Purpose: LRP1 (low-density lipoprotein receptor–related protein 1) is a broadly expressed receptor that binds multiple extracellular ligands and participates in protein clearance. It is expressed in numerous cancers, but its role in lung cancer has not been characterized. Here, we investigate the relationship between LRP1 and lung cancer.

Experimental Design:LRP1 mRNA levels were determined in lung tumors from several large, multicenter studies. LRP1 protein localization was determined by immunohistochemical analysis of lung tumor microarrays. Normal fibroblasts, fibroblasts treated with the LRP1 inhibitor RAP (receptor-associated protein), and Lrp1 null fibroblasts were cocultured with 3 independent lung cancer cell lines to investigate the role of LRP1 on tumor cell proliferation.

Results:LRP1 mRNA levels are significantly decreased in lung tumors relative to nontumorous lung tissue. Lower expression of LRP1 in lung adenocarcinomas correlates with less favorable clinical outcome in a cohort of 439 patients. Immunohistochemical analysis shows that LRP1 is primarily expressed in stromal cells in 94/111 lung cancers, with very little protein found in cancer cells. A growth-suppressive function of mouse embryonic fibroblast (MEF) cells was observed in 3 lung cancer cell lines tested (H460, H2347, and HCC4006 cells); growth suppression was blocked by the LRP1 inhibitor RAP. Lrp1 deletion in fibroblasts reduced the ability of MEF cells to suppress tumor cell mitosis. In a validation set of adenocarcinomas, we confirmed a significant, positive correlation between both LRP1 mRNA and protein levels and favorable clinical outcomes.

Conclusions:LRP1 expression is associated with improved lung cancer outcomes. Mechanistically, stromal LRP1 may non–cell autonomously suppress lung tumor cell proliferation. Clin Cancer Res; 17(8); 2426–33. ©2011 AACR.

Translational Relevance

LRP1 (low-density lipoprotein receptor–related protein 1) is an essential protein that participates in multiple physiologic functions, but its role in lung cancer pathogenesis is not known. We find that low LRP1 expression correlates with poor prognosis in a large, multicenter cohort of lung adenocarcinoma patients. LRP1 plays a direct role in suppression of cancer cell growth, since coculture experiments show that the presence of functional LRP1 in fibroblasts inhibits tumor cell proliferation. These findings are significant because they suggest that enhancement of LRP1 function could improve clinical outcomes in lung cancer. Moreover, since LRP1 is expressed in tumor stroma, which may be more stable than neoplastic tissues, controlling LRP1 function could be more effective than treatments that target rapidly adapting cancer cells.

The effects of the tumor stroma on the behavior of cancer are known to be dual-natured (1). On the one hand, early studies showed that reactive stroma in Rous sarcoma virus–infected chickens provides a receptive environment for cancer development (2). Factors such as VEGF are secreted into the stroma, providing an angiogenic environment with increased vascular permeability that facilitates matrix protein deposition and tumor propagation (3–5). Increased deposition of extracellular matrix components such as proteolycans and tenascin C is also strongly predictive of poor clinical prognosis in bladder and breast cancer (6, 7). On the other hand, other investigators have shown that stromal matrix components can repress cancer cells under specific circumstances. For example, inhibition of collagen fibril formation increases B16F10 melanoma tumor growth in a mouse model (8). Therefore, prior work supports the concept that tumor stroma exerts divergent and context-specific effects on cancer.

Newer data now suggest that the divergent effects of stroma on cancer progression could result from heterogeneity of the tumor stroma itself. In colon cancer, increased stromal myofibroblast content within the tumor predicts tumor recurrence (9). In a cohort of breast cancer patients, individuals whose tumors contained high levels of platelet-derived growth factor (PDGF)-B receptor within the stroma had less favorable outcomes (10). In non–small cell lung cancer, periostin expression in the stroma predicted poor clinical outcome (11). The association between stromal phenotypes and clinical outcomes has been further refined at the molecular level by recent studies that link breast cancer stromal gene expression patterns to patient outcomes (12) and tumor chemoresponsiveness (13).

Tumor fibroblasts have emerged as an important regulator within the stroma that may ultimately define whether the stroma promotes or inhibits cancer progression (14–17). Fibroblasts provide proteolytic enzymes that actively enhance growth and invasiveness (18) and increase the metastatic tumor size (19). In several mouse models, cancer-associated fibroblasts (as compared with normal fibroblasts) accelerate invasiveness of tumors (20), tumor growth (21), metastasis (22), and angiogenesis within the tumor (23) whereas normal fibroblasts have been shown to inhibit cell growth and recruit inflammatory defense systems. Tumor fibroblasts frequently secreted growth factors such as TGF-β and PDGF, whose levels of expression can stimulate mitogenic activity in cancer cells (24–26). Specific molecules expressed in lung cancer stromal fibroblasts have not been functionally characterized.

LRP1 (low-density lipoprotein receptor–related protein 1) is a large transmembrane receptor that is abundantly produced by fibroblasts. It acts as both a signaling receptor and a clearance receptor. Its substrates and ligands include more than 30 molecules with highly diverse function; therefore, LRP1 exerts multiple context-specific functions on normal cell physiology (27, 28).

Although early studies suggested that LRP1 was expressed in fibroblasts and excluded in cancers (29, 30), it has been found in a wide range of human malignancies. The expression of LRP1in vitro was lower in cell lines that exhibited increased invasiveness (31). But in other studies, increased LRP1 levels correlated with high levels of invasiveness and silencing of LRP1 prevented spread of malignant cells (32).

In addition, a number of studies have suggested a role of LRP1 in regulation of tumor growth. The expression levels of LRP1 were observed to decrease during the progression of melanoma (33). In gliomas, the magnitude of LRP1 expression in tumors greatly exceeds its levels in normal brain (34); the protein is produced by glioma cells, and its expression correlates with aggressiveness of the cancer (35).

In lung cancer, little is known about LRP1 and its potential function. Yamamoto and colleagues showed very low LRP1 mRNA expression in a small set of lung tumors and the protein was present in the stroma in 1 of 10 lung cancer samples (35). In this study, we conducted a more comprehensive analysis of the level of expression and stromal distribution of LRP1; we determined whether LRP1 expression is linked to clinical outcomes in a large gene expression array study (36), localized LRP1 protein to the stroma within lung tumors by using tissue microarrays (TMA), and characterized the functional effects of LRP1 on lung cancer cell proliferation.

Microarray data and statistical analysis

Our previously described Affymetrix microarray data set representing 439 lung adenocarcinomas (36) was used to test the relation between LRP1 expression and clinical variables and also to define genes coregulated with LRP1 in lung cancer. Characteristics of this set of clinical samples are summarized in Supplementary Table S1. t tests were used to identify statistically differences in mean gene expression levels between different clinical variables. Survival curves were constructed using the method of Kaplan–Meier, and survival differences assessed using the log-rank test. The multivariate (adjusted by sex, age, and stage) Cox proportional hazards model with continuous values of LRP1 mRNA was used to assess survival, censored at 5 years. P < 0.05 values were considered to indicate statistical significance. The Pearson correlation method was used to test the correlation between the expression of LRP1 and other genes in tumors. The hierarchical clustering with TreeView (37) was used for the representation of LRP1-correlated genes in 439 lung adenocarcinomas. Meta-analysis of LRP1 mRNA expression in multiple cancers was done by the Oncomine Web site (https://www.oncomine.org).

Immunohistochemistry

Lung tumor TMAs were constructed at the University of Michigan Cancer Center, Ann Arbor, MI. Sections were analyzed by immunohistochemical staining by standard techniques. Briefly, after deparaffinization and rehydration, antigen retrieval was done by microwave treatment; immunoperoxidase staining was conducted using a DAKO autostaining system. All sections were counterstained with hematoxylin. Digital images were captured using an Olympus system. Monoclonal antibody 8G1 (anti-LRP1; Santa Cruz Biotechnology) was applied at a 1:50 dilution.

Cell culture

Mouse embryonic fibroblasts (MEF) and PEA13 cells have been described before; these cells were propagated in Dulbecco's modied Eagle's medium (DMEM) with 10% FBS (Invitrogen). H460, H2347, and HCC4006 lung cancer lines were selected for studies because they showed very low LRP1 mRNA expression levels. Lung cancer cells were propagated in RPMI 1640 with 10% FBS. Cocultures were carried out in DMEM complete media. Cocultures were grown for 24 hours before the addition of BrDU (bromodeoxyuridine); after an additional 24 hours, cells were double stained for BrdU and TRA1-85 (70 ng/mL), which reacts against human (but not mouse) CD147. Double-stained cells were counted as human cancer cells that had undergone mitosis. Unless noted, each experiment was repeated at least 3 times.

Western blots

Cell monolayers were rinsed with PBS, harvested, and sonicated in lysis buffer (50 mmol/L Tris-HCl, pH 8; 200 mmol/L NaCl; 0.5% Nonidet-40; protease inhibitor cocktail; Pierce). Cell lysates were centrifuged at 14,000 rpm for 10 minutes at 4°C, and supernatants were used for Western blot analysis with the monoclonal antibody 11H4, which detects the 85-kDa B-chain of LRP1. IRDye-labeled secondary antibodies (Rockland; 1:10,000) were incubated with filters and then detected with an Odyssey imaging system (LI-COR Biosciences).

LRP1 mRNA expression and DNA copy number changes in a validation set of adenocarcinomas

An independent set of adenocarcinomas was analyzed for mRNA expression, DNA copy number change, and protein expression from patients at the University of Michigan. The characteristics of this set of clinical variables are summarized in Supplementary Table S1.

Total RNA was extracted using the RNeasy Mini Kit (Qiagen; catalogue no. 217004) according to the manufacturer's instructions. cDNA was prepared from RNA samples by using the High Capacity cDNA Reverse Transcription Kit (Applied Biosytems) according to the manufacturer's instructions.

Quantitative reverse transcriptase-PCR (qRT-PCR) was done by Power SYBR Green PCR Master Mix (Applied Biosystems; PN.4367659) on an ABI Prism 7900HT Sequence Detection system using a 2-temperature cycling protocol: 95°C for 10 minutes, followed by 40 cycles of 97°C for 30 seconds and 65°C for 1 minute. The oligonucleotide primers for LRP1 mRNA expression are shown in Supplementary Table S2. β-Actin expression was used to normalize the total RNA content. Relative mRNA levels were assessed using the 2ΔCt method.

Genomic DNA from frozen lung tissues was extracted using UltraPure buffer-saturated phenol (Invitrogen; catalogue no. 15513-047). Primers for LRP1 DNA amplification are shown in Supplementary Table S2. The qPCR was done using the same conditions as qRT-PCR.

LRP1 expression in multiple cancers

LRP1 expression in normal and neoplastic tissue was examined using the Oncomine database that contains gene expression studies of multiple types of cancers. Organ-specific differences in LRP1 expression were observed (Supplementary Fig. S1). In some tissues, such as brain cancers, a marked increase in LRP1 expression is observed, suggesting that LRP1 plays a role in the promotion of glioma growth (Supplementary Fig. S2, right); these findings are consistent with smaller individual pathologic studies (34, 35). In contrast, multiple cancers, including lung cancer, exhibit reduced expression of LRP1 as compared with normal tissues, suggesting that LRP1 could exert context-specific functions within tumors. Notably, 8 of 8 lung cancer studies in this database showed significantly reduced expression of LRP1 transcript levels in lung cancers relative to normal lung tissue (Supplementary Fig. S2, left).

LRP1 mRNA expression and lung adenocarcinoma outcomes

We examined the relationship between LRP1 mRNA expression and survival in lung adenocarcinomas, the most common subtype of lung cancer. We analyzed individual tumors for LRP1 expression along with clinical outcome measures and discovered a significant positive correlation between preserved LRP1 expression and patient survival (Fig. 1 and Table 1) that was independent of tumor stage, age, and sex by using multivariate Cox model analysis (Table 1). LRP1 mRNA expression was not related to other variables, including age, sex, tumor stage, p53 status, smoking, and tumor differentiation. LRP1 mRNA levels did not correlate with survival in lung squamous cell carcinomas (data not shown, samples described in ref. 38).

Figure 1.

The Kaplan–Meier survival curve indicates that high levels of LRP1 mRNA are favorable for patient survival in 439 lung adenocarcinomas (higher 2/3 vs. lower 1/3, log-rank test, P = 0.047). We used a multivariable Cox model adjusted by age, gender, and stage, using continuous LRP1 mRNA values with likelihood test, P = 0.024, β = −0.26.

Figure 1.

The Kaplan–Meier survival curve indicates that high levels of LRP1 mRNA are favorable for patient survival in 439 lung adenocarcinomas (higher 2/3 vs. lower 1/3, log-rank test, P = 0.047). We used a multivariable Cox model adjusted by age, gender, and stage, using continuous LRP1 mRNA values with likelihood test, P = 0.024, β = −0.26.

Close modal
Table 1.

LRP1 mRNA expression and patient survival in 439 adenocarcinomas

GroupnHR95% CIPa
Low LRP1 146   
High LRP1 293 0.73 0.56–0.98 0.03 
GroupnHR95% CIPa
Low LRP1 146   
High LRP1 293 0.73 0.56–0.98 0.03 

NOTE: Multivariable Cox model (age, gender, and stage adjusted) using continuous LRP1 mRNA value: P = 0.024, β = −0.26.

aLog-rank test was used, high 2/3 vs. low 1/3 samples.

Tissue distribution of LRP1 in lung tumors

One interpretation of these data is that LRP1 acts within neoplastic cells to cell autonomously regulate growth. If this were true, LRP1 should be expressed by tumor cells as observed in gliomas (34, 35). Surprisingly, however, immunohistochemistry using lung tumor TMAs showed either no or very low expression of LRP1 within neoplastic lung tumor cells of 68 adenocarcinomas and 43 squamous lung cancer samples. Rather, LRP1 was abundantly expressed in stromal cells of 37% of the tumors whereas it was only strongly expressed in 5% of neoplastic tumor cells (Fig. 2; Supplementary Table S3).

Figure 2.

Immunohistochemistry of lung cancer tissue array including squamous cell cancers (A) and adenocarcinomas (B) indicates that positive LRP1 protein staining, when present, is predominantly in stroma whereas malignant tumor cells fail to strongly stain.

Figure 2.

Immunohistochemistry of lung cancer tissue array including squamous cell cancers (A) and adenocarcinomas (B) indicates that positive LRP1 protein staining, when present, is predominantly in stroma whereas malignant tumor cells fail to strongly stain.

Close modal

LRP1 and fibroblast gene expression in lung cancer

A major component of the tumor stroma includes fibroblasts, which are known to express high levels of LRP1 (29). One possible explanation for our observations that link LRP1 in lung cancer to clinical outcome could therefore be that LRP1 is simply a marker for increased tumor fibroblast quantity that may then determine outcome. To test this, we examined whether other fibroblast-expressed genes correlated with LRP1 expression and whether expression of these genes was linked to clinical outcome. Five genes associated with tumor stromal fibroblasts were modestly correlated with LRP1, but 6 other genes were not correlated (refs. 39, 40; Supplementary Fig. S3 and Supplementary Table S4), suggesting that LRP1 is not simply a marker of overall stromal fibroblast content. Rather, the lack of association between LRP1 and most fibroblast markers suggests molecular diversity of tumor stromal fibroblasts, which has been suggested by Sugimoto, who examined murine cancer models (40). Among all the fibroblast genes examined, only LRP1 levels predicted clinical outcome (Supplementary Table S4).

Direct role of LRP1 expression in suppressing cancer cell proliferation

The findings that increased LRP1 levels correlate with more favorable outcomes and that LRP1 is expressed in the stroma of tumors suggest that LRP1 may regulate tumor cell proliferation via a non–cell autonomous mechanism. To test this, we used an in vitro system to study the effects of LRP1 on proliferation of lung cancer cells. We cocultured normal MEFs or PEA13 cells (Lrp1-knockout fibroblasts; ref. 41) with H460, H2347, and HCC4006 lung tumor cells lines (which express very low levels of LRP1; Fig. 3A) and determined the number of lung cancer cells undergoing mitosis by using BrdU incorporation (Fig. 3B–D). MEF cells significantly suppressed proliferation of H460 cells compared with Lrp1-knockout fibroblasts (PEA13). This growth-suppressive effect of MEF was blocked by incubation with RAP (receptor-associated protein), which binds and inactivates LRP1. In contrast, RAP did not affect the division of any of the lung cancer cells cultured with PEA13 cells. In sum, LRP1, expressed in nontumor cells, efficiently inhibited the proliferation of 3 lung cancer cell lines.

Figure 3.

Non–cell autonomous inhibition of tumor cell line growth by Lrp1 in fibroblasts. We co-cultured MEF cells or Lrp1-null PEA13 cells with 3 lung cancer cell lines. Only MEF cells expressed detectable levels of LRP1 (A). Proliferation was quantified by double staining for human cancer cells (expressing human CD147) and BrdU incorporated into the nucleus after pulse labeling of the coculture. Proliferation of all 3 lung cancer cell lines (B–D) was inhibited by cells containing functional LRP1 (MEF cells); inhibition was blocked by RAP or genetic inactivation of LRP1. Lung cancer cell lines that were grown without coculture incorporated BrDU at more than 8 and 2 times the level of cells cocultured with MEF and PEA13 cells, respectively (data not shown). Monocultures were not affected by RAP.

Figure 3.

Non–cell autonomous inhibition of tumor cell line growth by Lrp1 in fibroblasts. We co-cultured MEF cells or Lrp1-null PEA13 cells with 3 lung cancer cell lines. Only MEF cells expressed detectable levels of LRP1 (A). Proliferation was quantified by double staining for human cancer cells (expressing human CD147) and BrdU incorporated into the nucleus after pulse labeling of the coculture. Proliferation of all 3 lung cancer cell lines (B–D) was inhibited by cells containing functional LRP1 (MEF cells); inhibition was blocked by RAP or genetic inactivation of LRP1. Lung cancer cell lines that were grown without coculture incorporated BrDU at more than 8 and 2 times the level of cells cocultured with MEF and PEA13 cells, respectively (data not shown). Monocultures were not affected by RAP.

Close modal

Genes coregulated in LRP1 in lung cancer

To identify the additional markers that may be coregulated with LRP1, Pearson correlation analysis between LRP1 and other genes was done; the top 100 most significant correlated genes are graphically displayed in Supplementary Fig. S4 and listed in Supplementary Table S5. A fraction of these coregulated genes (35%) also showed significant favorable clinical outcomes, which are highlighted in Supplementary Table S5.

Validation of findings in an independent cohort of patients

To test the durability of our findings, we examined a validation set of 101 lung adenocarcinomas (clinical characteristics shown in Supplementary Table S1) and 39 normal lung samples. We confirmed that tumors in the validation set contained significantly less LRP1 mRNA than normal tissue (Supplementary Fig. S5). Moreover, the levels of LRP1 mRNA in this cohort were statistically associated with favorable outcome (Fig. 4A and Table 2). DNA amplification or deletion of the LRP1 gene was not common (Supplementary Fig. S6A), and DNA copy number did not correlate with mRNA expression in these samples (Supplementary Fig. S6B).

Figure 4.

The Kaplan–Meier survival curve indicates that higher LRP1 mRNA and protein expression favor survival in a validation set of lung adenocarcinomas [higher 2/3 vs. lower 1/3 samples, log-rank test, P = 0.04 for mRNA, n = 101 (A); P = 0.009 for protein, n = 96 (B)]. The levels of LRP1 expression were determined by qRT-PCR. In addition, protein expression scores were determined from TMAs stained for LRP1 protein (the average of stroma, neoplastic cell, and macrophage scores was used for survival analysis). We also applied a multivariable Cox model adjusted for age, gender, and stage, using LRP1 values with likelihood test, P = 0.04, β = −0.75 for mRNA and P = 0.01, β = −0.99 for protein.

Figure 4.

The Kaplan–Meier survival curve indicates that higher LRP1 mRNA and protein expression favor survival in a validation set of lung adenocarcinomas [higher 2/3 vs. lower 1/3 samples, log-rank test, P = 0.04 for mRNA, n = 101 (A); P = 0.009 for protein, n = 96 (B)]. The levels of LRP1 expression were determined by qRT-PCR. In addition, protein expression scores were determined from TMAs stained for LRP1 protein (the average of stroma, neoplastic cell, and macrophage scores was used for survival analysis). We also applied a multivariable Cox model adjusted for age, gender, and stage, using LRP1 values with likelihood test, P = 0.04, β = −0.75 for mRNA and P = 0.01, β = −0.99 for protein.

Close modal
Table 2.

Influence of LRP1 mRNA and protein expression on patient survival in a validation set of adenocarcinomas

GroupnHR95% CIPa
Low LRP1 mRNA 34   
High LRP1 mRNA 67 0.54 0.297–0.989 0.043 
Low LRP1 protein 34   
High LRP1 protein 62 0.45 0.248–0.833 0.009 
GroupnHR95% CIPa
Low LRP1 mRNA 34   
High LRP1 mRNA 67 0.54 0.297–0.989 0.043 
Low LRP1 protein 34   
High LRP1 protein 62 0.45 0.248–0.833 0.009 

NOTE: Protein scores were derived from the average of stroma, neoplastic cell, and macrophage scores.

aLog-rank test was used, high 2/3 vs. low 1/3 samples.

We evaluated LRP1 by immunohistochemistry in the validation set of lung tumors (Supplementary Table S6) and again identified strong LRP1 protein expression in the stroma in a significant fraction of the samples (98%). Strong neoplastic cell expression of LRP1 was seen only in 13% of the tumors. LRP1 protein staining levels in the validation set showed a strong correlation between LRP1 protein expression and favorable clinical outcomes (Fig. 4B and Table 2).

Our results provide new information supporting a role of LRP1 in lung cancer. Preservation of LRP1 expression correlates with improved clinical outcomes; importantly, expression of LRP1 protein localizes predominantly to the tumor stroma, suggesting that it may modulate tumor cells via a noncell autonomous mechanism. In vitro studies support a model in which stromal fibroblasts expressing LRP1 repress tumor cell growth or proliferation. As such, we propose that LRP1 may function as a stromal tumor growth inhibitory gene product.

The well-established expression of LRP1 in fibroblasts and its central role in extracellular protein catabolism logically position it as a functionally important protein in the tumor stroma; we show that, indeed, LRP1 expressed at low levels in transformed epithelial cancer cells yet is strongly expressed in stromal cells of most cancers. In this report, we also provide 3 pieces of evidence in support of a functional role of LRP1 in the stroma. First, there is a significant correlation between LRP1 expression and favorable lung cancer outcomes, suggesting that LRP1 function in stromal fibroblasts may be a key factor in defining the effects of stroma on tumor cell growth. Second, cell culture experiments strongly suggest a direct functional role of LRP1 in growth suppression. Finally, we emphasize that LRP1 is a unique determinant of stromal character, since our analysis of gene expression of other markers of cancer-associated fibroblasts showed no statistical association between clinical outcomes and expression of 11 other established cancer fibroblast genes.

The stromal expression pattern of LRP1 within lung tumors indicates that it may serve as a potential biomarker for clinically favorable tumors. Gene expression patterns within the stroma that influence the outcome have been elegantly described in breast cancer (12, 13) and have suggested that clusters of coregulated genes underlying specific cellular programs could influence tumor growth and behavior. Because fewer than half of the cancer-associated fibroblast markers analyzed were coregulated with LRP1, it is tempting to speculate, in analogy, that a small subset of coregulated genes in lung tumor stroma (which include LRP1) determines a favorable stroma in lung cancers; however, in opposition to this, among the cancer fibroblast genes, only LRP1 was linked to clinical outcome.

LRP1 binds to more than 30 different ligands and has both signaling properties and endocytic functions (27, 28). Therefore, a large array of mechanisms could account for its activity. Two specific examples should be mentioned. First, LRP1 functions in concert with thrombospondin 2 (TSP2; THBS2), whose expression is also significantly coregulated with LRP1 expression (Supplementary Table S5; last row), to clear matrix metalloproteinases (MMP2/9) from the extracellular space (42). Since MMPs have been shown to participate both in cancer cell growth (20) and in metastasis (43), it is tempting to speculate that increased levels of LRP1 and TSP2 in stromal cells enhance the clearance of progrowth MMP expression. In addition, recent studies show that LRP1, together with TSP2, regulates the strength of Notch3 signaling in tumor cells, which inhibits lung cancer cell growth in vitro (44, 45).

Second, alpha-2-macroglobulin (A2M), a protein that targets proteases for catabolism by LRP1, is also coregulated with LRP1 in lung cancers. Notably, increased levels of A2M are strongly associated with improved clinical outcomes in our initial patient cohort (439 tumors; Supplementary Fig. S7). Thus, A2M–LRP1 cooperative activity is potentially a very potent mechanism by which the tumor stroma may regulate patient outcomes.

Notably, a majority of the clinically significant genes coregulated with LRP1 (Supplementary Table S5) are molecules that are expressed in the extracellular space, where they may functionally interact with LRP1; of the 35 genes coregulated with LRP1 and positively associated with clinical outcome, 22 (63%) could potentially interact with LRP1 (17 encoded extracellular proteins and 5 encoded transmembrane proteins). This is consistent with the possibility that tumor microenvironment is important in the modulation of cancer outcome (noncell autonomous factors) and is perhaps as influential as intracellular gene products (which are more likely to mediate cell autonomous functions within transformed cells). More work will be needed to determine whether these gene products physically and functionally interact with LRP1 to influence tumor cells.

Finally, there are potential advantages of promoting LRP1 as a part of cancer therapy. First, therapies activating LRP1 in the stroma may produce a durable effect on tumor behavior, since the tumor stroma could be more genetically stable (46) and less likely to evolve drug resistance during therapy (47); analysis of the LRP1 locus in the validation set of tumors failed to show a high rate of genetic amplification or deletion (Supplementary Fig. S6A). Second, concerted activation of a broad range of potential tumor cell extracellular pathways affected by a multifunctional molecule such as LRP1 may hold promise since neoplastic cells are not likely to simultaneously adapt to multiple antiproliferative pathways.

No potential conflicts of interest were disclosed.

We thank Dudley Strickland for the 11H4 antibody and Liangyou Rui for use of key equipment. TRA1-85 was obtained from the Developmental Studies Hybridoma Bank at the University of Iowa. We appreciate the strong support of Nancy McAnsh at the University of Michigan Cancer Center Histology Core.

This work was supported by grants NIH grants NS054724 (M.M. Wang), NS052681 (M.M. Wang), and American Heart Association Fellowship 0726004Z (H. Meng).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Mueller
MM
,
Fusenig
NE
. 
Friends or foes—bipolar effects of the tumour stroma in cancer
.
Nat Rev Cancer
2004
;
4
:
839
49
.
2.
Dolberg
DS
,
Hollingsworth
R
,
Hertle
M
,
Bissell
MJ
. 
Wounding and its role in RSV-mediated tumor formation
.
Science
1985
;
230
:
676
8
.
3.
Brown
LF
,
Guidi
AJ
,
Schnitt
SJ
,
Van De Water
L
,
Iruela-Arispe
ML
,
Yeo
TK
, et al
Vascular stroma formation in carcinoma in situ, invasive carcinoma, and metastatic carcinoma of the breast
.
Clin Cancer Res
1999
;
5
:
1041
56
.
4.
Senger
DR
,
Galli
SJ
,
Dvorak
AM
,
Perruzzi
CA
,
Harvey
VS
,
Dvorak
HF
. 
Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid
.
Science
1983
;
219
:
983
5
.
5.
Dvorak
HF
,
Sioussat
TM
,
Brown
LF
,
Berse
B
,
Nagy
JA
,
Sotrel
A
, et al
Distribution of vascular permeability factor (vascular endothelial growth factor) in tumors: concentration in tumor blood vessels
.
J Exp Med
1991
;
174
:
1275
8
.
6.
Ishihara
A
,
Yoshida
T
,
Tamaki
H
,
Sakakura
T
. 
Tenascin expression in cancer cells and stroma of human breast cancer and its prognostic significance
.
Clin Cancer Res
1995
;
1
:
1035
41
.
7.
Brunner
A
,
Mayerl
C
,
Tzankov
A
,
Verdorfer
I
,
Tschorner
I
,
Rogatsch
H
, et al
Prognostic significance of tenascin-C expression in superficial and invasive bladder cancer
.
J Clin Pathol
2004
;
57
:
927
31
.
8.
Barsky
SH
,
Gopalakrishna
R
. 
Increased invasion and spontaneous metastasis of BL6 melanoma with inhibition of the desmoplastic response in C57 BL/6 mice
.
Cancer Res
1987
;
47
:
1663
7
.
9.
Tsujino
T
,
Seshimo
I
,
Yamamoto
H
,
Ngan
CY
,
Ezumi
K
,
Takemasa
I
, et al
Stromal myofibroblasts predict disease recurrence for colorectal cancer
.
Clin Cancer Res
2007
;
13
:
2082
90
.
10.
Paulsson
J
,
Sjoblom
T
,
Micke
P
,
Ponten
F
,
Landberg
G
,
Heldin
CH
, et al
Prognostic significance of stromal platelet-derived growth factor beta-receptor expression in human breast cancer
.
Am J Pathol
2009
;
175
:
334
41
.
11.
Soltermann
A
,
Tischler
V
,
Arbogast
S
,
Braun
J
,
Probst-Hensch
N
,
Weder
W
, et al
Prognostic significance of epithelial-mesenchymal and mesenchymal-epithelial transition protein expression in non-small cell lung cancer
.
Clin Cancer Res
2008
;
14
:
7430
7
.
12.
Finak
G
,
Bertos
N
,
Pepin
F
,
Sadekova
S
,
Souleimanova
M
,
Zhao
H
, et al
Stromal gene expression predicts clinical outcome in breast cancer
.
Nat Med
2008
;
14
:
518
27
.
13.
Farmer
P
,
Bonnefoi
H
,
Anderle
P
,
Cameron
D
,
Wirapati
P
,
Becette
V
, et al
A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer
.
Nat Med
2009
;
15
:
68
74
.
14.
Ostman
A
,
Augsten
M
. 
Cancer-associated fibroblasts and tumor growth—bystanders turning into key players
.
Curr Opin Genet Dev
2009
;
19
:
67
73
.
15.
Rasanen
K
,
Vaheri
A
. 
Activation of fibroblasts in cancer stroma
.
Exp Cell Res
2010
;
316
:
2713
22
.
16.
Franco
OE
,
Shaw
AK
,
Strand
DW
,
Hayward
SW
. 
Cancer associated fibroblasts in cancer pathogenesis
.
Semin Cell Dev Biol
2010
;
21
:
33
9
.
17.
Gonda
TA
,
Varro
A
,
Wang
TC
,
Tycko
B
. 
Molecular biology of cancer-associated fibroblasts: can these cells be targeted in anti-cancer therapy?
Semin Cell Dev Biol
2010
;
21
:
2
10
.
18.
Zhang
W
,
Matrisian
LM
,
Holmbeck
K
,
Vick
CC
,
Rosenthal
EL
. 
Fibroblast-derived MT1-MMP promotes tumor progression in vitro and in vivo
.
BMC Cancer
2006
;
6
:
52
.
19.
Coucke
P
,
De Leval
L
,
Leyh
P
,
Bonjean
K
,
Siwek
B
,
Noel
A
, et al
Influence of laminin or fibroblasts upon colony formation in the mouse by B16 melanoma cell spheroids: a morphometric analysis
.
In Vivo
1992
;
6
:
119
24
.
20.
Noel
A
,
Hajitou
A
,
L'Hoir
C
,
Maquoi
E
,
Baramova
E
,
Lewalle
JM
, et al
Inhibition of stromal matrix metalloproteases: effects on breast-tumor promotion by fibroblasts
.
Int J Cancer
1998
;
76
:
267
73
.
21.
Yang
F
,
Strand
DW
,
Rowley
DR
. 
Fibroblast growth factor-2 mediates transforming growth factor-beta action in prostate cancer reactive stroma
.
Oncogene
2008
;
27
:
450
9
.
22.
Grum-Schwensen
B
,
Klingelhofer
J
,
Grigorian
M
,
Almholt
K
,
Nielsen
BS
,
Lukanidin
E
, et al
Lung metastasis fails in MMTV-PyMT oncomice lacking S100A4 due to a T-cell deficiency in primary tumors
.
Cancer Res
2010
;
70
:
936
47
.
23.
Tuxhorn
JA
,
McAlhany
SJ
,
Dang
TD
,
Ayala
GE
,
Rowley
DR
. 
Stromal cells promote angiogenesis and growth of human prostate tumors in a differential reactive stroma (DRS) xenograft model
.
Cancer Res
2002
;
62
:
3298
307
.
24.
Akhurst
RJ
. 
TGF-beta antagonists: why suppress a tumor suppressor?
J Clin Invest
2002
;
109
:
1533
6
.
25.
Derynck
R
,
Akhurst
RJ
,
Balmain
A
. 
TGF-beta signaling in tumor suppression and cancer progression
.
Nat Genet
2001
;
29
:
117
29
.
26.
Bronzert
DA
,
Pantazis
P
,
Antoniades
HN
,
Kasid
A
,
Davidson
N
,
Dickson
RB
, et al
Synthesis and secretion of platelet-derived growth factor by human breast cancer cell lines
.
Proc Natl Acad Sci U S A
1987
;
84
:
5763
7
.
27.
Herz
J
,
Strickland
DK
. 
LRP: a multifunctional scavenger and signaling receptor
.
J Clin Invest
2001
;
108
:
779
84
.
28.
Lillis
AP
,
Mikhailenko
I
,
Strickland
DK
. 
Beyond endocytosis: LRP function in cell migration, proliferation and vascular permeability
.
J Thromb Haemost
2005
;
3
:
1884
93
.
29.
Van Leuven
F
,
Cassiman
JJ
,
Van Den Berghe
H
. 
Demonstration of an alpha2-macroglobulin receptor in human fibroblasts, absent in tumor-derived cell lines
.
J Biol Chem
1979
;
254
:
5155
60
.
30.
Foca
C
,
Moses
EK
,
Quinn
MA
,
Rice
GE
. 
Differential expression of the alpha(2)-macroglobulin receptor and the receptor associated protein in normal human endometrium and endometrial carcinoma
.
Mol Hum Reprod
2000
;
6
:
921
7
.
31.
Kancha
RK
,
Stearns
ME
,
Hussain
MM
. 
Decreased expression of the low density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor in invasive cell clones derived from human prostate and breast tumor cells
.
Oncol Res
1994
;
6
:
365
72
.
32.
Dedieu
S
,
Langlois
B
,
Devy
J
,
Sid
B
,
Henriet
P
,
Sartelet
H
, et al
LRP-1 silencing prevents malignant cell invasion despite increased pericellular proteolytic activities
.
Mol Cell Biol
2008
;
28
:
2980
95
.
33.
de Vries
TJ
,
Verheijen
JH
,
de Bart
AC
,
Weidle
UH
,
Ruiter
DJ
,
van Muijen
GN
. 
Decreased expression of both the low-density lipoprotein receptor-related protein/alpha(2)-macroglobulin receptor and its receptor-associated protein in late stages of cutaneous melanocytic tumor progression
.
Cancer Res
1996
;
56
:
1432
9
.
34.
Lopes
MB
,
Bogaev
CA
,
Gonias
SL
,
VandenBerg
SR
. 
Expression of alpha 2-macroglobulin receptor/low density lipoprotein receptor-related protein is increased in reactive and neoplastic glial cells
.
FEBS Lett
1994
;
338
:
301
5
.
35.
Yamamoto
M
,
Ikeda
K
,
Ohshima
K
,
Tsugu
H
,
Kimura
H
,
Tomonaga
M
. 
Increased expression of low density lipoprotein receptor-related protein/alpha2-macroglobulin receptor in human malignant astrocytomas
.
Cancer Res
1997
;
57
:
2799
805
.
36.
Shedden
K
,
Taylor
JM
,
Enkemann
SA
,
Tsao
MS
,
Yeatman
TJ
,
Gerald
WL
, et al
Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study
.
Nat Med
2008
;
14
:
822
7
.
37.
Eisen
MB
,
Spellman
PT
,
Brown
PO
,
Botstein
D
. 
Cluster analysis and display of genome-wide expression patterns
.
Proc Natl Acad Sci U S A
1998
;
95
:
14863
8
.
38.
Raponi
M
,
Zhang
Y
,
Yu
J
,
Chen
G
,
Lee
G
,
Taylor
JM
, et al
Gene expression signatures for predicting prognosis of squamous cell and adenocarcinomas of the lung
.
Cancer Res
2006
;
66
:
7466
72
.
39.
Kalluri
R
,
Zeisberg
M
. 
Fibroblasts in cancer
.
Nat Rev Cancer
2006
;
6
:
392
401
.
40.
Sugimoto
H
,
Mundel
TM
,
Kieran
MW
,
Kalluri
R
. 
Identification of fibroblast heterogeneity in the tumor microenvironment
.
Cancer Biol Ther
2006
;
5
:
1640
6
.
41.
Willnow
TE
,
Herz
J
. 
Genetic deficiency in low density lipoprotein receptor-related protein confers cellular resistance to Pseudomonas exotoxin A. Evidence that this protein is required for uptake and degradation of multiple ligands
.
J Cell Sci
1994
;
107
:
719
26
.
42.
Yang
Z
,
Strickland
DK
,
Bornstein
P
. 
Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2
.
J Biol Chem
2001
;
276
:
8403
8
.
43.
Kessenbrock
K
,
Plaks
V
,
Werb
Z
. 
Matrix metalloproteinases: regulators of the tumor microenvironment
.
Cell
2010
;
141
:
52
67
.
44.
Meng
H
,
Zhang
X
,
Hankenson
KD
,
Wang
MM
. 
Thrombospondin 2 potentiates notch3/jagged1 signaling
.
J Biol Chem
2009
;
284
:
7866
74
.
45.
Meng
H
,
Zhang
X
,
Lee
SJ
,
Strickland
DK
,
Lawrence
DA
,
Wang
MM
. 
Low density lipoprotein receptor-related protein-1 (LRP1) regulates thrombospondin-2 (TSP2) enhancement of Notch3 signaling
.
J Biol Chem
2010
;
285
:
23047
55
.
46.
Kopantzev
EP
,
Vayshlya
NA
,
Kopantseva
MR
,
Egorov
VI
,
Pikunov
M
,
Zinovyeva
MV
, et al
Cellular and molecular phenotypes of proliferating stromal cells from human carcinomas
.
Br J Cancer
2010
;
102
:
1533
40
.
47.
Kerbel
RS
. 
A cancer therapy resistant to resistance
.
Nature
1997
;
390
:
335
6
.