tumor-associated fibroblasts predominantly come …tumor-associated fibroblasts predominantly come...

6
Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3 , Elizabeth M. Hyjek a , Maria-Luisa Alegre b,c , Ying Wang b,c , Vytautas P. Bindokas d , Ralph R. Weichselbaum e,f , and Hans Schreiber a,b,g a Department of Pathology, The University of Chicago, Chicago, IL 60637; b Committee on Immunology, The University of Chicago, Chicago, IL 60637; c Section of Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL 60637; d Integrated Microscopy Core, Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, IL 60637; e The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637; f Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637; and g Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637 Edited by Douglas T. Fearon, University of Cambridge, Cambridge, United Kingdom, and approved May 16, 2016 (received for review January 12, 2016) Fibroblasts are common cell types in cancer stroma and lay down collagen required for survival and growth of cancer cells. Although some cancer therapy strategies target tumor fibroblasts, their origin remains controversial. Multiple publications suggest circulating mesenchymal precursors as a source of tumor-associated fibro- blasts. However, we show by three independent approaches that tumor fibroblasts derive primarily from local, sessile precursors. First, transplantable tumors developing in a mouse expressing green fluorescent reporter protein (EGFP) under control of the type I collagen (Col-I) promoter (COL-EGFP) had green stroma, whereas we could not find COL-EGFP + cells in tumors developing in the parabiotic partner lacking the fluorescent reporter. Lack of incorpo- ration of COL-EGFP + cells from the circulation into tumors was con- firmed in parabiotic pairs of COL-EGFP mice and transgenic mice developing autochthonous intestinal adenomas. Second, transplant- able tumors developing in chimeric mice reconstituted with bone mar- row cells from COL-EGFP mice very rarely showed stromal fibroblasts expressing EGFP. Finally, cancer cells injected under full-thickness COL- EGFP skin grafts transplanted in nonreporter mice developed into tumors containing green stromal cells. Using multicolor in vivo con- focal microscopy, we found that Col-Iexpressing fibroblasts con- stituted approximately one-third of the stromal mass and formed a continuous sheet wrapping the tumor vessels. In summary, tu- mors form their fibroblastic stroma predominantly from precursors present in the local tumor microenvironment, whereas the contribu- tion of bone marrow-derived circulating precursors is rare. stroma | origin | bone marrow | collagen | mesenchymal T umor-associated fibroblasts (TAFs) are a prominent cellular component of tumors, often assumed to be the most abun- dant cell type in tumor stroma, and there is an interest in TAFs as a therapeutic target in cancer. However, therapeutic interven- tions are limited by the lack of truly TAF-specific targets and un- certainty about their origins. If TAFs were predominantly recruited from circulating bone marrow (BM)-derived precursors, only sys- temic but not local therapies would prevent the recruitment of TAF precursors into tumors to suppress their growth. It is generally thought that at least a fraction of the fibroblasts in tumor stroma is derived from circulating BM-derived progenitors/stem cells. The idea that mononuclear blood cells could differentiate into fibro- blasts was suggested already in 1892 (13). A century later, several reports indicated that circulating CD14 + monocytes or CD45 + CD34 + collagen I + fibrocytes(4) could be progenitors for fibroblasts in vitro (57) or in vivo (8). Other recent reports also highlight the derivation of TAFs from circulating BM-derived progenitors of either hematopoietic or mesenchymal lineage (912). If TAFs were predominantly recruited from local sources, site- specific differences could be exploited to develop much more selec- tive therapeutics to target TAFs with fewer systemic side effects. It is increasingly clear that fibroblasts from different tissues can consti- tute well-specialized, tissue-specific populations. Fibroblasts from different anatomic sites have positional memory, and differ in transcriptional expression (1315), responsiveness to local stim- uli [e.g., steroid hormones (16)], and their capacity to inhibit cancer cell growth (17), which could well be linked to the organotropism of metastases (18, 19) and could represent a mechanistic basis for the seed and soilhypothesis (20). We therefore examined in this study the relative contributions of circulating cells and locally derived fi- broblasts to the development of tumor stroma. Fibroblasts are the most common cell type producing collagen, and over 90% of collagen in the body is type I (21). Genes for type I collagen (Col-I) are only active in mesenchymal cell types such as osteoblasts, odontoblasts, and, most relevant for this study, fi- broblasts and circulating mesenchymal progenitors (3, 22). We therefore chose expression of an EGFP reporter driven by the collagen α1(I) promoters/enhancers to unambiguously mark fibro- blasts (23). α-Smooth muscle actin (αSMA) does not exclusively mark fibroblasts, but can be up-regulated on activated fibroblasts (called myofibroblasts) in conditions involving active ECM deposition such as fibrosis, wound healing, and cancer (2426). Thus, we also studied the origin of αSMA + cells using the αSMA- Discosoma sp. Red (SMA-DsRed) mouse model (23). We found that Col-I + and αSMA + TAFs derive predominantly from local sources, whereas finding TAFs derived from circulating progenitors is a very rare event. Significance Fibroblasts constitute an important element of tumors and have received considerable attention in recent years due to their tu- mor-promoting and immunosuppressive properties. As a conse- quence, tumor-associated fibroblasts (TAFs) are considered an attractive target for cancer therapies. However, their origin re- mains controversial, with some evidence pointing at a local origin, whereas many publications suggest a significant contribution of progenitors from bone marrow. We found that TAFs derive al- most exclusively from local sources. Therefore, therapeutic strat- egies to target fibroblasts must exploit local recruitment and the unique transcriptional and response patterns of fibroblasts from different sites. Author contributions: A.A., M.-L.A., and H.S. designed research; A.A., C.I., E.M.H., and Y.W. performed research; R.R.W. and H.S. supervised research; E.M.H. and V.P.B. contributed new reagents/analytic tools; A.A. and V.P.B. analyzed data; and A.A., R.R.W., and H.S. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. Data deposition: Imaging data reported in this paper have been deposited in The Cell Image Library www.cellimagelibrary.org/. 1 To whom correspondence should be addressed. Email: [email protected]. 2 Present address: The Ludwig Center for Metastasis Research and Department of Radia- tion and Cellular Oncology, The University of Chicago, Chicago, IL 60637. 3 Present address: Department of Otorhinolaryngology, University Hospital of Schleswig- Holstein, 23562 Luebeck, Germany. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1600363113/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1600363113 PNAS | July 5, 2016 | vol. 113 | no. 27 | 75517556 CELL BIOLOGY Downloaded by guest on June 23, 2020

Upload: others

Post on 16-Jun-2020

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

Tumor-associated fibroblasts predominantly come fromlocal and not circulating precursorsAinhoa Arinaa,b,1,2, Christian Idela,b,3, Elizabeth M. Hyjeka, Maria-Luisa Alegreb,c, Ying Wangb,c, Vytautas P. Bindokasd,Ralph R. Weichselbaume,f, and Hans Schreibera,b,g

aDepartment of Pathology, The University of Chicago, Chicago, IL 60637; bCommittee on Immunology, The University of Chicago, Chicago, IL 60637; cSectionof Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL 60637; dIntegrated Microscopy Core, Department of Neurobiology,Pharmacology and Physiology, The University of Chicago, Chicago, IL 60637; eThe Ludwig Center for Metastasis Research, The University of Chicago,Chicago, IL 60637; fDepartment of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637; and gCommittee on Cancer Biology, TheUniversity of Chicago, Chicago, IL 60637

Edited by Douglas T. Fearon, University of Cambridge, Cambridge, United Kingdom, and approved May 16, 2016 (received for review January 12, 2016)

Fibroblasts are common cell types in cancer stroma and lay downcollagen required for survival and growth of cancer cells. Althoughsome cancer therapy strategies target tumor fibroblasts, their originremains controversial. Multiple publications suggest circulatingmesenchymal precursors as a source of tumor-associated fibro-blasts. However, we show by three independent approaches thattumor fibroblasts derive primarily from local, sessile precursors. First,transplantable tumors developing in a mouse expressing greenfluorescent reporter protein (EGFP) under control of the type Icollagen (Col-I) promoter (COL-EGFP) had green stroma, whereaswe could not find COL-EGFP+ cells in tumors developing in theparabiotic partner lacking the fluorescent reporter. Lack of incorpo-ration of COL-EGFP+ cells from the circulation into tumors was con-firmed in parabiotic pairs of COL-EGFP mice and transgenic micedeveloping autochthonous intestinal adenomas. Second, transplant-able tumors developing in chimeric mice reconstituted with bone mar-row cells from COL-EGFP mice very rarely showed stromal fibroblastsexpressing EGFP. Finally, cancer cells injected under full-thickness COL-EGFP skin grafts transplanted in nonreporter mice developed intotumors containing green stromal cells. Using multicolor in vivo con-focal microscopy, we found that Col-I–expressing fibroblasts con-stituted approximately one-third of the stromal mass and formeda continuous sheet wrapping the tumor vessels. In summary, tu-mors form their fibroblastic stroma predominantly from precursorspresent in the local tumor microenvironment, whereas the contribu-tion of bone marrow-derived circulating precursors is rare.

stroma | origin | bone marrow | collagen | mesenchymal

Tumor-associated fibroblasts (TAFs) are a prominent cellularcomponent of tumors, often assumed to be the most abun-

dant cell type in tumor stroma, and there is an interest in TAFsas a therapeutic target in cancer. However, therapeutic interven-tions are limited by the lack of truly TAF-specific targets and un-certainty about their origins. If TAFs were predominantly recruitedfrom circulating bone marrow (BM)-derived precursors, only sys-temic but not local therapies would prevent the recruitment of TAFprecursors into tumors to suppress their growth. It is generallythought that at least a fraction of the fibroblasts in tumor stroma isderived from circulating BM-derived progenitors/stem cells. Theidea that mononuclear blood cells could differentiate into fibro-blasts was suggested already in 1892 (1–3). A century later,several reports indicated that circulating CD14+ monocytes orCD45+CD34+ collagen I+ “fibrocytes” (4) could be progenitors forfibroblasts in vitro (5–7) or in vivo (8). Other recent reports alsohighlight the derivation of TAFs from circulating BM-derivedprogenitors of either hematopoietic or mesenchymal lineage (9–12). If TAFs were predominantly recruited from local sources, site-specific differences could be exploited to develop much more selec-tive therapeutics to target TAFs with fewer systemic side effects. It isincreasingly clear that fibroblasts from different tissues can consti-tute well-specialized, tissue-specific populations. Fibroblasts fromdifferent anatomic sites have positional memory, and differ in

transcriptional expression (13–15), responsiveness to local stim-uli [e.g., steroid hormones (16)], and their capacity to inhibit cancercell growth (17), which could well be linked to the organotropism ofmetastases (18, 19) and could represent a mechanistic basis for the“seed and soil” hypothesis (20). We therefore examined in this studythe relative contributions of circulating cells and locally derived fi-broblasts to the development of tumor stroma.Fibroblasts are the most common cell type producing collagen,

and over 90% of collagen in the body is type I (21). Genes for type Icollagen (Col-I) are only active in mesenchymal cell types suchas osteoblasts, odontoblasts, and, most relevant for this study, fi-broblasts and circulating mesenchymal progenitors (3, 22). Wetherefore chose expression of an EGFP reporter driven by thecollagen α1(I) promoters/enhancers to unambiguously mark fibro-blasts (23). α-Smooth muscle actin (αSMA) does not exclusivelymark fibroblasts, but can be up-regulated on activated fibroblasts(called myofibroblasts) in conditions involving active ECMdeposition such as fibrosis, wound healing, and cancer (24–26).Thus, we also studied the origin of αSMA+ cells using the αSMA-Discosoma sp. Red (SMA-DsRed) mouse model (23). We foundthat Col-I+ and αSMA+ TAFs derive predominantly from localsources, whereas finding TAFs derived from circulating progenitorsis a very rare event.

Significance

Fibroblasts constitute an important element of tumors and havereceived considerable attention in recent years due to their tu-mor-promoting and immunosuppressive properties. As a conse-quence, tumor-associated fibroblasts (TAFs) are considered anattractive target for cancer therapies. However, their origin re-mains controversial, with some evidence pointing at a local origin,whereas many publications suggest a significant contribution ofprogenitors from bone marrow. We found that TAFs derive al-most exclusively from local sources. Therefore, therapeutic strat-egies to target fibroblasts must exploit local recruitment and theunique transcriptional and response patterns of fibroblasts fromdifferent sites.

Author contributions: A.A., M.-L.A., and H.S. designed research; A.A., C.I., E.M.H., and Y.W.performed research; R.R.W. and H.S. supervised research; E.M.H. and V.P.B. contributed newreagents/analytic tools; A.A. and V.P.B. analyzed data; andA.A., R.R.W., andH.S. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Data deposition: Imaging data reported in this paper have been deposited in The CellImage Library www.cellimagelibrary.org/.1To whom correspondence should be addressed. Email: [email protected] address: The Ludwig Center for Metastasis Research and Department of Radia-tion and Cellular Oncology, The University of Chicago, Chicago, IL 60637.

3Present address: Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, 23562 Luebeck, Germany.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1600363113/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1600363113 PNAS | July 5, 2016 | vol. 113 | no. 27 | 7551–7556

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

June

23,

202

0

Page 2: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

ResultsTumor Growth Is Associated with the Appearance of αSMA and theContinuous Presence of Collagen α1(I)-Expressing Fibroblasts. Tostudy the distribution of TAFs during tumor development, wegenerated dual COL-EGFP/SMA-DsRed reporter mice (23) (Fig.S1) and implanted dorsal skinfold windows and cancer cells for lon-gitudinal analysis of tumors (27). As can be seen in Fig. 1A, healthyskin (day 0) in dual reporter mice shows normal dermal fibroblastsexpressing Col-I (COL-EGFP+), whereas the expression of αSMA israre. After cancer cell inoculation, SMA-DsRed+ cells appearedaround days 5–8 and became significantly more abundant arounddays 11–15 (Fig. 1B, Fig. S2, and Table S1). Expression of COL-EGFP showed an increasing trend between the same time points,although it was not statistically significant (Fig. 1B). The appear-ance of SMA-DsRed+ cells coincided with the development of tu-mor vasculature (Fig. S2). Several fibrosarcoma (MC57, Pro4L,and 8101Pro) and one carcinoma (MC38 colon cancer) celllines growing in immunodeficient or immunocompetent micerendered similar results (Table S1).

To determine whether αSMA and Col-I were expressed by thesame or different cells, colocalization studies were performed inconfocal high-magnification images (20×) from established tumors(Fig. S3). More than half of the cells that were positive for Col-Ialso expressed αSMA and vice versa at days 14–22 of tumor growth(average Mander’s coefficients varied between 0.56 ± 0.04 and0.77 ± 0.29 for COL-EGFP and 0.56 ± 0.1 and 0.83 ± 0.21 forSMA-DsRed). Therefore, a majority of cells were positive for bothmarkers, but there was always a fraction of cells that expressed onlyone or the other.

Tumor-Associated Fibroblasts Derive Predominantly from Local, andNot Circulating, Precursors. To determine the origin of TAFs, twotypes of BM chimeric mice were generated from COL-EGFP orSMA-DsRed transgenic mice, in which either only the BM or onlythe lethally irradiated host had the potential to generate fluorescentlylabeled fibroblasts. In four independent experiments using fourtumor models, TAFs were visible during the first weeks followingtumor implantation exclusively in the tumors of the chimerasexpressing the transgene in the non-BM (sessile) compartment(Fig. 2 A, D, and F and Fig. S4). After 2–4 wk of tumor growth,only a few BM-derived TAFs were observed in the chimeras thathad been reconstituted with BM from COL-EGFP transgenic mice(Fig. 2 B, C, and F and Fig. S4). No BM-derived SMA-DsRed+ cellswere found in mice reconstituted with BM from SMA-DsRedtransgenic mice in two independent experiments (Fig. S5).Because the BM chimera model could be limited by an incom-

plete deletion of mesenchymal stromal cells and engraftment of thenewly transferred mesenchymal progenitors in the BM of irradiatedmice (28), we generated parabiotic mouse pairs (Fig. 3 A and B). Insuch pairs, one mouse was COL-EGFP transgenic and the othermouse expressed DsRed ubiquitously under the chicken beta-actinpromoter. Around 2 wk after the surgery, the presence of similarpercentages of DsRed+ cells in both animals indicated that circu-lation was shared (Fig. S6). Cancer cells were injected into the distalflanks in both mice. No COL-EGFP+ cells could be found in thetumors grown in the DsRed mice parabiosed to COL-EGFP mice(Fig. 3 A and B). These data in parabiotic mice prove that thecollagen α1-expressing TAFs are derived from local sessile pre-cursors. To further investigate the local origin of TAFs, we madeuse of DsRed mice transplanted with full-thickness skin grafts fromCOL-EGFP mice (Fig. 3C). Upon s.c. injection of cancer cellsunderneath the grafts, abundant COL-EGFP+ cells were detectedin freshly explanted sections of the tumors. We performed twoexperiments, one of which is shown in Fig. 3B. In five of seven totalmice from two independent experiments, COL-EGFP+ cells couldbe detected in tumor sections cut 2.5–3 mm below the graft surfaceand, in three of seven mice, some COL-EGFP+ cells could be foundeven 4–6 mm from the surface. COL-EGFP+ cells were found inthe tumors when cancer cells had been injected either under thecenter of the grafted skin (4/5 total mice) or into its margin (2/2mice). Therefore, EGFP+ TAFs had derived from local precursorspresent in the original COL-EGFP skin grafts.

Circulating Cells Do Not Contribute Significantly to TAFs in ParabiosedMice Developing Autochthonous Intestinal Tumors. To confirm ourresults from transplantable models, we parabiosed fibroblast reportermice (COL-EGFP or SMA-DsRed) with ApcMin (Apc, adenomatosispolyposis coli; Min, multiple intestinal neoplasia) transgenic micethat develop intestinal adenomas starting at puberty (29). This tumormodel recreates more closely some aspects of the process of tu-morigenesis in humans because tumors are autochthonous. For theCOL-EGFP/ApcMin pairs, as can be seen in Fig. 4 A and C, Col-Icould readily be detected by immunofluorescence (IF) in the in-testine from the COL-EGFP mouse and the ApcMin mouse; how-ever, EGFP could exclusively be found in the COL-EGFP mousesample. Similarly, no DsRed+, but many αSMA+, cells could bedetected in the polyps from an ApcMin mouse parabiosed with

Day

0

Day

12

SMA-DsRed

A

B

500 m

500 m

COL-EGFP

COL-EGFP Cerulean

SMA-DsRedCerulean

100 m

COL-EGFP SMA-DsRedCerulean

2010

86420

5-8 11-15

Are

a (x

105

m2 )

Days after tumor cell inoculation

Dual reporter mouse COL-EGFP/SMA-DsRed

Cerulean

SMA-DsRedCOL-EGFP

Fig. 1. Tumor growth is associated with the appearance of αSMA and thecontinuous presence of collagen α1(I)-expressing fibroblasts. (A) Expressionof COL-EGFP and SMA-DsRed on healthy skin (day 0) and 12 d after MC57-Cerulean tumor cells were injected in a dual reporter COL-EGFP/SMA-DsRedRag−/− mouse bearing a dorsal skinfold window chamber. Dotted lines in-dicate window and tumor boundary. (B) Quantification of the area (meanand SD) occupied by COL-EGFP+, SMA-DsRed+, and Cerulean+ (cancer) cells inimages obtained from tumors developing in dual reporter mice. Data werepooled from five independent experiments using COL-EGFP/SMA-DsRedRag−/− mice injected with MC57, Pro4L, and 8101Pro fibrosarcomas (n = 1mouse each) and COL-EGFP/SMA-DsRed Rag+/+ mice injected with MC38colon carcinoma and 8101Pro fibrosarcoma (n = 1 each); all cell linesexpressed Cerulean. Two to eight tumor regions were averaged per mouseand time point before pooling the data from individual mice. *P = 0.031.

7552 | www.pnas.org/cgi/doi/10.1073/pnas.1600363113 Arina et al.

Dow

nloa

ded

by g

uest

on

June

23,

202

0

Page 3: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

an SMA-DsRed mouse for 10 wk (Fig. 4 B and D). Thoroughscanning of IF sections from ApcMin mice “Swiss gut rolls,” con-taining both normal gut and adenomatous tissue, showed that fi-broblasts were not recruited from circulation into either healthy ormalignant tissue (Fig. 4 C and D). Thus, our results with autoch-thonous tumors in the parabiosis model support the conclusion thatTAFs originate mainly from local sessile precursors.

TAFs Constitute One-Third of the Tumor Stroma. To determine thecontribution of each component to the total tumor mass by livemicroscopy image quantification, mice were used in which the fol-lowing compartments were color-coded: cancer cells (Cerulean),BM-derived stroma [enhanced yellow fluorescent protein (EYFP)],and non–BM-derived stroma (DsRed) including COL-EGFP+

type I collagen-expressing fibroblasts (DsRed and EGFP) (Figs.S1 and S7 and Movies S1 and S2). Using this approach, collagen-expressing fibroblasts constituted the third most prominent cell

type in four different tumors. Cancer cells and BM-derived stromalcells constituted most of the cellular volume, their ratio beingabout 1:1 (Fig. S7 A and D). The non–BM-derived cells that werenegative for COL-EGFP included cells lining the vessels (probablyendothelial cells) and most likely some COL-EGFP−αSMA+ fi-broblasts (Fig. S7 B and C and Movies S1 and S2).

TAFs Form Perivascular Structures. In the “multicolor” tumor modeldescribed above, many DsRed+COL-EGFP+ cells wrapped thevessels once the tumor was established (Fig. 5A and Movie S1).Consistently, 66–83% of the total COL-EGFP+ cells were foundin close proximity to, or in direct contact with, an endothelial cellby two-color immunohistochemistry (Fig. S8). In dual reporterCOL-EGFP/SMA-DsRed mice, COL-EGFP+ cells often formedtubular structures around vessels in well-established tumors (Fig. 5Band Fig. S2). In the same tumors, αSMA+ cells could also be foundnext to vessels (Fig. S2, day 11), although were less abundant at late

Day

1-1

3 D

ay 1

4-36

Host: COL-EGFP BM:DsRed

500 μm 500 μm

500 μm 500 μm

100 μm

Host:DsRed BM: COL-EGFP

MC57-Cerulean

MC57-Cerulean 8101Pro-Cerulean

COL-EGFP HostCOL-EGFP BM

MC57-Cerulean

Day

s af

ter t

umor

cel

l in

ocul

atio

n

0 0.2 10 20 Percent EGFP+ area

5

A D

B

C F

E

Fig. 2. Newly transplanted BM precursors do not significantly contribute to formTAFs. (A–E) Representative images from BM chimeric mice in which only the BM(A–C) or only the irradiated host (D and E) expressed the COL-EGFP transgeneand were injected with Cerulean-expressing tumor cells in dorsal windows. EGFP-expressing cells were almost exclusively observedwhen COL-EGFP was expressedin the irradiated host. A few cells derived from BM precursors could be ob-served only at late time points (yellow dotted line in C). The white dotted lineindicates tumor boundary. (F) Quantification of the area (mean and SD) oc-cupied by COL-EGFP+ cells in images from BM chimera experiments. Data havebeen pooled from four independent longitudinal experiments usingMC57 andPro4L cell lines growing in Rag−/− chimeras and 8101Pro and MC38 growing inRag+/+ chimeras (n = 1 mouse per cell line). Two to 11 tumor regions werepooled per mouse and time point before pooling the data from individualmice. *P = 0.029, **P = 0.01.

GFP

+ ar

ea

fract

ion

(%)

at 2

.5-3

mm

fro

m s

urfa

ce

2.5

mm

Excised tumor

COL-EGFP skin graft

Ds-Red host

Injection MC38 Cerulean

EGFP+ area fraction (%)

Full thickness skin grafts

Parabiosis

Mouse #1 # 2 #3 #4

MC57 MC38

EGFP+ area fraction (%)

A

B

C

Fig. 3. TAFs derive predominantly from local sessile precursors. (A) Parabioticpairs of mice were created where only one mouse expressed COL-EGFP and theother mouse expressed DsRed ubiquitously. Two pairs were created: pair 1 con-tained immunocompetent mice with MC38-Cerulean tumors; pair 2 had MC57-Cerulean tumors in Rag−/− mice. On each mouse of the parabiotic pairs, cancercells were injected into the distal flank. (A) Representative images are shownfrom the freshly explanted MC57 tumors from pair 2 at week 3 of tumor growth(n = 6–9 areas per tumor; mean and SD). (B) Quantification of the EGFP+ areafraction (mean and SD) in the tumors from parabiosed mice. **P = 0.0022,***P = 0.001. (C) Full-thickness skin grafts from COL-EGFP mice were trans-planted into four DsRed Rag−/− mice. One month later, MC38-Cerulean cancercells were injected s.c. into the center of the green skin grafts with the help of aUV lamp. At day 12 (mouse 1) or 25 (mice 2–4) of tumor growth, mice were killedand 1- to 2-mm-thick slices were cut from freshly explanted tumors at a distanceof 2.5–3mm from the surface. The area fraction occupied by COL-EGFP+ cells wasquantified in 4–17 optical regions per mouse (mean and SD). A second experi-ment in which cancer cells were injected 2 mo after skin graft transplantationgave similar results (n = 3 mice). N.D., nondetectable. (Scale bars, 50 μm.)

Arina et al. PNAS | July 5, 2016 | vol. 113 | no. 27 | 7553

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

June

23,

202

0

Page 4: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

stages (Fig. 5B and Fig. S2, MC38, day 25). These data suggest thatTAFs associate spatially and temporally with the development oftumor vasculature.

DiscussionIn contrast to other stromal cells [e.g., pericytes (30), macro-phages (31), and endothelial cells (32)], the origin of TAFs hasremained unresolved. Here we show that TAFs derive predominantly

from local sessile precursors and only rarely fromBM circulating cells.Recruitment of BM-derived circulating cells to the tumor wasuncompromised, as the majority of the stromal compartmentwas BM-derived.Three lines of experimentation support the main conclusion of

this study. (i) In BM chimeric mice, fibroblast reporter genes wereexpressed almost exclusively in cells derived from the non-BMcompartment. (ii) In parabiotic pairs of COL-EGFP and DsRedmice, DsRed cells were abundant in the tumor grown in the COL-EGFP transgenic partner, whereas COL-EGFP+ cells could not bedetected in the tumor grown in the DsRed mouse; similarly, noCOL-EGFP+ cells were found in autochthonous tumors fromApcMin mice parabiosed to COL-EGFP mice. (iii) COL-EGFP+

TAFs were found in tumors developing in skin grafts from COL-EGFP mice transplanted into nonreporter mice.

SMA-DsRed gut ApcMin polyp B

COL-EGFP gut ApcMin polyp A

C

SMA-DsRed/ApcMin parabiosis

COL-EGFP/ApcMin parabiosis

COL-EGFP Collagen DAPI

50 μm

SMA SMA-DsRed

DAPI

μm

50 μm

COL-EGFP Collagen

DAPI

50 μm

SMA SMA-DsRed

DAPI

aSMA:DAPIDsRed:DAPI

Rat

io

0

1

2 EGFP:DAPICollagen I:DAPI

Rat

io

0

1

2 D

50

Fig. 4. TAFs originating from circulating cells cannot be detected in au-tochthonous ApcMin intestinal adenomas. ApcMin mice that develop in-testinal adenomas were parabiosed with fibroblast reporter mice. Animalswere killed when the ApcMin mice became moribund, and intestinal sectionswere stained with antibodies against EGFP and Col-I (A and C) or DsRed andαSMA (B and D). Representative immunofluorescence images are shownfrom (A) a COL-EGFP/ApcMin pair parabiosed for 5 wk and (B) an SMA-DsRed/ApcMin pair parabiosed for 10 wk. Dotted lines indicate polyp boundary. Datafrom experiments shown in A and B are quantified in C and D, respectively, asthe ratio of (C) EGFP+ and collagen I+ or (D) DsRed+ and αSMA+ area to nucleararea (DAPI) in positive control sections from normal reporter mouse gut versussections of Swiss gut rolls containing both normal gut and polyps or onlyApcMin polyps from the ApcMin mouse (mean and SD). Quantitative data werederived using 5×magnification images from scanned IF slides, comprising mostof the sample (at least 8–11 5× regions per slide or its entirety). Data are fromthree ApcMin Swiss roll slides and five slides containing only polyps (C), threeApcMin Swiss roll slides, and one slide containing only polyps (D), plus onepositive control slide in each case. Two additional parabiosis experiments inthe COL-EGFP model showed consistent results. **P < 0.01, ***P < 0.001.

MC38 (day 25)

MC57 (day 22)

MC57 (day 22)

MC38 (day 12)

50 μm

50 μm

50 μm

50 μm

Non BM-DsRed COL-EGFP Merge Vasculature

SMA-DsRed COL-EGFP DiD-RBC Merge

A

B

Fig. 5. Col-I–expressing cells form perivascular structures. (A) COL-EGFP+ cellsare found in close association with tumor vessels. COL-EGFP+ structures wrap theDsRed+ vessels in MC57 fibrosarcoma and MC38 carcinoma growing in DsRed+

COL-EGFP+ chimeric mice with an EYFP+ BM. Representative images are shownfrom two out of three independent longitudinal experiments performed withMC57, MC38, and Pro4L-Cerulean tumors (n = 1 mouse each). COL-EGFP+ cellswrapping vessels were observed in normal (non-BM chimeric) COL-EGFP mice infive more independent experiments. (B) SMA-DsRed+ cells do not form tubularstructures around vessels in late-stage established tumors. In day 22–25 MC57andMC38 tumors grown in COL-EGFP/SMA-DsRed dual reporter mice, structuresthat are COL-EGFP+ but SMA-DsRed− wrap the vessels. DiD-labeled red bloodcells (DiD-RBC) were injected i.v. to visualize blood flow. Representative imagesare shown from two out of three independent longitudinal experiments per-formed using Cerulean-expressing MC57, Pro4L, and MC38 (n = 1 mouse each).The host was Rag−/− for the MC57 and Pro4L tumors.

7554 | www.pnas.org/cgi/doi/10.1073/pnas.1600363113 Arina et al.

Dow

nloa

ded

by g

uest

on

June

23,

202

0

Page 5: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

The process of recruitment and activation of fibroblasts ingrowing tumors is not well-understood. Leukocytes play an im-portant role in promoting skin wound healing by secreting IL-22,which activates extracellular matrix production and expression ofαSMA by dermal fibroblasts (33). Importantly, in wound healing,as well as in renal fibrosis, fibroblasts are also predominantlyderived from local tissues (22, 34–37). It is plausible that tumorstromal BM-derived cells could similarly secrete factors thatrecruit local fibroblast precursors and induce their activation.Consistently, we found an almost immediate (14-h) infiltration ofleukocytes into cancer cell inoculation sites (27) and a delay inthe expression of αSMA compared with the earlier observationof αSMA−COL-EGFP+ TAFs (Fig. 1 and Fig. S2). The close prox-imity of BM-derived stromal cells and fibroblasts in tumors (Fig. S7)would allow easy access of leukocyte-produced factors to neigh-boring cells. Alternatively, resident fibroblasts may convert intotumor-promoting TAFs through TGF-β– and SDF-1–mediatedautocrine signaling loops (38).The partial overlap between expression of αSMA+ and Col-

EGFP observed by us (Fig. S3) and others (23, 35), may reflectdifferent states of activation of the promoters. The expression ofCol-I seems to be more stable and probably detects most of thefibroblasts, whereas αSMA could be induced or decreased undercertain circumstances such as active tumor formation and/orneovascularization. Interestingly, many COL-EGFP+ cells in thetumor wrapped vessels, forming “sheaths.” This is somewhatsurprising, because Col-I is not generally considered a compo-nent of the vessel wall. This wrapping was not so evident withαSMA-expressing cells, maybe because of the more transientexpression of αSMA (Fig. 5B and Fig. S2). However, αSMA ex-pression began with tumor vascularization, therefore temporallycorrelating with the development of tumor vessels. These findingsare consistent with abundant in vitro experimental evidence show-ing the role of fibroblasts in endothelial cell tubulogenesis by pro-moting vessel sprouting and, especially, formation of intercellularlumens (reviewed in ref. 39). Endothelial cells also produce extra-cellular matrix metalloproteinase inducer (EMMPRIN), whichdrives induction of αSMA and activation of fibroblasts (40). Theproduction of ECM and VEGF by living fibroblasts in close asso-ciation with endothelial cells seems to be required for tubulogenesisto occur (41). Our conclusions are also consistent with the sug-gested local origin of pericytes in tumors (30). It is therefore pos-sible that some of the αSMA- and/or Col-I–expressing cells wedescribe are pericytes, although costaining with other markers (42)would be necessary to properly characterize them.Although we do not know the precise identity of the local

precursors of type I collagen- and αSMA-expressing cells, mes-enchymal progenitors (30) or mesenchymal stem cells (MSCs) arelikely candidates. MSCs are radioresistant (43, 44), reside inmultiple organs in perivascular locations (45), and have migratorycapability (45). It has even been speculated that all MSCs could bepericytes, although not all pericytes are MSCs (46). Therefore,MSCs, pericytes, and most fibroblasts in tumors may well repre-sent various states of differentiation of the same cellular lineage.Multiple factors contribute to the contradictory reports on the

origin of TAFs. First is the lack of specificity of most of themarkers used to identify TAFs (24). Second, there is someconfusion about the terminology referring to circulating mesen-chymal precursors. This terminology has sometimes been appliedto hematopoietic (nonmesenchymal) precursors that differenti-ate into cells with fibroblastoid morphology (47). It is thoughtthat the number of TAF precursors circulating in blood might beextremely low (47–49). Third, many studies highlight the BMcontribution to mesenchymal cells in tumors without a side-by-sidecomparison of the relative contributions of circulating versus localsources (10, 12, 50). In our hands, the BM contribution is minimalcompared with the contribution of the non-BM compartment. In-terestingly, we observed occasional BM-derived TAFs only at later

time points during tumor development and only in the BM chimerasetting, whereas we could not detect any BM-derived TAFs in theexperiments in parabiotic mice. We cannot exclude the possibilitythat under certain pathophysiological conditions, such as the injuryinduced by total-body irradiation, the contribution of circulatingprecursors to TAFs can be increased. Finally, identification of TAFsin most previous studies relied on morphology and immunofluo-rescence of tumor sections. This is reminiscent of the controversyinvolving the origin of endothelial cells in tumors, in that markercolocalization on a single cell in tumor sections can be easily mis-taken for two juxtaposing/overlapping cells, as shown previously forendothelial cells (32). The predominant origin of endothelial cellswas eventually shown to be local (32). Here, we reach the sameconclusion about TAFs through the direct observation of live orfreshly explanted tumors.We have shown previously that both BM and non-BM com-

partments of tumor stroma must be targeted for T cell-mediatedtumor eradication (51, 52). TAFs are also targets for other typesof cancer therapy in preclinical studies (53–57) and even inclinical trials (58–60) because of their role in tumor promotionand resistance to drug therapy (reviewed in ref. 26) and immu-nosuppression (61). A better understanding of the origins anddevelopment of TAFs is essential for successful targeting. Ourresults should not discourage strategies to target tumor stromasuch as genetic manipulation of mesenchymal stem cells. How-ever, this manipulation has to occur at the site of tumor growth.If mesenchymal stem cells were to be used as “Trojan horses”(62), we would need to learn to attract them efficiently from distantsources. Until this is achieved, local therapies such as radiotherapymay be more appropriate than systemic treatments to target theformation of mesenchymal stroma. Our findings should also en-courage the search for more selective targets for destroying orinhibiting TAFs, based on the unique transcriptional and responsepatterns of fibroblasts from different sites/organs of the body.

Experimental ProceduresCOL-EGFP (63) and SMA-DsRed (αSMA-RFP) mice (23) were a generousgift from D. A. Brenner (University of California, San Diego, La Jolla, CA).Tg(ACTB-DsRed*MST)1Nagy/J and 129-Tg(ACTB-EYFP)7AC5Nagy/J mice express-ing the red fluorescent protein variant DsRed.MST or EYFP gene, respectively,under the control of the chicken beta-actin promoter coupled with the cyto-megalovirus immediate-early enhancer were backcrossed to C57BL/6 mice for 20generations and then crossed to Rag-1 KOmice to obtain DsRed and EYFP Rag-1KO mice, respectively. All strains were purchased from The Jackson Laboratory.Mice were bred andmaintained in a specific pathogen-free barrier facility at TheUniversity of Chicago according to Institutional Animal Care and UseCommittee (IACUC) guidelines. All animal experiments were approvedby the IACUC of The University of Chicago. Methylcholanthrene-inducedMC57G fibrosarcoma was provided by P. Ohashi (University of Toronto,Toronto, Canada), with permission of H. Hengartner (University HospitalZurich, Zurich, Switzerland). The fibrosarcoma cell lines Pro4L and 8101Prowere induced by UV light in C3H/HeN and C57BL/6 mice, respectively (64, 65).MC38 colon adenocarcinoma was induced by s.c. injection of dimethylhy-drazine in C57BL/6 mice (66), and was generously provided by A. Schietingerand P. Greenberg (University of Washington, Seattle, WA). MC57, Pro4L,and 8101Pro fibrosarcomas and MC38 carcinoma were retrovirally infectedwith pMFG-Cerulean (27) and FACS-sorted to generate lines expressingthe fluorescent protein Cerulean. More methods are available in SIExperimental Procedures.

ACKNOWLEDGMENTS. We thank David A. Brenner for the generous dona-tion of the COL-EGFP and SMA-DsRed mice, and F. Gounari for the generousdonation of the ApcMin mice. We also thank Christine Labno, Shirley Bond,Rolando Torres, Dorothy Kane, Christian Friese, and Khashayarsha Khazaiefor expert assistance with technical protocols; Karin Schreiber for providinganimals for experiments; Sydeaka Watson from the University of ChicagoBiostatistics Core for help with statistical analysis; and Donald A. Rowley,Boris Engels, David Binder, and Douglas Fearon for productive discussions.This work was supported by National Institutes of Health Grants P01-CA97296, R01-CA22677, and R01-CA37516; the Berlin Institute of Healthand Einstein Foundation (H.S.); and the Ludwig Foundation (R.R.W.). A.A.had a fellowship from Fundación Alfonso Martín Escudero (Spain).

Arina et al. PNAS | July 5, 2016 | vol. 113 | no. 27 | 7555

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

June

23,

202

0

Page 6: Tumor-associated fibroblasts predominantly come …Tumor-associated fibroblasts predominantly come from local and not circulating precursors Ainhoa Arina a,b,1,2 , Christian Idel a,b,3

1. Metchnikoff E (1892) Leçons sur la pathologie comparée de l’inflammation. Faites àl’Institut Pasteur en Avril et Mai 1891 (Masson, Paris).

2. Maximov A (1928) Culture of blood leukocytes. From lymphocytes and monocytes toconnective tissue. Arch Exp Zellforsch 5:169–268.

3. Friedenstein A (1989) Stromal-hematopoietic interrelationships: Maximov’s ideas andmodern models. Haematol Blood Transfus 32:159–167.

4. Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A (1994) Circulating fibrocytesdefine a new leukocyte subpopulation that mediates tissue repair. Mol Med 1(1):71–81.

5. Labat ML, et al. (1991) Cystic fibrosis: Production of high levels of uromodulin-likeprotein by HLA-DR blood monocytes differentiating towards a fibroblastic pheno-type. Biomed Pharmacother 45(9):387–401.

6. Kuwana M, et al. (2003) Human circulating CD14+ monocytes as a source of pro-genitors that exhibit mesenchymal cell differentiation. J Leukoc Biol 74(5):833–845.

7. Abe R, Donnelly SC, Peng T, Bucala R, Metz CN (2001) Peripheral blood fibrocytes:Differentiation pathway and migration to wound sites. J Immunol 166(12):7556–7562.

8. Phillips RJ, et al. (2004) Circulating fibrocytes traffic to the lungs in response toCXCL12 and mediate fibrosis. J Clin Invest 114(3):438–446.

9. LaRue AC, et al. (2006) Hematopoietic origins of fibroblasts: I. In vivo studies of fi-broblasts associated with solid tumors. Exp Hematol 34(2):208–218.

10. Reddy K, Zhou Z, Schadler K, Jia SF, Kleinerman ES (2008) Bone marrow subsets dif-ferentiate into endothelial cells and pericytes contributing to Ewing’s tumor vessels.Mol Cancer Res 6(6):929–936.

11. Spaeth EL, et al. (2009) Mesenchymal stem cell transition to tumor-associated fibro-blasts contributes to fibrovascular network expansion and tumor progression. PLoSOne 4(4):e4992.

12. Quante M, et al. (2011) Bone marrow-derived myofibroblasts contribute to themesenchymal stem cell niche and promote tumor growth. Cancer Cell 19(2):257–272.

13. Chang HY, et al. (2002) Diversity, topographic differentiation, and positional memoryin human fibroblasts. Proc Natl Acad Sci USA 99(20):12877–12882.

14. Sági B, et al. (2012) Positional identity of murine mesenchymal stem cells resident indifferent organs is determined in the postsegmentation mesoderm. Stem Cells Dev21(5):814–828.

15. Higuchi Y, et al. (2015) Gastrointestinal fibroblasts have specialized, diverse tran-scriptional phenotypes: A comprehensive gene expression analysis of human fibro-blasts. PLoS One 10(6):e0129241.

16. Mukudai S, et al. (2015) Differential responses to steroid hormones in fibroblasts fromthe vocal fold, trachea, and esophagus. Endocrinology 156(3):1000–1009.

17. Flaberg E, et al. (2012) The architecture of fibroblast monolayers of different origindifferentially influences tumor cell growth. Int J Cancer 131(10):2274–2283.

18. Klein G (2014) Evolutionary aspects of cancer resistance. Semin Cancer Biol 25:10–14.19. Klein-Goldberg A, Maman S, Witz IP (2014) The role played by the microenvironment

in site-specific metastasis. Cancer Lett 352(1):54–58.20. Paget S (1889) Distribution of secondary growths in cancer of the breast. Lancet

133(3421):571–573.21. Cormack DH (2001) Essential Histology (Lippincott Williams & Wilkins, Philadelphia),

2nd Ed.22. Roufosse C, et al. (2006) Bone marrow-derived cells do not contribute significantly to

collagen I synthesis in a murine model of renal fibrosis. J Am Soc Nephrol 17(3):775–782.

23. Magness ST, Bataller R, Yang L, Brenner DA (2004) A dual reporter gene transgenicmouse demonstrates heterogeneity in hepatic fibrogenic cell populations. Hepatology40(5):1151–1159.

24. Kalluri R, Zeisberg M (2006) Fibroblasts in cancer. Nat Rev Cancer 6(5):392–401.25. De Wever O, Demetter P, Mareel M, Bracke M (2008) Stromal myofibroblasts are

drivers of invasive cancer growth. Int J Cancer 123(10):2229–2238.26. Polanska UM, Orimo A (2013) Carcinoma-associated fibroblasts: Non-neoplastic tu-

mour-promoting mesenchymal cells. J Cell Physiol 228(8):1651–1657.27. Schietinger A, et al. (2013) Longitudinal confocal microscopy imaging of solid tumor

destruction following adoptive T cell transfer. OncoImmunology 2(11):e26677.28. Morikawa S, et al. (2009) Prospective identification, isolation, and systemic trans-

plantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med206(11):2483–2496.

29. Moser AR, Pitot HC, Dove WF (1990) A dominant mutation that predisposes to mul-tiple intestinal neoplasia in the mouse. Science 247(4940):322–324.

30. De Palma M, et al. (2005) Tie2 identifies a hematopoietic lineage of proangiogenicmonocytes required for tumor vessel formation and a mesenchymal population ofpericyte progenitors. Cancer Cell 8(3):211–226.

31. Cortez-Retamozo V, et al. (2012) Origins of tumor-associated macrophages andneutrophils. Proc Natl Acad Sci USA 109(7):2491–2496.

32. Purhonen S, et al. (2008) Bone marrow-derived circulating endothelial precursors donot contribute to vascular endothelium and are not needed for tumor growth. ProcNatl Acad Sci USA 105(18):6620–6625.

33. McGee HM, et al. (2013) IL-22 promotes fibroblast-mediated wound repair in the skin.J Invest Dermatol 133(5):1321–1329.

34. Ross R, Everett NB, Tyler R (1970) Wound healing and collagen formation. VI. Theorigin of the wound fibroblast studied in parabiosis. J Cell Biol 44(3):645–654.

35. Lin SL, Kisseleva T, Brenner DA, Duffield JS (2008) Pericytes and perivascular fibro-blasts are the primary source of collagen-producing cells in obstructive fibrosis of thekidney. Am J Pathol 173(6):1617–1627.

36. Barisic-Dujmovic T, Boban I, Clark SH (2010) Fibroblasts/myofibroblasts that partici-pate in cutaneous wound healing are not derived from circulating progenitor cells.J Cell Physiol 222(3):703–712.

37. Rinkevich Y, et al. (2015) Skin fibrosis. Identification and isolation of a dermal lineagewith intrinsic fibrogenic potential. Science 348(6232):aaa2151.

38. Kojima Y, et al. (2010) Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1)signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts.Proc Natl Acad Sci USA 107(46):20009–20014.

39. Hughes CC (2008) Endothelial-stromal interactions in angiogenesis. Curr OpinHematol 15(3):204–209.

40. Huet E, et al. (2008) Extracellular matrix metalloproteinase inducer/CD147 promotesmyofibroblast differentiation by inducing alpha-smooth muscle actin expression andcollagen gel contraction: Implications in tissue remodeling. FASEB J 22(4):1144–1154.

41. Berthod F, Germain L, Tremblay N, Auger FA (2006) Extracellular matrix deposition byfibroblasts is necessary to promote capillary-like tube formation in vitro. J Cell Physiol207(2):491–498.

42. Morikawa S, et al. (2002) Abnormalities in pericytes on blood vessels and endothelialsprouts in tumors. Am J Pathol 160(3):985–1000.

43. Dickhut A, et al. (2005) Mesenchymal stem cells obtained after bone marrow trans-plantation or peripheral blood stem cell transplantation originate from host tissue.Ann Hematol 84(11):722–727.

44. Bartsch K, et al. (2009) Mesenchymal stem cells remain host-derived independent ofthe source of the stem-cell graft and conditioning regimen used. Transplantation87(2):217–221.

45. Crisan M, et al. (2008) A perivascular origin for mesenchymal stem cells in multiplehuman organs. Cell Stem Cell 3(3):301–313.

46. Caplan AI (2008) All MSCs are pericytes? Cell Stem Cell 3(3):229–230.47. He Q, Wan C, Li G (2007) Concise review: Multipotent mesenchymal stromal cells in

blood. Stem Cells 25(1):69–77.48. Wexler SA, et al. (2003) Adult bone marrow is a rich source of human mesenchymal

‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol121(2):368–374.

49. da Silva Meirelles L, Chagastelles PC, Nardi NB (2006) Mesenchymal stem cells reside invirtually all post-natal organs and tissues. J Cell Sci 119(Pt 11):2204–2213.

50. Direkze NC, et al. (2004) Bone marrow contribution to tumor-associated myofibro-blasts and fibroblasts. Cancer Res 64(23):8492–8495.

51. Spiotto MT, Rowley DA, Schreiber H (2004) Bystander elimination of antigen lossvariants in established tumors. Nat Med 10(3):294–298.

52. Zhang B, Karrison T, Rowley DA, Schreiber H (2008) IFN-gamma- and TNF-dependentbystander eradication of antigen-loss variants in established mouse cancers. J ClinInvest 118(4):1398–1404.

53. Schüler T, Körnig S, Blankenstein T (2003) Tumor rejection by modulation of tumorstromal fibroblasts. J Exp Med 198(10):1487–1493.

54. Ostermann E, et al. (2008) Effective immunoconjugate therapy in cancer modelstargeting a serine protease of tumor fibroblasts. Clin Cancer Res 14(14):4584–4592.

55. Huang S, et al. (2011) Evaluation of the tumor targeting of a FAPα-based doxorubicinprodrug. J Drug Target 19(7):487–496.

56. Olive KP, et al. (2009) Inhibition of Hedgehog signaling enhances delivery of che-motherapy in a mouse model of pancreatic cancer. Science 324(5933):1457–1461.

57. Provenzano PP, et al. (2012) Enzymatic targeting of the stroma ablates physicalbarriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21(3):418–429.

58. Hofheinz RD, et al. (2003) Stromal antigen targeting by a humanised monoclonalantibody: An early phase II trial of sibrotuzumab in patients with metastatic co-lorectal cancer. Onkologie 26(1):44–48.

59. Narra K, et al. (2007) Phase II trial of single agent Val-boroPro (talabostat) inhibitingfibroblast activation protein in patients with metastatic colorectal cancer. Cancer BiolTher 6(11):1691–1699.

60. Eager RM, et al. (2009) Phase II trial of talabostat and docetaxel in advanced non-smallcell lung cancer. Clin Oncol (R Coll Radiol) 21(6):464–472.

61. Kraman M, et al. (2010) Suppression of antitumor immunity by stromal cells expressingfibroblast activation protein-alpha. Science 330(6005):827–830.

62. Studeny M, et al. (2004) Mesenchymal stem cells: Potential precursors for tumorstroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst 96(21):1593–1603.

63. Yata Y, et al. (2003) DNase I-hypersensitive sites enhance alpha1(I) collagen geneexpression in hepatic stellate cells. Hepatology 37(2):267–276.

64. Singh S, Ross SR, Acena M, Rowley DA, Schreiber H (1992) Stroma is critical forpreventing or permitting immunological destruction of antigenic cancer cells. J ExpMed 175(1):139–146.

65. Dubey P, et al. (1997) The immunodominant antigen of an ultraviolet-induced re-gressor tumor is generated by a somatic point mutation in the DEAD box helicasep68. J Exp Med 185(4):695–705.

66. Rosenberg SA, et al. (1986) A new approach to the therapy of cancer based on thesystemic administration of autologous lymphokine-activated killer cells and recombinantinterleukin-2. Surgery 100(2):262–272.

67. Schindelin J, et al. (2012) Fiji: An open-source platform for biological-image analysis.Nat Methods 9(7):676–682.

68. Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL (2001) Physiological mi-gration of hematopoietic stem and progenitor cells. Science 294(5548):1933–1936.

7556 | www.pnas.org/cgi/doi/10.1073/pnas.1600363113 Arina et al.

Dow

nloa

ded

by g

uest

on

June

23,

202

0