Leukemia (2011) 25, 1502–1509 & 2011 Macmillan Publishers Limited All rights reserved 0887-6924/11 www.nature.com/leu ORIGINAL ARTICLE The absolute monocyte and lymphocyte prognostic score predicts survival and identifies high-risk patients in diffuse large-B-cell lymphoma RA Wilcox1,2, K Ristow1, TM Habermann1, DJ Inwards1, INM Micallef1, PB Johnston1, JP Colgan1, GS Nowakowski1, SM Ansell1, TE Witzig1, SN Markovic1,2 and L Porrata1 1 Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN, USA and 2Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA Despite the use of modern immunochemotherapy regimens, almost 50% of patients with diffuse large-B-cell lymphoma will relapse. Current prognostic models, including the International Prognostic Index, incorporate patient and tumor characteristics. In contrast, recent observations show that variables related to host adaptive immunity and the tumor microenvironment are significant prognostic variables in non-Hodgkin lymphoma. Therefore, we retrospectively examined the absolute monocyte and lymphocyte counts as prognostic variables in a cohort of 366 diffuse large-B-cell lymphoma patients who were treated between 1993 and 2007 and followed at a single institution. The absolute monocyte and lymphocyte counts in univariate analysis predicted progression-free and overall survival when analyzed as continuous and dichotomized variables. On multivariate analysis performed with factors included in the IPI, the absolute monocyte and lymphocyte counts remained independent predictors of progression-free and overall survival. Therefore, the absolute monocyte and lymphocyte counts were combined to generate a prognostic score that identified patients with an especially poor overall survival. This prognostic score was independent of the IPI and added to its ability to identify high-risk patients. Leukemia (2011) 25, 1502–1509; doi:10.1038/leu.2011.112; published online 24 May 2011 Keywords: monocyte; absolute lymphocyte count; diffuse largeB-cell lymphoma; prognosis Introduction The International Prognostic Index (IPI) is currently the standard prognostic tool used to predict clinical outcomes for patients with diffuse large-B-cell lymphoma (DLBCL). Developed in the pre-rituximab era, the IPI was able to stratify patients into four discrete groups with 5-year overall survival (OS) ranging from approximately 25 to 75%. However, with improved outcomes attributed to the addition of rituximab, the identification of a high-risk subset of patients with an anticipated 5-year survival of less than 50% remains a challenge with use of the IPI alone.1,2 Gene-expression profiling,3–7 immunohistochemistry-based detection of prognostic biomarkers8–16 and early interim analysis with positron emission tomography17,18 following the initiation of immunochemotherapy have all been explored as predictors that may identify high-risk patients. Although promising, many of these methods are costly, difficult to obtain, not easily Correspondence: Dr RA Wilcox, Department of Internal Medicine, Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA. E-mail: [email protected] Received 5 October 2010; revised 26 April 2011; accepted 27 April 2011; published online 24 May 2011 interpreted and require further validation. Therefore, the identification of prognostic factors that are inexpensive, widely available and easily interpreted by clinicians engaged in the care of patients with DLBCL is needed. Most of the proposed prognostic factors utilized in DLBCL include solely patient or tumor characteristics. However, geneexpression profiling studies in non-Hodgkin lymphomas (NHL) show that gene expression by tumor-infiltrating lymphocytes and myeloid-derived cells predict clinical outcomes.3,19 Lymphocytes have an important role in immune surveillance in NHL, a view supported by the observation that lymphopenia is an adverse prognostic factor in NHL of various subtypes, including DLBCL,20–23 and is widely considered an important surrogate marker of immunological reconstitution following stem cell transplantation in NHL.24 Monocytes and lymphomaassociated macrophages (LAMs) contribute to the suppression of host anti-tumor immunity and the promotion of tumor angiogenesis. Furthermore, monocytes may also provide trophic factors that directly promote the growth and survival of malignant lymphocytes, as recently shown in both B- and T-cell-derived NHL.25,26 Therefore, the aim of this study was to devise a prognostic model that is comprised of immunologically relevant host factors that is simple, applicable for routine clinical use, and sufficient to identify high-risk patients. As myeloid cells, including monocytes and their progeny, promote lymphomagenesis both directly, via the provision of trophic factors, and indirectly via the suppression of host immunity and stimulation of tumor angiogenesis, we examined the peripheral blood monocyte count at diagnosis as a prognostic factor for survival in DLBCL patients. Patients and methods Patients Consecutive patients with DLBCL who were evaluated and treated with CHOP (cyclophosphamide, hydroxydaunorubicin, oncovin, prednisone) or R-CHOP (rituximab-cyclophosphamide, hydroxydaunorubicin, oncovin, prednisone) at the Mayo Clinic, Rochester between 1993 and 2007 were considered for study participation. HIV-positive patients were excluded from this study. The laboratory values were obtained from routine automated complete blood count (CBC) determination at the time of diagnosis.27 Patient data was collected and entered into the Mayo Clinic Lymphoma Database. Our study sample included 366 DLBCL patients, none of whom declined to participate in this retrospective review. An additional 26 patients were eligible for study participation, but were excluded owing to missing laboratory data. Study approval was granted by the AMC and ALC predict survival in DLBCL RA Wilcox et al 1503 Mayo Clinic Institutional Review Board, in accordance with US federal regulations and the Declaration of Helsinki. Table 1 Patient characteristics (n ¼ 366) Characteristic Study objective The primary objective of the study was to determine the prognostic significance of the absolute monocyte and lymphocyte counts at diagnosis with both OS and progression-free survival (PFS). The absolute monocyte count (AMC) and absolute lymphocyte count (ALC) were obtained from a standard CBC obtained at the time of diagnosis. The secondary study objective was to determine whether the AMC or ALC are independently significant prognostic factors when compared with the IPI. Patient and disease characteristics included in the IPI (age (X60 vs o60 years), Ann Arbor stage (III/IV vs I/II), Eastern Cooperative Oncology Group performance status (41 vs p1), lactate dehydrogenase (4normal vs normal for age/sex) and the number of extranodal sites of disease (41 vs p1)) were utilized.28 Statistical analysis PFS and OS were estimated using the Kaplan–Meier method and two-tailed log-rank test.29 Response criteria were based on the criteria from the International Harmonization Project.30 PFS was calculated from the date of diagnosis to the date of disease progression or last follow-up. OS was calculated from the date of diagnosis to the date of death or last follow-up. The AMC was analyzed as both a continuous and dichotomized variable using a median split. Receiver operating characteristics analysis was also performed (Cox’s regression analysis) to determine the optimal cut-point for both the AMC and ALC. However, the optimal AMC cut-point determined by receiver operating characteristics analysis for the AMC (610/ml) was nearly identical to the median AMC (data not shown). Therefore, a median split was selected for subsequent analyses using the AMC. The optimal cut-point for the ALC by receiver operating characteristic analysis was 1470/ml. However, as an ALC cutpoint of 1000/ml has been utilized in previous studies,23,31,32 and failed to yield a significant difference in the ability to risk stratify patients by OS when compared with the cut-point selected by receiver operating characteristic analysis (data not shown), we chose to dichotomize the ALC using a cut-point of 1000/ml. The Cox proportional hazards model was used to evaluate the AMC and ALC as prognostic factors for PFS and OS and to adjust for other known prognostic variables included in the IPI.33 P-values were not adjusted for multiple comparisons. All two-sided P-values o0.05 were determined to be statistically significant. Results Patient characteristics The median age at diagnosis for this cohort of 366 DLBCL patients was 64 years (range, 20–92 years), as summarized in Table 1. Of these, 58% of patients were men. Median follow-up following diagnosis was 50 months for the entire cohort (range, o1 month to 146 months) and 5.5 years for censored patients. At diagnosis, the median AMC was 630/ml (range 30–4040, interquartile range 470–833) and the median ALC was 1230/ml (range 140–9200, interquartile range 870–1770). All patients were treated initially with CHOP or R-CHOP. The estimated 5-year OS for the entire cohort was 64%. No. Age (years) Median (range) % 64 (20–92) Gender Male 214 58 Ann Arbor stage I II III IV 85 96 38 147 23 26 10 40 LDH 4Normal pNormal 151 215 41 59 Extranodal sites of disease 41 p1 73 293 20 80 ECOG PS 41 p1 44 322 12 88 IPI 0 1 2 3 4 5 47 111 94 75 34 5 13 30 26 20 9 1 Treatment CHOP R-CHOP Radiation 110 256 88 30 70 24 Absolute monocyte count Median (interquartile range) 630/ml (470–833) Absolute lymphocyte count Median (interquartile range) 1230/ml (870–1770) Abbreviations: CHOP, cyclophosphamide, hydroxydaunorubicin, oncovin, prednisone; R-CHOP, rituximab-cyclophosphamide, hydroxydaunorubicin, oncovin, prednisone; ECOG, Eastern Cooperative Oncology Group; IPI, International Prognostic Index; LDH, lactate dehydrogenase; PS, performance status. Elevation in the AMC and relative lymphopenia are adverse prognostic factors To determine the prognostic significance of the AMC and ALC, both values were obtained from a CBC at the time of initial diagnosis. On univariate analysis, the AMC predicted OS as a continuous variable, whereas the relationship between the ALC and OS approached, but did not reach, statistical significance (Table 2). An elevated AMC, analyzed as a dichotomized variable using a median split, was associated with inferior OS on univariate analysis (hazard ratio 2.77, 95% confidence interval 1.92–4.09, Po0.0001). Lymphopenia defined as an ALC p1000/ml, was associated with inferior OS (hazard ratio 1.74, 95% confidence interval 1.23–2.43, P ¼ 0.002). For comparison, each of the five factors that comprise the IPI was included in the analysis. Of these, performance status (X2), lactate dehydrogenase (4normal), number of extranodal sites (41) and Ann Arbor stage (stage III/IV) were of prognostic significance on univariate analysis (Table 2). Leukemia AMC and ALC predict survival in DLBCL RA Wilcox et al 1504 Table 2 Univariate and multivariate analyses for overall survival Prognostic factors Univariate analysis HR (95% CI) AMC (continuous variable) AMC X630/ml ALC (continuous variable) ALC p1000/ml Age X60 years ECOG PS X2 LDH 4normal Number of extranodal sites 41 Ann Arbor stage III/IV 1.0008 2.77 0.80 1.74 1.25 1.64 2.14 1.60 2.04 (1.0005–1.001) (1.92–4.09) (0.62–1.01) (1.23–2.43) (0.88–1.81) (1.02–2.54) (1.52–3.00) (1.07–2.33) (1.44–2.90) Multivariate analysis P-value o0.0001 o0.0001 0.06 0.002 0.2 0.04 o0.0001 0.02 o0.0001 HR (95% CI) P-value 2.65 (1.82–3.92) o0.0001 1.56 1.21 1.07 1.61 1.12 1.45 (1.10–2.22) (0.85–1.76) (0.65–1.70) (1.12–2.33) (0.72–1.70) (0.98–2.15) 0.01 0.30 0.77 0.01 0.61 0.06 Abbreviations: AMC, absolute monocyte count; ALC, absolute lymphocyte count; CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; HR, hazard ratio; LDH, lactate dehydrogenase; PS, performance status. As components of the IPI are important and commonly used prognostic factors in DLBCL, we included components of the IPI in a multivariate analysis with both the AMC and ALC as dichotomized variables. As summarized in Table 2, only the AMC, ALC and lactate dehydrogenase were independently significant prognostic factors for OS, with hazard ratios of 2.65 (95% confidence interval 1.82–3.92, Po0.0001), 1.56 (95% confidence interval 1.10–2.22, P ¼ 0.01) and 1.61 (95% confidence interval 1.12–2.33, P ¼ 0.01), respectively. Both the AMC and ALC remained independent predictors of survival when adjusted for the IPI alone (high risk vs low/intermediate risk) on multivariate analysis, with hazard ratios of 2.61 (95% confidence interval 1.83–3.80, Po0.0001) and 1.76 (95% confidence interval 1.25–2.47, P ¼ 0.001), respectively. Similarly, the AMC and ALC were independently significant predictors of PFS when adjusted for components of the IPI on multivariate analysis, with hazard ratios of 2.5 (95% confidence interval 1.6–4.0, Po0.0001) and 3.2 (95% confidence interval 2.0–5.1, Po0.0001), respectively. An AMC X630/ml was associated with 4.8 years median OS (94 events observed; 95% confidence interval 3.7–6.8 years) and an estimated 5-year OS of 49% (95% confidence interval 42–57%). In contrast, median OS has not been reached (43 events observed; 95% confidence interval 8.3–12.2 years) for patients with an AMC o630/ml, with an estimated 5-year OS of 80% (95% confidence interval 74–86%; Figure 1a). Similarly, an elevated AMC was associated with inferior PFS (89 vs 46 events observed; median PFS 5.0 years vs not reached; 5-year PFS 50 vs 74%, Po0.0001), as shown in Figure 1b. Lymphopenia (ALC p1000/ml) was associated with inferior survival, with a median OS of 6.3 years (59 events observed; 95% confidence interval 2.9–11.6 years) and PFS of 2.4 years (66 events observed; 95% confidence interval 1.3–11.6 years) with estimated 5-year OS and PFS of 53% (95% confidence interval 44–62%) and 44% (95% confidence interval 35–54%), respectively. In contrast, the median OS (78 events observed) and PFS (69 events observed) had not been reached, with estimated 5-year OS and PFS of 70% (95% confidence interval 64–76%) and 71% (95% confidence interval 66–78%), respectively, for patients with an ALC 41000/ ml (Figures 1c and d). The AMC/ALC prognostic score identifies poor-risk patients and provides additional prognostic information when superimposed on the IPI Like the IPI, use of the AMC or ALC as single variables was unable to identify a subset of truly high-risk patients, even Leukemia though each variable alone was able to identify poor-risk patients with 5-year OS comparable to poor-risk patients identified by the IPI. Furthermore, factors included in the IPI may be co-dependent variables. For example, patients in this cohort with advanced stage disease were more likely to have an elevated lactate dehydrogenase (Po0.0001). In contrast, the AMC and ALC are each independent from the other (data not shown). Therefore, we combined the dichotomized AMC and ALC to generate the AMC/ALC prognostic score and stratified patients into three risk groups: low- (AMC o630/ml and ALC 41000/ml), intermediate- (AMC X630/ml or ALC p1000/ml) and high-risk (AMC X630/ml and ALC p1000/ml) populations. PFS and OS were analyzed using either the AMC/ALC prognostic score or the IPI. The estimated 5-year PFS among low- and intermediate-risk patients stratified by the AMC/ALC prognostic score was 83 and 59%, respectively (Figure 2a). Similarly, the estimated 5-year PFS among low-, low-intermediate- and high-intermediate-risk patients was 80, 53 and 46%, respectively, using the IPI (Figure 2b). The AMC/ALC prognostic score identified a group of poor-risk patients with a median PFS of 14 months (95% confidence interval 10–23 months) and an estimated 5-year PFS of 30%. Similarly, the median PFS was 14 months (95% confidence interval 7–95 months) and the estimated 5-year PFS was 37% among high-risk patients identified using the IPI. For both low- and intermediate-risk patients, identified by either the AMC/ALC prognostic score or the IPI, median OS either had not been reached or exceeded 6 years (in low-intermediate-risk IPI). In contrast, the median OS was 2.2 years (95% confidence interval 1.7–3.8 years), with an estimated 5-year OS of 32%, among high-risk patients identified using the AMC/ALC prognostic score (Figure 2c). By comparison, a median OS of 2.1 years (95% confidence interval 1.2–7.9 years) was observed in high-risk patients identified by the IPI, with a 38% 5-year OS (Figure 2d). Clearly, the AMC/ALC prognostic score was able to risk-stratify patients in a manner comparable to the IPI. We next examined the AMC/ALC prognostic score’s performance in the rituximab era by confining our analysis to only those patients treated with R-CHOP (n ¼ 256). As shown in Figure 3, the AMC/ALC prognostic score was able to identify low-, intermediate- and high-risk patients with 5-year PFS ranging from 26 to 88% and 5-year OS ranging from 28 to 88% (Figures 3a and c). In contrast, high-risk patients identified by the IPI experienced a 44% 5-year PFS and OS (Figures 3b and d). The AMC/ALC prognostic score not only remained prognostically significant in the rituximab era, but was at least as proficient as the IPI in identifying high-risk patients. AMC and ALC predict survival in DLBCL RA Wilcox et al 1505 Figure 1 Kaplan–Meier estimates of overall survival (a, c) and progression-free survival (b, d) in 366 diffuse large-B-cell NHL patients stratified by the absolute monocyte count (X630/ml (dotted line) vs o630/ml (solid line), in a and b) or the absolute lymphocyte count (p1000/ml (dotted line) vs 41000/ml (solid line), in c and d) observed at the time of diagnosis are shown. Figure 2 Kaplan–Meier estimates of progression-free (a, b) and overall (c, d) survival for the entire cohort of patients stratified by the AMC/ALC prognostic score (a, c) and the International Prognostic Index (b, d) are shown. The AMC/ALC score remains an independently significant prognostic factor when adjusting for the IPI. Therefore, we sought to determine whether it may provide additional prog- nostic information when combined with the IPI. To test this possibility, both low- (Figure 4a) and intermediate-risk (Figure 4b) patients were segregated by the IPI. There were too Leukemia AMC and ALC predict survival in DLBCL RA Wilcox et al 1506 Figure 3 Kaplan–Meier estimates of progression-free (a, b) and overall (c, d) survival for R-CHOP-treated patients stratified by the AMC/ALC prognostic score (a, c) and the International Prognostic Index (b, d) are shown. Figure 4 Patients identified by the IPI as either low- (a) or low-intermediate/high-intermediate risk (b) were further stratified into low- (solid line), intermediate- (dotted line) or high-risk (dashed line) groups by the AMC/ALC prognostic score (Kaplan–Meier estimates of overall survival are shown). few high-risk patients to make a similar analysis in this subgroup meaningful. The 158 low-risk and 169 intermediate-risk (highintermediate and low-intermediate were combined) patients identified by the IPI were subsequently risk stratified using the AMC/ALC prognostic score. In both cases, the AMC/ALC score was able to further risk-stratify these patients, although this was most significant in patients identified as intermediate risk by the IPI (Figure 4b). Among these patients, 5-year OS ranged from 64 to 77% among low- and intermediate-risk patients identified by the AMC/ALC prognostic score. In contrast, 21% of patients identified by the IPI as ‘intermediate-risk’, upon further riskstratification by the AMC/ALC prognostic score, were found to have dismal outcomes, with a median OS of only 19 months Leukemia (95% confidence interval 12–32 months) and 5-year OS of 21% (Figure 4b). Similar results were obtained when intermediaterisk patients treated with R-CHOP were risk-stratified by the AMC/ALC prognostic score (Figure 5). In this case, the high-risk subgroup experienced a median OS of 17 months (95% confidence interval 11–26 months) and 5-year OS of 17%. The AMC/ALC prognostic score, derived from values easily obtained from a CBC, is comparable to the IPI in its ability to risk stratify patients in the pre- and post-rituximab era. Furthermore, it provides additional prognostic information when used to sub-stratify risk groups obtained from the IPI, leading to the identification of high-risk patients otherwise classified as intermediate risk by the IPI alone. AMC and ALC predict survival in DLBCL RA Wilcox et al 1507 Figure 5 Patients treated with R-CHOP identified by the IPI as lowintermediate/high-intermediate risk were further stratified into low(solid line), intermediate- (dotted line) or high-risk (dashed line) groups by the AMC/ALC prognostic score (Kaplan–Meier estimates of overall survival are shown). Discussion Recent work, based on large gene-expression profiling and immunohistochemistry-based studies in a variety of histological subtypes of NHL, shows that the quality of tumor-infiltrating lymphocytes and myeloid-derived cells provide valuable prognostic information that, in some cases, may exceed that provided by consideration of the tumor characteristics alone.3,19,34 This implies that a prognostic system that considers features of the tumor-bearing host and the tumor microenvironment may provide prognostic information that is independent from conventional prognostic models. Therefore, we sought to investigate whether the AMC and ALC, as biomarkers for host immunity and the tumor microenvironment, affect survival in DLBCL. Myeloid-lineage cells, including monocytes and their progeny, promote tumorigenesis.35–38 These myeloid-derived cells contribute to the suppression of host antitumor immunity and have an important role in tumor angiogenesis. Not surprisingly then, development of peripheral blood neutrophilia or monocytosis are adverse prognostic factors in multiple solid tumors.39–46 Multiple mechanisms, none of which are mutually exclusive, may be invoked to explain the prognostic significance of a relative monocytosis in DLBCL. First, monocytes and monocytic myeloid-derived suppressor cells suppress host antitumor immunity by a variety of different mechanisms.47,48 For example, we have previously shown that both peripheral blood monocytes and their progeny within the tumor microenvironment inducibly express the T-cell co-inhibitory ligand B7-H1 (PD-L1).47 B7-H1 expression was found to directly impair the expansion and effector functions of tumor-specific T cells, and when expressed by myeloid-lineage cells within the tumor microenvironment led to the expansion of suppressive regulatory T cells. Next, a considerable body of evidence highlights the role of myeloid-lineage cells in supporting tumor angiogenesis, including a subpopulation of monocytes expressing the angiopoietin-2 receptor Tie2.49,50 Given the role of macrophages in lymphomagenesis, peripheral blood monocytes may represent an important reservoir of macrophage precursors. To address this possibility, the density of CD68 þ lymphomaassociated macrophages in lymph nodes was examined in this study (data not shown). Although a direct correlation between the AMC and the density of LAMs may have been anticipated, no such correlation was observed. This suggests that the heterogeneity of LAM density observed may be explained by differences in monocyte recruitment and the retention of tissueresident macrophages, as opposed to the AMC. Finally, monocytes are an important source of soluble mediators, like B-lymphocyte stimulator, that support the growth and survival of both normal and malignant B cells.51–53 Not surprisingly then, monocytes have been convincingly shown to support the growth of malignant lymphocytes in both T- and B-cell NHL.26,54 The vast majority of DLBCL originate from either germinalcenter B cells (that is, GCB type) or from B cells with a geneexpression profile resembling activated B cells (that is, ABC type). Each DLBCL subtype may preferentially exploit a particular pathway during lymphomagenesis. For example, both the STAT3 and nuclear factor-kB pathways have been implicated in the pathogenesis of ABC-DLBCL and chemokine and cytokine genes targeted by STAT3 and nuclear factor-kB regulate monocyte recruitment and ultimately LAM function and polarity. Similarly, inflammatory and hematopoietic cytokines that contribute to monocyte expansion (and the ‘leukemoid reaction’) may be differentially expressed in DLBCL, raising the possibility that differences in the AMC may, at least in part, be explained by differences in the cell of origin (either GCB or non-GCB). To examine this possibility, DLBCL specimens, in which immunohistochemical staining for CD10, BCL-6 and MUM-1 was performed at diagnosis (n ¼ 42), were classified as either GCB or non-GCB in origin using the Hans algorithm (data not shown).14 No significant difference in the mean AMC was observed between GCB and non-GCB DLBCL and the AMC remained an independent prognostic factor when adjusting for the cell of origin on multivariate analysis (data not shown). Although this observation, limited by the small sample size, suggests that the cell of origin in DLBCL may not be an important determinant of monocyte expansion in DLBCL, it certainly does not exclude the real possibility that genes differentially expressed in GCB- or non-GCB-type DLBCL regulate the biology of monocytes and their progeny within the tumor microenvironment. The contribution of immune suppression in lymphomagenesis, illustrated by post-transplant lymphoproliferative disease and HIV-associated lymphomas, highlights the important contribution of T and NK cells in immune surveillance. Therefore, lymphopenia is considered a surrogate marker of host immunological incompetence.24,55 In addition, lymphocytes (including natural killer cells) are important mediators of antibody-dependent cell-mediated cytotoxicity, and may be required for rituximab-mediated, antibody-dependent cellmediated cytotoxicity-dependent destruction of malignant B cells.56 Not surprisingly then, lymphopenia is an adverse prognostic factor in indolent and aggressive NHL, including DLBCL. For example, lymphopenia is a poor-risk feature in DLBCL patients treated with either CHOP or R-CHOP and in GCB- and non-GCB-type DLBCL.23,31 More recently, the development of new-onset lymphopenia following autologousperipheral blood stem cell transplantation was associated with a significant increase in the risk of disease relapse.32 Patients with an ALC o1000/ml following autologous-peripheral blood stem cell transplantation had an incidence of disease relapse of 92%, compared with a relapse rate of 19% among patients without lymphopenia. The evidence linking both myeloid-lineage cells and lymphopenia with lymphomagenesis, and in this study with poor PFS and OS, raises the possibility that strategies which deplete tumorigenic myeloid-lineage cells or reverse lymphopenia may Leukemia AMC and ALC predict survival in DLBCL RA Wilcox et al 1508 represent novel therapeutic strategies in DLBCL. Therapeutic strategies targeting myeloid-lineage cells, including myeloidderived suppressor cells, are currently being investigated in solid tumors and may warrant further investigation in NHL.57 In contrast, manipulation of the lymphocyte content administered with autologous stem cells during autologous-peripheral blood stem cell transplantation, in hopes of raising the ALC and improving survival, is the subject of an ongoing phase III clinical trial in relapsed NHL (NCT00566228). Given the need for improved prognostic models in DLBCL, other predictors have been investigated. Some of these, initially identified before the introduction of rituximab, no longer provide useful prognostic information in the rituximab era.8,9 Other predictive models based on gene-expression profiling, while prognostically useful, are cumbersome, difficult to interpret, require the availability of suitable biopsy material and are cost prohibitive.3,5 Although the introduction of interim positron emission tomography analysis and risk-adapted therapy is being explored, this will require further validation, as the interpretation of interim positron emission tomography scans is not always reproducible and the rate of false-positive interim scans is not insignificant.17,18 Despite the growing body of evidence highlighting the relationship between the tumor microenvironment and lymphomagenesis, the incorporation of prognostic variables associated with the tumor microenvironment into routine clinical practice has remained a challenge, as most studies exploring this relationship are based on geneexpression profiling or immunohistochemistry data. In contrast, the AMC/ALC prognostic score, derived from a CBC at diagnosis, is widely available and may be easily incorporated into clinical practice. Furthermore, the AMC/ALC prognostic score provided prognostic information independently of that included in the IPI and was able to provide additional prognostic information when used in conjunction with the IPI. This was particularly true for low-intermediate- and high-intermediaterisk patients, which collectively accounted for nearly 50% of the patients in our series. When patients identified as intermediate risk by the IPI were further risk stratified by the AMC/ALC prognostic score, approximately 20% of these patients were identified as high risk, as evidenced by the fewer than 20% longterm survivors, even in the rituximab era. Given the limited number of patients included in this retrospective study, the AMC/ALC prognostic score will require validation in an independent cohort of patients. Despite the need for independent validation, the AMC/ALC prognostic score demonstrates that variables related to the host immune response and the tumor microenvironment are worth consideration when generating prognostic indices in NHL, as these variables may provide additional prognostic information independently from conventional patient- and tumor-specific factors. Conflict of interest The authors declare no conflict of interest. Author contributions RAW designed study, performed research, analyzed data and wrote the manuscript; KR performed research, provided technical assistance and reviewed the manuscript; TMH, DJI, INMM, PBJ, JPC, GSN, SMA, TEW and SNM assisted with data analysis and manuscript preparation; LP assisted with study design, data analysis and manuscript preparation. Leukemia References 1 Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K, Hoskins P et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007; 109: 1857–1861. 2 Ziepert M, Hasenclever D, Kuhnt E, Glass B, Schmitz N, Pfreundschuh M et al. Standard International Prognostic Index remains a valid predictor of outcome for patients with aggressive CD20+ B-cell lymphoma in the rituximab era. J Clin Oncol 2010; 28: 2373–2380. 3 Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 2008; 359: 2313–2323. 4 Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000; 403: 503–511. 5 Lossos IS, Czerwinski DK, Alizadeh AA, Wechser MA, Tibshirani R, Botstein D et al. Prediction of survival in diffuse large-B-cell lymphoma based on the expression of six genes. N Engl J Med 2004; 350: 1828–1837. 6 Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 2002; 346: 1937–1947. 7 Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar RC et al. Diffuse large B-cell lymphoma outcome prediction by geneexpression profiling and supervised machine learning. Nat Med 2002; 8: 68–74. 8 Hermine O, Haioun C, Lepage E, d’Agay MF, Briere J, Lavignac C et al. Prognostic significance of bcl-2 protein expression in aggressive non-Hodgkin’s lymphoma. Groupe d’Etude des Lymphomes de l’Adulte (GELA). Blood 1996; 87: 265–272. 9 Mounier N, Briere J, Gisselbrecht C, Emile JF, Lederlin P, Sebban C et al. Rituximab plus CHOP (R-CHOP) overcomes bcl-2-associated resistance to chemotherapy in elderly patients with diffuse large Bcell lymphoma (DLBCL). Blood 2003; 101: 4279–4284. 10 Ansell SM, Stenson M, Habermann TM, Jelinek DF, Witzig TE. Cd4+ T-cell immune response to large B-cell non-Hodgkin’s lymphoma predicts patient outcome. J Clin Oncol 2001; 19: 720–726. 11 Tzankov A, Meier C, Hirschmann P, Went P, Pileri SA, Dirnhofer S. Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin’s lymphoma. Haematologica 2008; 93: 193–200. 12 Lee NR, Song EK, Jang KY, Choi HN, Moon WS, Kwon K et al. Prognostic impact of tumor infiltrating FOXP3 positive regulatory T cells in diffuse large B-cell lymphoma at diagnosis. Leuk Lymph 2008; 49: 247–256. 13 Hasselblom S, Sigurdadottir M, Hansson U, Nilsson-Ehle H, Ridell B, Andersson PO. The number of tumour-infiltrating TIA-1+ cytotoxic T cells but not FOXP3+ regulatory T cells predicts outcome in diffuse large B-cell lymphoma. Br J Haematol 2007; 137: 364–373. 14 Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 2004; 103: 275–282. 15 Nyman H, Jerkeman M, Karjalainen-Lindsberg ML, Banham AH, Leppa S. Prognostic impact of activated B-cell focused classification in diffuse large B-cell lymphoma patients treated with R-CHOP. Mod Pathol 2009; 22: 1094–1101. 16 Nyman H, Adde M, Karjalainen-Lindsberg ML, Taskinen M, Berglund M, Amini RM et al. Prognostic impact of immunohistochemically defined germinal center phenotype in diffuse large B-cell lymphoma patients treated with immunochemotherapy. Blood 2007; 109: 4930–4935. 17 Mikhaeel NG. Interim fluorodeoxyglucose positron emission tomography for early response assessment in diffuse large B cell lymphoma: where are we now? Leuk Lymph 2009; 50: 1931–1936. 18 Duhrsen U, Huttmann A, Jockel KH, Muller S. Positron emission tomography guided therapy of aggressive non-Hodgkin lymphomas Fthe PETAL trial. Leuk Lymph 2009; 50: 1757–1760. AMC and ALC predict survival in DLBCL RA Wilcox et al 1509 19 Dave SS, Wright G, Tan B, Rosenwald A, Gascoyne RD, Chan WC et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 2004; 351: 2159–2169. 20 Oki Y, Yamamoto K, Kato H, Kuwatsuka Y, Taji H, Kagami Y et al. Low absolute lymphocyte count is a poor prognostic marker in patients with diffuse large B-cell lymphoma and suggests patients’ survival benefit from rituximab. Eur J Haematol 2008; 81: 448–453. 21 Behl D, Ristow K, Markovic SN, Witzig TE, Habermann TM, Colgan JP et al. Absolute lymphocyte count predicts therapeutic efficacy of rituximab therapy in follicular lymphomas. Br J Haematol 2007; 137: 409–415. 22 Cox MC, Nofroni I, Laverde G, Ferrari A, Amodeo R, Tatarelli C et al. Absolute lymphocyte count is a prognostic factor in diffuse large B-cell lymphoma. Br J Haematol 2008; 141: 265–268. 23 Kim DH, Baek JH, Chae YS, Kim YK, Kim HJ, Park YH et al. Absolute lymphocyte counts predicts response to chemotherapy and survival in diffuse large B-cell lymphoma. Leukemia 2007; 21: 2227–2230. 24 Fruehauf S, Tricot G. Comparison of unmobilized and mobilized graft characteristics and the implications of cell subsets on autologous and allogeneic transplantation outcomes. Biol Blood Marrow Transplant 2010; 16: 1629–1648. 25 Shivakumar L, Ansell S. Targeting B-lymphocyte stimulator/B-cell activating factor and a proliferation-inducing ligand in hematologic malignancies. Clin Lymph Myel 2006; 7: 106–108. 26 Wilcox RA, Wada DA, Ziesmer SC, Elsawa SF, Comfere NI, Dietz AB et al. Monocytes promote tumor cell survival in T-cell lymphoproliferative disorders and are impaired in their ability to differentiate into mature dendritic cells. Blood 2009; 114: 2936–2944. 27 Cox CJ, Habermann TM, Payne BA, Klee GG, Pierre RV. Evaluation of the Coulter Counter model S-Plus IV. Am J Clin Pathol 1985; 84: 297–306. 28 Anonymous. A predictive model for aggressive non-Hodgkin’s lymphoma. The International Non-Hodgkin’s Lymphoma Prognostic Factors Project. N Engl J Med 1993; 329: 987–994. 29 Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958; 53: 457–481. 30 Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Horning SJ et al. Revised response criteria for malignant lymphoma. J Clin Oncol 2007; 25: 579–586. 31 Song MK, Chung JS, Seol YM, Kim SG, Shin HJ, Choi YJ et al. Influence of low absolute lymphocyte count of patients with nongerminal center type diffuse large B-cell lymphoma with R-CHOP therapy. Ann Oncol 2010; 21: 140–144. 32 Porrata LF, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Hogan WJ et al. New-onset lymphopenia assessed during routine follow-up is a risk factor for relapse postautologous peripheral blood hematopoietic stem cell transplantation in patients with diffuse large B-cell lymphoma. Biol Blood Marrow Transplant 2010; 16: 376–383. 33 Cox DR. Regression models and life tablets. J R Statist Soc Ser C 1972; 34: 187–202. 34 Ansell SM, Stenson M, Habermann TM, Jelinek DF, Witzig TE. Cd4+ T-cell immune response to large B-cell non-Hodgkin’s lymphoma predicts patient outcome. J Clin Oncol 2001; 19: 720–726. 35 Young MR, Newby M, Wepsic HT. Hematopoiesis and suppressor bone marrow cells in mice bearing large metastatic Lewis lung carcinoma tumors. Cancer Res 1987; 47: 100–105. 36 Young MR, Ellis NK, Young ME, Wepsic HT. Stimulation of hematopoiesis and bone marrow suppressor cells by the subcutaneous injection of linoleic acid. Cell Immunol 1987; 107: 238–248. 37 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9: 162–174. 38 Seung LP, Rowley DA, Dubey P, Schreiber H. Synergy between T-cell immunity and inhibition of paracrine stimulation causes tumor rejection. Proc Natl Acad Sci USA 1995; 92: 6254–6258. 39 Mandrekar SJ, Schild SE, Hillman SL, Allen KL, Marks RS, Mailliard JA et al. A prognostic model for advanced stage nonsmall cell lung cancer. Pooled analysis of North Central Cancer Treatment Group trials. Cancer 2006; 107: 781–792. 40 Schmidt H, Bastholt L, Geertsen P, Christensen IJ, Larsen S, Gehl J et al. Elevated neutrophil and monocyte counts in peripheral blood are associated with poor survival in patients with metastatic melanoma: a prognostic model. Br J Cancer 2005; 93: 273–278. 41 Schmidt H, Suciu S, Punt CJ, Gore M, Kruit W, Patel P et al. Pretreatment levels of peripheral neutrophils and leukocytes as independent predictors of overall survival in patients with American Joint Committee on Cancer Stage IV Melanoma: results of the EORTC 18951 Biochemotherapy Trial. J Clin Oncol 2007; 25: 1562–1569. 42 Michael M, Goldstein D, Clarke SJ, Milner AD, Beale P, Friedlander M et al. Prognostic factors predictive of response and survival to a modified FOLFOX regimen: importance of an increased neutrophil count. Clin Colorectal Cancer 2006; 6: 297–304. 43 Choueiri TK, Garcia JA, Elson P, Khasawneh M, Usman S, Golshayan AR et al. Clinical factors associated with outcome in patients with metastatic clear-cell renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. Cancer 2007; 110: 543–550. 44 Donskov F, von der Maase H. Impact of immune parameters on long-term survival in metastatic renal cell carcinoma. J Clin Oncol 2006; 24: 1997–2005. 45 Paesmans M, Sculier JP, Lecomte J, Thiriaux J, Libert P, Sergysels R et al. Prognostic factors for patients with small cell lung carcinoma: analysis of a series of 763 patients included in 4 consecutive prospective trials with a minimum follow-up of 5 years. Cancer 2000; 89: 523–533. 46 Takasaki Y, Iwanaga M, Tsukasaki K, Kusano M, Sugahara K, Yamada Y et al. Impact of visceral involvements and blood cell count abnormalities on survival in adult T-cell leukemia/lymphoma (ATLL). Leuk Res 2007; 31: 751–757. 47 Wilcox RA, Feldman AL, Wada DA, Yang ZZ, Comfere NI, Dong H et al. B7-H1 (PD-L1, CD274) suppresses host immunity in T-cell lymphoproliferative disorders. Blood 2009; 114: 2149–2158. 48 Wilcox RA. Cancer-associated myeloproliferation: old association, new therapeutic target. Mayo Clin Proc 2010; 85: 656–663. 49 De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi M et al. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 2005; 8: 211–226. 50 Venneri MA, De Palma M, Ponzoni M, Pucci F, Scielzo C, Zonari E et al. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood 2007; 109: 5276–5285. 51 He B, Chadburn A, Jou E, Schattner EJ, Knowles DM, Cerutti A. Lymphoma B cells evade apoptosis through the TNF family members BAFF/BLyS and APRIL. J Immunol 2004; 172: 3268–3279. 52 Novak AJ, Grote DM, Stenson M, Ziesmer SC, Witzig TE, Habermann TM et al. Expression of BLyS and its receptors in B-cell non-Hodgkin lymphoma: correlation with disease activity and patient outcome. Blood 2004; 104: 2247–2253. 53 Oki Y, Georgakis GV, Migone TS, Kwak LW, Younes A. Elevated serum BLyS levels in patients with non-Hodgkin lymphoma. Leuk Lymph 2007; 48: 1869–1871. 54 Seiffert M, Schulz A, Ohl S, Dohner H, Stilgenbauer S, Lichter P. Soluble CD14 is a novel monocyte-derived survival factor for chronic lymphocytic leukemia cells, which is induced by CLL cells in vitro and present at abnormally high levels in vivo. Blood 2010; 116: 4223–4230. 55 Porrata LF, Markovic SN. Timely reconstitution of immune competence affects clinical outcome following autologous stem cell transplantation. Clin Exp Med 2004; 4: 78–85. 56 Weiner GJ. Rituximab: mechanism of action. Semin Hematol 2010; 47: 115–123. 57 Ko JS, Bukowski RM, Fincke JH. Myeloid-derived suppressor cells: a novel therapeutic target. Curr Oncol Rep 2009; 11: 87–93. Leukemia
© Copyright 2025 Paperzz