CD 24 0100
CD 24 0100
Significance: This study provides new insights into the mechanisms underlying the programming
of CAFs and shows that during this process, expression of the cytokine IL33 is induced. CAF-derived
IL33 has pleiotropic effects on the tumor microenvironment, supporting its potential as a thera-
peutic target.
Furthermore, pancreatic CAFs are heterogenous—although Beyond this characterization of epithelial IL33 (34–39), the
their classification continues to evolve, a general framework role of stromal IL33 has been largely ignored, despite previ-
for CAF heterogeneity describes three groups: “myofibro- ous descriptions of IL33 in this compartment in mouse and
blastic CAFs” (myCAF), “inflammatory CAFs” (iCAF), and human PDA and in other pancreas diseases, such as pancre-
“antigen-presenting CAFs” (apCAF; refs. 22, 23). myCAFs atitis (12, 40, 41). Furthermore, the mechanisms underlying
exist in close proximity to tumor cells and are marked by high the activation of stromal IL33 during carcinogenesis remain
expression of α-smooth muscle actin (α-SMA); these cells are unknown.
major sources of ECM and ECM-remodeling proteins (22). Here, we show that pancreatic PAFs/CAFs are a dominant
Conversely, iCAFs express little-to-no α-SMA and instead source of IL33 in PanIN and PDA. We show that deletion of
produce and secrete a wide variety of signaling molecules, compartment-specific stromal IL33 in an orthotopic model of
including many immunoregulatory chemokines and growth PDA alleviates immunosuppression and suppresses tumor
factors (22). Of note, these classifications are largely based on growth. Additionally, we investigate the mechanisms of IL33
single-cell RNA sequencing (scRNA-seq) and are not fully re- induction and show that its expression by PAFs/CAFs is depen-
flected at the protein level (e.g., most pancreatic CAFs have dent on tumor and fibroblast-derived signaling factors. In sum-
10 1. CK19+ ECAD+
Patient #1 Patient #2 2. Fibroblast
3. Pericyte
4. Endothelial
UMAP_2
5. Myeloid
Adj. Normal
0
6. T Lymphocyte
7. NK Cell
8. B Lymphocyte
9. Acinar
−10 10. Endocrine
11. Platelet
12. RBC
IL33 50 µm IL33 13. Proliferating
−10 0 10 −10 0 10
UMAP_1
C hAdj. Normal hPDA
IL33
3 10 10
T 2
UMAP_2
UMAP_2
0 0
1
IL33 IL33
0 3 3
−10 −10
4 1 4 1
2 2
−10 0 10 −10 0 10
D mHealthy mPanIN mPDA
UMAP_1 UMAP_1
n = 3; cells = 6,859 n = 3; cells = 14,207 n = 3; cells = 18,834
10
E mPDA
5 Lymphocyte mPanIN
UMAP_2
Percent
mWT
0 Expressed
0 mPDA
−5 20 Myeloid mPanIN
40
−10 60 mWT
80 mPDA
−15
−10 0 10 −10 0 10 −10 0 10 Fibroblast mPanIN
Average
UMAP_1
Expression
1. CK19 ECAD
+ +
3. Tuft Cell 5. Mesothelial 7. Myeloid 9. RBC mWT
2. Acinar 4. Endothelial 6. Fibroblast 8. Lymphocyte 10. Proliferating 2
mPDA
1
F Mesothelial mPanIN
mHealthy (WT) mPanIN (KC) mPDA (KPC) 0
mWT
IL-33/PDGFRD/E/E-Cadherin
mPDA
Endothelial mPanIN
<0.0001
mWT
<0.0001 mPDA
IL-33 CTCF per PDGFRD/E+ ROI
Tuft Cell
<0.0001 mPanIN
60,000
50 µm A mPDA
mWT
20,000
mPDA
mWT
18
38
KC = 2
(n
(n
Figure 1. IL33+ stromal cells are abundant in human and mouse PDA. A, Human IHC staining of IL33 in matched adjacent normal (“Adj. Normal”) and
PDA regions. S, stromal area; T, tumor area. B, UMAP visualization of human scRNA-seq dataset split into adjacent normal and PDA groups. n = number of
patients in each dataset. C, Feature plot of IL33 transcription levels in human scRNA-seq. D, UMAP visualization of murine scRNA-seq dataset split into
healthy, PanIN, and PDA groups. E, Dot plot representation of Il33 transcription levels across cell types in the murine scRNA-seq dataset. F, Co-IF staining
of murine tissues [healthy (wildtype) aka WT, PanIN aka KC (Ptf1a-Cre; LSL-KrasG12D), and PDA aka KPC (Ptf1a-Cre; Trp53R172H/+;LSL-KrasG12D)]. IL33
(green), PDGFRα/β (red), E-Cadherin (white), DAPI (blue). IL33 CTCF was quantified per individual ROI; each ROI encompasses one PDGFRα/β+ cell. N = 3
mice were quantified per group. N in the figure represents the number of ROIs measured per group. P values represent one-way ANOVA testing between
groups. Line = mean CTCF.
these cell types may all give rise to PAFs/CAFs in the pancre- orthotopic treatment schedule whereby we gavaged mice
atic disease context. We observed an abundance of IL33 in once a day for 5 days prior to surgery, but then let mice rest
ECAD− PDGFRα/β+ cells in the PanIN and PDA microenvi- for 2 days before tumor implantation, withholding tamoxifen
ronments, as well as occasional expression in the healthy pan- chow for the entirety of the experiment (Fig. 2G). We took this
creas (Fig. 1F). Taken together, our findings show that IL33 is approach to prevent systemic toxicity of prolonged tamoxifen
robustly expressed in transformed pancreatic tissues and that treatment and to assess the relative contribution of PDGFRA+
the stroma is a major source of IL33 in the TME. healthy cells in total CAF IL33 expression. The tumors from
this treatment model exhibited a comparable trend in tumor
Stromal IL33 Promotes PDA Growth size reduction between groups, as seen in our tamoxifen chow
Our observations revealed that the PAF/CAF compartment model (Supplementary Fig. S2C). Two experimental and two
is the highest expressor of stromal IL33 in pancreatic disease. control tumors (one male and one female per group) were
Therefore, to decipher the impact of stromal IL33 in PDA, we pooled and submitted for scRNA-seq. The resulting data re-
utilized the Pdgfra-CreERT2/+ mouse, which targets fibroblasts, vealed a complex TME that included tumor cells as well as
PSCs, and mesothelial cells (the cell types that ultimately fibroblasts, immune cells, and other stromal compartments
A B OT
C CreER CreER;Il33f/f
#1 #2 #1 #2
Injection
Pdgfra +Tamoxifen +Tamoxifen IL-33 (light)
Cre ER
Il33 Il33 IL-33 (dark)
Exons 5-7 EGFP EGFP +Tamoxifen Chow Harvest
LoxP LoxP LoxP −7 days
−2 days 0 day PdgfrD
CreER 21 days
or
CreER;Il33f/f Vinculin
D E F
Ki67/PDGFRD/E/E-Cadherin CC3
Tumor Weight/Body Weight
0.0443
1.5
%Ki67+ of E-Cadherin+
60 0.0257
1.0
40
CreER
CreER
0.5
20 1.5
%Ki67+ of PDGFRD/E+
400 40 0.0790
Tumor Weight (mg)
CreER;Il33f/f
CreER;Il33f/f
300 30 0.0
CreER CreER;Il33f/f
200 20
100 10
0 50 µm 0 50 µm
Ki67 CreER CreER;Il33f/f
CreER CreER;Il33f/f
OT
G Injection H
+Tamoxifen CreER CreER;Il33f/f
n = 2; cells = 5,371 n = 2; cells = 4,816
Single Cell
−7 days Sequencing 19 19
CreER −2 days 0 days
21 days
or
CreER;Il33f/f 10 9 9
PANCREAS_BETA_CELLS 2 2
MITOTIC_SPINDLE 0 4 4
16 10 16 10
EPITHELIAL_MESENCHYMAL_TRANSITION
15 15
APICAL_JUNCTION
ESTROGEN_RESPONSE_EARLY 12 1 7 12 1 7
MTORC1_SIGNALING
P53_PATHWAY
3 3
−10 18 18
APOPTOSIS
INTERFERON_GAMMA_RESPONSE 8 8
UV_RESPONSE_UP 21 21
HYPOXIA 5 5
CHOLESTEROL_HOMEOSTASIS
padj −10 −5 0 5 10 15 −10 −5 0 5 10 15
ADIPOGENESIS
XENOBIOTIC_METABOLISM 0.020
UMAP_1
FATTY_ACID_METABOLISM 1. Tumor 5. Endothelial 9. Macrophage 13. CD8+ T Cell 17. Mast Cell
0.015
ESTROGEN_RESPONSE_LATE
0.010 2. EMT-like 6. Mesothelial 10. Dendritic Cell 14. NK Cell 18. B Cell
INTERFERON_ALPHA_RESPONSE
TNFA_SIGNALING_VIA_NFKB 0.005 3. Acinar 7. Fibroblast 11. Monocytic Mac. 15. CD4+ T Cell 19. Plasma Cell
OXIDATIVE_PHOSPHORYLATION 4. Ductal 8. Pericyte 12. Granulocyte 16. ILC2 20. Proliferating
−2 −1 0 1 21. RBC
Normalized Enrichment Score
J Il1rl1 (ST2) K L
Left: Cre Right: Cre;Il33f/f OT
Tumor Weight/Body Weight
800 2.0
Expression Level
Injection 0.3575
0.0469
Tumor Weight (mg)
3 600 1.5
2
1 Harvest 400 1.0
0 Il1rl1+⁄+ 0 day 21days 200 0.5
or
Naive CD4
Exhausted CD4
Th1
Th17
Treg
ILC2
Mast Cell
Il1rl1−⁄−
0 0.0
Il1rl1+⁄+ Il1rl1−⁄− Il1rl1+⁄+ Il1rl1−⁄−
Figure 2. Stromal IL33 promotes PDA growth. A, Genetic scheme of Pdgfra-CreERT2/+;Il33f/f murine model. Tamoxifen induces activation of the Cre-
ERT2 fusion protein, allowing recombination to occur. B, Experimental design for the Pdgfra-CreERT2/+;Il33f/f orthotopic tumor model. OT, orthotopic,
CreER = Pdgfra-CreERT2/+, CreER;Il33f/f = Pdgfra-CreERT2/+;Il33f/f. C, Western blot of PDGFRα+ cells sorted from CreER and CreER;Il33f/f orthotopic
tumors. Two tumors/mice were pooled in each lane. D, Relative and absolute tumor sizes from CreER and CreER;Il33f/f orthotopic tumors. (E + F) Immu-
nostainings of CreER and CreER;Il33f/f tumors: E, = Co-IF staining of Ki67 (green), PDGFRα/β (red), E-Cadherin (white), and DAPI (blue), F, = IHC staining
of Cleaved Caspase-3 (CC3). In quantification, each dot represents one animal. G, Treatment schedule for the Pdgfra-CreERT2/+;Il33f/f orthotopic tumor
model adapted for scRNA-seq. H, UMAP visualization of orthotopic scRNA-seq dataset split into CreER and CreER;Il33f/f groups. I, Waterfall plot depict-
ing differential pathway enrichment in tumor cells based on the Hallmark collection of annotations. Positive normalized enrichment scores are enriched
in the control group. Pathways of interest are bolded. padj = Bonferroni-corrected P value. J, Violin plot depicting expression of Il1rl1 (ST2) in select
leukocytes from scRNA-seq. K, Experimental design for Il1rl1+/+ and Il1rl1−/− orthotopic tumor experiment. L, Relative and absolute tumor sizes from
Il1rl1+/+ and Il1rl1−/− orthotopic tumors. Tumor weight/body weight ratios are relative to the control group. Histogram data are mean ± standard deviation.
Experiments with two conditions were compared using a two-tailed Student t test.
improved and worse survival in PDA, respectively (51). We Fig. S2D). To parse out which CD4+ T cells expressed ST2,
found that the tumor cells of CreER;Il33f/f mice had an enrich- we subclustered them and discovered that Tregs, but not
ment for the “classical” gene signature, whereas the basal sig- other helper T-cell populations present in the tumors, were
nature score was unchanged across the two groups (Supple- ST2-positive (Fig. 2J; Supplementary Fig. S3D). Interestingly,
mentary Fig. S2H). We then took an unbiased approach and IL33 activation of mast cells, ILC2s, and Tregs has been linked
assessed differential pathway activation between tumor cells to disease progression in solid tumors, including IL33-ILC2/
in the control and experimental animals using the Hallmark Treg activation in pancreatic cancer (35, 36, 39, 60). Given
collection of molecular signatures (Fig. 2I). In accordance with the presence of ST2+ immune cell populations in the tumors,
our staining result (Fig. 2E and F; Supplementary Fig. S2B), we performed an additional orthotopic PDA implantation ex-
we found enrichment for the “MITOTIC_SPINDLE” signature periment using Il1rl1+/+ and Il1rl1−/− mice (Fig. 2K). Tumors in
in the control group and “P53_PATHWAY” and “APOPTOSIS” Il1rl1−/− mice were ∼30% smaller than controls, further impli-
in the experimental group. We additionally detected enrich- cating IL33-ST2 signaling as protumorigenic in PDA (Fig. 2L).
ment for the “EPITHELIAL_MESENCHYMAL_TRANSITION” Overall, these data suggest that CAF IL33 supports PDA.
signature in the control tumor cells, indicating that tumor The loss of IL33 from PDGFRA+ stromal cells correlates with
Try4/GM10334
b1
Macrophage Acinar
Lama4
Cd a s 3
Itg
G bs
Gdf10
fb
Cxcl1 Pos xb
CD8+ T
22 6
Thb tga9/
Th
P dg
Igf1
6
Treg CreER;Il33 b NK
f/f
d gf 9 Mast
2
P gals
I
CreER
s1
L
Tn
tn
Fibroblast
Mast Cell CreER;Il33
f/f
Ptn
p1
Sp
CreER Fibroblast 1
am
Nc
ILC2 CreER;Il33
f/f
CreER Tncfb1
Tg b
Il1 rres2
Ra a5a
Klrg1
Areg
Csf2
Ccl3
Il13
80 Il4
Il5
Il6
Lif
Sem
Entpd1
Percent
20
40
60
Fibroblast Thbs1
0
Expressed
receiving
Col1
Il1a/b
Average 2 1 0 Tnf
a1/a
Expression Macrophage
Th
B
bs
CreER vs. CreER;Il33f/f
1
Endothelial
Il1a/b
Sell
EGF
Col4a1/a2
Inhba
ILC2 Monocytic Mac.
S p eg
gf
Areg
p1
APOPTOSIS
Ar
N 1r Tumor
Agrn
MYC_TARGETS_V1 1 Granulocyte
Ductal F Treg
GLYCOLYSIS EMT
MYOGENESIS
HYPOXIA D F CreERT2/+
TGF_BETA_SIGNALING 1 CreERT2/+;Il33f/f
CHOLESTEROL_HOMEOSTASIS 2 1. iCAF 4
HEDGEHOG_SIGNALING 2 2. iCAFhi, myCAFlo
MTORC1_SIGNALING 5 6 4
UMAP_2
P53_PATHWAY Il33
3. myCAF
TNFA_SIGNALING_VIA_NFKB
0
−1.5 −1 −0.5 0.0 4. myCAF , iCAFhi lo
lo
lo
lo
−2
0.04
0.03
0.02
0.01
AF
AF
AF
AF
AF
AF
3
iC
yC
C
yC
pC
6. myCAFhi, apCAFlo
ap
,i
padj
m
hi
,m
,a
AF
hi
−8 −4 0 4
hi
AF
AF
yC
UMAP_1
yC
iC
m
E
m
3 4 5 7
Pdpn Clec3b Acta2 Saa3
Pan-Fibroblast
3 4 4 4
myCAF
apCAF
iCAF
3 4 5 5
Pdgfrb Col14a1 Ccn2 Slpi
, i F lo
AF my F lo
ap F lo
F lo
AF my F lo
ap F lo
, i F lo
AF my F lo
ap F lo
, i F lo
AF y F lo
ap F lo
F
pC F
AF
pC F
AF
pC F
AF
yC m AF
pC F
AF
m h,i iCA
, a CA
m h,i iCA
, a CA
m h,i iCA
, a CA
, a CA
hi A
hi A
hi A
hi A
A
C
iC
C
AF yC
C
AF yC
C
AF yC
C
AF yC
C
i
m
yC m
yC m
yC m
,
hi
hi
hi
hi
hi
m ,
AF
AF
AF
AF
yC
yC
yC
yC
iC
iC
iC
iC
m
35.9
59.4% 22.1 Tumor Granulocyte Tumor CD8+ T
CD8+ T NK
NK CD4+ T
Kng2 CD4+ T Treg
Gdf10 Treg Il1b Mast
50% 9.6 B
Clec2d ILC2 Cx3cl1 Plasma
myCAF hi
Vegfd Mast Tgfb1
31.4 Igf1 B Vegfa
20.0 63.1% 2 Plasma
Cxcl5/1stn Cxcl1
/2
myCAF hi Po nxb m1
25% T Nca
c
35.7%
4.2 Ptn Tn Th
bs
11.5 1
10.2 C3
apCAF
p1
0% apCAF 4.9
Sp
Co
Fibroblast Fibroblast
l1a
Mif
1/
Figure 3. Loss of stromal IL33 alters the ST2+ immune cell secretome, resulting in a shift in CAF differentiation. A, Gene expression of activation
markers split by CreER and CreER;Il33f/f from scRNA-seq. B, Waterfall plot depicting differential pathway enrichment in fibroblasts based on the Hallmark
collection of annotations. Negative normalized enrichment scores are enriched in the experimental group. Pathways of interest are bolded. No genesets
were enriched in the control group with Bonferroni-corrected P value (padj) of < 0.05. C, Chord diagram visualizing differentially enriched (Bonferroni-
corrected P value < 0.05 and fold-change ≥0.25) predicted to interact with fibroblasts. Edge widths are proportional to predicted interaction strength.
D, UMAP visualization of fibroblasts from the CreER and CreER;Il33f/f scRNA-seq datasets. E, Gene expression of markers representing CAF subtypes.
F, Il33 expression in each CAF population split by experimental group. G, Histogram depicting the frequency of each CAF population across the CreER
and CreER;Il33f/f scRNA-seq datasets. H, Chord diagram visualizing differentially enriched (Bonferroni-corrected P value < 0.05 and fold-change ≥0.25)
fibroblast-derived ligands and their predicted interaction partners. Edge widths are proportional to predicted interaction strength.
analyses (Fig. 3B–H) include only the “fibroblast” population changes in NF-κB signaling or TGFβ signaling between the
and not the “mesothelial” or “EMT-like” cells). Interestingly, the IL33 WT and KO cells, either at baseline or upon treatment
top enriched pathway in CreER;Il33f/f fibroblasts was “TNFA_ with PDA CM (Supplementary Fig. S4E). As these pathways
SIGNALING_VIA_NFKB,” which is downstream of AREG define myCAF/iCAF/apCAF differentiation in vivo, it seems un-
activation (Fig. 3B). Other pathways differentially activated likely that the nuclear activity of IL33 plays a role in regulating
in the IL33-deficient fibroblasts included “HEDGEHOG_ CAF polarization. A more likely explanation is that a change
SIGNALING” and “TGF_BETA_SIGNALING” (both linked in factors secreted by ST2-expressing immune cells affects
to myCAF differentiation; refs. 11, 22), as well as “P53_ fibroblast differentiation status. Importantly, we also mea-
PATHWAY,” “HYPOXIA,” and “APOPTOSIS” (the latter of sured proliferation in these cell lines and found no differ-
which complements our CC3 IHC staining; Fig. 2F). To un- ences between the IL33 WT or KO groups, under DMEM or
derstand the drivers of these changes in fibroblasts, we used PDA CM treatment (Supplementary Fig. S4F). These data
our scRNA-seq data to plot an unbiased differential predicted suggest that intrinsic IL33 is dispensable to CAF survival
interaction analysis between cells across the TME and fibro- and cell growth.
blasts in the control and CreER;Il33f/f context (Fig. 3C). Our As an important function of PDA CAFs is to activate protu-
Inactivation of Stromal IL33 Enables Cytotoxic changes in CD8+ T-cell function (Fig. 4C). We found that the
T-cell Activity strongest predicted interactions in control tumors were be-
Tumor-infiltrating myeloid cells play a central role in the ini- tween CD8+ T cells and collagens originating from fibroblasts
tiation, progression, and maintenance of PDA through their and EMT-like cells; in KRASG12D-driven murine models of lung
secretion of tumor-promoting factors and their suppression cancer, collagen interactions directly induce CD8+ T-cell ex-
of CD8+ T cells (28). Given the changes in multiple immune- haustion and correlate with low CD8+ T-cell infiltration (77).
regulatory factors in CreER;Il33f/f tumors, we sought to de- Conversely, the strongest signal received by CD8+ T cells in
termine whether loss of PDGFRA+ stromal cell IL33 caused the CreER;Il33f/f model was MHC-I presentation from tumor
changes in myeloid populations within the TME. We first cells (Fig. 4C), suggesting a potential for increased tumor
stained tumors for macrophages (F4/80) and granulocytes cell killing and correlating with our increase in CC3 staining
(myeloperoxidase) to measure cell abundance (Fig. 4A; Sup- (Fig. 2F). We also observed multiple CD8+ T-cell recruiting fac-
plementary Fig. S5A). Although macrophage levels were tors enriched in CreER;Il33f/f tumors, including Cxcl9, Ccl7, and
Ccl2 from EMT-like cells, endothelial cells, mesothelial cells,
unchanged, granulocyte infiltration exhibited a dramatic in-
and macrophages (Fig. 4C). Beyond the interaction analysis, the
crease; the latter was predicted by the upregulation of fibro-
A CreER CreER;Il33f/f
B
CreER;Il33f/f
Monocytic Mac.
CreER
CreER;Il33f/f
Macrophage
F4/80
CreER
Il1a
Il1b
Cxcl10
Cd80
Cd86
Tlr2
Il6
Cxcl9
H2−Ab1
H2−Eb1
Ccl5
Cxcl16
Tlr4
Tgfb1
Vegfa
Ccl24
Chil3
Arg1
Mrc1
Cd163
Tnf
Percent Average
50 µm Expressed Expression
−0.5
−1.0
1.0
0.5
0.0
25
50
75
Pro-Inflammatory Immunosuppressive
%F4/80+ Area per FOV
15
0.2166 C
10
Enriched Signaling in CreER Enriched Signaling in CreER;Il33
5 CD8+ T receiving CD8+ T receiving
0
CreER CreER;Il33f/f
M
/T2
Fibroblast
HC
Endothelial 3) f
Tn 9
−I
Collagen (Col1a1 Macrophage Cxcl
(H
/a2)
En hbs l2
2-
tp 1
T Cc
D1
Sp d1
Th p1
/K
1
Cc 4/Gm1
Ccl7
1)
Pericyte
bs
Try
Lama4
Icam2
Cxcl9
MHC−
EMT
l2
Fibroblast
I (H2-Q
Mesothelial
033
Endothelial Acinar
4
6)
D E
CD8/Granzyme-B/E-Cadherin CD4/Foxp3
CreER
CreER
50 µm 25 µm 50 µm 25 µm
CreER;Il33f/f
CreER;Il33f/f
80 150 80
Total CD4+ Cells per FOV
4 0.0451
%CD8+ Area per FOV
0.3765
%GzmB+ of CD8+ Area
0.0475 0.0007
3 60 60
100
2 40 40
50
1 20 20
0 0 0 0
CreER CreER;Il33f/f CreER CreER;Il33f/f CreER CreER;Il33f/f CreER CreER;Il33f/f
Figure 4. Inactivation of stromal IL33 enables cytotoxic T-cell activity. A, IHC staining of F4/80 in CreER and CreER; Il33f/f tumors. B, scRNA-seq gene
expression of curated proinflammatory and immunosuppressive markers, grouped by cell type and split by experimental group. C, Chord diagram visualiz-
ing ligands differentially enriched (Bonferroni-corrected P value < 0.05 and fold-change ≥0.25) in CreER and CreER;Il33f/f tumors that interact with CD8+
T cells. Edge widths are proportional to predicted interaction strength. Chemokines are bolded. D and E, Co-IF staining of CreER and CreER;Il33f/f tumors:
(D) = CD8 (green), Granzyme-B (red), E-Cadherin (white) and DAPI (blue), (E) = CD4 (yellow), Foxp3 (magenta), and DAPI (cyan). For staining quantification,
each dot represents one animal, and values were compared using a two-tailed Student t test. Histogram data are mean ± standard deviation.
express KrasG12D in a doxycycline-dependent, inducible and re- although the mechanism by which this occurs remains un-
versible manner (referred to as “KRASG12D ON” and “KRASG12D known. As such, we first endeavored to understand whether
OFF,” respectively; Fig. 5A). Using the iKRASG12D model, we stromal IL33 expression remains dependent on oncogenic KRAS
have shown that expression of PAF Il33 is dependent on epithe- throughout tumorigenesis. We generated an “atlas” of pre-
lial oncogenic KRAS at the onset of tumor initiation (12), viously published and newly generated iKRASG12D scRNA-seq
A rtTa
B
p48
Cre –Dox “KRASG12D OFF”
+cae
KRASG12D “3 or 5 weeks
Rosa26 TetO OFF
Early PanIN
STOP rtTa EGFP rtTa EGFP ON
LoxP LoxP LoxP +dox KRASG12D ON or +3 days OFF”
Dox “KRAS G12D
ON”
72 hours 48 3 or 5 weeks
+Dox rtTa iKRASG12D hours ON “3 or
KRASG12D 5 weeks ON”
TetO 3 days
C
iKrasG12D ON iKrasG12D OFF “14 weeks ON”
Late PanIN
10
“15 weeks ON
10 +dox KRASG12D ON ON OFF +1 week OFF”
iKRASG12D; 14 weeks
5 1 week 1 week
2 Trp53R172H/+
9
UMAP_2
6
0 OT injection
iKRASG12D;Trp53R172H/+
“OT PDA ON
4
OFF +3 days OFF”
PDA
−10 8 1 +dox KRAS ON
G12D or
7 11 3
6 FVBN 2 weeks
ON “OT PDA ON”
Expression Level
4
D 3
iKrasG12D ON iKrasG12D OFF 2
1
10 10 Il33 0
5 5
4 KrasG12D: ON OFF ON OFF ON OFF ON OFF
UMAP_2
UMAP_2
3
0 0 3 week 5 week 14+ week OT PDA
2
1
−10
4
−10
4
0
G +cae +/– JAK1/2i
F Fibroblast ON vs OFF
H
HALLMARK “IL6_JAK_STAT3_SIGNALING” IL-33/PDGFRD/E/E-Cadherin
NES = 1.58
enrichment score
0.4
20,000
0.25
0.2
0.00 10,000
0.0
50 µm
0 5,000 10,000 15,000 20,000
rank 0 5,000 10,000 15,000 20,000 0
rank
Control
(n = 305)
JAK1/2i
(n = 270)
14+ week iKRASG12D;Trp53R172H/+ OT PDA iKRASG12D;Trp53R172H/+ I
FDR = 2.92e–2 FDR = 6.51e–4 Pdgfra
Cre ER
+Tamoxifen
K Il33
enrichment score
+Tamoxifen
0.0
CAF
CreER;Stat3f/f
CreER
Sorting
CreER +
−7 days 0 days
or 21 days RNA
Extraction
CreER;Stat3f/f
Figure 5. Expression of fibroblast IL33 is extrinsically induced by epithelial KRASG12D and requires JAK1/2-STAT3 activation throughout tumorigenesis.
A, Genetic scheme of the iKRASG12D mouse. Doxycycline induces reversible expression of KRASG12D in pancreatic epithelial cells. B, Diagram representing
the various iKRASG12D treatment models and collection points across tumorigenesis. Cae, caerulein; OT, orthotopic. C, UMAP visualization of iKRASG12D
scRNA-seq dataset. Projection on the left is colored by cell type (all datasets merged). Projections on the right are split by iKRASG12D “ON” and “OFF” status
and are colored by timepoint. D, Feature plot representation of Il33 expression levels split by iKRASG12D “ON” and “OFF” status (all timepoints merged). E, Violin
plots depicting fibroblast Il33 expression level per timepoint and split by iKRASG12D “ON” and “OFF” status. Wilcoxon rank sum tests were performed between
iKRASG12D “ON” and “OFF” pairings per each timepoint, and Bonferroni adjusted P values are displayed above violins. F, GSEA enrichment plots of the Hallmark
“IL6_JAK_STAT3_SIGNALING” pathway based on fibroblast iKRASG12D “ON” and “OFF” differential gene expression analysis within each timepoint. G, Treatment
scheme for iKRASG12D “ON” model + JAK1/2 inhibitor. H, Co-IF staining of IL33 (green), PDGFRα/β (red), E-Cadherin (white), DAPI (blue). IL33 CTCF was
quantified per individual ROI; each ROI encompasses one PDGFRα/β+ cell. N = 3 mice were quantified per group. N in the figure represents the number of ROIs
measured per group. P values represent a two-tailed Student t test. Line = Mean CTCF. I, Genetic scheme of Pdgfra-CreERT2/+;Stat3f/f (CreER;Stat3f/f) murine
model. Tamoxifen induces activation of the Cre-ERT2 fusion protein, allowing recombination to occur. J, Diagram representing the treatment schedule for the
CreER;Stat3f/f orthotopic tumor model. K, Expression levels of Il33 in CAFs from J as measured by RT-qPCR. Values are normalized to Ppia (Cyclophilin A) and
relative to the CreER group. Two-tailed Student t test was performed to compare groups; data are mean ± standard deviation.
data representing distinct stages of PDA development. This even in the presence of oncogenic KRAS. Next, to condition-
included two “Early PanIN” timepoints (3 and 5 weeks post- ally disrupt stromal JAK1/2-STAT3 signaling, we bred Pdgfra-
pancreatitis; refs. 7, 12, 79), a “Late PanIN” timepoint wherein CreERT2/+;Stat3f/f (CreER;Stat3f/f) mice, which lose exons 18
mice also have a full-body Trp53R172H/+ knock-in mutation to to 20 of Stat3 upon activation of recombination by tamoxi-
accelerate tumorigenesis (55), and a “PDA” model of synge- fen (Fig. 5I). We gavaged mice with tamoxifen once a day for
neic orthotopically injected iKRASG12D;Trp53R172H/+ tumor cells 5 days to suppress Stat3 in Pdgfra+ cells and then implanted
(Fig. 5B; refs. 55, 79). We also included matched “KRASG12D syngeneic tumor cells orthotopically to model mature PDA
OFF” groups for comparison at each timepoint (7, 12, 55). (Fig. 5J; Supplementary Fig. S6G). The resulting tumors
These datasets were batch-corrected and analyzed collectively. also displayed a notable decrease in growth (Supplementary
The resulting Uniform Manifold Approximation and Pro- Fig. S6H), in accordance with our CreER;Il33f/f and Il1rl1−/−
jection for Dimension Reduction (UMAP) visualization re- orthotopic models. We sorted PDGFRα+ cells from these
vealed a diverse cellular landscape including CK19+ ECAD+ tumors, extracted RNA, and assessed via RT-qPCR to reveal
cells (encompassing ductal/malignant-ductal cells), stromal a reduction in Il33 mRNA levels in the CreER;Stat3f/f model
cells including fibroblasts and mesothelial cells, and immune (Fig. 5K). Collectively, these findings suggest that KRASG12D-
A B
M
EM
C
+ ON CM
FF
iKRASG12D ON
M
“ON CM”
+O
+dox JAK1/2i (µmol/L) – – – 0.03 0.3 3
Harvest IL-33
CAFs
iKRASG12D
iKRASG12D; IL-33/D-Tubulin 1 1.5 9.2 6.4 2.2 1.1
Tumor Cells
Trp53R172H/+ iKRASG12D OFF “OFF CM” p-STAT3 (Y705)
–dox pSTAT3/STAT3 1 1.1 2.5 2.4 1.0 0.1
Harvest
STAT3
Fibroblasts CAFs
Healthy
D-Tubulin
C Il33 Il6 D
0.0006 0.0124 Acta2 rIL-6 (ng/mL)
+ON +ON rLIF (ng/mL)
<0.0001 0.0003
0.0002 DMEM CM 0.03 0.3 3 30 DMEM CM 0.06 0.6 6
5 0.0017 5 0.0024 2.0 0.0032 <0.0001
IL-33
Relative mRNA
Relative mRNA
Relative mRNA
4 4
1.5 IL-33/D-Tubulin 1 2.8 1.0 1.2 2.5 2.6 1 3.6 1.6 1.5 3.2
3 3
1.0 p-STAT3 (Y705)
2 0.9430 2 0.3300 pSTAT3/STAT3 1 1.6 1.0 1.3 1.7 3.2 1 2.4 1.6 1.8 3.0
0.6028
Il6
E F Il33 <0.0001
<0.0001 Acta2
<0.0001 <0.0001
Log10(Relative mRNA)
Log10(Relative mRNA)
Log10(Relative mRNA)
<0.0001 <0.0001
100 10 10 <0.0001
+ON CM (hours) – 2 4 8 24 – 0.0005
<0.0001
IL-33
10 1
p-STAT3 (Y705) 1
STAT3 1 0.1
D-Tubulin
0.1 0.1 0.01
Hours: 0 1 2 4 8 16 24 48 8 48 Hours: 0 1 2 4 8 16 24 48 8 48 Hours: 0 1 2 4 8 16 24 48 8 48
+ON CM +OFF CM
pSTAT3 Relative Band Intensity
0.9496 0.8223
15 <0.0001
6 0.0113 G DMEM Group 1: Group 2:
0.1106
Collect Add Back Add Fresh Treatment
4 Fibro. CM Matching Fibro. CM + GolgiStop
10 0.0038
0.3296
+“ON CM” Harvest
0.0709
RNA
5 2 0.9860 +
0.9624
18 hours 6 hours Protein
+rLIF (6 ng/mL) Wash plates,
0 0 & Re-Treat:
Hours: 0 2 4 8 24 24 Hours: 0 2 4 8 24 24
+ON CM +OFF +ON CM +OFF
CM CM
H Lif I
0.0768
Il6
0.4021 Acta2
Il33 0.3782
+ON CM
DMEM
0.0072 0.0417 0.4945 +ON CM
<0.0001
+rLIF
0.8757
DMEM
Relative mRNA
Relative mRNA
Relative mRNA
20 6 3
15
IL-33/D-Tubulin 1 9.3 11.8 0.4 1.6 1.7
15
10 4 2 p-STAT3 (Y705)
10 pSTAT3/STAT3 1 2.0 1.9 0.5 0.9 2.2
5 2 1
5 STAT3
0 0 0 0 D-Tubulin
+GolgiStop +GolgiStop +GolgiStop +GolgiStop
DMEM
+ON CM
+rLIF
DMEM
+ON CM
+rLIF
DMEM
+ON CM
+rLIF
DMEM
+rLIF
+ON CM
DMEM
+ON CM
+rLIF
DMEM
+ON CM
+rLIF
DMEM
+ON CM
+rLIF
DMEM
+ON CM
+rLIF
Figure 6. Tumor cell-initiated autocrine signaling drives IL33 upregulation in pancreatic fibroblasts. A, Ex vivo culture scheme for iKRASG12D; Trp53R172H/+
(cell line 9805) CM generation and healthy pancreatic fibroblasts (cell line CD1WT). B, Western blot of CD1WT whole cell lysates after 24 hours of treatment
with DMEM, iKRASG12D “OFF” CM, iKRASG12D “ON” CM, or concurrent iKRASG12D “ON” CM and JAK1/2i. C, RT-qPCR of CD1WT after treatment with DMEM,
JAK1/2i (4 hours, 0.3 μmol/L), iKRASG12D “ON” CM (24 hours), or pretreatment of iKRASG12D “ON” CM for 20 hours followed by spike-in of JAK1/2i (0.3 μmol/L)
for an additional 4 hours (24 hours total iKRASG12D “ON” CM treatment). Groups were compared with ordinary one-way ANOVA. D, Western blot of CD1WT
whole cell lysates after 24 hours of treatment with DMEM, iKRASG12D “ON” CM, rIL6 (left) or rLIF (right). E, Representative western blot of CD1WT whole
cell lysates after treatment with DMEM, iKRASG12D “OFF” CM, or iKRASG12D “ON” CM for increasing intervals of time. Densitometry quantification for IL33
normalized to loading control (α-tubulin) and pSTAT3 normalized to total STAT3 are shown. Quantification is relative to the 0-hour timepoint. Ordinary
one-way ANOVA was performed to compare each timepoint to the control. F, RT-qPCR of CD1WT after treatment with DMEM, iKRASG12D “OFF” CM, or
iKRASG12D “ON” CM for increasing intervals of time. Values are log10 transformed to better visualize large changes in gene expression level. Ordinary
one-way ANOVA was performed to compare each timepoint to the 0-hour timepoint. Only comparisons with P value < 0.05 are shown. G, Experimental
scheme to block autocrine signaling in CD1WT. CD1WT were treated with DMEM, iKRASG12D “ON” CM, or rLIF for 18 hours, and then, the resulting CM was
set aside. Cells were washed with PBS and then given back their original 18-hour CM or given GolgiStop (1.3 μL/2 mL) + fresh DMEM, iKRASG12D “ON” CM,
or rLIF media. Cells were incubated for an additional 6 hours before harvesting CD1WT RNA and protein. H, RT-qPCR of CD1WT after autocrine blocking
experiment. Two-tailed Student t test was performed to compare groups of interest (all tested comparisons shown). I, Western blot of CD1WT whole cell
lysates after autocrine blocking experiment. In all experiments with iKRASG12D CM, doxycycline is used as a vehicle control. In all experiments with JAK1/2i
(ruxolitinib), DMSO was used as a vehicle control. All replicates represent complete, independent experiments. RT-qPCR values are normalized to Ppia
(Cyclophilin A) and relative to the untreated DMEM group. Histogram data are mean ± standard deviation.
We then queried whether JAK1/2 was also required to main- Interestingly, we found that TGFβ induced heightened expres-
tain IL33 expression after its initial induction and subsequent sion of the myCAF marker Acta2 (as predicted given the rela-
CAF polarization. We tested this by pretreating CD1WT with tionship between TGFβ and the myCAF phenotype; ref. 22) and
“KRASG12D ON” CM for 20 hours and then supplementing moderate upregulation of Il6 but suppressed Il33 expression.
cultures with JAK1/2i for an additional 4 hours (Fig. 6C). By Lif was also unaffected by rTGFβ at this timepoint. We further
RT-qPCR, we found that this acute spike-in of the JAK1/2i investigated the relationship between TGFβ and IL33 by com-
suppressed Il33 and Il6 expression, corroborating our in vivo bining the treatment of CD1WT with “KRASG12D ON” CM and
findings that JAK1/2 activation is continuously required to a TGFβ receptor inhibitor (Supplementary Fig. S7G). The ad-
maintain IL33 upregulation in PDA CAFs (Fig. 5H–K). Next, dition of the inhibitor did not impact the ability of “KRASG12D
we investigated the relationship between CAF polarization, ON” CM to upregulate IL33, despite dose-dependent loss of
JAK1/2 activation, and IL33 expression in a human model. We pSMAD2/3 in this assay. Therefore, our data indicate that
generated CM from primary patient-derived PDA organoids TGFβ is not involved in the upregulation of fibroblast IL33.
and provided it to primary fibroblasts collected from either In fact, Il33 expression may be antagonized by high TGFβ ac-
adjacent normal (normal-like fibroblasts) or tumor regions tivity, similar to the generation of the iCAF phenotype.
endothelial cells express IL33, only a subset of murine endo- found a potential immune cell-fibroblast paracrine signaling
thelial cells show IL33 expression; ref. 31). This observation axis. Beyond its effect on fibroblasts, AREG might act on the
may also be reflected in our scRNA-seq data: although murine pancreatic epithelium to reinforce the KRAS-MAPK signaling
endothelial cells have little-to-no Il33, our human endothelial axis, which is important in the onset of pancreatic carcino-
cells have substantial IL33 expression in adjacent normal and genesis even in the presence of oncogenic KRAS (88, 89). CAFs
PDA tissues, suggesting that IL33 expression in this compart- in CreER;Il33f/f tumors also displayed increased activation of
ment may be disease agnostic (it should be noted, however, Hedgehog and TGFβ signaling, both of which are linked to
that some level of dysplasia is expected in adjacent normal myCAF differentiation (11, 22). In addition to an increase
tissues due to their close localization with the tumor, making in Areg, stromal IL33 deletion results in increased NF-κB li-
them a limited control for such a comparison). Although we gands, namely Il1a, Il1b, and Tnf from myeloid cells and fibro-
found that the fibroblast compartment was the highest ex- blasts, as well as increased fibroblast Tgfb1, all of which may
pressor of IL33 across multiple GEMMs of PanIN and PDA, act on the CAF compartment. The overarching result of these
with minimal expression of epithelial IL33 by RNA and pro- pleiotropic changes was a shift in fibroblasts toward a myofi-
tein staining, our human PDA data show robust expression broblastic phenotype and a reduction in immunosuppressive
component Cd80 in the CreER;Il33f/f context, adding to the adhesion kinase activation has been implicated in IL33 ex-
proinflammatory milieux of the TME. Given that we did not pression (92, 93), and in PSCs, PDGF-BB and IFNγ can both
detect ST2 expression in these myeloid cell populations by cause IL33 upregulation (40).
scRNA-seq, these changes are likely due to indirect effects of Together, our findings shed light on the mechanisms
IL33 loss, potentially stemming from changes in the produc- through which cancer cells reprogram the stroma, particularly
tion of other cytokines from ST2+ cells. CAFs—mechanisms that could be targeted therapeutically to
In the context of stromal IL33 loss, tumor cell/TME to “normalize” fibroblasts in the setting of pancreatic cancer,
T-cell interactions were altered in such a way that the overall as proposed (94). Our data demonstrate the rapid nature by
result was an increase in CD8+ T-cell recruitment and activa- which CAF polarization can shift due to the addition or loss of
tion, with likely cytotoxic activity, as evidenced by increased extracellular stimuli, further complicating potential efforts to
CC3 and Granzyme-B. To this point, we also found increased establish sustained CAF reprograming as a therapeutic ap-
infiltration by helper T cells, which are necessary for driving proach. Importantly, our use of the pharmacologic KRASG12D
an antitumoral cytotoxic response (91). We were surprised to inhibitor, MRTX1133, showed a reduction of CAF IL33 expres-
find no change in the relative abundance of Th2s or ILC2s sion that mirrored the genetic ablation of oncogenic KRAS,
mice and Pdgfra-CreERT2/+;Stat3f/f mice, respectively. These mice were Histology and Immunohistochemistry
aged 8 to 12 weeks and then treated with tamoxifen (4 mg/day, Murine tissues were incubated in 10 mL of 10% neutral-buffered
#T5648, Sigma-Aldrich) dissolved in corn oil via oral gavage once a formalin at room temperature (RT) for 24 hours before being sub-
day for 5 days. Two days after their last gavage, mice underwent orthot- mitted to the University of Michigan Tissue & Molecular Pathology
opic tumor implantation surgery. Experimental and control animals Shared Resource core facility for paraffin embedding and sectioning.
used in Pdgfra-CreERT2/+;Il33f/f-eGFP experiments were additionally Gomori trichrome staining was performed following the manufac-
switched to tamoxifen chow (400 mg/kg, #TD.130860, Teklad Cus- turer’s guidelines (#87021, Epredia).
tom Diets) immediately following the last administration of tamox- For IHC staining, slides were deparaffinized and rehydrated
ifen gavage and remained on tamoxifen chow until experiment end by serial incubation in Xylene (5 minutes, two times), 100% eth-
(except in the case of animals intended for scRNA-seq, which received anol (5 minutes, two times), 95% ethanol (1 minute, two times), and
standard chow diet). diH2O (5 minutes). Slides were then subjected to antigen retrieval
Il1rl1−/− mice were originally generated by Dr. Shizuo Akira’s lab with Antigen Retrieval Citra Solution (#HK086-9K, BioGenex) and
and backcrossed to C57BL/6 by Dr. Stefan Writz’s group (97, 98). Mice microwaving as per the manufacturer’s instructions. Upon cooling,
were aged to 8 weeks old before undergoing orthotopic tumor im- slides were quenched in 0.3% H2O2 in methanol for 15 minutes
plantation surgery. Il1rl1+/+ C57BL/6 mice were used as controls. before blocking in 1% BSA (#BP1600-100, Fisher Scientific) in PBS
(Leica Microsystems). Investigators were blind to the experimental and a scale factor of 10,000. Variable genes were identified using the
condition of the slide when taking images, and at least three mice per FindVariableFeatures function. When scRNA-seq datasets from dif-
group were assessed. fering run dates were merged, batch correction was performed using
IHC and IF images were quantified using Fiji (102). To quantify the IntegrateData workflow in Seurat (105). All genes were scaled and
% positive staining per field of view (FOV), three to eight images per centered using ScaleData. Principal component analysis (PCA) was
slide (depending on the size of the tissue) were taken on the Olym- performed using RunPCA. Cell clusters were identified using Find-
pus system at 20× magnification with care to capture only tissue Neighbors and FindClusters with dimensions that captured ∼90%
area within the FOV. The fraction of stained area for each image was variance as defined by the PCA result. UMAP clustering was complet-
averaged within each mouse, and the resulting % positive scores are ed using RunUMAP. To compare gene expression profiles between
reported in the text (each datapoint is a single mouse). To quanti- groups, we used FindMarkers and Wilcoxon rank sum test with
fy the % positive cells, E-Cadherin+ or PDGFRα/β+ cells (∼100–150 Bonferroni correction. In addition to Seurat, the R package scCus-
cells each per FOV) or CD4 cells (all cells per FOV) were manually tomize version 1.1.1 (RRID:SCR_024675) was used for visualization.
categorized as Ki67/Foxp3 positive or negative; at least three images Pathway analysis was performed with the R package fgsea version
per mouse were quantified, and total % positive scores were averaged 1.26.0 (bioRxiv 060012) and the MSigDB mouse-ortholog hallmark
within each mouse and represented in the figure. To quantify the gene sets were downloaded using the R package msigdbr version 7.5.1
fluorescence intensity of IL33 and/or pSTAT3 within fibroblasts, (RRID:SCR_022870). Pathway enrichment scores were also calculated
and then, the following day, washed with PBS and switched to RNA Extraction and RT-qPCR
doxycycline-deficient low-serum DMEM (for “KRASG12D OFF” CM) RNA was extracted using the RNeasy Plus Mini Kit (#74134,
or doxycycline-proficient low-serum DMEM (for “KRASG12D ON” QIAGEN) following the manufacturer’s protocol. RNA levels and
CM; d2). On d3, cells were again washed and replenished with low- quality were assessed via nanodrop, and cDNA was generated using
serum DMEM (for “KRASG12D OFF” CM), low-serum DMEM + doxy- the High-capacity cDNA Reverse Transcription Kit (#4368814, Ap-
cycline (for “KRASG12D ON” CM), or low-serum DMEM + doxycycline + plied Biosystems) supplemented with RNAse Inhibitor (#N8080119,
small molecule inhibitor: MRTX1133 (0.5 μmol/L, #HY134813, Applied Biosystems). Samples were prepared for qPCR using Fast
MedChemExpress), Sotorasib (0.5 μmol/L, #HY114277, MedChe- SYBR Green Master Mix (#4385612, Applied Biosystems). Primers are
mExpress), or Trametinib (50 nmol/L, #S2673, Selleck Chemicals). listed in the Supplementary Table S2. Reactions were run on a Quant-
On d4, inhibitors were again spiked into the media at equivalent con- Studio 6 Pro (Applied Biosystems). Cyclophilin A (Ppia) and GAPDH
centrations to ensure continued inhibition of KRAS. Tumor CM was were used as the housekeeping genes in all mouse and human
collected as described above on d5. DMSO was used as a vehicle con- RT-qPCR experiments, respectively.
trol for the untreated conditions.
CD1WT cells were plated at 3 × 105 cells per well in six-well dishes
Bulk RNA Sequencing
with DMEM + 10% FBS + 1% PS. The next day, cells were washed with
PBS and treated with 2 mL of fresh, low-serum media (DMEM + 1% Extracted RNA was submitted to the University of Michigan
grants from NIH during the conduct of the study. J. Shi reports Received January 30, 2024; revised May 18, 2024; accepted July 1,
grants from NIH during the conduct of the study. E.S. Carpenter re- 2024; published first July 3, 2024.
ports grants from Cornerstone Pharmaceutical and nonfinancial sup-
port from Beigene outside the submitted work. No disclosures were
reported by the other authors. References
1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer
Acknowledgments J Clin 2024;74:12–49.
2. Sarantis P, Koustas E, Papadimitropoulou A, Papavassiliou AG,
The authors would like to thank Dr. Gregory Beatty (University
Karamouzis MV. Pancreatic ductal adenocarcinoma: treatment hur-
of Pennsylvania) for generously sharing the murine pancreatic cancer dles, tumor microenvironment and immunotherapy. World J Gas-
cell line 7940b. We would like to thank Dr. Nicholas Lukacs (Uni- trointest Oncol 2020;12:173–81.
versity of Michigan) for providing the Il1rl1−/− mouse model. The 3. Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J,
Ptf1a-Cre mouse was generously gifted by Dr. Chris Wright (Vander- et al. Whole-exome sequencing of pancreatic cancer defines genetic
bilt University). We thank Lee Olsen for her editing services as well diversity and therapeutic targets. Nat Commun 2015;6:6744.
as Michael Mattea and Christopher Strayhorn for providing his- 4. Buscail L, Bournet B, Cordelier P. Role of oncogenic KRAS in the
topathology support. We thank Matthew Felten for his assistance diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev
21. Sherman MH, Magliano MPD. Cancer-associated fibroblasts: les- 42. Steele NG, Carpenter ES, Kemp SB, Sirihorachai VR, The S,
sons from pancreatic cancer. Annu Rev Cancer Biol 2023;7:43–55. Delrosario L, et al. Multimodal mapping of the tumor and periph-
22. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL1- eral blood immune landscape in human pancreatic cancer. Nat
Induced JAK/STAT signaling is antagonized by TGFβ to shape CAF Cancer 2020;1:1097–112.
heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov 43. Hosein AN, Huang H, Wang Z, Parmar K, Du W, Huang J, et al. Cel-
2019;9:282–301. lular heterogeneity during mouse pancreatic ductal adenocarcinoma
23. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, progression at single-cell resolution. JCI Insight 2019;5:e129212.
et al. Cross-species single-cell analysis of pancreatic ductal adeno- 44. K emp SB, Carpenter ES, Steele NG, Donahue KL, Nwosu ZC,
carcinoma reveals antigen-presenting cancer-associated fibroblasts. Pacheco A, et al. Apolipoprotein E promotes immune suppression in
Cancer Discov 2019;9:1102–23. pancreatic cancer through NF-κB-Mediated production of CXCL1.
24. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Cancer Res 2021;81:4305–18.
et al. Targeting CXCL12 from FAP-expressing carcinoma-associated 45. Du W, Menjivar RE, Donahue KL, Kadiyala P, Velez-Delgado A,
fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic Brown KL, et al. WNT signaling in the tumor microenvironment
cancer. Proc Natl Acad Sci U S A 2013;110:20212–7. promotes immunosuppression in murine pancreatic cancer. J Exp
25. Zhang Y, Yan W, Collins MA, Bednar F, Rakshit S, Zetter BR, et al. Med 2023;220:e20220503.
Interleukin-6 is required for pancreatic cancer progression by pro- 46. Jin G, Hong W, Guo Y, Bai Y, Chen B. Molecular mechanism of pan-
62. Li A, Herbst RH, Canner D, Schenkel JM, Smith OC, Kim JY, et al. 82. Peñuelas S, Anido J, Prieto-Sánchez RM, Folch G, Barba I, Cuartas I,
IL-33 signaling alters regulatory T cell diversity in support of tumor et al. TGF-beta increases glioma-initiating cell self-renewal through
development. Cell Rep 2019;29:2998–3008.e8. the induction of LIF in human glioblastoma. Cancer Cell 2009;
63. Mucciolo G, Araos Henríquez J, Jihad M, Pinto Teles S, Manansala JS, 15:315–27.
Li W, et al. EGFR-activated myofibroblasts promote metastasis of 83. Zhang F, Yan Y, Cao X, Guo C, Wang K, Lv S. TGF-β-driven LIF ex-
pancreatic cancer. Cancer Cell 2024;42:101–18.e11. pression influences neutrophil extracellular traps (NETs) and con-
64. Ali S, Mohs A, Thomas M, Klare J, Ross R, Schmitz ML, et al. The tributes to peritoneal metastasis in gastric cancer. Cell Death Dis
dual function cytokine IL-33 interacts with the transcription factor 2024;15:218.
NF-κB to dampen NF-κB–Stimulated gene transcription. J Immunol 84. Shi Y, Gao W, Lytle NK, Huang P, Yuan X, Dann AM, et al. Target-
2011;187:1609–16. ing LIF-mediated paracrine interaction for pancreatic cancer therapy
65. Park JH, Ameri AH, Dempsey KE, Conrad DN, Kem M, Mino- and monitoring. Nature 2019;569:131–5.
Kenudson M, et al. Nuclear IL-33/SMAD signaling axis promotes 85. Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, Maitra A. Pancre-
cancer development in chronic inflammation. EMBO J 2021;40: atic cancer: advances and challenges. Cell 2023;186:1729–54.
e106151. 86. Helms E, Onate MK, Sherman MH. Fibroblast heterogeneity in
66. Gautier V, Cayrol C, Farache D, Roga S, Monsarrat B, Burlet-Schiltz O, the pancreatic tumor microenvironment. Cancer Discov 2020;10:
et al. Extracellular IL-33 cytokine, but not endogenous nuclear IL-33, 648–56.
104. Hao Y, Hao S, Andersen-Nissen E, Mauck WM III, Zheng S, Butler A, 108. Miyoshi H, Stappenbeck TS. In vitro expansion and genetic modifi-
et al. Integrated analysis of multimodal single-cell data. Cell 2021; cation of gastrointestinal stem cells in spheroid culture. Nat Protoc
184:3573–87.e29. 2013;8:2471–82.
105. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck 109. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, et al.
WM III, et al. Comprehensive integration of single-cell data. Cell Organoid models of human and mouse ductal pancreatic cancer.
2019;177:1888–902.e21. Cell 2015;160:324–38.
106. Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan C-H, 110. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S,
et al. Inference and analysis of cell-cell communication using Cell- et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics
Chat. Nat Commun 2021;12:1088. 2013;29:15–21.
107. Carpenter ES, Kadiyala P, Elhossiny AM, Kemp SB, Li J, Steele NG, 111. Li B, Dewey CN. RSEM: accurate transcript quantification from
et al. KRT17high/CXCL8+ tumor cells display both classical RNA-Seq data with or without a reference genome. BMC Bioinfor-
and basal features and regulate myeloid infiltration in the pan- matics 2011;12:323.
creatic cancer microenvironment. Clin Cancer Res 2024;30:2497– 112. Love MI, Huber W, Anders S. Moderated estimation of fold change and
513. dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.