cd13-positive bone marrow-derived myeloid cells promote ...€¦ · therapy, the university of...

13
CD13-positive bone marrow-derived myeloid cells promote angiogenesis, tumor growth, and metastasis Eleonora Dondossola a , Roberto Rangel a,b , Liliana Guzman-Rojas a,b , Elena M. Barbu a , Hitomi Hosoya a,c , Lisa S. St. John d , Jeffrey J. Molldrem d , Angelo Corti e , Richard L. Sidman f,g,1 , Wadih Arap a,1,2,3 , and Renata Pasqualini a,1,2,3 a David H. Koch Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; b Cancer Research Program, The Methodist Hospital Research Institute, Houston, TX 77030; c The US Naval Hospital Yokosuka, Fleet Post Ofce, AP, 96350-1600; d Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; e Division of Molecular Oncology, San Raffaele Scientic Institute, 20132 Milan, Italy; f Harvard Medical School, Boston, MA 02215; and g Department of Neurology, Beth Israel-Deaconess Medical Center, Boston, MA 02215 Contributed by Richard L. Sidman, November 9, 2013 (sent for review June 13, 2013) Angiogenesis is fundamental to tumorigenesis and an attractive target for therapeutic intervention against cancer. We have recently demonstrated that CD13 (aminopeptidase N) expressed by nonmalignant host cells of unspecied types regulate tumor blood vessel development. Here, we compare CD13 wild-type and null bone marrow-transplanted tumor-bearing mice to show that host CD13 + bone marrow-derived cells promote cancer progression via their ef- fect on angiogenesis. Furthermore, we have identied CD11b + CD13 + myeloid cells as the immune subpopulation directly regulating tumor blood vessel development. Finally, we show that these cells are spe- cically localized within the tumor microenvironment and produce proangiogenic soluble factors. Thus, CD11b + CD13 + myeloid cells con- stitute a population of bone marrow-derived cells that promote tu- mor progression and metastasis and are potential candidates for the development of targeted antiangiogenic drugs. mouse models | protease | stromal cells | vascular pericytes A ngiogenesis is a rate-limiting step in the development of many solid tumors, making it an attractive target for ther- apeutic intervention (1). Although pharmacologic modulation of angiogenesis has shown some clinical success, negative factors such as treatment-related plasticity, drug resistance, and tumor diversity have underscored the need for a better understanding of tumor-associated blood vessel formation at a mechanistic level (2). Tumor angiogenesis is a multifactorial process involving several different cell subtypes, especially tumor stromal endo- thelial cells, pericytes, carcinoma-associated broblasts (CAFs), and bone marrow-derived cells (BMDCs) (3, 4). A variety of immune cells directly support angiogenesis, including mast cells, tumor-associated macrophages (TAMs), and Tie2-expressing macrophages (TEMs) (57). Endothelial cells recruit in- ammatory cells to the extravascular tissue by the expression of different leukocyte adhesion molecules. In turn, immune cells produce soluble factors, such as chemokines, cytokines, and proteases, that inuence endothelial cell function and angiogenesis in a paracrine fashion (5). Other studies have shown that BMDCs have the ability to differentiate into en- dothelial cells, possibly by conversion rst to endothelial progenitors (810). Proteases activate growth factors and inhibit suppressive fac- tors within tumors and are central to the angiogenic process within the tumor microenvironment (11). Studies on the in- volvement of aminopeptidases in tumor progression and an- giogenesis have revealed a role for aminopeptidase N (CD13) expressed by stromal cells (12). Originally identied as a sur- face marker on myeloid cells (13), CD13 is a widely expressed membrane-bound metalloprotease involved in pleiotropic functions, including enzymatic cleavage of peptides, antigen presentation, and signal transduction that ultimately mediate downstream biological phenomena such as cell adhesion, proliferation, and motility (14). Diverse cell subpopulations (e.g., broblasts, pericytes, epithelial cells, tumor-initiating cells, and stem cells) express CD13 (15-19). Our group has shown that CD13 is a functional biomarker of angiogenesis (20) and is also expressed by tumor-associated blood vessels (21). Furthermore, by using CD13-null neonatal mice, we demonstrated an impaired angiogenic response in the oxygen-induced retinopathy-of-prematurity model, which quanti- es retinal pathological neovascularization in response to hypoxia (22). We have also shown that CD13-null mice displayed dose- dependent reduced tumor growth after implantation with either B16-F10 melanoma or Lewis lung carcinoma (LLC) cells, largely because of impaired angiogenesis (12). These data indicate that CD13 expressed by host stromal cells mediates a pathological angiogenic response, but the specic cell subtypes involved in this phenomenon have yet to be identied. Here, we unravel the type and role of CD13 + stromal cells in tumor progression. By using CD13 wild-type (WT) and null (KO) mice in the setting of paired bone marrow transplantations, we demonstrate that CD13 + BMDCs affect the progression of LLC, B16-F10 melanoma, and TSA mammary adenocarcinoma via their regulation of tumor angiogenesis. Notably, we show direct involvement of a CD11b + CD13 + myeloid subset of BMDCs in this process. Finally, we show that these cells produce angio- genic factors and specically localize within the tumor micro- environment. Our results indicate that CD11b + CD13 + cells represent a population of proangiogenic BMDCs and that CD13 is crucial for their activity. Signicance The progression of many solid tumors is associated with in- creased vascularization. We previously recognized involvement in tumor development and angiogenesis of tumor stromal cells expressing the CD13 protease aminopeptidase. The basic bi- ological concept of participation of nontumor cells in the cancer stroma microenvironment is strengthened in the present study by our nding that a CD11b + CD13 + myeloid subset of bone marrow-derived cells affects pericyte biology and angiogenesis and thereby inuences tumor growth and metastasis. Thera- peutic implications of the identication of specic CD11 + CD13 + myeloid bone marrow-derived cells as participants in the mechanism of tumor angiogenesis merit further investigation. Author contributions: E.D., R.R., L.G.-R., R.L.S., W.A., and R.P. designed research; E.D., R.R., L.G.-R., E.M.B., H.H., and L.S.S.J. performed research; E.D., R.R., L.G.-R., L.S.S.J., J.J.M., A.C., R.L.S., W.A., and R.P. analyzed data; and E.D., A.C., R.L.S., W.A., and R.P. wrote the paper. The authors declare no conict of interest. 1 To whom correspondence may be addressed. E-mail: [email protected], [email protected], or [email protected]. 2 W.A. and R.P. contributed equally to this work. 3 Present address: Department of Internal Medicine, The University of New Mexico Cancer Center, Albuquerque, NM 87106. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1321139110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1321139110 PNAS | December 17, 2013 | vol. 110 | no. 51 | 2071720722 MEDICAL SCIENCES

Upload: others

Post on 14-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

CD13-positive bone marrow-derived myeloid cellspromote angiogenesis, tumor growth, and metastasisEleonora Dondossolaa, Roberto Rangela,b, Liliana Guzman-Rojasa,b, Elena M. Barbua, Hitomi Hosoyaa,c, Lisa S. St. Johnd,Jeffrey J. Molldremd, Angelo Cortie, Richard L. Sidmanf,g,1, Wadih Arapa,1,2,3, and Renata Pasqualinia,1,2,3

aDavid H. Koch Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; bCancer Research Program, The Methodist Hospital ResearchInstitute, Houston, TX 77030; cThe US Naval Hospital Yokosuka, Fleet Post Office, AP, 96350-1600; dDepartment of Stem Cell Transplantation and CellularTherapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology, San Raffaele Scientific Institute, 20132Milan, Italy; fHarvard Medical School, Boston, MA 02215; and gDepartment of Neurology, Beth Israel-Deaconess Medical Center, Boston, MA 02215

Contributed by Richard L. Sidman, November 9, 2013 (sent for review June 13, 2013)

Angiogenesis is fundamental to tumorigenesis and an attractivetarget for therapeutic intervention against cancer. We haverecently demonstrated that CD13 (aminopeptidase N) expressed bynonmalignant host cells of unspecified types regulate tumor bloodvessel development. Here, we compare CD13 wild-type and null bonemarrow-transplanted tumor-bearing mice to show that host CD13+

bone marrow-derived cells promote cancer progression via their ef-fect on angiogenesis. Furthermore, we have identified CD11b+CD13+

myeloid cells as the immune subpopulation directly regulating tumorblood vessel development. Finally, we show that these cells are spe-cifically localized within the tumor microenvironment and produceproangiogenic soluble factors. Thus, CD11b+CD13+ myeloid cells con-stitute a population of bone marrow-derived cells that promote tu-mor progression and metastasis and are potential candidates for thedevelopment of targeted antiangiogenic drugs.

mouse models | protease | stromal cells | vascular pericytes

Angiogenesis is a rate-limiting step in the development ofmany solid tumors, making it an attractive target for ther-

apeutic intervention (1). Although pharmacologic modulation ofangiogenesis has shown some clinical success, negative factorssuch as treatment-related plasticity, drug resistance, and tumordiversity have underscored the need for a better understandingof tumor-associated blood vessel formation at a mechanistic level(2). Tumor angiogenesis is a multifactorial process involvingseveral different cell subtypes, especially tumor stromal endo-thelial cells, pericytes, carcinoma-associated fibroblasts (CAFs),and bone marrow-derived cells (BMDCs) (3, 4). A variety ofimmune cells directly support angiogenesis, including mast cells,tumor-associated macrophages (TAMs), and Tie2-expressingmacrophages (TEMs) (5–7). Endothelial cells recruit in-flammatory cells to the extravascular tissue by the expressionof different leukocyte adhesion molecules. In turn, immunecells produce soluble factors, such as chemokines, cytokines,and proteases, that influence endothelial cell function andangiogenesis in a paracrine fashion (5). Other studies haveshown that BMDCs have the ability to differentiate into en-dothelial cells, possibly by conversion first to endothelialprogenitors (8–10).Proteases activate growth factors and inhibit suppressive fac-

tors within tumors and are central to the angiogenic processwithin the tumor microenvironment (11). Studies on the in-volvement of aminopeptidases in tumor progression and an-giogenesis have revealed a role for aminopeptidase N (CD13)expressed by stromal cells (12). Originally identified as a sur-face marker on myeloid cells (13), CD13 is a widely expressedmembrane-bound metalloprotease involved in pleiotropic functions,including enzymatic cleavage of peptides, antigen presentation, andsignal transduction that ultimately mediate downstream biologicalphenomena such as cell adhesion, proliferation, and motility (14).Diverse cell subpopulations (e.g., fibroblasts, pericytes, epithelialcells, tumor-initiating cells, and stem cells) express CD13 (15-19).

Our group has shown that CD13 is a functional biomarker ofangiogenesis (20) and is also expressed by tumor-associatedblood vessels (21). Furthermore, by using CD13-null neonatalmice, we demonstrated an impaired angiogenic response in theoxygen-induced retinopathy-of-prematurity model, which quanti-fies retinal pathological neovascularization in response to hypoxia(22). We have also shown that CD13-null mice displayed dose-dependent reduced tumor growth after implantation with eitherB16-F10 melanoma or Lewis lung carcinoma (LLC) cells, largelybecause of impaired angiogenesis (12). These data indicate thatCD13 expressed by host stromal cells mediates a pathologicalangiogenic response, but the specific cell subtypes involved in thisphenomenon have yet to be identified.Here, we unravel the type and role of CD13+ stromal cells in

tumor progression. By using CD13 wild-type (WT) and null (KO)mice in the setting of paired bone marrow transplantations, wedemonstrate that CD13+ BMDCs affect the progression of LLC,B16-F10 melanoma, and TSA mammary adenocarcinoma viatheir regulation of tumor angiogenesis. Notably, we show directinvolvement of a CD11b+CD13+ myeloid subset of BMDCs inthis process. Finally, we show that these cells produce angio-genic factors and specifically localize within the tumor micro-environment. Our results indicate that CD11b+CD13+ cellsrepresent a population of proangiogenic BMDCs and that CD13is crucial for their activity.

Significance

The progression of many solid tumors is associated with in-creased vascularization. We previously recognized involvementin tumor development and angiogenesis of tumor stromal cellsexpressing the CD13 protease aminopeptidase. The basic bi-ological concept of participation of nontumor cells in the cancerstroma microenvironment is strengthened in the present studyby our finding that a CD11b+CD13+ myeloid subset of bonemarrow-derived cells affects pericyte biology and angiogenesisand thereby influences tumor growth and metastasis. Thera-peutic implications of the identification of specific CD11+CD13+

myeloid bone marrow-derived cells as participants in themechanism of tumor angiogenesis merit further investigation.

Author contributions: E.D., R.R., L.G.-R., R.L.S., W.A., and R.P. designed research; E.D., R.R.,L.G.-R., E.M.B., H.H., and L.S.S.J. performed research; E.D., R.R., L.G.-R., L.S.S.J., J.J.M., A.C.,R.L.S., W.A., and R.P. analyzed data; and E.D., A.C., R.L.S., W.A., and R.P. wrote the paper.

The authors declare no conflict of interest.1To whom correspondence may be addressed. E-mail: [email protected],[email protected], or [email protected].

2W.A. and R.P. contributed equally to this work.3Present address: Department of Internal Medicine, The University of New Mexico CancerCenter, Albuquerque, NM 87106.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1321139110/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1321139110 PNAS | December 17, 2013 | vol. 110 | no. 51 | 20717–20722

MED

ICALSC

IENCE

S

Page 2: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

ResultsIn Vivo Characterization of CD13-Expressing Tumor Stromal Cells.Previous molecular and cellular analysis of LLC and B16-F10 melanoma tumor growth in CD13-null mice revealed thatCD13 expression on nonmalignant host cells mediates tumorgrowth and angiogenesis (12). Given that endothelial cells,fibroblasts, pericytes, and immune cells (in particular, myeloidcells) display CD13 (13, 18, 19, 21), but no comprehensivestudies on CD13 expression by tumor stromal cells have beenperformed, we first used FACS to quantitatively analyze LLC-,B16-F10–, and TSA-derived endothelial cells (CD31+), pericytes/CAFs [α smooth muscle actin (αSMA)+] (23), myeloid cells(CD45+CD11b+), and other immune cells (CD45+CD11b−).Myeloid cells were the most frequent stromal cell population inthe three tumor models studied (Fig. 1A). The majority of per-icytes/CAFs (82–94%) expressed CD13, whereas about 40% ofmyeloid cells and 20% of endothelial cells expressed CD13 in alltumor models. Only a small subset of nonmyeloid immune cellsexpressed CD13 (Fig. 1B). Finally, we analyzed the cell surfaceexpression among CD13-expressing stromal cell componentswithin the tumor microenvironment. Myeloid cells represented50–72% of CD13-expressing cells in all of the models analyzed,whereas endothelial cells varied between 6% and 30%, pericytes/CAFs were ∼10%, and other inflammatory cells were ∼3–9%(Fig. 1C). Thus, myeloid cells are the most frequently representedCD13-expressing stromal cells in these tumors.

Experimental Generation of CD13 WT and KO Bone Marrow-TransplantedMouse Models. To study the contribution of CD13-expressing stro-mal cells to tumor growth and potentially to angiogenesis, we de-veloped bone marrow transplant (BMT) mouse models with CD13WT and KO mice. We transplanted lethally irradiated mice of bothgenotypes with total nucleated bone marrow cells obtained fromWT or KO mice to produce: (i) WTwt mice (CD13 WT micetransplanted with bone marrow cells derived from CD13WTmice);(ii) WTko mice (CD13 WT mice transplanted with bone marrowcells derived from CD13 KO mice); (iii) KOwt mice (CD13 KOmice transplanted with bone marrow cells derived from CD13 WTmice); and (iv) KOko mice (CD13 KO mice transplanted with bonemarrow cells derived from CD13 KOmice; Fig. S1A). These modelswere designed to evaluate the relative contribution of CD13+

BMDCs versus other stromal components to tumor growth andangiogenesis (Fig. S2).Four weeks later, mice were genotyped by PCR (for both DNA

extracted from nucleated blood cells and from tail tips), and suc-cessful engraftment of either WT- or KO-BMDCs was evaluatedand confirmed (Fig. S1B). Hematological parameters (i.e., red andwhite blood cells, hemoglobin, and hematocrit) from the sets ofmice analyzed were similar, indicating that bone marrow re-constitution did not alter the complete blood count composition(Fig. S1C). Long-term bone marrow reconstitution (demonstratedby H&E staining of postmortem femur sections) showed no de-tectable differences between WTwt, KOko, WTko, or KOwt mice(Fig. S1D). Finally, the levels of interleukin-2, tumor necrosisfactor-α, and IFN-γ released by phorbol-12-myristate-13-acetate/ionomycin-stimulated lymphocytes (CD45+CD3+) and myeloidcells (CD45+CD11b+) obtained from WTwt, KOko, WTko, orKOwt mice were similar, suggesting that the cells obtainedfrom the various reconstituted mice are also functionally sim-ilar (Fig. S1E).In summary, transplanted mice recovered from the BMT and

did not show hematological or functional abnormalities.

CD13+ BMDCs Affect Experimental Tumors, Angiogenesis, andMetastasis. We next studied the role of CD13-expressing stromalcells in tumor development. After recovery, BMT mice were ad-ministered s.c. with LLC, B16-F10, or TSA tumor cells. Tumorgrowth in LLC and B16-F10 models was significantly impaired inKOko mice (displaying no CD13-expressing stromal component),compared with WTwt mice, as also previously shown with LLC andB16-F10 transplanted tumors (12); tumor growth was severelyimpaired in the TSA tumor model as well. The growth of LLC,TSA, or B16-F10 tumors was also significantly reduced in WTko

mice, which specifically lack only CD13+ BMDCs (Fig. 2 A and B).Consistent with these observations, tumor growth was restored inboth LLC- and TSA-bearing KOwt mice, which express CD13 onlyon BMDCs but not on pericytes, endothelial cells, or CAFs (Fig. 2A and B). Only B16-F10 tumor growth remained unrescued in thepresence of CD13+ BMDCs (Fig. 2 A and B). We also analyzedthe role of CD13+ BMDCs on tumor growth at secondary sites bytail vein injection of tumor cells in BMT mice. Similar to theresults for primary tumor growth, the absence specifically ofCD13+ BMDCs clearly reduced the formation of lung colonies(Fig. 2C). We conclude that CD13+ BMDCs are an importantstromal component for growth at both the primary tumor site andsecondary experimental metastasis sites.

CD13+ BMDCs Affect Blood Vessel Development in Tumors. To un-derstand how CD13+ BMDCs regulate tumor growth, we analyzedangiogenesis by quantification of CD31 (endothelial cells) andNG2 chondroitin sulfate proteoglycan (NG2, pericytes) immu-noreactive blood vessels in LLC, TSA, and B16-F10 tumorsections obtained from WTwt, KOko, WTko, and KOwt tumor-bearing mice. Tumors from mice specifically lacking CD13+

BMDCs (i.e., WTko and KOko) exhibited lower blood vessel

Fig. 1. Characterization of CD13-expressing stromal cells. (A) TSA, LLC,and B16-F10 tumor cells (shown and labeled in the vertically positionedgraphs) were analyzed by FACS for expression of several cell type-specificmarkers: αSMA for pericytes/CAFs, CD31 (endothelial cells), and CD45(immune cells), separated into CD11b− and CD11b+ groups to distinguishnonmyeloid and myeloid cells. Pericytes and endothelial, myeloid, andnonmyeloid stromal cells are represented as percentage of total tumorcells. Means ± SEM are shown. (B) CD13-expressing cells in the three tumorcell types represented as a percentage of each stromal cell component.Means ± SEM are shown. (C ) Distribution of CD13-expressing cells amongstromal cells in the three tumor cell types. Means ± SEM are shown. In eachcase, three to five tumors per experiment were analyzed, and each ex-periment was performed at least three times. Data from one representa-tive experiment are shown.

20718 | www.pnas.org/cgi/doi/10.1073/pnas.1321139110 Dondossola et al.

Page 3: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

densities (Fig. 3 A and C and Fig. S3) and lesser coverage withNG2-immunoreactive pericytes (Fig. 3 B and C and Fig. S3), incomparison with tumors from WTwt mice. Notably, transplantationof CD13+ BMDCs into CD13 KO mice (KOwt) increased thenumber of blood vessels in LLC- and TSA-derived tumors andblood vessel coverage with NG2-immunoreactive pericytes incomparison with tumors from KOko mice (Fig. 3 and Fig. S3A).Similar to the effect on tumor growth, no rescue was observed inB16-F10–bearing KOwt mice (Fig. S3B). Taken together, theseresults establish that CD13+ BMDCs represent a subset of immunecells whose absence impairs tumor-associated angiogenesis.

CD13+ BMDCs and Angiogenic Blood Vessels Express Different CD13Isoforms. CD13 has multiple isoforms, and the isoform found onangiogenic blood vessels is recognized specifically by the Asn-Gly-Arg (NGR) (single-letter amino acid code) motif (24). To de-termine whether the same isoform was also expressed on CD13+

BMDCs, we performed subtractive cell-binding assays with a bac-teriophage (phage) clone displaying an NGR motif (NGR-targeted phage) through the biopanning and rapid analysis ofselective interactive ligands methodology (25). Tumor-asso-ciated BMDCs with and without CD13 expression (CD45+

CD13+ and CD45+CD13−, respectively) and angiogenic endo-thelial cells with and without CD13 expression (CD31+CD13+ andCD31+CD13−, respectively) were isolated by FACS from TSA-derived tumors and incubated with NGR-targeted phage oruntargeted control phage. Only CD13-expressing endothelial cells

specifically bound NGR-targeted phage (Fig. S4). These resultsindicate that BMDCs express an isoform of CD13 different fromthat displayed by angiogenic endothelial cells.

CD13+ Myeloid Cells Regulate Angiogenesis. Several subsets ofBMDCs are well-established contributors to tumor angiogenesis(3, 5). Given that our BMT-based experimental models do notaddress the contribution of specific subpopulations of BMDCs, wedirectly analyzed the effect of isolated subsets of CD13+ BMDCson tumor blood vessel development. CD45+CD11b+CD13+,CD45+CD11b+CD13−, or CD45+CD11b−CD13+ (identified asCD11b+CD13+ myeloid cells, CD11b+CD13− myeloid cells, andCD11b−CD13+ nonmyeloid cells, respectively) were sorted byFACS from TSA-derived tumors grown in WT mice and coad-ministered with TSA cells into CD13 KO mice (3 × 104 and 4 ×105 cells, respectively). Sorted cells were also administered directlyinto the tumors at days 5 and 9 posttumor challenge, and tumorangiogenesis was quantified by immunofluorescence on day 12.Administration of CD11b+CD13+ myeloid cells rescued angiogen-esis, as evaluated by quantification of the number of endothelialcell-containing (CD31+) blood vessels, and markedly restoredNG2+ pericyte coverage. The other cell populations did not rescueangiogenesis and pericyte coverage (Fig. 4A). Therefore, CD11b+

CD13+ myeloid cells can selectively promote angiogenesis in vivo.To assess whether CD11b+CD13+ myeloid cells can regulate an-giogenesis by affecting endothelial cell migration and organization,we incubated the sorted cells with carboxyfluorescein succinimidyl

Fig. 2. Experimental tumor growth in BMT mice. (A and B) LLC, TSA, and B16-F10 tumor growth (mean ± SEM), tumor weight (mean ± SEM), and repre-sentative photographs of tumors recovered from BMT mice administered s.c. with tumor cells. (C) Lung weight (LLC) or number of lung colonies (TSA, B16-F10) (mean ± SEM) and representative photographs of lungs recovered from BMT mice administered with tumor cells i.v. *P < 0.05; **P < 0.01; ***P < 0.001by two-tailed Student t test.

Dondossola et al. PNAS | December 17, 2013 | vol. 110 | no. 51 | 20719

MED

ICALSC

IENCE

S

Page 4: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

ester (CFSE)-labeled human dermal microvascular endothelial cells(HDMECs) and analyzed the formation of endothelial tubes after 8h (“tube-formation” assay). In parallel, we cocultured sorted cellswith scratched CFSE-labeled HDMEC monolayers and moni-tored “wound” closure for 3 d (“wound-healing” assay). Noeffects were observed in either assay (Fig. 4B and Fig. S5).These data suggest that CD11b+CD13+ myeloid cells do notinhibit angiogenesis in tumors by directly affecting endothelialcell migration and organization and more likely affect the complexinterplay between pericytes and endothelial cells within the tumormicroenvironment.

CD13+ Myeloid Cells Produce Soluble Angiogenic Factors. To assesswhether CD11b+CD13+ myeloid cells could affect angiogenesis byproducing soluble proangiogenic factors, we sorted CD11b+

CD13+, CD11b+CD13−, and CD11b−CD13+ from LLC tumorsand cultured them in vitro for 5 d. Then, we analyzed the presenceof soluble pro-/antiangiogenic factors in their supernatants withthe Mouse Angiogenesis Array Kit (R&D). The results showedthat (i) CD11b+CD13+ cells secreted matrix metalloproteinase(MMP)-9 and osteopontin (OPN); (ii) CD11b+CD13− producedOPN, CXCL16, and CXCL4 (a potent inhibitor of angiogenesis);and (iii) CD11b−CD13+ secreted OPN and stromal cell-derivedfactor-1 (Fig. 4C). Thus, CD11b+CD13+ could selectively releaseMMP-9, a protease known to play a role in angiogenesis (26).A lysate of CD11b+CD13+ myeloid cells was then analyzed

using the same angiogenesis array. Besides confirming the pro-duction of osteopontin and MMP-9 by CD11b+CD13+ cells, thisassay could also detect the production of monocyte chemo-attractant protein (MCP)-1 (Fig. S6A). Notably, both MCP-1 andMMP-9 are known to regulate vascular architecture by pro-moting pericyte recruitment (26, 27).Finally, we analyzed the release of angiogenic factors by per-

icytes alone or cocultured with sorted BMDCs. Although various

factors were released by these cells, no significant changes wereinduced by BMDCs in this experimental system (Fig. S6B).Collectively, these data indicate that CD11b+CD13+ mye-

loid cells secrete proteins known to affect pericyte biologyand angiogenesis.

CD13+ Myeloid Cells Specifically Localize Within Tumors andMacrophages Are the Most Represented CD13+ Population. Toinvestigate the location of CD11b+CD13+ myeloid cells, we per-formed a biodistribution study in LLC, B16-F10, and TSA tumor-bearing mice. Single-cell suspensions obtained from blood, bonemarrow, brain, spleen, liver, lung, kidney, and tumor were analyzedby FACS. CD11b+CD13+ myeloid cells were found only in tumors,where they represented 10.5%, 2.7%, and 5.11% of LLC, B16-F10,and TSA tumor cells, respectively (Fig. S7 A and B). Finally,we analyzed the percentage of CD13+ cells in different tumor-infiltrating myeloid and nonmyeloid cell populations. CD13-expressing cells were identified as monocytes (CD45+CD11b+

Ly6G−Ly6C+CD13+), macrophages (CD45+CD11b+F4/80+

CD13+), granulocytes (CD45+CD11b+Ly6G+Ly6C+CD13+),dendritic cells (CD45+CD11c+CD13+), CD4 T lymphocytes(CD45+CD4+CD13+), and CD8 T lymphocytes (CD45+CD8+

CD13+). Although CD13 was expressed by monocytes andgranulocytes, macrophages were the most represented pop-ulation, comprising 18% of the BMDCs (Fig. S7C). FewerCD13+ dendritic cells and CD4 T lymphocytes, and no CD8 Tlymphocytes, were identified.

DiscussionIn this report, we provide evidence that CD13+ myeloid BMDCsrepresent a population of proangiogenic immune cells implicatedin tumor-associated blood vessel formation. We also show thatCD13, a membrane-bound protease, has a central role in theiractivity. To dissect the relative contributions of different CD13+

cells of the tumor microenvironment, we have generated syngeneictumor models based on CD13 WT and KOmice transplanted withCD13-positive or -negative bone marrow recovered from the samemice to obtain different combinations called WTwt, WTko KOwt,and KOko (see Fig. S2 for a schematic representation of the ex-perimental system). The growth of different tumor cells (namelyB16-F10 melanoma, TSA adenocarcinoma, and LLC lung carci-noma) in these mice was subsequently investigated in vivo. Theresults showed that growth of all these tumors and angiogenesiswithin them is severely impaired in mice lacking CD13+ BMDCs(KOko and WTko), and this impairment is associated with a re-duction in both vascular density and pericyte coverage in tumortissues. Restoring CD13+ BMDCs in CD13-negative mice (KOwt)rescued tumor growth and angiogenesis in the TSA and LLCmodels but not in the B16-F10 model.These results clearly indicate that CD13+ BMDCs have a role

in tumor-related angiogenesis and blood vessel stabilization. Thedifferent results, in quantitative terms, obtained with differenttumor models (particularly in the “rescue” experiment) suggestthat tumor cells have a role in the cross-talk between CD13+

BMDCs and the tumor microenvironment during angiogenesis.The role of BMDCs in tumor angiogenesis has been increasingly

investigated in recent years. Proangiogenic BMDCs include dif-ferent myeloid subsets: granulocytes, monocytes/macrophages,dendritic cells, mast cells, myeloid suppressor cells, and otherpopulations (e.g., progenitor cells) (3). Monocytes (circulating andresident) can differentiate into various functional subpopulationsof TAMs, whose phenotypes are regulated by the milieu of cyto-kines present within the tumor microenvironment (3). Althoughthe developmental relationships between monocyte/macrophagessubsets are still poorly defined, monocytes have been shown toproduce at least two distinct macrophage populations: the CD11c+

TAMs and TEMs (28). Both populations can support angio-genesis by producing different proangiogenic cytokines, such as

Fig. 3. Evaluation of tumor angiogenesis in LLC tumor-bearing BMT mice.(A) Immunohistochemical detection of CD31 (endothelial cells, red) and NG2(pericytes, green) in LLC tumors recovered from tumor-bearing BMT mice.DAPI, blue. (B) Higher magnification of NG2+ and CD31+ blood vessels isshown. (C) The number of CD31+ blood vessels per field and the number ofNG2+ blood vessels (mean ± SEM) averaged from 10 random fields areshown. Three tumors for each of the BMT group were analyzed. *P < 0.05;**P < 0.01; ***P < 0.001 by two-tailed Student t test. (Scale bar, 20 μm.)

20720 | www.pnas.org/cgi/doi/10.1073/pnas.1321139110 Dondossola et al.

Page 5: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

basic fibroblast growth factor, vascular endothelial growth factor(VEGF), and MMP-9 (7, 29, 30). The depletion of proangio-genic macrophages (e.g., induced by ablation or pharmacologi-cal neutralization or suppression of their recruitment) caninhibit tumor growth by preventing production of proangio-genic, tumor-promoting, and immunosuppressive factors (3).Granulocytes, mast cells, and myeloid-derived suppressor cellsalso can play a role in angiogenesis by producing proangiogenicfactors, such as VEGF-A and MMP-9 (30). Neutrophils, in par-ticular, together with monocytes and macrophages, were identifiedas the most important producers of MMP-9, a protease that affectsthe interactions of VEGF with VEGFR2 by promoting MMP-9bioavailability, thereby inducing angiogenesis (30). Our resultsshow that CD13+ myeloid BMDCs cells, but not CD13− myeloidcells or CD13+ nonmyeloid immune cells, can rescue tumor an-giogenesis in vivo when administered to KO mice. However, noneof these cell populations could promote capillary-like tube for-mation by endothelial cells in vitro. This result suggests that in-teraction of CD13+ myeloid BMDCs with other cells of the tumormicroenvironment is necessary for angiogenesis stimulation. In-terestingly, we found that CD13+ myeloid cells can specificallyproduce detectable levels of MCP-1 and MMP-9. Given that bothMMP-9 and MCP-1 are key factors for pericyte recruitment, a celltype known to consolidate vascular architecture and prevent blood

vessel regression (26, 27, 31), this finding suggests that CD13+

myeloid BMDCs cells can promote angiogenesis and vascularmaturation by regulating pericyte recruitment.The identification of CD13+ myeloid BMDCs as participants in

tumor angiogenesis could have important implications for de-velopment of therapeutic strategies against cancer. First, the ob-servation that these cells are localized almost exclusively withintumors makes them an attractive potential deliverer of therapeu-tics to the tumor microenvironment. Second, CD13+ myeloidBMDCs may represent a target for antiangiogenic therapeutics.Given the crucial role of CD13 in the activity of these cells,compounds inhibiting this ectoenzyme could have therapeuticbenefit. In addition, CD13 ligands could be exploited for deliveringdrugs to these cells within the tumor microenvironment. CD13peptide ligands containing the CNGRC motif, identified by phagedisplay technology, have previously been exploited to delivercytokines, cytotoxins, apoptotic peptides, and even liposomes totumor blood vessels (20, 32–34). Unfortunately, our results ofCNGRC peptide-binding experiments show that CD13+ BMDCsare not targeted by this peptide, likely owing to expression ofdifferent CD13 isoforms in CD13+ BMDCs and endothelial cells.Indeed, it has been demonstrated previously by histochemicalanalysis that differentially immunoreactive forms of CD13 areexpressed in tumor-associated blood vessels, myeloid cells, and

Fig. 4. Effect of sorted BMDCs on angiogenesis. (A) Immunohistochemical detection of CD31 (endothelial cells, red) and NG2 (pericytes, green) in TSA tumors. ThenumberofCD31+bloodvesselsperfieldandthenumberofNG2+bloodvessels (mean±SE)averagedfrom10randomfieldsare shown.Threetumorspereachgroupwereanalyzed. (B) In vitro tube-formation assay. CFSE-labeled HDMECs were incubated with sorted CD13-expressing myeloid cells (CD11b+CD13+), CD13-expressing non-myeloid immune cells (CD11b−CD13+), and CD13-negative myeloid cells (CD11b+CD13−). Images were taken after 8 h, and tube length was analyzed by WimTubesoftware. Representative images are shown (mean± SEM). Each experimentwas performed three times in quadruplicate. Data fromone representative experiment areshown. (C)Analysisofangiogenicproteins secretedbyCD11b+CD13+,CD11b+CD13−andCD11b−CD13+cells.Cells (1.5×105)were isolatedbyFACSfromLLCtumorsgrownin CD13WTmice and cultured for 5 d inDMEM, 2%FBS. The supernatantswere thenanalyzedwith theMouseAngiogenesis AntibodyArrayKit (R&D). Immunoreactivespots (in duplicate) are shown. Immunoreactive control spots are also shown at the corners. **P < 0.01; ***P < 0.001 by two-tailed Student t test. (Scale bar, 20 μm.)

Dondossola et al. PNAS | December 17, 2013 | vol. 110 | no. 51 | 20721

MED

ICALSC

IENCE

S

Page 6: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

epithelia (24). The structural determinants of this selectivity are stillunknown. It has been postulated that the use of alternative glyco-sylation sites or changes in the composition of oligosaccharides maymask antibody or peptide-binding sites (35). Peptide-binding se-lectivity might also be related to differential formation of quaternarycomplexes with other proteins in different tissues. Furtherstudies are necessary to clarify this issue. In any case, it shouldbe possible, in principle, to develop other peptides or antibodyligand selective for the CD13+ form expressed by BMDCs or forother markers of these proangiogenic cells, as accomplished pre-viously with the endothelial cells. A valid and realistic approach isto exploit phage display libraries to identify ligands and/or to fin-gerprint the BMDCs surface to identify receptors expressed asthese cells home to tumors.In summary, our results suggest that CD13+ myeloid BMDCs

are the most represented CD13-expressing cells in the tumormicroenvironment and have mechanistic roles in tumor growthand metastasis by affecting pericyte biology and angiogenesis.

Materials and MethodsCell Lines, Drugs, Antibodies, and Reagents. We obtained mouse B16-F10 cellsand LLC cells from the American Type Culture Collection and HDMECs andhuman pericytes from PromoCell. TSA mammary adenocarcinoma cells werea gift from Giulia Casorati (San Raffaele Scientific Institute, Milan). Rabbitpolyclonal anti-cleaved caspase 3, anti-NG2 chondroitin sulfate proteoglycan,and anti-αSMA were purchased from Cell Signaling, Chemicon, and Abcam,respectively. Rat monoclonal anti-CD31 was from Abcam, and CFSE wasfrom Sigma.

Generation of Bone Marrow-Transplanted Mice. All of the animal studies wereapproved through the Institutional Animal Care and Use Committee of theUniversity of Texas M. D. Anderson Cancer Center. CD13 WT and KO BALB/cand C57BL/6 mice were generated as described (12, 22). Recipient 8- to 12-

wk-old CD13 WT and KO mice were whole body-irradiated in a cobalt irra-diator at 12 Gy per mouse (two rounds of radiation at 6 Gy each, with a 2-hrecovery interval). Donor CD13 WT and KO mice were killed, and bonemarrow cells from femur and tibia were flushed with PBS containing 2%(vol/vol) FBS and washed in PBS. The cells were resuspended in hypotonicsolution to lyse red blood cells, and subsequently washed again in PBS.Three hours after the last round of radiation, recipient mice received 5–10 ×106 total nucleated BMDCs i.v. Following BMT, mice received enrofloxacin(Bayer) in the water for 4 wk. Control mice that were radiated but nottransplanted died within 12–14 d.

Tumor and Experimental Metastasis Studies. BMT BALB/c mice received TSAcells (4 × 105) s.c. BMT C57BL/6 received either LLC cells (1 × 106 per mouse)s.c. or B16-F10 cells (5 × 105 per mouse) s.c. Tumor sizes were measured witha caliper until mice were killed. Tumors were collected and immersed in OCT.For experimental metastasis models, BMT BALB/c mice received TSA cells (7 ×104 per mouse) i.v. BMT C57BL/6 mice received either LLC (1.5 × 105 permouse) i.v. or B16-F10 cells (1.2 × 105 per mouse) i.v. Lungs were removedand weighed, and metastatic lung colonies were counted under a stereo-microscope. For experiments involving coadministration of tumor and sortedcells, we first purified the populations of interest (CD45+CD11b+CD13+,CD45+CD11b+CD13−, CD45+CD11b−CD13+ cells) from TSA-derived tumorsrecovered from CD13 WT mice by the FACSAria cell sorter as describedabove. Sorted cells (3 × 104) were coadministered s.c. with TSA cells (4 × 105)to CD13 KO BALB/c mice and then administered intratumor 5 and 10 d, re-spectively, after tumor implantation.

FACS analyses; in vitro assays on evaluation of bone marrow re-constitution and lymphocyte and myeloid cell function in BMT mice; im-munofluorescence of tumor blood vessels; tube-formation, cell-migration,and angiogenic factor measurements; and cell-binding studies are reportedin SI Materials and Methods.

ACKNOWLEDGMENTS. This work was supported by grants from the NationalInstitutes of Health and National Cancer Institute and by awards fromAngelWorks and the Gillson-Longenbaugh Foundation (all to W.A. and R.P.).

1. Kerbel RS (2008) Tumor angiogenesis. N Engl J Med 358(19):2039–2049.2. Ebos JM, Kerbel RS (2011) Antiangiogenic therapy: Impact on invasion, disease pro-

gression, and metastasis. Nat Rev Clin Oncol 8(4):210–221.3. Coffelt SB, et al. (2010) Elusive identities and overlapping phenotypes of proangio-

genic myeloid cells in tumors. Am J Pathol 176(4):1564–1576.4. Hanahan D, Coussens LM (2012) Accessories to the crime: Functions of cells recruited

to the tumor microenvironment. Cancer Cell 21(3):309–322.5. De Palma M, Naldini L (2006) Role of haematopoietic cells and endothelial progeni-

tors in tumour angiogenesis. Biochim Biophys Acta 1766(1):159–166.6. De Palma M, Murdoch C, Venneri MA, Naldini L, Lewis CE (2007) Tie2-expressing

monocytes: Regulation of tumor angiogenesis and therapeutic implications. TrendsImmunol 28(12):519–524.

7. 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.

8. Shi Q, et al. (1998) Evidence for circulating bone marrow-derived endothelial cells.Blood 92(2):362–367.

9. Lyden D, et al. (2001) Impaired recruitment of bone-marrow-derived endothelial andhematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 7(11):1194–1201.

10. Nolan DJ, et al. (2007) Bone marrow-derived endothelial progenitor cells are a majordeterminant of nascent tumor neovascularization. Genes Dev 21(12):1546–1558.

11. van Hinsbergh VW, Engelse MA, Quax PH (2006) Pericellular proteases in angiogen-esis and vasculogenesis. Arterioscler Thromb Vasc Biol 26(4):716–728.

12. Guzman-Rojas L, et al. (2012) Cooperative effects of aminopeptidase N (CD13) ex-pressed by nonmalignant and cancer cells within the tumor microenvironment. ProcNatl Acad Sci USA 109(5):1637–1642.

13. Favaloro EJ, et al. (1988) Further characterization of human myeloid antigens (gp160,95;gp150; gp67): Investigation of epitopic heterogeneity and non-haemopoietic distributionusing panels of monoclonal antibodies belonging to CD-11b, CD-13 and CD-33.Br J Haematol 69(2):163–171.

14. Mina-Osorio P (2008) The moonlighting enzyme CD13: Old and new functions totarget. Trends Mol Med 14(8):361–371.

15. Lai A, Ghaffari A, Li Y, Ghahary A (2011) Paracrine regulation of fibroblast amino-peptidase N/CD13 expression by keratinocyte-releasable stratifin. J Cell Physiol226(12):3114–3120.

16. Amoscato AA, Sramkoski RM, Babcock GF, Alexander JW (1990) Neutral surfaceaminopeptidase activity of human tumor cell lines. Biochim Biophys Acta 1041(3):317–319.

17. Calloni R, Cordero EA, Henriques JA, Bonatto D (2013) Reviewing and updating themajor molecular markers for stem cells. Stem Cells Dev 22(9):1455–1476.

18. Lai A, Ghaffari A, Ghahary A (2010) Inhibitory effect of anti-aminopeptidase N/CD13antibodies on fibroblast migration. Mol Cell Biochem 343(1-2):191–199.

19. Alliot F, Rutin J, Leenen PJ, Pessac B (1999) Pericytes and periendothelial cells of brainparenchyma vessels co-express aminopeptidase N, aminopeptidase A, and nestin.J Neurosci Res 58(3):367–378.

20. Arap W, Pasqualini R, Ruoslahti E (1998) Cancer treatment by targeted drug deliveryto tumor vasculature in a mouse model. Science 279(5349):377–380.

21. Pasqualini R, et al. (2000) Aminopeptidase N is a receptor for tumor-homing peptidesand a target for inhibiting angiogenesis. Cancer Res 60(3):722–727.

22. Rangel R, et al. (2007) Impaired angiogenesis in aminopeptidase N-null mice. ProcNatl Acad Sci USA 104(11):4588–4593.

23. Sugimoto H, Mundel TM, Kieran MW, Kalluri R (2006) Identification of fibroblastheterogeneity in the tumor microenvironment. Cancer Biol Ther 5(12):1640–1646.

24. Curnis F, et al. (2002) Differential binding of drugs containing the NGR motif to CD13isoforms in tumor vessels, epithelia, and myeloid cells. Cancer Res 62(3):867–874.

25. Giordano RJ, Cardó-Vila M, Lahdenranta J, Pasqualini R, Arap W (2001) Biopanningand rapid analysis of selective interactive ligands. Nat Med 7(11):1249–1253.

26. Chantrain CF, et al. (2004) Stromal matrix metalloproteinase-9 regulates the vasculararchitecture in neuroblastoma by promoting pericyte recruitment. Cancer Res 64(5):1675–1686.

27. Ma J, Wang Q, Fei T, Han JD, Chen YG (2007) MCP-1 mediates TGF-beta-inducedangiogenesis by stimulating vascular smooth muscle cell migration. Blood 109(3):987–994.

28. Pucci F, et al. (2009) A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophagessuggests common functions and developmental relationships. Blood 114(4):901–914.

29. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization:Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phag-ocytes. Trends Immunol 23(11):549–555.

30. Nozawa H, Chiu C, Hanahan D (2006) Infiltrating neutrophils mediate the initial an-giogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad SciUSA 103(33):12493–12498.

31. Salcedo R, et al. (2000) Human endothelial cells express CCR2 and respond to MCP-1:Direct role of MCP-1 in angiogenesis and tumor progression. Blood 96(1):34–40.

32. Ellerby HM, et al. (1999) Anti-cancer activity of targeted pro-apoptotic peptides. NatMed 5(9):1032–1038.

33. Curnis F, et al. (2000) Enhancement of tumor necrosis factor alpha antitumor im-munotherapeutic properties by targeted delivery to aminopeptidase N (CD13). NatBiotechnol 18(11):1185–1190.

34. Pastorino F, et al. (2006) Targeting liposomal chemotherapy via both tumor cell-specificand tumor vasculature-specific ligands potentiates therapeutic efficacy. Cancer Res66(20):10073–10082.

35. Corti A, Curnis F, Arap W, Pasqualini R (2008) The neovasculature homing motif NGR:More than meets the eye. Blood 112(7):2628–2635.

20722 | www.pnas.org/cgi/doi/10.1073/pnas.1321139110 Dondossola et al.

Page 7: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Supporting InformationDondossola et al. 10.1073/pnas.1321139110SI Materials and MethodsFACS Analysis. We used an LSRII FACS (Becton-Dickinson)equipped with Diva Software. Tumors were mechanically dis-aggregated on a 70-μm cell strainer to generate a suspension ofsingle cells, resuspended in PBS containing 2% (vol/vol) FCSand 2 mM EDTA and incubated with the following rat mono-clonal antibodies (BD Pharmingen): PE-Cy 7 anti-CD45; PerCP-Cy 5.5 anti-CD11b; FITC-conjugated anti-CD13, anti-Ly6G, andanti-CD11c; PE-conjugated anti-CD4 and anti-CD13; and APC-conjugated anti-F4/80, anti-CD31, anti-Ly6G, and anti-CD8b.Anti-smooth muscle actin α was used in combination with PB-conjugated goat anti-rabbit antibody (Invitrogen). Dead cellsstained with 4′,6-diamidino-2-phenylindole (DAPI) were ex-cluded. We set up fluorescence minus one controls for gatinganalysis to distinguish staining of negative from positive cellpopulations. For experiments involving sorted cells, we purifiedcell populations with a FACSAria cell sorter.

Evaluation of Bone Marrow Reconstitution. We used PCR geno-typing, analysis of hematological parameters, and H&E stainingof bones. Four weeks after bone marrow transplant (BMT),blood and tail tip samples from transplanted mice were collectedin EDTA tubes to prevent blood clotting. DNA from nucleatedblood cells and tail tips was extracted with the DNeasy Blood andTissue Extraction Kit (Qiagen) and with DirectPCR lysis reagent(Viagen Biotech), respectively. DNA was analyzed by PCR asdescribed (1, 2). Briefly, a combination of three primers was used:forward, 5′-CACCCCCATCCCCCATCCCTTAC-3′; reverse, 5′-GTGCCCACGCCCTTGAACCTTACTT-3′; and internal ribo-somal entry site reversed (IRES-rev), 5′-ACAAACGCACACC-GGCCTTATTCC-3′. PCR with the forward/reverse and forward/IRES-rev primer pairs generated an 879-bp WT product anda 320-bp KO product, respectively. Blood samples were analyzedon an ABX Micros 60 instrument (Horiba). Femurs were re-moved from BMT mice, decalcified in 5% formic acid solution,and paraffin-embedded, and slides were stained with H&E.

Evaluation of Lymphocytes and Myeloid Cells Function in BMT Mice.Long bones from posterior legs of reconstituted BMT mice wereflushed with DMEMand 10% FBS to isolate bone marrow-derivedcells (BMDCs). BMDCs were then stimulated for 4 h with phorbol-12-myristate-13-acetate (5 ng/mL; Sigma) and ionomycin (500 ng/mL; Sigma) in the presence of a protein transport inhibitor (bre-feldin A; 3 μg/mL; Sigma). Cells were washed with PBS andstained with anti-CD45/APC-eFluor780, anti-CD11b/PerCP-Cy5.5, and anti-CD3/FITC antibody/fluorochrome conjugates. Cellswere fixed with 4% paraformaldehyde, permeabilized with BDPerm/Wash buffer (BD Pharmingen) for 15 min, and stained forintracellular cytokines with anti-IL-2/APC, anti-TNFα/Pacificblue, and anti IFNγ/PE-Cy7 antibody/fluorochrome conjugates.Samples were then analyzed by FACS.

Immunofluorescence of Tumor Blood Vessels. To assess the tumorvasculature in TSA, Lewis lung carcinoma (LLC), and B16-F10tumors recovered from BMT mice, we performed immunofluo-rescence on frozen tumor sections. Tissue on microscope slideswas fixed in PBS containing 2% paraformaldehyde and blocked inPBS containing with 5% normal goat serum, 1% BSA, and 0.3%Triton X-100. Sections were incubated with a rat monoclonalanti-CD31 antibody and a rabbit polyclonal anti-NG2 chondroitinsulfate proteoglycan antibody in 1% normal goat serum, 1%BSA, and 0.3% Triton X-100 in PBS, followed by Alexa Fluor 546

anti-rat or Alexa Fluor 488 anti-rabbit antibodies (Invitrogen).Images were acquired on an Olympus IX70 inverted fluorescencemicroscope/MagnaFire software and were analyzed with Photo-shop CS4 software (Adobe). Three tumors were acquired for eachgroup of BMT mice, with at least 10 images per tumor.

Tube-Formation Assay. Human dermal microvascular endothelialcells (HDMECs) were detached in PBS containing 0.25% trypsin,washed three times, and incubated with 20 μMcarboxyfluoresceinsuccinimidyl ester (CFSE) for 8 min. After addition of 100%FBS for 1 min, cells were washed again and incubated at 37 °C and5% CO2 overnight (ON). CD45+CD11b+CD13+, CD45+CD11b+

CD13−, and CD45+CD11b−CD13+ cells were sorted in a FACSA-ria cell sorter. Growth factor-reduced Matrigel (BD Biosciences)was thawed at 4 °C, added to 96-well plates (60 μL per well), andpolymerized for 30 min at 37 °C, 5% CO2. HDMECs were de-tached in PBS containing 0.25% trypsin and washed twice in PBS. Atotal of 104 sorted cells and 104 HDMECs in basal medium con-taining 2% FCS were plated on top of the Matrigel layers. Tubeformation was monitored for 10 h. Pictures were taken at 8 h andanalyzed through the WimTube software (Wimasis).

Migration Assay.Confluent CFSE-labeled HDMECs were scratchedwith a 10-μL pipet tip. Sorted CD45+CD11b+CD13+, CD45+

CD11b+CD13−, and CD45+CD11b−CD13+ cells were added tothe cell monolayer, and wound closure was monitored for 3 d bymeasuring the gap area at days 0, 1, and 2 with the Photoshop CS4program (Adobe).

Angiogenic Factor Assay. The presence of pro-/antiangiogenic fac-tors in cell supernatants and cell lysates was assessed by using theProteome Profiler Antibody Array (R&D) specific for mouse orhuman angiogenesis. Cell lysates and cell supernatants were pre-pared as follows: (i) sorted CD45+CD13+CD11b+ cells (1.5 × 106

cells, obtained by pooling 10 LLC tumors) were lysed in 1% TritonX-100, 2 mM EDTA, and 50 mM Tris·HCl in PBS (pH 7.4),supplemented with protease inhibitors (CompleteMini; Roche);(ii) sorted CD45+CD11b+CD13+, CD45+CD11b+CD13−, andCD45+CD11b−CD13+ (1.5 × 105) cells were plated in DMEM and2% FBS, and, after 5 d, their supernatants were collected;and (iii) CD45+CD11b+CD13+, CD45+CD11b+CD13−, andCD45+CD11b−CD13+ (1 × 104) cells were incubated for 5 d withhuman pericytes in pericyte growth medium and 2% FBS, withoutgrowing factors. The supernatants were collected and stored at–20 °C until analysis.

Cell-Binding Studies.We used the biopanning and rapid analysis ofselective ligands (BRASIL) assay as described (3). Briefly, CD45+

CD13+, CD45+CD13−, CD31+CD13+, and CD31+CD13− cellswere sorted as described above. Cells (6 × 103) were incubatedwith NGR-targeted phage or empty (fd-tet) control phage (2 ×108) on ice for 2 h. The cell–phage admixture was carefullylayered on top of oil (dibutyl pthalate-5 cyclohexane,1:50 so-lution) in microcentrifuge tubes and centrifuged at 1 × 104 × gfor 10 min, to separate cell-bound from unbound phage. Tubeswere snap-frozen at −80 °C. The bottoms of the tubes were cutout and incubated with K91kan Escherichia coli cells for15 min. Bacteria were plated on dual-antibiotic selection(kanamycin plus tetracycline) plates at different dilutions andwere incubated ON at 37 °C, after which time, the number ofcolonies was counted.

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 1 of 7

Page 8: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

1. Guzman-Rojas L, et al. (2012) Cooperative effects of aminopeptidase N (CD13)expressed by nonmalignant and cancer cells within the tumor microenvironment. Proc NatlAcad Sci USA 109(5):1637–1642.

2. Rangel R, et al. (2007) Impaired angiogenesis in aminopeptidase N-null mice. Proc NatlAcad Sci USA 104(11):4588–4593.

3. Giordano RJ, Cardó-Vila M, Lahdenranta J, Pasqualini R, Arap W (2001) Biopanning andrapid analysis of selective interactive ligands. Nat Med 7(11):1249–1253.

Fig. S1. Characterization of CD13 WT and KO bone marrow transplanted mice. (A) Scheme of the experimental BMT models. WT and KO labels designate WTor KO mice untransplanted; WTwt and KOwt designate irradiated WT or KO mice, respectively, transplanted with bone marrow derived from WT mice; andWTko and KOko designate irradiated WT or KO mice, respectively, transplanted with bone marrow derived from KO mice. Yellow lightning bolts representradiation. (B) Representative PCR analysis of CD13 genotypes from tail tip (T) and blood (B) samples; 879-and 320-bp bands indicate WT- or KO-derived DNA,respectively. (C) Blood cell counts and hematological analysis in transplanted and control animals. HCT, hematocrit; HGB, hemoglobin; RBC, red blood cells;WBC, white blood cells. Each circle, square, or dot represents one mouse. Mean ± SEM is shown. (D) H&E-stained femur sections of representative mice. (E)Intracellular production of interleukin-2 (IL-2), tumor necrosis factor-α (TNFα), and interferon-γ (IFNγ) by lymphocytes (CD45+CD3+) and myeloid cells (CD45+

CD11b+) from reconstituted BMT mice, as measured by FACS analysis. Mean ± SEM is shown (n = 3).

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 2 of 7

Page 9: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Fig. S2. Schematic representation of CD13 expression by stromal cells within the tumor microenvironment of BMT mice. CAFs, carcinoma-associated fibroblasts.

Fig. S3. Evaluation of tumor angiogenesis in TSA and B16-F10 tumor-bearing BMT mice. Immunohistochemical analysis of CD31 (endothelial cells, red) andNG2 (pericytes, green) expression in blood vessels of B16-F10 tumors (A) and TSA tumors (B). The number of CD31+ blood vessels per field and the number ofNG2+ blood vessels per field (mean ± SE) averaged from 10 random fields are shown. DAPI, blue. *P < 0.05; **P < 0.01; ***P < 0.001 by two-tailed Studentt test. (Scale bar, 20 μm.)

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 3 of 7

Page 10: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Fig. S4. Analysis of the binding of NGR-targeted phage to sorted cells. Sorted CD45+CD13+, CD45+CD13−, CD31+CD13+, and CD31+CD13− cells were incubatedwith NGR phage. Bound phage was recovered by the BRASIL methodology (3). Results are displayed as the number of colonies obtained for each cell pop-ulation incubated with NGR-targeted phage normalized on the same population incubated with control fd-tet phage. CFU, colony forming units. Experimentswere performed twice in quadruplicate. Representative results from one experiment are shown as mean ± SEM.

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 4 of 7

Page 11: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Fig. S5. Effect of sorted BMDCs on HDMEC migration (wound-healing assay). Confluent CFSE-labeled HDMECs were scratched by using a pipet tip and in-cubated with sorted CD13+ myeloid cells (CD11b+CD13+), CD13+ nonmyeloid immune cells (CD11b−CD13+), and CD13− myeloid cells (CD11b+CD13−). Imageswere taken at days 0, 1, 2, and 3 after scratching (10× objective). A quantification of the wound area at days 0, 1, and 2 is reported (mean ± SE). Experimentswere performed two times in quadruplicate. Data from one representative experiment are shown.

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 5 of 7

Page 12: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Fig. S6. Analysis of angiogenic proteins expressed by CD11b+CD13+ myeloid cells and by pericytes cocultured with sorted BMDCs. (A) CD11b+CD13+ myeloidcells were isolated by FACS from LLC tumors grown in CD13 WT mice. Cell lysate was analyzed with the Mouse Angiogenesis Antibody Array Kit (R&D). Im-munoreactive spots (in duplicates) are shown. Immunoreactive control spots are also shown at the corners. (B) Human pericytes were cocultured with CD11b+

CD13+, CD11b+CD13−, and CD11b−CD13+ cells isolated by FACS from LLC tumors grown in CD13 WT mice and cultured for 5 d. The supernatants were analyzedas indicated above.

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 6 of 7

Page 13: CD13-positive bone marrow-derived myeloid cells promote ...€¦ · Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030; eDivision of Molecular Oncology,

Fig. S7. Distribution and phenotype of CD13+CD11b+ myeloid cells. (A) LLC, B16-F10, or TSA tumors were disaggregated into single cell suspensions andCD13+CD11b+ cells were detected by using FACS. (B) Biodistribution of CD13+CD11b+ cells in organs. bm, bone marrow. Experiments were performed threetimes, and at least three tumors were analyzed per experiment. Mean ± SEM of a representative experiment is shown. (C) Single cell suspensions wereprepared from TSA-derived tumors and were analyzed by using FACS. CD13-expressing cells were identified as CD45+CD11b+Ly6G−Ly6C+CD13+ monocytes,CD45+CD11b+F4/80+CD13+ macrophages, CD45+CD11b+Ly6G+Ly6C+ granulocytes, CD45+CD11c+CD13+ dendritic cells, and CD45+CD4+CD13+ CD4 and CD45+

CD8+CD13+ CD8 T lymphocytes (n = 4).

Dondossola et al. www.pnas.org/cgi/content/short/1321139110 7 of 7