Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

CD163 Expression Was Associated with Angiogenesis and Shortened Survival in Patients with Uniformly Treated Classical Hodgkin Lymphoma

  • Young Wha Koh,

    Affiliation Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, South Korea

  • Chan-Sik Park,

    Affiliation Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea

  • Dok Hyun Yoon,

    Affiliation Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea

  • Cheolwon Suh,

    Affiliation Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea

  • Jooryung Huh

    jrhuh@amc.seoul.kr

    Affiliation Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea

Abstract

Background

Recent studies have reported the prognostic value of tissue-associated magrophages (TAMs) in classical Hodgkin lymphoma (cHL). In addition, TAMs are implicated in the tumor angiogenesis. In this study, we examined the prognostic relevance of TAMs in relation to vascular endothelial growth factor (VEGF) expression and angiogenesis in uniformly treated cases of cHL.

Methods

Diagnostic tissue from 116 patients with ABVD-treated cHL was evaluated retrospectively by immunohistochemical analysis for CD68, CD163 and VEGF expression and for CD31 expression as a measure of microvessel density (MVD).

Results

High CD163 expression (≥35% of cellularity) correlated with VEGF expression (Pearson’s Chi-square test, P = 0.008) and MVD (Spearman correlation coefficient 0.310, P<0.001). High CD163 expression was associated with inferior event-free survival (EFS, P = 0.005) and overall survival (OS, P<0.001) in univariate analysis. In multivariate analysis, high CD163 expression was strongly associated with inferior EFS (P = 0.043) and OS (P = 0.008). Patients with high MVD had a lower OS than those with low MVD, but the difference was not significant (P = 0.071, respectively). While high expression of CD68 was also associated with inferior EFS (P = 0.007), it showed no correlation with VEGF or MVD.

Conclusions

Our data confirms that CD163 expression provides independent prognostic information in cHL. The correlation of CD163 with VEGF expression and MVD suggests the role of CD163-positive cells in tumor angiogenesis of cHL.

Introduction

Classical Hodgkin lymphoma (cHL) is characterized by the disruption of the normal lymph node architecture by the presence of few of Hodgkin/Reed-Sternberg (HRS) cells in a background of reactive bystander cells mainly composed of T and B lymphocytes, macrophages and other cell types [1]. cHL is associated with high cure rates; However, despite significant advances in treatment, there remains a significant minority of patients with refractory disease in whom prolonged exposure to initial therapy induces chemo-resistance and unnecessary toxicity [2]. The major challenge remains to tailor treatments to eradicate cHL with minimal side-effects and to find biological predictive markers for patients who need intensive therapy [2].

The tumor microenvironment is emerging as an important player in the progression of malignant tumors including cHL. Recently, tumor-associated macrophages (TAMs) in lesional tissues have been shown to be a strong prognostic indicator of cHL by gene expression profile analysis and subsequent immunohistochemical detection using CD68 and CD163 as markers [3][6]. Furthermore, the peripheral blood lymphocyte/monocyte ratio at diagnosis in cHL was reported to be a prognostic factor of clinical outcome [7], [8].

Vascular endothelial growth factor (VEGF) plays an important role in physiologic and pathologic angiogenesis, including neoangiogenesis in tumors [9], [10]. VEGF expression has also demonstrated prognostic value in several solid malignancies [11], [12]. Bevacizumab is a humanized VEGF antagonist approved by the Food and Drug Administration for use in the treatment of human solid cancers [13], [14]. By blocking VEGF binding to the VEGF receptor, bevacizumab interferes with tumor angiogenesis. VEGF inhibition has shown significant survival benefit in several types of solid malignancies. Previous studies have established that VEGF is expressed in cHL [15], [16].

CD31 is a reliable marker of the vascular endothelium. Quantification of CD31 stained vessels in tumors is a standard method of measuring of intra-tumoral microvessel density (MVD) [17], a useful prognostic indicator in various malignant tumors. [18], [19] MVD is correlated with the biologic behavior of non-Hodgkin’s lymphoma [20][22] as well as cHL [16], [23], [24].

Although the mechanisms by which TAMs affect cancer progression are still unclear and are probably multifactorial, one of the potential tumor-promoting functions of TAMs is as a proangiogenetic factor via the expression of angiogenic factors, such as VEGF [25], [26]. A recent study reported a positive correlation between CD68-positive TAMs and MVD in cHL [27]. However, CD163 may be a superior marker of TAMs due to its higher specificity, as compared to CD68, for M2 macrophages [28], which are involved in tumor angiogenesis and progression, [29], [30]. While associations between TAMs, VEGF and MVD have been observed in several malignancies [31][34], no study has examined the relationship and prognostic implication of CD68, CD163, and VEGF expression and MVD in cHL patients. This retrospective study evaluated CD68, CD163, and VEGF expression and MVD in cHL patients to determine correlations between these markers and assess their prognostic significance.

Materials and Methods

Patients

The present research was approved by the Internal Review Board of the Asan Medical Center. No informed consent (written or verbal) was obtained for use of retrospective tissue samples from the patients within present study, the IRB waived the need for written informed consent. This retrospective study reviewed histological and immunohistochemical data from 116 consecutive patients diagnosed with cHL at Asan Medical Center, Seoul, South Korea, between 1990 and 2012. All patients were ≥15 years of age at diagnosis, had pathologically confirmed cHL, no previous treatment, no history of malignancy and had been treated with doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) therapy regimens, with or without radiation. Paraffin-embedded tumor tissues and follow-up data were available for all included patients.

The median follow-up time was 6.167 years (interquartile range: 3.75–10.33 years). Response criteria were based on standard guidelines. Routine follow-up imaging analyses were performed every 3 months for the first 2 years, every 6 months for the next 3 years, and then annually (or whenever clinically indicated) thereafter.

Histopathological Analysis and Immunohistochemistry

All histological and immunophenotypic data of the 116 patients with cHL were reviewed by two pathologists (JH and YWK). According to the World Health Organization (WHO) criteria, the cases were subtyped as follows: nodular sclerosis (NS), lymphocyte-rich (LR), mixed cellularity (MC), lymphocyte-depleted (LD), or not otherwise specified cHL. A tissue microarray (TMA) was generated with three 1 mm diameter tumor cores from selected areas of each formalin-fixed paraffin-embedded tumor sample. TMA section were stained using an automatic immunohistochemistry staining device (Benchmark XT, Ventana Medical System, Tucson, AZ, USA). Briefly, 5 µm thick sections were transferred onto poly-L-lysine-coated adhesive slides and dried at 62°C for 30 minutes. After standard heat epitope retrieval for 30 minutes in ethylenediaminetetraacetic acid (pH 8.0), the samples were incubated with antibodies against cleaved CD68 (dilution 1∶2000, DAKO, Glostrup, Denmark), CD163 (dilution 1∶400, NOVO, Newcastle, UK), VEGF (dilution 1∶500; Pharmingen, New Jersey, USA), CD31 (dilution 1∶100; Novo, Newcastle, UK) and CD30 (1∶25 dilution, clone BER-H2, mouse monoclonal; DAKO). The sections were then incubated with biotinylated anti-mouse immunoglobulins, peroxidase-conjugated streptavidin (LSAB kit, DAKO, Glostrup, Denmark), and 3,3′-diaminobenzidine. Slides were counterstained with Harris hematoxylin.

Each case was represented by three tissue cores in the TMA, with at least ten HRS cells detected in at least one of the three core cylinders from each patient. To minimize the counting of non-specific staining in cells other than macrophages, we only counted staining in cells that were morphologically compatible with macrophages, avoiding fibroblast, endothelial cell and Hodgkin-Reed-Sternberg cells on the basis of their size, shape, and CD30 staining. We examined protein expression levels of CD68, CD163 and VEGF in 5% increment. The relative percentage of CD68-positive or CD163-positive cells relative to overall cellularity was reported as mean scores. In cases with >10% difference in scores awarded by the two pathologists, re-evaluation was performed using a double-headed microscope. The pathologists agreed on the level of CD68 expression (<30% vs. ≥30%) in 99 out of 116 cases (85.3%, k = 0.644), on the level of CD163 expression (<35% vs. ≥35%) in 102 out of 116 cases (87.9%, k = 0.662). Cutoff values for CD68, CD163, and VEGF expression that showed the most significant differences in OS were selected (Table S1). A sample was considered high-CD68 expression if positive cells made up 30% or more of the overall cellularity and high-CD163 expression if positive cells made up 35% or more of the overall cellularity. A sample was considered VEGF-positive if 25% or more of the HRS cells showed reactivity to the VEGF antibody, while a sample was considered VEGF-negative if VEGF expression was detected in <25% of the HRS cells, or in bystander cells only. CD30 stain was used as a guide to identify HRS cells for the interpretation of VEGF.

To quantify MVD, the areas of highest vascularization were selected at low power (×100), and quantified at high magnification (×400). Three hot spots were selected per case and quantified simultaneously by two pathologists. The final MVD for each case was the mean of the number of vessels counted in the three hot spots that were scored. Microvessels with a clearly defined lumen or with a well-defined linear vessel shape were included in the counts. For statistical analysis, the cases were divided into low and high MVD groups, with high MVD tumors having ≥15.33 vessels/field according to the maximal chi-square method.

In situ hybridization (ISH) analysis for EBV-encoded RNA-1 and RNA-2 (EBER) was performed and scored as described elsewhere [35].

Statistical Analysis

OS was defined as the interval between the date of diagnosis and the date of death from any cause. Follow-up of living patients (with or without events) was censored at their last follow-up date. Event-free survival (EFS) was defined as the interval between the date of diagnosis and the date of disease progression, relapse, or death from any cause. Cumulative OS and EFS were analyzed by the Kaplan-Meier method, with comparisons analyzed by log-rank testing.

Multivariate prognostic analyses were performed on OS and EFS with the Cox proportional hazards regression model using the enter method. Categorical variables were compared using the chi-square test. Continuous variables were compared using the Mann–Whitney U test and Spearman’s correlation coefficients were used to evaluate associations for continuous variables. The maximal chi-square method was used to determine the cutoff of MVD. The maximal chi-square method was adopted to evaluate which cutoff point in each data set best segregated patients into poor and good prognosis subgroups (based on the likelihood of survival), with the log-rank test as the method used to measure the strength of the grouping [36], [37]. All statistical analyses were performed using the SPSS statistical software program (version 18.0; SPSS, Chicago, IL) or R 2.15.2. All P values are two-sided associations and P<0.05 is considered statistically significant.

Results

Patient Characteristics

The clinical characteristics of the 116 patients included in the study are summarized in Table 1. Patient age ranged from 15 to 77 years (median: 35 years). Forty-four patients experienced relapse, disease progression, or death, and 20 patients died. Median OS and EFS were not reached. The estimated 5-year OS and EFS were 83.7% and 58.9%, respectively.

thumbnail
Table 1. Demographic and clinical characteristics of patients.

https://doi.org/10.1371/journal.pone.0087066.t001

CD68, CD163, VEGF, and CD31 Expression in cHL Tissues

Correlations of CD68, CD163, VEGF, and MVD with clinical variables are summarized in Table S2.

The high-CD68 expression group (CD68≥30%, n = 32, Fig. 1A) included more men (78.1% vs. 51.2%, P = 0.011), high risk IPS patients (56.3% vs. 32.1%, P = 0.02), and cases of EBER positivity (53.1% vs. 31%, P = 0.033) compared to the low-CD68 group (CD68<30%, n = 84, Fig. 1B).

thumbnail
Figure 1. CD68, CD163, vascular endothelial growth factor (VEGF), and CD31 expression in cHL tissues.

(A) High CD68 expression (≥30%). (B) Low CD68 expression (<30%). (C) High CD163 expression (≥35%). (D) Low CD163 expression (<35%). (E) High VEGF expression (≥25%). (F) Low VEGF expression (<25%). (G) High microvessel density with CD31 expression. (H) Low microvessel density with CD31 expression.

https://doi.org/10.1371/journal.pone.0087066.g001

The high-CD163 group (CD163≥35%, n = 26, Fig. 1C) included more patients who were older (69.2% vs. 32.2%, P<0.001), of the male gender (84.6% vs. 51.1%, P = 0.003), had high risk IPS (69.2% vs. 30%, P<0.001), and who showed EBER positivity (61.5% vs. 30%, P = 0.005) compared to the low-CD163 group (CD163<35%, n = 90, Fig. 1D). A statistically significant correlation was observed between high CD68 and CD163 expression (P<0.001).

Neither the high-VEGF expression (VEGF ≥25%, n = 33, Fig. 1E) nor the low-VEGF expression (VEGF <25%, n = 83, Fig. 1F) groups were associated with clinical variables.

The mean MVD of all of cases was 13 (standard deviation [SD] = 6.82), with a range of 1–40. Forty-three samples had high MVD (MVD≥15.33, Fig. 1G), while the remaining 73 had low MVD (MVD<15.33, Fig. 1H). Low MVD was significantly associated with high levels of LDH (65.8% vs. 44.2%, P = 0.032).

Correlations of CD68, CD163 and VEGF Expression with MVD

A statistically significant correlation was observed between high CD163 and VEGF expression (P = 0.008, Table 2), and between high MVD and VEGF expression (P = 0.019, Table 2). There was no correlation between CD68 index and VEGF expression (P = 0.106).

thumbnail
Table 2. Correlations among CD68 expression, CD163 expression, VEGF expression, and microvessel density.

https://doi.org/10.1371/journal.pone.0087066.t002

We performed a correlation study on the relationship between CD68 expression, CD163 expression and MVD. There was a positive correlation between CD163 index and MVD in cHL tissues as assessed by Spearman correlation analysis regression (rho = 0.310, P<0.001, Fig. S1A). No correlation between MVD and CD68 expression was identified by Spearman correlation analysis regression (P = 0.176, Fig. S1B).

Prognostic Significance of CD68, CD163, and VEGF Expression and MVD

High-CD68 groups had lower 5-year EFS (31.7% vs. 67.7%, P<0.001; Fig. 2A) and 5-year OS (62.8% vs. 89.4%, P = 0.012; Fig. 2B) rates than low-CD68 patients. The high-CD163 group had lower 5-year EFS (31.4% vs. 65.7%, P = 0.005; Fig. 2C) and 5-year OS (60.1% vs. 89.8%, P<0.001; Fig. 2D) rates than low-CD163 patients. VEGF expression was not significantly associated with either EFS or OS (P = 0.342 and P = 0.339, respectively). Patients with high MVD had worse OS than those with low MVD (5-year OS, 77.2% vs. 87.4%; P = 0.071, Fig. 2F), although statistical significance was not reached. MVD was not significantly associated with EFS (P = 0.326, Fig. 2E).

thumbnail
Figure 2. Comparison of survival rates according to CD68, CD163, VEGF expression and MVD.

Event-free survival (EFS) (A) or overall survival (OS) (B) was significantly worse in high-CD68 group. EFS (C) or OS (D) was significantly worse in high-CD163 group. MVD was not significantly associated with EFS (E). Patients with high MVD had worse OS (F) than those with low MVD, although the statistical significance was not reached.

https://doi.org/10.1371/journal.pone.0087066.g002

Patients with high risk IPS (≥3) had lower 5-year OS rates compared to patients with low risk (71.6% vs. 91.2%, P = 0.01; Fig. S2A), however high risk IPS (≥3) was not associated with EFS rates (P = 0.098; Fig. S2B).

By univariate analysis, both OS and EFS were associated with IPS (≥3). CD68 and CD163 indices were associated with EFS and OS (Table 3). By multivariate analysis, CD68 and CD163 expression were independent prognostic markers for EFS (P = 0.007 and P = 0.034, respectively, Table 4). High CD163 expression was independent prognostic marker for OS (P = 0.026 Table 4), along with high risk IPS (≥3).

thumbnail
Table 3. Univariate analysis for overall survival (OS) and event-free survival (EFS).

https://doi.org/10.1371/journal.pone.0087066.t003

thumbnail
Table 4. Multivariate analysis for overall survival (OS) and event-free survival (EFS).

https://doi.org/10.1371/journal.pone.0087066.t004

Discussion

Inflammatory cells such as macrophages, neutrophils, and lymphocytes interact with cancer cells and express angiogenic factors [25], [38], [39]. Specifically, TAMs release a vast variety of proteolytic enzymes, cytokines, inflammatory mediators and growth factors [40]. Of these, members of the VEGF family and angiogenic peptides induce direct angiogenic effects on target endothelial cells or their bone marrow-derived precursors. TAMs also act as ‘bridge cells’ or ‘cellular chaperones’ that guide the fusion of endothelial tip cells for vascular anastomosis and facilitate vascular sprouting [41], [42]. Co-culture with macrophages promote the expressions of VEGF in lung cancer cell lines [38], [43]. In addition, TAMs are closely associated with VEGF expression and MVD in solid tumors [33], [34], [38].

In this study, a significant association of MVD with the expression of CD163 and VEGF was demonstrated in uniformly treated cHL, suggesting that the interaction between host macrophages and HRS cells may synergistically increase angiogenesis in cHL, leading to poor clinical outcome. High CD163 expression was associated with shorter EFS and OS. In contrast, VEGF or MVD did not show significant correlations with survival. Panico et al. also reported the absence of a correlation of MVD with clinical outcomes of cHL [27]. There may be several explanations for this lack of association. Firstly, TAMs may contribute to disease progression through mechanisms other than VEGF secretion or angiogenesis, which may overshadow or negate the effects of angiogenesis. In fact, TAMs contribute to extracellular matrix remodeling, promote cancer cell proliferation, invasion and metastasis; suppress the adaptive immune response [25], [40]. Secondly, VEGF-positive patients are more likely to have the NS or MC disease subtypes, which are associated with a better overall prognosis than other cHL subtypes [44], whereas CD163 expression did not show any such predilection. Thirdly, the relatively small size of the present cohort may preclude the power needed to fully demonstrate the effect of increased TAMs, thereby limiting the interpretation of the present results and calling for further validation.

Our findings confirm the superiority of CD163 as a marker of TAMs. We have shown a correlation between MVD and CD163 expression, but not with CD68 expression, which stands in contrast to the findings by Panico et al [27]. However, Panio et al. used a CD34 antibody for MVD; while we used a CD31 antibody, which is a more sensitive and specific marker of endothelial cell differentiation [45]. In most cancers, TAMs express the M2-like phenotype [25], [46], while CD68 is expressed in both M1 or M2 macrophages [30]. Previous studies yielded conflict results on the effectiveness of CD68 and CD163 expression as a measure of macrophages in cHL tissue. Kamper et al found CD163 to be less effective of CD68 [4]; however Zaki et al found prognostic value in CD163 only [47]. Other studies including the present one found independent prognostic significance for both markers [6], [48]. However, the absence of an association between CD68 expression and MVD in the present study suggests that CD68-positive macrophages have an impact on cHL progression via additional mechanisms other than angiogenesis.

The TAM phenotype is reversible, and these cells can be re-educated to exert antitumor activity [49][51]. In a recent study, re-educated CD40-activated macrophages rapidly infiltrated tumors and became tumoricidal in pancreas carcinoma [50]. In addition, angiogenic monocyte subsets can be eliminated by biotherapeutics antibodies as shown in a xenograft model [52], [53]. Anti-VEGF therapy is extensively used in solid malignant tumors [14], [54], [55], and the anti-tumor efficacy of anti-VEGF antibodies has been demonstrated in relapsed HL patients [56]. However, combining chemotherapy with bevacizumab increases the toxicities in patients with diffuse large B-cell lymphoma and peripheral T cell lymphoma [57], [58]. Further studies are warranted to determine the effect of bevacizumab in on cHL patients.

To interpret CD68 and CD163immunostaining, we used a measure of percent positivity as described in most reports [3], [59], rather than an overall visual volume estimation, which is more subject to over- or underestimation due to non-specific staining [48]. It is to be noted, however, that the cutoff values vary among previous studies, albeit in a rather narrow range for most reports. In the pioneering study by Steidl et al., patients were classified into three subgroups based on CD68 expression (<5% positive cells, IHC score 1; 5–25% positive cells, IHC score 2; and >25% positive cells, IHC score 3). While they used 5% as the cutoff in the final analysis, survival was significantly lower in patients with a score of 3 than in those with a score of 2 or 1 [3]. Four other studies also reported inferior survival using a cutoff of 25–30%[59][62], which demonstrated the reproducibility of using a 25–30% cutoff. Possible reasons for the inter-study discrepancy include disparate study populations, technical differences, use of tissue microarray vs. whole sections, inter-observer variability, and disparate use of the index of outcome. For example, Steidl et al. used a population enriched with poor-risk patients, whereas we studied a series of consecutive patients in one hospital. While the chemotherapy regimens in most studies were varied, we limited our analysis to patients treated with an ABVD regimen. For survival analysis, while survival was measured in EFS in our previous study [6], here we used OS, as the accuracy of EFS may be limited by innate limitations of radiologic examination. Our use of a more rigorous definition of the limited stage disease, the exclusion of pediatric patients, and the relatively high prevalence of EBV in our study may have contributed to the poorer outcome compared to those of Western studies [63][65]. A previous multi-center study in Korea also reported similarly poor survival rates, which may reflect ethnic and socioeconomic differences [66].

Of 32 high-CD68 cases, 20 cases had the high-CD163 expression. In the 32 cases with high-CD68 expression, cases with high-CD163 expression showed inferior OS rate than cases with low-CD163 expression, although there was no statistical significance due to the small sample size (5 year OS rate, 51.3% vs. 91.7%, P = 0.183). In multivariate analysis including CD68 and CD163, CD163 index was an independent prognostic marker for or OS (P = 0.045), however CD68 index was not an independent prognostic marker for or OS (P = 0.582). These results suggest that CD163 positive cells may be a subpopulation of CD68 positive cells although some cases showed more staining CD163 than CD68. CD163 expression particularly was associated with poor prognosis.

Limitations of this study include the retrospective design, short follow-up period, relatively small sample size and TMA-based design of the specimen preparation. TMA design cannot reflect the entire distribution of TAMs because of heterogeneity in TAMs expression with regional variation. Non-specific staining of the inflammatory background by immunophenotypic markers of TAMs also remains a formidable challenge in the clinical quantification of TAMs according to cHL risk stratification. As noted previously [67] background staining was found for both CD68 and CD163, but more so for CD68. Although every effort was made to avoid counting false positives, both CD68 and CD163 counts undoubtedly included minor populations of lymphocytes, basophils, mast cells, and other cell types.

In summary, this study is one of the first to examine the prognostic significance of TAM content in relation to VEGF expression and MVD in a uniformly treated population. Our results show that CD163 expression is associated with poor prognosis and correlates with VEGF expression and MVD, which suggests a role for TAMs in tumor angiogenesis. However, the absence of the prognostic impact of VEGF or MVD suggests that mechanisms other than angiogenesis may also be involved in the contribution of TAMs to tumor progression of cHL. Further studies are warranted to delineate the mechanism of TAM in tumor progression of cHL. Our findings provide evidence supporting new therapeutic approaches, including anti-TAM or anti-VEGF therapy in addition to the current ABVD regimen.

Supporting Information

Figure S1.

Spearman correlation among CD68, CD163, and MVD. (A) a positive correlation between CD163 index and MVD (rho = 0.310 and P<0.001). (B) No correlation between MVD and indices of CD68.

https://doi.org/10.1371/journal.pone.0087066.s001

(TIF)

Figure S2.

Comparison of survival rates according to international prognostic score (IPS). (A) Overall survival (OS) was significantly worse in the high risk IPS (≥3) group. (B) High risk IPS was not associated with EFS rates.

https://doi.org/10.1371/journal.pone.0087066.s002

(TIF)

Table S1.

CD68, CD163 and VEGF index vs. overall survival (OS).

https://doi.org/10.1371/journal.pone.0087066.s003

(DOCX)

Table S2.

Correlations between clinical variables and CD68 expression, CD163 expression, VEGF expression, and MVD.

https://doi.org/10.1371/journal.pone.0087066.s004

(DOCX)

Author Contributions

Conceived and designed the experiments: YWK JH. Performed the experiments: YWK JH. Analyzed the data: YWK JH. Contributed reagents/materials/analysis tools: YWK CP DHY CS JH. Wrote the paper: YWK JH.

References

  1. 1. Habermann TM (2013) Hodgkin’s Disease. In: Elsevier/Saunders Kellerman BI, Editor. Conn’s Current Therapy. 2013: 810–813.
  2. 2. Quddus F, Armitage JO (2009) Salvage therapy for Hodgkin’s lymphoma. Cancer J 15: 161–163.
  3. 3. Steidl C, Lee T, Shah SP, Farinha P, Han G, et al. (2010) Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med 362: 875–885.
  4. 4. Kamper P, Bendix K, Hamilton-Dutoit S, Honore B, Nyengaard JR, et al. (2011) Tumor-infiltrating macrophages correlate with adverse prognosis and Epstein-Barr virus status in classical Hodgkin’s lymphoma. Haematologica 96: 269–276.
  5. 5. Greaves P, Clear A, Coutinho R, Wilson A, Matthews J, et al. (2013) Expression of FOXP3, CD68, and CD20 at diagnosis in the microenvironment of classical Hodgkin lymphoma is predictive of outcome. J Clin Oncol 31: 256–262.
  6. 6. Yoon DH, Koh YW, Kang HJ, Kim S, Park CS, et al. (2011) CD68 and CD163 as prognostic factors for Korean patients with Hodgkin lymphoma. Eur J Haematol 88: 292–305.
  7. 7. Porrata LF, Ristow K, Colgan J, Habermann T, Witzig T, et al. (2012 ) Peripheral blood lymphocyte/monocyte ratio at diagnosis and survival in classical Hodgkin lymphoma. Haematologica 97: 262–269.
  8. 8. Koh YW, Kang HJ, Park C, Yoon DH, Kim S, et al. (2012) The ratio of the absolute lymphocyte count to the absolute monocyte count is associated with prognosis in Hodgkin’s lymphoma: correlation with tumor-associated macrophages. Oncologist 17: 871–880.
  9. 9. Folkman J, Merler E, Abernathy C, Williams G (1971) Isolation of a tumor factor responsible for angiogenesis. J Exp Med 133: 275–288.
  10. 10. Ferrara N (2000) VEGF: an update on biological and therapeutic aspects. Curr Opin Biotechnol 11: 617–624.
  11. 11. Ishigami SI, Arii S, Furutani M, Niwano M, Harada T, et al. (1998) Predictive value of vascular endothelial growth factor (VEGF) in metastasis and prognosis of human colorectal cancer. Br J Cancer 78: 1379–1384.
  12. 12. Uchida S, Shimada Y, Watanabe G, Tanaka H, Shibagaki I, et al. (1998) In oesophageal squamous cell carcinoma vascular endothelial growth factor is associated with p53 mutation, advanced stage and poor prognosis. Br J Cancer 77: 1704–1709.
  13. 13. Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, et al. (2003) Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol 21: 60–65.
  14. 14. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, et al. (2003) A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 349: 427–434.
  15. 15. Passam FH, Alexandrakis MG, Kafousi M, Fotinou M, Darivianaki K, et al. (2009) Histological expression of angiogenic factors: VEGF, PDGFRalpha, and HIF-1alpha in Hodgkin lymphoma. Pathol Res Pract 205: 11–20.
  16. 16. Doussis-Anagnostopoulou IA, Talks KL, Turley H, Debnam P, Tan DC, et al. (2002) Vascular endothelial growth factor (VEGF) is expressed by neoplastic Hodgkin-Reed-Sternberg cells in Hodgkin’s disease. J Pathol 197: 677–683.
  17. 17. Hlatky L, Hahnfeldt P, Folkman J (2002) Clinical application of antiangiogenic therapy: microvessel density, what it does and doesn’t tell us. J Natl Cancer Inst 94: 883–893.
  18. 18. Tanaka F, Otake Y, Yanagihara K, Kawano Y, Miyahara R, et al. (2001) Evaluation of angiogenesis in non-small cell lung cancer: comparison between anti-CD34 antibody and anti-CD105 antibody. Clin Cancer Res 7: 3410–3415.
  19. 19. Weidner N, Carroll PR, Flax J, Blumenfeld W, Folkman J (1993) Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma. Am J Pathol 143: 401–409.
  20. 20. Ruan J, Hyjek E, Kermani P, Christos PJ, Hooper AT, et al. (2006) Magnitude of stromal hemangiogenesis correlates with histologic subtype of non-Hodgkin’s lymphoma. Clin Cancer Res 12: 5622–5631.
  21. 21. Gratzinger D, Zhao S, Marinelli RJ, Kapp AV, Tibshirani RJ, et al. (2007) Microvessel density and expression of vascular endothelial growth factor and its receptors in diffuse large B-cell lymphoma subtypes. Am J Pathol 170: 1362–1369.
  22. 22. Ruan J, Hajjar K, Rafii S, Leonard JP (2009) Angiogenesis and antiangiogenic therapy in non-Hodgkin’s lymphoma. Ann Oncol 20: 413–424.
  23. 23. Korkolopoulou P, Thymara I, Kavantzas N, Vassilakopoulos TP, Angelopoulou MK, et al. (2005) Angiogenesis in Hodgkin’s lymphoma: a morphometric approach in 286 patients with prognostic implications. Leukemia 19: 894–900.
  24. 24. Mainou-Fowler T, Angus B, Miller S, Proctor SJ, Taylor PR, et al. (2006) Micro-vessel density and the expression of vascular endothelial growth factor (VEGF) and platelet-derived endothelial cell growth factor (PdEGF) in classical Hodgkin lymphoma (HL). Leuk Lymphoma 47: 223–230.
  25. 25. Mantovani A, Schioppa T, Porta C, Allavena P, Sica A (2006) Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev 25: 315–322.
  26. 26. Murdoch C, Muthana M, Coffelt SB, Lewis CE (2008) The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 8: 618–631.
  27. 27. Panico L, Ronconi F, Lepore M, Tenneriello V, Cantore N, et al. (2013) The prognostic role of tumor associated macrophages and angiogenesis in classical Hodgkin lymphoma. Leuk Lymphoma 54: 2418–2425.
  28. 28. Lau SK, Chu PG, Weiss LM (2004) CD163: a specific marker of macrophages in paraffin-embedded tissue samples. Am J Clin Pathol 122: 794–801.
  29. 29. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23: 549–555.
  30. 30. Ohri CM, Shikotra A, Green RH, Waller DA, Bradding P (2011) The tissue microlocalisation and cellular expression of CD163, VEGF, HLA-DR, iNOS, and MRP 8/14 is correlated to clinical outcome in NSCLC. PLoS One 6: e21874.
  31. 31. Leek RD, Hunt NC, Landers RJ, Lewis CE, Royds JA, et al. (2000) Macrophage infiltration is associated with VEGF and EGFR expression in breast cancer. J Pathol 190: 430–436.
  32. 32. Wu H, Xu JB, He YL, Peng JJ, Zhang XH, et al. (2012) Tumor-associated macrophages promote angiogenesis and lymphangiogenesis of gastric cancer. J Surg Oncol 106: 462–468.
  33. 33. Guzman-Medrano R, Arreola-Rosales RL, Shibayama M, Silva-Olivares DA, Bologna-Molina R, et al. (2012) Tumor-associated macrophages and angiogenesis: a statistical correlation that could reflect a critical relationship in ameloblastoma. Pathol Res Pract 208: 672–676.
  34. 34. Shieh YS, Hung YJ, Hsieh CB, Chen JS, Chou KC, et al. (2009) Tumor-associated macrophage correlated with angiogenesis and progression of mucoepidermoid carcinoma of salivary glands. Ann Surg Oncol 16: 751–760.
  35. 35. Huh J, Cho K, Heo DS, Kim JE, Kim CW (1999) Detection of Epstein-Barr virus in Korean peripheral T-cell lymphoma. Am J Hematol 60: 205–214.
  36. 36. Miller R, Siegmund D (1982) Maximally selected x2 statistics. Biometrics 38: 1011–1016.
  37. 37. Halpern J (1982) Maximally selected x2 statistics for small samples. Biometrics 38: 1017–1023.
  38. 38. Chen JJ, Yao PL, Yuan A, Hong TM, Shun CT, et al. (2003) Up-regulation of tumor interleukin-8 expression by infiltrating macrophages: its correlation with tumor angiogenesis and patient survival in non-small cell lung cancer. Clin Cancer Res 9: 729–737.
  39. 39. Kimura YN, Watari K, Fotovati A, Hosoi F, Yasumoto K, et al. (2007) Inflammatory stimuli from macrophages and cancer cells synergistically promote tumor growth and angiogenesis. Cancer Sci 98: 2009–2018.
  40. 40. Galdiero MR, Garlanda C, Jaillon S, Marone G, Mantovani A (2012) Tumor associated macrophages and neutrophils in tumor progression. J Cell Physiol 228: 1404–1412.
  41. 41. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, et al. (2010) Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood 116: 829–840.
  42. 42. Schmidt T, Carmeliet P (2010) Blood-vessel formation: Bridges that guide and unite. Nature 465: 697–699.
  43. 43. White ES, Strom SR, Wys NL, Arenberg DA (2001) Non-small cell lung cancer cells induce monocytes to increase expression of angiogenic activity. J Immunol 166: 7549–7555.
  44. 44. Yahalom J (2006) Favorable early-stage Hodgkin lymphoma. J Natl Compr Canc Netw 4: 233–240.
  45. 45. Pusztaszeri MP, Seelentag W, Bosman FT (2006) Immunohistochemical expression of endothelial markers CD31, CD34, von Willebrand factor, and Fli-1 in normal human tissues. J Histochem Cytochem 54: 385–395.
  46. 46. Torroella-Kouri M, Silvera R, Rodriguez D, Caso R, Shatry A, et al. (2009) Identification of a subpopulation of macrophages in mammary tumor-bearing mice that are neither M1 nor M2 and are less differentiated. Cancer Res 69: 4800–4809.
  47. 47. Zaki MA, Wada N, Ikeda J, Shibayama H, Hashimoto K, et al. (2011) Prognostic implication of types of tumor-associated macrophages in Hodgkin lymphoma. Virchows Arch 459: 361–366.
  48. 48. Tan KL, Scott DW, Hong F, Kahl BS, Fisher RI, et al. (2012) Tumor-associated macrophages predict inferior outcomes in classic Hodgkin lymphoma: a correlative study from the E2496 Intergroup trial. Blood 120: 3280–3287.
  49. 49. Watkins SK, Li B, Richardson KS, Head K, Egilmez NK, et al. (2009) Rapid release of cytoplasmic IL-15 from tumor-associated macrophages is an initial and critical event in IL-12-initiated tumor regression. Eur J Immunol 39: 2126–2135.
  50. 50. Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, et al. (2011) CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331: 1612–1616.
  51. 51. Duluc D, Corvaisier M, Blanchard S, Catala L, Descamps P, et al. (2009) Interferon-gamma reverses the immunosuppressive and protumoral properties and prevents the generation of human tumor-associated macrophages. Int J Cancer 125: 367–373.
  52. 52. Huang H, Lai JY, Do J, Liu D, Li L, et al. (2011) Specifically targeting angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltration, and tumor growth. Clin Cancer Res 17: 1001–1011.
  53. 53. De Palma M, Murdoch C, Venneri MA, Naldini L, Lewis CE (2007) Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic implications. Trends Immunol 28: 519–524.
  54. 54. Johnson DH, Fehrenbacher L, Novotny WF, Herbst RS, Nemunaitis JJ, et al. (2004) Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 22: 2184–2191.
  55. 55. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350: 2335–2342.
  56. 56. Reiners KS, Gossmann A, von Strandmann EP, Boll B, Engert A, et al. (2009) Effects of the anti-VEGF monoclonal antibody bevacizumab in a preclinical model and in patients with refractory and multiple relapsed Hodgkin lymphoma. J Immunother 32: 508–512.
  57. 57. Advani RH, Hong F, Horning SJ, Kahl BS, Manola J, et al. (2012) Cardiac toxicity associated with bevacizumab (Avastin) in combination with CHOP chemotherapy for peripheral T cell lymphoma in ECOG 2404 trial. Leuk Lymphoma 53: 718–720.
  58. 58. Stopeck AT, Unger JM, Rimsza LM, LeBlanc M, Farnsworth B, et al. (2012) A phase 2 trial of standard-dose cyclophosphamide, doxorubicin, vincristine, prednisone (CHOP) and rituximab plus bevacizumab for patients with newly diagnosed diffuse large B-cell non-Hodgkin lymphoma: SWOG 0515. Blood 120: 1210–1217.
  59. 59. Casulo C, Arcila M, Bohn OL, Teruya-Feldstein J, Maragulia J, et al. (2013) Tumor associated macrophages in relapsed and refractory Hodgkin lymphoma. Leuk Res 37: 1178–1183.
  60. 60. Jakovic LR, Mihaljevic BS, Perunicic Jovanovic MD, Bogdanovic AD, Andjelic BM, et al. (2011) The prognostic relevance of tumor associated macrophages in advanced stage classical Hodgkin lymphoma. Leuk Lymphoma 52: 1913–1919.
  61. 61. Jakovic LR, Mihaljevic BS, Jovanovic MD, Bogdanovic AD, Andjelic BM, et al. (2012) Prognostic significance of Bcl-2, tumor-associated macrophages, and total neoplastic and inflammatory lymph node involvement in advanced stage classical Hodgkin’s lymphoma. Onkologie 35: 733–739.
  62. 62. Sanchez-Espiridion B, Martin-Moreno AM, Montalban C, Medeiros LJ, Vega F, et al. (2012) Immunohistochemical markers for tumor associated macrophages and survival in advanced classical Hodgkin’s lymphoma. Haematologica 97: 1080–1084.
  63. 63. Enblad G, Sandvej K, Sundstrom C, Pallesen G, Glimelius B (1999) Epstein-Barr virus distribution in Hodgkin’s disease in an unselected Swedish population. Acta Oncol 38: 425–429.
  64. 64. Keegan TH, Glaser SL, Clarke CA, Gulley ML, Craig FE, et al. (2005) Epstein-Barr virus as a marker of survival after Hodgkin’s lymphoma: a population-based study. J Clin Oncol 23: 7604–7613.
  65. 65. Koh YW, Yoon DH, Suh C, Huh J (2012) Impact of the Epstein-Barr virus positivity on Hodgkin’s lymphoma in a large cohort from a single institute in Korea. Ann Hematol 91: 1403–1412.
  66. 66. Won YW, Kwon JH, Lee SI, Oh SY, Kim WS, et al. (2012) Clinical features and outcomes of Hodgkin’s lymphoma in Korea: Consortium for Improving Survival of Lymphoma (CISL). Ann Hematol 91: 223–233.
  67. 67. Harris JA, Jain S, Ren Q, Zarineh A, Liu C, et al. (2012) CD163 versus CD68 in tumor associated macrophages of classical Hodgkin lymphoma. Diagn Pathol 7: 12.