in pre-dc C-C motif chemokine receptor (Ccr) expression in tumor-bearing mice (Supplementary Fig. i). Together, these data suggest that tumor-induced repression of cdc development results in a reduced pool of circulating pre-dcs and a subsequent reduction in recruitment of cdcs to new sites of inflammation and impaired priming of CD + T cells. Tumors impair cdc developmental potential of progenitors. The contrasting increases in granulocytes and decreases in cdc progenitors led us to hypothesize that tumors alter the differentiation potential of common myeloid precursor cells. To test this, we isolated CD. + Lin ckit + ScaI myeloid progenitors (MPs). This cell population maintains granulocyte, monocyte, and cdc potential and contains granulocyte-macrophage progenitors (GMPs). The MPs were transferred into tumor-free controls or mice bearing PyMT-B tumors. We found that CD. + MPs transferred into tumor-bearing mice differentiated into fewer CDPs, pre-dcs, and cdcs in the BM, but more granulocytes in both the BM and blood (Fig. a). Fewer CDα + cdcs were included in the CD. + products in the spleens of tumor-bearing hosts. CD.+ granulocyte products did not accumulate in the spleen of tumor-bearing mice relative to tumor-free controls. Although CD. + monocytes were not consistently increased in each organ, they accumulated in the spleen (Supplementary Fig. a). These data suggest that tumors preferentially drive the differentiation of shared progenitors to granulocytes at the expense of cdc development. To determine if exposure to tumors in vivo programs BM progenitor cells to resist cdc differentiation, we isolated MPs, MDPs, or CDPs from tumor-free and PyMT-B tumor-bearing mice. When cultured in FMS-like tyrosine kinase ligand (FltL), we found MPs, MDPs, and CDPs isolated from tumor-bearing mice were less able to differentiate into cdcs compared to those cells isolated from tumor-free mice (Fig. b). Of these progenitors, CDPs appeared to be most impaired in their capacity to differentiate into cdcs. Of note, MPs from tumor-bearing mice but not from tumor-free mice differentiated into granulocytes even in the absence of granulocyte differentiation factors (Supplementary Fig. b). This result suggests MPs from tumor-bearing mice are Fig. Primary mammary and pancreatic tumors systemically decrease cdcs. a Representative flow cytometry gating strategy for mouse BM pre-dcs, CD + cdcs, and Sirpα + cdcs. b Number of BM MDPs, CDPs, pre-dcs, immature granulocytes, granulocytes, CD + cdcs, and Sirpα + cdc from mice bearing end-stage orthotopic PyMT-B mammary tumors relative to tumor-free controls; n = /group. Data are representative of three independent experiments. c Number of BM pre-dcs, CD + cdc, and Sirpα + cdc in end-stage genetically engineered mouse models (GEMM) of BC (MMTV- PyMT) and PDAC (KPC) relative to tumor-free controls; tumor-free MMTV-PyMT controls n = ; MMTV-PyMT, n = ; tumor-free KPC controls, n = 7; KPC, n =. d Alternative representative gating strategy using Zbtb-GFP mice continued from Fig. a for BM committed pre-cdcs, including representative plots from tumor-free and PyMT-B tumor-bearing mice. Number of BM pre-cdcs in Zbtb-GFP + mice bearing end-stage orthotopic PyMT-B mammary tumors relative to tumor-free Zbtb-GFP + controls; n = /group. Data are representative of two independent experiments. e Representative flow cytometry gating strategy for mouse blood pre-dcs. f Number of blood pre-dcs and granulocytes from mice bearing end-stage orthotopic PyMT-B mammary tumors relative to tumor-free controls; n = /group. Data are representative of three independent experiments. g Number of blood pre-dcs in end-stage genetic mouse models of BC (MMTV-PyMT) and PDAC (KPC) relative to tumor-free controls, n = 7/group. h Number of uninvolved LN CD + cdcs from mice bearing end-stage orthotopic PyMT-B mammary tumors relative to tumor-free controls; n = /group. Data are representative of two independent experiments. i Mice with. cm diameter orthotopic PyMT-B tumors and tumor-free controls were implanted with matrigel plugs containing poly I:C+OVA-TxRd peptide in the upper mammary fat pad. Number of CD + cdcs in the plug and draining LN and OVAspecific CD + T cells (CD + CD + Dextamer + ) in the plug days after implant; n = /group. Data are representative of two independent experiments. End-stage for each model is defined in the Methods. Error bars represent mean +/ s.e.m.; p <., p <., p <., n.s., not significant by unpaired two-sided Student s t test NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

Hierarchical clustering ARTICLE NATURE COMMUNICATIONS DOI:./s7--- intrinsically primed to differentiate into granulocytes at the expense of cdcs. As expected, MDPs and CDPs from tumorfree or tumor-bearing mice did not have granulocyte differentiation potential (Supplementary Fig. b, c). Taken together, these data suggest that tumors alter myeloid progenitor fate by increasing their potential to differentiate into granulocytes at the expense of cdc production. To further investigate the differences in progenitors from tumor-free and tumor-bearing mice, we profiled the gene expression in GMPs, MDPs, and CDPs. We found GMPs, MDPs, and CDPs from tumor-bearing mice failed to upregulate proteins indicative of the cdc program (e.g., Irf, Zbtb, Cd9a, Spib, Batf, and Bcla). This result is in contrast to the increased expression of markers indicative of a granulocyte or monocyte a CD.+ MP % CD. Bone marrow MDPs CDPs...... % CD. % CD... % CD..... CD + cdcs % CD. Granulocytes % CD. Blood Granulocytes. host. PyMT-B host. b MPs cdcs MDPs cdcs CDPs cdcs.. CD.+ MP, MDP, or CDP CD.+ Bone marrow feeder layer + FltL % CD. % CD...... % CD..... donor PyMT-B donor c Dendritic cell identity Granulocyte/monocyte identity d Irf Cd9a................ Zbtb SpiB..... Cebpe...... Cebpa......... Gfi Id PyMT-B MDP MDP PyMT-B CDP CDP........ Batf.... Bcla.... Cebpb.... Mafb.... PyMT-B e. Tumor burden cm.. Wild-type BATF / BATF / +Normal cdc progenitors BATF / +Tumor cdc progenitors. 7 9 Days after implant.. NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

NATURE COMMUNICATIONS DOI:./s7--- ARTICLE fate (e.g., Cebpe, Gfi, Cebpa, Id, Cebpb, and Mafb) (Fig. c). We further profiled MDPs and CDPs from tumor-bearing mice by microarray to look for global changes in gene expression. We found the CDPs from tumor-bearing mice clustered closer with MDPs than did CDPs from tumor-free mice (Fig. d, Supplementary Tables, ). DAVID (database for annotation, visualization and integrated discovery) analysis showed that the upregulated genes were enriched for biologic process terms like inflammatory response, myeloid cell differentiation, cell cycle regulation, and cytokine secretion, whereas downregulated genes were enriched for biologic process terms like transcriptional regulation, major histocompatibility complex (MHC) class II protein complex binding, differentiation, and covalent chromatin modifications (Supplementary Table ). Overall, these data suggest a failure in progenitors to upregulate the cdc program, while favoring the granulocytic and monocytic programs in tumor-bearing mice. To determine if the reduction in cdc differentiation potential could impact tumor immunity and tumor progression, we measured the ability of progenitors isolated from tumor-bearing or tumor-free mice to control tumor growth in BATF / mice. We chose BATF / mice because they lack functional cdcs but have no defect in granulocyte number (Supplementary Fig. d). First, we observed that tumor growth of PyMT-mCh- OVA, a variant of PyMT-B expressing antigenic mcherry and ovalbumin, was restrained in wild-type, but not in BATF / mice (Fig. e). MDPs and CDPs were sorted from PyMT-B tumor-bearing or tumor-free controls and transferred into BATF / mice. The recipients were then implanted with PyMT-mCh-OVA. Mice that received MDPs and CDPs from wild-type mice were able to restrain PyMT-mCh-OVA tumor growth, similar to wild-type mice, whereas mice that received MDPs and CDPs from tumor-bearing mice were not able to restrain tumor growth (Fig. e). These data suggest that progenitors influenced by tumor burden are no longer able to act as a source of cdcs and thus fail to induce anti-tumor immunity and control antigenic tumor progression. Tumor-derived GCSF inhibits cdc development. To understand which factors inhibit cdc development during cancer progression, we profiled cytokines known to influence myeloid and cdc differentiation in the blood and BM of tumor-bearing mice. We found that both granulocyte colony-stimulating factor (GCSF) and interleukin- (IL-) were highly upregulated in the blood of PyMT-B tumor-bearing mice relative to tumor-free controls (Fig. a). Additionally, GCSF, but not IL-, was upregulated in the BM of tumor-bearing mice relative to controls (Fig. a) and in the blood serum of BC patients (Fig. b). To determine if either of these cytokines was necessary to alter cdc differentiation, we neutralized both GCSF and IL- in the context of PyMT-B tumors. As above, the presence of PyMT-B tumors reduced BM pre-dc and CD + cdc and blood pre-dc numbers while increasing granulocytes in both tissues. After treatment with GCSF-neutralizing antibody, we observed that pre-dcs and CD + cdcs in the BM and pre-dcs in the blood were restored to levels in tumor-free mice. This result suggests that tumor-induced GCSF is necessary to reduce cdc numbers in tumor-bearing mice. Additionally, GCSF neutralization reduced immature granulocytes in the BM and granulocytes in the blood, as expected,7. Neutralization of GCSF was also sufficient to increase CD + T cells in the tumor (Fig. c). Although IL- neutralization reduced immature granulocytes and inflammatory monocytes as previously described, it did not reverse tumor-induced reductions in pre-dcs or CD + cdcs in the BM (Supplementary Fig. a). To determine if GCSF alone is sufficient to disrupt cdc differentiation, we dosed mice with recombinant GCSF. We used μg/day of recombinant GCSF to achieve steady-state serum GCSF concentrations comparable to those in tumor-bearing mice (Supplementary Fig. b). We found that changes in BM progenitor numbers in tumor-free mice dosed with GCSF mice paralleled those observed in tumor-bearing mice. These changes included GCSF-induced reduction in MDPs, CDPs, pre-dcs, and cdcs in the BM and reduced pre-dcs in the blood as compared to untreated mice (Fig. d). As expected, GCSF increased immature granulocytes in the BM and granulocytes in the blood (Fig. d). These data implicate a role for GCSF in reducing cdc numbers during cancer progression. To determine if GCSF could act directly on cdc progenitors to inhibit cdc differentiation, we cultured MPs, MDPs, and CDPs isolated from tumor-free mice in the presence of FltL+/ GCSF. As expected, FltL drove cdc differentiation in all cell types, whereas GCSF prevented FltL-mediated cdc expansion, and in MPs instead drove granulocyte differentiation (Fig. e). These results are consistent across a wide range of FltL and GCSF concentrations (Supplementary Fig. c). To understand if GCSF alone could prime progenitors to become unresponsive to FltLmediated cdc differentiation, consistent with the ex vivo results shown in Fig. b, progenitors were pre-treated with GCSF for h prior to plating for differentiation in FltL. Pre-treatment with GCSF inhibited cdc differentiation even in CDPs (Fig. f), suggesting that GCSF can alter the ability of a progenitor to respond to cdc differentiation cues even in progenitors lacking granulocyte differentiation capacity. To ensure GCSF was able to signal in cdc progenitors directly, we analyzed the expression of the GCSF receptor, Csfr, in vivo. We found that Csfr was expressed in GMPs, MDP, and CDPs and unmodified by the Fig. Tumor burden alters the fate of myeloid progenitors. a CD. + Lin Sca ckit + MPs were transferred into mice bearing. cm diameter orthotopic PyMT-B tumors or tumor-free controls. BM was analyzed for CD. + populations after weeks. BM MDPs, CDPs, pre-dcs, CD + cdcs, and granulocytes, and blood granulocytes displayed as frequency of CD.; n = /group. b CD. + Lin Sca ckit + MPs, CD. + MDPs, and CD. + CDPs were isolated from end-stage orthotopic PyMT-B tumor-bearing or tumor-free donors. Progenitors were cultured on CD. + BM feeder culture for days in the presence of ng/ml FltL. Final cultures were analyzed for cdc (Live CD. + CD. - MHCII + CDc + Sirpα - CD + ). End stage is defined in the Methods. Data are representative of three independent experiments consisting of three wells per condition. GMPs, MDPs, and CDPs were sorted from mice bearing end-stage orthotopic PyMT-B mammary tumors and tumor-free controls. c GMPs, MDPs, and CDPs were analyzed by RT-qPCR. d MDPs and CDPs were analyzed by microarray. Cluster analysis was performed with a differential genes list generated from gene with.-fold at p <. and FDR q <. in MDP or CDP comparison from PyMT-B tumor bearing to tumor free. Four samples, each consisting of two mice, were analyzed per group. e MDPs and CDPs were isolated from PyMT-B tumor-bearing mice or tumor-free controls. Progenitors were adoptively transferred into BATF / mice. PyMT-mCh-OVA was implanted after days into wild-type mice, BATF / mice without adoptive transfer, and BATF / mice with adoptive transfer from tumor-free or tumor-bearing mice. Tumor growth was monitored. Error bars represent mean +/ s.e.m.; p <., p <., p <. by unpaired two-sided Student s t test or two-way ANOVA NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications 7

ARTICLE NATURE COMMUNICATIONS DOI:./s7--- presence of tumors (Supplementary Fig. d). Together, these data suggest that tumor-induced GCSF is both necessary and sufficient to reduce cdc differentiation and can act directly on cdc progenitors. We next sought to determine the source of GCSF. First we analyzed both tumor tissues and BM samples from the transplantable PyMT-B and the genetic KPC models for Csf by in situ hybridization (ISH). We found that although BM cells expressed Csf, levels were not elevated by the presence of PyMT- B and KPC tumors (Supplementary Fig. e). By contrast, tumor tissues from PyMT-B and KPC models had higher Csf levels compared to normal mammary and pancreas tissues (Fig. a). We also observed that the majority of the Csf ISH signal was present in the tumor cells and not the stroma (Fig. a). To verify this result in human BC and PDAC tissues, we stained for GCSF protein by immunohistochemistry (IHC). Similar to our mouse models, we found that GCSF was highly expressed in breast and pancreas tumor tissues (Fig. b). Additionally, although GCSF was expressed in both tumor and stroma, tumor levels of GCSF were considerably higher than stroma levels in most cases. Together, these data suggest that tumor-derived GCSF may be a dominant source of GCSF in tumor-bearing animals. To directly a Cytokine pg/ml c PyMT-B Blood 7 GCSF Tumor Free IL-β VEGF IL- IL- TNFα MCSF PyMT-B IL-α IFNγ GMCSF FltL Cytokine pg/ml Bone marrow,,,,,,, GCSF IL-β Days Analysis VEGF IL- IL- d TNFα MCSF IL-α IFNγ GMCSF FltL b Patient serum GCSF pg/ml Healthy controls n= Breast cancer n= Days Analysis μg Anti-GCSF IgG Bone marrow Number per bone (x ) Blood Number per ml (x ) Number per bone (x ) Number per ml (x ) Granulocytes CD + cdcs Immature granulocytes Tumor CD + T cells PyMT-B PyMT-B + Anti-GCSF IgG Number per bone (x ) Number per gram (x ) Number per bone (x ) μg GCSF Bone marrow MDPs Blood Number per ml (x ) Vehicle Number per bone (x ) CDPs Number per ml (x ) Granulocytes GCSF Number per bone (x ) Number per bone (x ) CD + cdcs Number per bone (x ) Immature granulocytes e % CD. % CD. MPs cdcs MPs Granulocytes % CD. MDPs cdcs CDPs cdcs...... CD. + MP MDP or CDP % CD. FltL GCSF FltL+GCSF.. CD. + bone marrow feeder layer + cytokines f % CD. % CD. MPs MDPs CD. + MP MDP or CDP cdcs GCSF or no cytokines h % CD. cdcs CDPs cdcs.. % CD. MPs FltL GCSF GCSF FltL... Granulocytes CD. + bone marrow feeder layer + cytokines NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

NATURE COMMUNICATIONS DOI:./s7--- ARTICLE test if tumor-derived GCSF was responsible for impaired cdc development, we deleted the Csf gene using CRISPR CAS9 in the PyMT-B cell line (PyMT-B GCSFKO). Both BM and blood profiling showed pre-dcs and cdc populations were recovered and immature granulocytes were reduced in PyMT-B GCSFKO tumor-bearing mice relative to PyMT-B tumor-bearing controls (Fig. c). These results were repeated with a second independent clone (Supplementary Fig. f). These data demonstrate that GCSF derived from tumor cells is impairing cdc differentiation. GCSF inhibits cdc development by impairing IRF expression. Research has shown that GCSF preferentially expands immature granulocytes and monocytes by suppressing IRF expression through activation of signal transducer and activator of transcription (STAT) in granulocytic-monocytic precursors 9,. Because IRF is an important transcription factor for cdc development, we hypothesized GCSF might drive reduced IRF expression in cdc progenitors leading to impaired cdc differentiation 9,,. In support of this notion, our expression profiling demonstrated that Irf messenger RNA (mrna) was downregulated in cdc progenitors from tumor-bearing mice (Fig. c). Additionally, we found IRF protein was reduced in tumor-bearing mouse MDPs, CDPs, and pre-dcs (Fig. a). Further, we analyzed IRF target gene expression in GMPs, MDPs, and CDPs and found that, relative to controls, tumorbearing mice had decreased mrna expression of multiple MHC molecules, as well as TapBP, Tap, Batf, and Pml (Figs. c, b). In contrast, Id, another regulator of cdc development, was not reduced (Supplementary Fig. a) 7. To understand if downregulation of IRF is responsible for some of the gene expression changes in the microarray analysis (Fig. d), differentially expressed genes between control and tumor-bearing mice were compared to genes that are differentially expressed in IRF / myeloid progenitors 9,. We found tumor-regulated genes were enriched for genes regulated in IRF / myeloid progenitors (Supplementary Fig. b). In keeping with the ability of GCSF to downregulate IRF through STAT activation 9, we observed that phosphorylated STAT (pstat) was elevated in MDPs, CDPs, and pre-dcs from tumor-bearing mice (Fig. c). To determine if these changes also occur in human cancer patients, we analyzed IRF expression in human BM and found that IRF was downregulated in both BC and PDAC patients compared to healthy controls (Fig. d). Additionally, the extent of IRF downregulation correlated with the decrease in BM pre-dcs in patients (Fig. e). We also found IRF expression was decreased in the blood pre-dcs of pancreatic cancer patients (Fig. f) and the extent of IRF downregulation correlated with blood pre-dc numbers (Fig. g). Because IRF is a molecular marker of pre-dcs committed to the cdc lineage, these data also suggest that patients have reduced numbers of circulating pre-cdcs relative to healthy controls. We next assessed if downregulation of IRF in circulating pre-dcs impacted patient outcome and found patients with low IRF expression in circulating pre-dcs had decreased recurrence-free and overall survival (Fig. h). These results suggest that IRF expression is modulated in human cancers and may be an indicator of cdc development and patient outcome. To confirm that GCSF was upstream of IRF suppression, we measured IRF expression in cdc progenitors from tumorbearing mice following GCSF neutralization. We found IRF expression was recovered in the MDP, CDP, and pre-dcs following anti-gcsf immunoglobulin G (IgG) treatment (Fig. 7a). Furthermore, IRF expression was, again, downregulated in progenitors from PyMT-B tumor-bearing mice but was not downregulated in progenitors from PyMT-B GCSFKO mice (Supplementary Fig. c). To show GCSF was acting directly on progenitors to modulate IRF expression, we treated MPs, MDPs, and CDPs alone in vitro with GCSF or GCSF+FltL. We found Irf, as well as its target gene Batf, were downregulated in both conditions relative to controls (Fig. 7b). Interestingly, FltL treatment did not drive IRF expression above controls (Supplementary Fig. d), suggesting FltL and GCSF act through different mechanisms to modulate cdc differentiation. To determine if downregulation of IRF is necessary for GCSF to impair cdc differentiation, we investigated if IRF overexpression would make MPs insensitive to GCSF. Without IRF overexpression, GCSF drove down FltL-mediated cdc differentiation, similar to the results shown in Fig. e. However, IRF overexpression rendered MPs insensitive to GCSF, and their cdc differentiation was comparable to FltL-alone conditions (Fig. 7c). These data suggest GCSF must modulate IRF expression to impact cdc differentiation. IRF and cdcs support anti-tumor CD + T-cell responses. To determine whether suppression of cdc differentiation was detrimental to anti-tumor immunity, we measured tumor-specific T-cell activation in the presence or absence of an established tumor. To accomplish this, PyMT-B tumors, referred to here as the primary tumor, were established in the lower mammary fat pad. A secondary tumor using the PyMT-mCh-OVA cell line was then implanted into the upper mammary fat pad of the same mouse. We observed reduced recruitment of CD + cdcs to the secondary tumor site and draining LNs of primary tumorbearing mice (Fig. a). This result is consistent with the reduction in the available pool of circulating pre-cdcs in tumor-bearing animals (Fig. i). To test if reduced recruitment of CD + cdcs was functionally relevant, we measured OVA-specific CD + T cells within the secondary tumor site and the secondary tumor-draining LNs and found the OVA-specific CD + T cells Fig. GCSF disrupts cdc differentiation. a Blood and BM serum cytokines in orthotopic PyMT-B end-stage tumor-bearing mice relative to tumor-free controls; tumor-free, n = ; PyMT-B, n = 7. b GCSF in human BC patient blood serum relative to healthy controls; healthy controls, n = ; BC, n =. c Number of BM pre-dcs, CD + cdcs, and immature granulocytes; numbers of blood pre-dcs and granulocytes, and tumor CD + T cells (CD + CD + CD + ) in tumor-free mice, orthotopic PyMT-B tumor-bearing end-stage mice, and orthotopic PyMT-B tumor-bearing end-stage mice treated for weeks with μg anti-gcsf IgGs /week; n = 7/group. Data are representative of two independent experiments. d Number of BM MDPs, CDPs, pre-dcs, CD + cdcs, and immature granulocytes, and number of blood pre-dcs and granulocytes in C7BL/ mice treated with μg GCSF for days; n = /group. Data are representative of two independent experiments. e CD. + Lin Sca ckit + MPs, MDPs, and CDPs isolated from tumor-free mice were cultured on CD. + BM feeder culture in the presence of ng/ml FltL, ng/ml GCSF, or ng/ml FltL and ng/ml GCSF for days. Final cultures were analyzed for cdcs (Live CD. + CD. - MHCII + CDc + Sirpα - CD + ) and granulocytes (Live CD. + CD. - CDb + LyG + ). Data are representative of three independent experiments consisting of three wells per condition. f Experiment similar to that in Fig. d, but CD. + cells were pre-treated with ng/ml GCSF or media alone for h prior to plating on CD. + BM feeder layer. Data are representative of two independent experiments consisting of three wells per condition. End stage for each model is defined in the Methods. Error bars represent mean+/ s.e.m. or box plot; p <., p <., p <. by unpaired two-sided Student s t test NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications 9

ARTICLE NATURE COMMUNICATIONS DOI:./s7--- a Mouse models Normal mammary PyMT-B tumor/stroma In situ Csf mrna Csf Avg. foci CSF per cell..... Normal pancreas KPC tumor/stroma In situ Csf mrna Csf Avg. foci CSF per cell..... PyMT-B KPC b Breast cancer Low staining Medium staining High staining GCSF Human PDAC Adjacent normal pancreas PDAC Breast cancer n= GCSF Low Medium High c Bone marrow Blood Number per bone ( ) Number per bone ( ) PyMT-B CD + cdcs Number per bone ( ) PyMT-B GCSFKO Immature granulocytes Number per ml ( ) Fig. Tumor-derived GCSF inhibits cdc development. a ISH for Csf on end-stage PyMT-B and KPC tumor tissue and tumor-free mammary or pancreas tissue; n = per group. Scale bar μm. b Human patient BC and PDAC tumor tissue stained for GCSF. BC graded for low, medium, and high staining per tumor cell; BC, n = ; PDAC, n =. Scale bar μm. c Number of BM pre-dcs, CD + cdcs, and immature granulocytes and blood pre-dcs from mice bearing end-stage orthotopic PyMT-B or PyMT-B GCSFKO mammary tumors relative to tumor-free controls, n = /group. End stage for each model is defined in the Methods. Error bars represent mean +/ s.e.m.; p <., p <. by unpaired two-sided Student s t test NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

NATURE COMMUNICATIONS DOI:./s7--- ARTICLE were reduced in both sites of primary tumor-bearing mice compared to controls (Fig. a). To specifically implicate IRF and cdcs in this decreased CD + T-cell response, we employed IRF / and Batf / mice, both of which have impaired cdc differentiation,9,. In these knockouts, we observed reduced numbers of tumor-specific CD + T cells comparable to the primary tumor-bearing mice. In addition, this reduction was not further decreased by the presence of primary tumors (Fig. a). These data suggest that tumor-induced reductions in cdc development have a functional consequence on tumor-specific CD + T-cell responses that could contribute to reduction in antitumor immunity. a PyMT-B bone marrow b IRF target genes CDPs MDPs IRF MFI IRF MFI % Max % Max IRF IRF H-Ke..... TapBP............ PyMT-B H-Dma Tap.......... H-Q Pml GMPMDP CDP IRF MFI Unstained % Max IRF PyMT-B c p-stat MFI PyMT-B bone marrow MDPs CDPs p-stat MFI p-stat MFI PyMT-B d Patient bone marrow IRF e Patient bone marrow h Pancreatic cancer patient blood pre-dcs % Max IRF Unstained Breast cancer MFI Healthy controls Pancreatic cancer IRF (MFI)... (% total) r =.9 p =... % recurrence-free survival IRF high IRF low p =. Years f % Max Pancreatic cancer patient blood pre-dcs IRF MFI IRF Unstained Healthy controls Pancreatic cancer g Pre-DC IRF (MFI) Pancreatic cancer patient blood (% total) r =.9 p =..... % overall survival IRF high IRF low p =. Years NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

ARTICLE NATURE COMMUNICATIONS DOI:./s7--- Tumor-induced reductions in IRF also expand immature granulocytes that are known to have immune-suppressive functions and can otherwise promote tumor progression,7,. We sought to determine the contributions of immature granulocyte expansion to reduced number of cdcs and reduced anti-tumor CD + T-cell responses. To test if cdc depletion was regulated through granulocyte expansion, mice were treated with anti-lyg IgGs. LyG-depletion did not reverse the defect in BM pre-dcs and cdcs and did not recover IRF expression in BM progenitors (Supplementary Fig. 7a). To show that immature granulocytes were not mediating CD + T-cell suppression beyond the loss of cdcs in this model, we neutralized LyG in the context of the secondary tumor experiment described above. We found LyG depletion did not recover tumor-specific CD + T cells in the secondary tumor nor the draining LN (Fig. a). Similarly, we showed anti-lyg depletions did not recover CD + cdc recruitment to a matrigel plug containing poly I:C and OVA-TxRd conjugate, a secondary site of inflammation, in primary tumor-bearing mice. Additionally, CD + cdcs were not recovered in the draining LN and OVA-specific CD + T cells were not recovered in the matrigel plug (Supplementary Fig. 7b). To test whether the loss of IRF had a functional consequence on tumor outgrowth by acting through the loss of cdcs rather than the expansion of granulocytes, we measured the growth of PyMT-mCh-OVA tumors in IRF / mice, which lack cdcs and have expanded granulocytes, and Batf / mice, which only lack functional cdc (Supplementary Fig. 7c),. We found that tumors grew at the same rate in Batf / as IRF / mice, but that this rate was faster than wild-type controls (Fig. b). Also, we neutralized LyG in IRF / mice and measured PyMT-mCh-OVA tumor growth and found tumors grew at similar rates (Fig. c). These findings suggest that the loss of IRF can promote tumor growth by suppressing cdcs rather than solely through the expansion of granulocytes. Together, these results show granulocytes do not act as an intermediate to inhibit cdc differentiation and the functional effects on antigen specific CD + T cells and tumor control are primarily mediated by cdcs, not granulocytes, in this model. GCSF neutralization overcomes resistance to immunotherapy. Our data suggest that exposure to tumor-derived GCSF can impair BM progenitors in their ability to generate cdcs. As FltL therapy has been developed for use in patients to bolster cdc numbers and function, we asked if the impairment in cdc development could be rescued by FltL treatment in vivo or if GCSF neutralization was necessary for maximum efficacy of this therapy. To accomplish this we treated tumor-free or mice bearing a cm+ diameter PyMT-B tumor with FltL+/ anti- GCSF IgGs for weeks. Consistent with previous reports,we observed that in tumor-free mice treated with FltL expanded pre-dcs, cdc, and cdc in the BM and pre-dcs in the blood (Fig. 9a, b). We also observed that combined treatment with FltL and anti-gcsf IgGs did not further upregulate pre-dc or cdc numbers in tumor-free mice (Fig. 9a, b). This result is consistent with the finding that GCSF is expressed at lower levels under non-pathologic conditions (Fig. a). Similar to our other experiments, pre-dc and cdc numbers were decreased in the BM and blood of PyMT-B tumor-bearing mice compared to controls (Fig. 9a, b). We also found that although FltL could expand pre-dc and cdc numbers in the BM and blood in the presence of tumors, this expansion was limited compared to tumor-free mice in that pre-dc or cdc numbers only reached levels comparable to untreated tumor-free mice (Fig. 9a, b). These data suggest that BM progenitors in tumor-bearing mice are resistant to FltL-induced cdc differentiation, which is consistent with our in vitro results (Fig. b). Furthermore, anti-gcsf IgGs and FltL synergized to recover pre-dc and cdc numbers in tumor-bearing mice (Fig. 9a, b). Because our data suggest that GCSF impacts IRF levels, we analyzed the impact of FltL and anti-gcsf IgGs on IRF levels in DC progenitors. In tumorbearing mice, in which IRF is downregulated, neutralizing GCSF restores IRF levels to tumor-free mouse conditions in BM progenitors. Importantly, neutralizing GCSF also increased IRF levels in pre-dcs (Fig. 9c, Supplementary Fig. 7d). These findings suggest neutralizing tumor-induced GCSF is required to recover IRF expression, which is necessary for cdc differentiation. To determine if these changes in lymphoid tissues translated to increased tumor immunity, we analyzed tumor tissues. As previously reported, FltL increases both tumor-infiltrating CD + cdcs and CDb + cdcs, but this expansion of cdcs was modest in this model (Fig. 9d). When FltL was combined with anti-gcsf IgGs, tumor infiltration of CD + cdcs, but not CDb + cdcs, were increased by more than twofold and this increase correlated with substantially more CD + T cells. Together, these data show that GCSF neutralization supports FltL-mediated expansion and mobilization of cdc and suggest this activity is facilitated in part by restoring IRF expression and commitment to the cdc lineage. Given these results, we hypothesized FltL+anti-GCSF IgGs would bolster cdc-based immunotherapeutic strategies in established tumors. We employed poly I:C, which supports cdc maturation, and anti-programmed cell death protein (PD) IgGs, which block a T-cell checkpoint. Mice bearing wellestablished cm+ diameter PyMT-B tumors were treated with FltL+poly I:C+anti-PD IgGs+/ anti-gcsf IgGs. Mice treated with combined FltL+poly I:C+anti-PD IgGs did not have an increase in survival relative to vehicle-treated controls. By contrast, survival was extended in mice treated with anti-gcsf IgGs+FltL+poly I:C+anti-PD IgGs compared with mice treated with vehicle alone or FltL+poly I:C+anti-PD IgGs Fig. IRF expression is reduced during breast and pancreatic cancer. a IRF measured in BM MDPs, CDPs, and pre-dcs of end-stage orthotopic PyMT- B tumor-bearing mice and tumor-free controls; n = 7/group. Data are representative of three independent experiments. b RT-qPCR analysis of BM GMPs, MDPs, and CDPs sorted from mice bearing end-stage orthotopic PyMT-B mammary tumors and tumor-free controls for IRF target genes, four samples consisting of two mice each were analyzed per group. c pstat measured in BM MDPs, CDPs, and pre-dcs of end-stage orthotopic PyMT-B tumorbearing mice and tumor-free controls; n =. Data are representative of two independent experiments. d IRF expression in total BM from BC and PDAC patients relative to healthy controls; n = /group. Data are from BC cohort and PDAC cohort. e Correlation between BM pre-dcs and BM IRF expression in BC and PDAC patients. Data are from BC cohort and PDAC cohort ; n =. f IRF expression in blood pre-dcs from PDAC patients relative to healthy controls; healthy controls n =, PDAC n =. Data are from PDAC cohort. g Correlation between blood pre-dcs and blood pre-dc IRF expression in PDAC patients; n =. Data are from PDAC cohort. h Kaplan Meier estimate of recurrence free survival and overall survival of patients +/ median blood pre-dcs IRF expression. Log-rank (Mantel Cox) p value is denoted for differences in recurrence-free survival and overall survival. Data are from PDAC cohort. Spearman s correlation (r, correlation coefficient). End stage for each model is defined in the Methods. Error bars represent mean +/ s.e.m or box plot; p <., p <., p <. by unpaired two-sided Student s t test NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications

NATURE COMMUNICATIONS DOI:./s7--- ARTICLE a % Max PyMT-B bone marrow MDPs CDPs p =. IRF IRF IRF IRF MFI Unstained PyMT-B PyMT-B + Anti-GCSF IgG % Max IRF MFI % Max IRF MFI b Irf MPs MDPs CDPs c MP cdc %CD9. Batf..... MPs..... MDPs..... CDPs IRF overexpression FltL + + FltL+GCSF... Control GCSF FltL+GCSF Fig. 7 GCSF regulates IRF in dendritic cell progenitors. a IRF measured in BM MDPs, CDPs, and pre-dcs of tumor-free mice, orthotopic PyMT-B tumorbearing end-stage mice, and orthotopic PyMT-B tumor-bearing end-stage mice treated for weeks with μg anti-gcsf IgGs /week; n = /group. Data are representative of two independent experiments. End stage is defined in the Methods. b MPs, MDPs, and CDPs treated with ng/ml GCSF and/ or ng/ml FltL for h. Analyzed by RT-qPCR for Irf and Batf. Data are representative of two independent experiments consisting of three wells per condition. c CD. + Lin - Sca ckit + MPs transduced with CD9. IRF overexpression vector or CD9. empty vector control were cultured on CD. + BM feeder culture in the presence of ng/ml FltL and/or ng/ml GCSF for days. Final cultures were analyzed for cdcs (Live CD. + CD9. + CD. - MHCII + CDc + Sirpα - CD + ). Data are representative of three independent experiments consisting of three wells per condition. Error bars represent mean +/ s.e.m. or box plot; p <., p <., p <. by unpaired two-sided Student s t test (Fig. 9e, Supplementary Fig. 7e). This effect was not seen in similarly treated IRF / and BATF / mice, suggesting that this treatment is reliant on IRF and cdc activity (Fig. 9e, Supplementary Fig. 7e). Together, these data suggest that GCSF signaling may need to be inhibited to employ immunotherapy effectively in some established tumor settings. Discussion Breast and pancreas cancers have had limited responses to single agent immunotherapy in the clinic 7. Although this limited response might be explained in part by poor antigenicity, BC and PDAC are also known to expand systemic populations of immune-suppressive myeloid cells by increasing differentiation from myeloid progenitors in the BM 7,,. Our findings suggest that tumor-induced inflammatory cytokines expand potentially immune-suppressive myeloid cells and simultaneously suppress anti-tumor cdc development from BM progenitors. Beyond committing to the granulocyte, monocyte, or cdc lineages, cdc progenitors further commit to the cdc subset before leaving the BM 7. These findings suggest that the antigen-presenting capacity can be defined before cells traffic into the periphery and is not entirely reliant on cues experienced at the tumor site. Therefore, it is important to understand how tumors alter BM myeloid differentiation to overcome tumor-induced immune surveillance. Others have shown suppression of the transcription factor IRF during tumor progression expands the immature granulocyte and monocyte populations that are known to suppress anti-tumor immune responses 9. We further demonstrated IRF downregulation during cancer reduces cdc development in the BM. This is an important finding because IRF expression in progenitors as early as the hematopoietic stem cell primes transcriptional networks and influences lineage bias towards cdc and cdc fate,9. Additionally, tumor-induced depletion of cdcs has a functional implication on anti-tumor immunity beyond the effect of granulocyte expansion. Even in the context of granulocyte depletion, reduced cdc differentiation inhibits CD + T cell-mediated anti-tumor responses, leading to a loss of tumor control in this model. Given that previous work has shown granulocyte depletion is sufficient to recover anti-tumor immunity in some cases 9,, we speculate there is a balance between the expansion of granulocytes as immune suppressors and depletion of cdcs as immune stimulators that varies with tumor type, disease progression, and tumor models. In addition NATURE COMMUNICATIONS () 9: DOI:./s7--- www.nature.com/naturecommunications