Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2019

Open Access 01.12.2019 | Research

Prognostic or predictive value of circulating cytokines and angiogenic factors for initial treatment of multiple myeloma in the GIMEMA MM0305 randomized controlled trial

verfasst von: Ilaria Saltarella, Fortunato Morabito, Nicola Giuliani, Carolina Terragna, Paola Omedè, Antonio Palumbo, Sara Bringhen, Lorenzo De Paoli, Enrica Martino, Alessandra Larocca, Massimo Offidani, Francesca Patriarca, Chiara Nozzoli, Tommasina Guglielmelli, Giulia Benevolo, Vincenzo Callea, Luca Baldini, Mariella Grasso, Giovanna Leonardi, Manuela Rizzo, Antonietta Pia Falcone, Daniela Gottardi, Vittorio Montefusco, Pellegrino Musto, Maria Teresa Petrucci, Franco Dammacco, Mario Boccadoro, Angelo Vacca, Roberto Ria

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2019

Abstract

Background

Several new drugs are approved for treatment of patients with multiple myeloma (MM), but no validated biomarkers are available for the prediction of a clinical outcome. We aimed to establish whether pretreatment blood and bone marrow plasma concentrations of major cytokines and angiogenic factors (CAFs) of patients from a phase 3 trial of a MM treatment could have a prognostic and predictive value in terms of response to therapy and progression-free and overall survival and whether these patients could be stratified for their prognosis.

Methods

Blood and bone marrow plasma levels of Ang-2, FGF-2, HGF, VEGF, PDGF-β, IL-8, TNF-α, TIMP-1, and TIMP-2 were determined at diagnosis in MM patients enrolled in the GIMEMA MM0305 randomized controlled trial by an enzyme-linked immunosorbent assay (ELISA). These levels were correlated both reciprocally and with the type of therapy and patients’ characteristics and with a group of non-MM patients as controls.

Results

No significant differences were detected between the blood and bone marrow plasma levels of angiogenic cytokines. A cutoff for each CAF was established. The therapeutic response of patients with blood plasma levels of CAFs lower than the cutoff was better than the response of those with higher levels in terms of percentage of responding patients and quality of response.

Conclusion

FGF-2, HGF, VEGF, and PDGF-β plasma levels at diagnosis have predictive significance for response to treatment. The stratification of patients based on the levels of CAFs at diagnosis and their variations after therapy is useful to characterize different risk groups concerning outcome and response to therapy.

Trial registration

Clinical trial information can be found at the following link: NCT01063179
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13045-018-0691-4) contains supplementary material, which is available to authorized users.
Abkürzungen
ANG-2
Angiopoietin-2
CAFs
Cytokines and angiogenic factors
CI
Confidence interval
D&S
Durie and Salmon
ECOG
Eastern Cooperative Oncology Group
FGF-2
Fibroblast Growth Factor-2
HGF
Hepatocyte Growth Factor
HR
Hazard ratio
IL-8
Interleukin-8
ISS
International Staging System
MM
Multiple myeloma
OS
Overall Survival
PDGF-BB
Platelet-Derived Growth Factor-BB
PFS
Progression-Free Survival
TIMP
Tissue Inhibitor of Matrix Metalloproteinase
TNF-α
Tumor Necrosis Factor-alpha
VEGF
Vascular Endothelial Growth Factor
VGPR
Very Good Partial Response
VMP
Bortezomib-melphalan-prednisone
VMPT-VT
Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide

Background

Multiple myeloma (MM) is the second most common hematologic cancer after non-Hodgkin’s lymphoma, with a higher incidence in the elderly. Patients older than 70 years account for 56% of new cases and for 73% of all deaths from MM [1]. Combined melphalan-prednisone has been the standard of care for more than 40 years and has been found to be associated with a median survival of 29 to 37 months [24]. Today, the availability of novel agents, such as the first-in-class proteasome inhibitor bortezomib and the immunomodulatory drugs thalidomide and lenalidomide, has significantly improved the clinical outcome of these patients [513].
Accurate identification of high-risk patients and risk stratification are crucial in improving outcomes for MM patients, but considerable heterogeneity exists in their overall survival. Although a large number of prognostic markers have been described, including disease burden (Durie-Salmon staging system, International Staging System, magnetic resonance imaging, (18F)fluorodeoxyglucose positron emission tomography, presence of extramedullary disease or plasma-cell leukemia), host factors (age, performance status, and renal function), tumor biology (proliferation rate, conventional cytogenetics, interphase fluorescence in situ hybridization, and gene expression profiling), and depth of response to therapy [1418], none of them completely explains the heterogeneity seen in this tumor.
To further complicate matters, some of the new treatments appear to overcome the high risk defined by one or more of these prognostic factors [19, 20]. With the increased treatment options, the ability of some treatments to overcome certain risk factors, and the availability of markers to define risk categories, risk stratification in the management of MM is becoming an important issue [21]. The achievement of a uniform risk stratification system would also allow a better comparison of patient groups across different trials [22].
Angiogenesis is a constant hallmark of MM progression and has prognostic potential. The pathophysiology of MM-induced angiogenesis involves both direct production of angiogenic cytokines by plasma cells and their induction within the bone marrow microenvironment [23]. Moreover, inhibitors of the vascular endothelial growth factor (VEGF) pathway, including bevacizumab and tyrosine kinase inhibitors such as sorafenib, sunitinib, and pazopanib, have been shown to prolong progression-free survival (PFS) and overall survival (OS) [2428] and are in fact approved for the treatment of solid cancer.
It has been previously demonstrated that the plasma levels of cytokines and angiogenic factors (CAFs) decrease after therapy in patients with cancer, and this may be relevant for treatment response and PFS [2932]. Here, we demonstrate that high levels of CAFs are negative prognostic factors in patients with MM and seem to be predictive of relative benefit from therapy. Moreover, the stratification of patients based on CAF levels at diagnosis is useful to detect different risk groups for outcome and response to therapy.

Methods

Patients

Patient characteristics are reported in Table 1. This study has been carried out on MM patients enrolled in the multicenter clinical trial GIMEMA-MM0305, with the participation of 61 centers in Italy from May 2006 to January 2009. The study compared the combination bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide (VMPT-VT) with bortezomib-melphalan-prednisone (VMP) administered for nine cycles without maintenance. The details and results of the trial have been published previously [3335]. Clinical protocol and informed consent documents were approved by the participating local institution’s review boards, and the trial was undertaken in accordance with the International Conference on Harmonization Guidelines for Good Clinical Practice and the amended Declaration of Helsinki. Patients without MM or other tumors (patients with stage I arterial hypertension without organ damage and without other diseases) who gave their consent were used as controls.
Table 1
Patients’ characteristics
 
Patients
Controls
Total
VMP
VMPT
n
124
53
71
54
Age
71 (56–85)
71 (60–85)
71 (56–85)
69 (54–88)
Sex (M/F)
58/66
24/29
34/37
24/30
Type of MM
NA
 IgG (%)
71 (57.2)
31 (58.5)
46 (64.8)
 
 IgA (%)
28 (22.6)
12 (22.6)
14 (19.8)
 
 BJ (%)
25 (20.2)
10 (18.9)
11 (15.4)
 
Stage (D&S)
NA
 IIA (%)
14 (11.3)
5 (9.4)
9 (12.7)
 
 IIIA (%)
102 (82.3)
45 (84.9)
57 (80.3)
 
 IIIB (%)
8 (6.4)
3 (5.7)
5 (7)
 
ISS stage
NA
 1
26
8
18
 
 2
42
21
21
 
 3
21
6
15
 
 Missing data
35
18
17
 
Cytogenetics
NA
 High risk
28
11
17
 
 Standard risk
36
16
20
 
 Missing data
60
26
34
 
Response
 CR (%)
47 (37.9)
14 (26.4)
33 (46.6)
 
 VGPR (%)
27 (21.8)
11 (20.8)
16 (22.6)
NA
 PR (%)
38 (30.6)
19 (35.8)
19 (26.8)
 
 SD (%)
12 (9.7)
9 (17)
3 (4.2)
 
Relapse (Y/N)
80/44
39/14
41/30
NA
Death (Y/N)
54/70
22/31
32/39
NA
NA not applicable

Methods

Before starting the treatment, peripheral blood and bone marrow plasma (the initial 1 ml of bone marrow aspirate) samples were collected into EDTA-containing tubes. Both blood and bone marrow plasma samples from 124 of 511 patients enrolled in the study (24%) were available for analysis. Plasma was separated by centrifugation (2,000 rpm for 20 min at 4 °C) within 1 h from blood drawing and aliquoted into multiple cryovials. Plasma samples were stored at − 80 °C until use. Before analysis, plasma samples were thawed slowly in an ice bath and all analyses were done from a one-off thaw sample. CAFs were measured by using Q-Plex™ Array Human Angiogenesis Antigen (Quansys Biosciences, Logan, Utah) allowing the simultaneous quantification of the following factors: angiopoietin-2 (ANG-2), fibroblast growth factor-2 (FGF-2), hepatocyte growth factor (HGF), interleukin-8 (IL-8), platelet-derived growth factor-BB (PDGF-BB), tissue inhibitor of matrix metalloproteinase-1 and 2 (TIMP-1, TIMP-2), tumor necrosis factor-alpha (TNF-α), and vascular endothelial growth factor (VEGF), according to the manufacturer’s instructions. Secreted levels of CAFs were quantified through Q-View Software (Quansys Biosciences, Logan, Utah) in triplicate samples, and the mean results were used in biomarker analysis.

Statistical analysis

In the first step, CAF levels were measured in the blood and bone marrow plasma samples of MM patients to assess whether significant differences could be detected in the two compartments by Student’s t test (p values less than 0.05 was considered significant). Since no significant differences between the two compartments were detected (see the “Results” section), subsequent analyses were carried out using plasma samples from the peripheral blood. With the Student t test, the CAF levels in the plasma samples of the MM patients were compared with those of controls.
In the second phase, plasma levels of CAFs were measured as an independent variable to predict binary response status (≥ VGPR vs < VGPR) by logistic regression analysis. CAF plasma levels were also correlated as a continuous variable with tumor response by linear regression and logarithmic transformation to normalize CAF values. The correlation between log-CAFs and tumor response was approximately linear. Selection of individual CAF markers from the screening phase was done on the basis of results of median cutoff, ROC curve estimation of cutoff, and logistic regression analysis between dichotomized tumor response and CAF plasma levels. We assessed the association between CAF plasma levels and progression-free survival (PFS) with the Cox proportional hazard model.
To establish whether plasma levels of any CAF might have prognostic or predictive significance, the Kaplan-Meier method was used to analyze PFS and OS. We used the Cox regression model to verify significant differences noted in Kaplan-Meier curves for both treatment groups between a high- and a low-CAF subgroup, defined by the respective median CAF plasma levels. Sensitivity analyses confirmed that median cutoff achieved the most significant segregation of clinical benefit. To assess the potential differential effects of baseline CAF concentrations between two treatment groups, a treatment versus CAF status interaction term was included in the Cox model analysis for PFS, with treatment group and CAF status as two additional independent variables. CAF plasma levels with a significant interaction value with treatment were regarded as predictive. A post hoc analysis was done to adjust for multiple testing of CAF markers with the Bonferroni test. Exploratory analyses included correlation between CAF plasma levels and Eastern Cooperative Oncology Group (ECOG) performance status, Durie & Salmon stage, International Staging System stage, Cytogenetic risk, and age and sex hierarchical clustering (unweighted pair group method with arithmetic mean, unweighted average, and Euclidean distance for similarity measure) to assess a multiple-CAF signature association with PFS or OS (Kaplan-Meier method for PFS and OS, and Cox regression models to assess differences). All statistical analyses were done with SPSS software.

Results

Biological samples (blood and bone marrow) from 124 MM patients randomly assigned to receive VMPT-VT (53 pts) or VMP (71 pts) in the GIMEMA MM0305 phase III clinical trial and blood plasma samples from 54 control subjects were available for this study. Baseline demographic and disease characteristics are reported in Table 1.
We evaluated the concentration of ANG-2, FGF-2, HGF, IL-8, PDGF-BB, TIMP-1, TIMP-2, TNF-α, and VEGF that are the major cytokines involved in angiogenesis in MM and other cancers [36] and, as previously demonstrated [37] in MM patients, directly correlate with disease activity and increase with progression. Moreover, plasma levels of CAFs are directly related to disease response to therapy in hematologic and solid tumors [2932].
Our results showed that there were no differences in the levels of the studied CAFs between the peripheral blood and bone marrow plasma samples of MM patients (Fig. 1, Additional file 1: Table S1), indicating that the concentrations of circulating cytokines well reflect those of the bone marrow and could be used for all subsequent analyses. As expected, the plasma levels of CAFs in MM patients were significantly higher compared to controls (p < 0.0001 for all CAFs). In addition, the levels of CAFs were found to be significantly related to MM response to therapy (Fig. 2) with the exception of TIMP-1 and TIMP-2. More precisely, low levels of ANG-2 (p < 0.05), FGF-2 (p < 0.005), HGF (p < 0.05), IL-8 (p < 0.05), PDGF-BB (p < 0.005), TNF-α (p < 0.05), and VEGF (p < 0.005) were indicative of more profound response (very good partial response [VGPR] or better in all patients, with no evident differences between the two therapeutic regimens (VMPT-VT vs VMP: p = 0.1).
On the basis of ROC curve estimation of cutoff and logistic regression analysis between dichotomized tumor response and CAF plasma levels, a cutoff for each cytokine was established that could be used to discriminate the probability of response to therapy of MM patients with high sensitivity and specificity (Additional file 1: Table S2). Seventy-four patients had the best degree of tumor response (≥ VGPR) and were termed good responders, whereas 50 had the smallest degree of tumor response (< VGPR) and were termed poor responders. Among the seven CAFs that were shown to be significantly related to MM response to therapy (Fig. 2), low (relative to median) concentrations of ANG-2 (p < 0.0001), FGF-2 (p < 0.0001), HGF (p < 0.0001), IL-8 (p < 0.0001), PDGF-BB (p < 0.0001), TNF-α (p < 0.001), and VEGF (p < 0.0001) highly correlated with best response (Additional file 1: Table S2).
In terms of survival, PFS correlated with CAF levels as a dichotomous variable, and low levels of FGF-2 (p < 0.0001), HGF (p < 0.05), IL-8 (p < 0.05), TNF-α (p < 0.05), and VEGF (p < 0.005) were associated with better PFS. Instead, only low levels of FGF-2 (p < 0.001) and VEGF (p < 0.004) were associated with prolonged OS. Similar results were obtained when CAF levels were related to PFS and OS according to the combination therapy administered. A better PFS was in fact related to low levels of FGF-2 (p < 0.0001), HGF (p < 0.0001), TNF-α (p < 0.005), and VEGF (p < 0.005) in the VMP arm and FGF-2 (p < 0.005) and VEGF (p < 0.05) in the VMPT-VT arm. When OS was taken into consideration, only FGF-2 (p < 0.001 in the VMP arm, p < 0.005 in the VMPT-VT arm), and VEGF (p < 0.05 for both arms) were demonstrated (Additional file 1: Table S3).
Hierarchical clustering analysis (Fig. 3) showed three distinct risk groups of patients, based on the concentrations of two CAF levels (FGF-2 and VEGF). Patients with elevation of both CAF levels had a worse prognosis with significantly shorter PFS and OS (high risk) compared with those with high level of only one (p < 0.0001, intermediate risk) and those of low blood levels of both CAFs (p < 0.0001, low risk) (Fig. 3). No significant differences were detected depending on the type of therapy received. Trying to construct a survival model, a three-stage system using FGF-2 and VEGF blood levels provided the highest statistically significant results (Table 2). Median survivals of the risk groups were as follows: low risk: PFS 44 months, OS 70 months; intermediate risk: PFS 23.5 months, OS 62 months; and high risk: PFS 14.5 months, OS 34 months (p < 0.0001 for differences). Patient numbers were well distributed across the three groups (low risk, 40%; intermediate risk, 29%; and high risk, 31%).
Table 2
Patient stratification based on CAF circulating levels
Risk group
Risk factors
Criteria
Median PFS (months)
Median OS
(months)
Low
0
FGF-2 ≤ 950 pg/dL and
VEGF ≤ 19,000 pg/dL
38
67
Intermediate
1
FGF-2 > 950 pg/dL or
VEGF > 19,000 pg/dL
24
55
High
2
FGF-2 > 950 pg/dL and
VEGF > 19,000 pg/dL
15
37
By multivariate analysis (Table 3), the other variables significantly associated with better outcome were age < 65 years in the intermediate- and high-risk groups (HR 0.61, CI 0.44–0.85, p = 0.0047 and HR 0.60, CI 0.38–0.89, p = 0.0391, respectively) for OS; age < 65 years and best response to induction therapy (≥ VGPR) only in the high-risk group (HR 0.68, CI 0.48–0.96, p = 0.0342) for PFS. Durie and Salmon (D&S) stage 1 or 2 impacted only on PFS for the high-risk group (HR 0.64, CI 0.49–0.89, p = 0.0169). No significant differences were demonstrated regarding both PFS and OS for sex, isotype of the M-component, renal failure, and type of induction therapy. The administration of maintenance therapy significantly impacted on PFS in all the risk groups (low: HR 0.70, CI 0.44–0.89, p = 0.0210; intermediate: HR 0.54, CI 0.38–0.80, p = 0.0008; high: HR 0.71, CI 0.56–0.92, p = 0.0249) and OS (low: HR 0.70, CI 0.48–0.88, p = 0.0210; intermediate: HR 0.74, CI 0.57–1.01, p = 0.0330; high: HR 0.52, CI 0.36–0.75, p = 0.0003).
Table 3
Multivariate analysis of risk stratification of patients based on the CAFs FGF-2 and VEGF
Variable
PFS
OS
Risk group
Risk group
Low
Intermediate
High
Low
Intermediate
High
HR
(95 CI)
p
HR
(95 CI)
p
HR
(95 CI)
p
HR
(95 CI)
p
HR
(95 CI)
p
HR
(95 CI)
p
Sex
0.82
(0.59–1.18)
0.3698
0.74
(0.59–1.11)
0.9645
0.86
(0.62–1.08)
0.3438
0.80
(0.57–1.05)
0.3698
0.96
(0.68–1.33)
0.6905
0.86
(0.59–1.16)
0.5713
Age (< 65 years)
0.96
(0.69–1.35)
0.7647
0.87
(0.55–1.20)
0.2027
0.73
(0.59–0.98)
0.0455
0.97
(0.71–1.32)
0.7647
0.61
(0.44–0.85)
0.0047
0.60
(0.38–0.89)
0.0391
Isotype
0.91
(0.56–1.32)
0.2957
0.91
(0.64–1.31)
0.6951
0.96
(0.68–1.33)
0.6419
0.82
(0.51–1.21)
0.2957
0.83
(0.57–1.15)
0.3627
0.98
(0.81–1.22)
0.8179
D&S stage
0.84
(0.60–1.16)
0.3218
0.82
(0.57–1.22)
0.3505
0.64
(0.49–0.89)
0.0169
0.83
(0.59–1.11)
0.3218
0.98
(0.67–1.44)
0.7874
0.74
(0.59–1.12)
0.0593
ISS stage
0.94
(0.66–1.29)
0.8163
0.91
(0.55–1.24)
0.2107
0.84
(0.58–1.28)
0.0619
0.79
(0.61–1.07)
0.1087
0.97
(0.66–1.34)
0.5221
0.87
(0.62–1.27)
0.0665
Cytogenetic risk
0.93
(0.64–1.28)
0.6358
0.87
(0.56–1.22)
0.3451
0.91
(0.55–1.33)
0.6670
0.86
(0.71–1.32)
0.2399
0.89
(0.69–1.41)
0.6133
0.92
(0.71–1.37)
0.6112
Renal failure
0.94
(0.61–1.25)
0.5866
0.92
(0.59–1.44)
0.6957
0.87
(0.60–1.21)
0.2410
0.92
(0.66–1.41)
0.5866
0.94
(0.71–1.35)
0.6108
0.97
(0.74–1.25)
0.8902
Induction therapy
0.84
(0.61–1.28)
0.3520
0.93
(0.66–1.34)
0.6838
0.88
(0.58–1.18)
0.3356
0.84
(0.57–1.19)
0.3520
0.93
(0.70–1.32)
0.6075
0.89
(0.66–1.28)
0.6015
Best response to induction therapy (≥ VGPR)
0.76
(0.59–1.18)
0.0984
0.68
(0.48–0.96)
0.0342
0.71
(0.55–1.08)
0.0533
0.74
(0.59–1.09)
0.0984
0.95
(0.69–1.31)
0.5031
0.88
(0.61–1.29)
0.0583
Maintenance
0.70
(0.44–0.89)
0.0210
0.54
(0.38–0.80)
0.0008
0.71
(0.56–0.92)
0.0249
0.70
(0.48–0.88)
0.0210
0.74
(0.57–1.01)
0.0330
0.52
(0.36–0.75)
0.0003
p values less than 0.05 was considered significant
As regards the cytogenetic risk, because many data relative to the cytogenetic characteristics of the patients were missing in the database of the trial, the statistical power of the relative analysis was low. For this reason, it is not possible to reach a correct conclusion on the value of CAFs stratification risk in correlation to this risk parameter.

Discussion

The emergence of new treatment options for MM has extended the patients’ survival [5] and the need to prospectively identify those patients who are likely to benefit from a specific treatment and understand the mechanisms underlying therapeutic resistance. Several adverse prognostic factors have been identified in MM at diagnosis and before initiation of treatment [38, 39], including an advanced stage in the international staging system (ISS) based on plasma albumin [40]. The dual activity of the new and newest drugs active both on MM plasma cells and bone marrow stromal cells, and in particular on angiogenesis [4143], obviously indicates that any new prognostic markers cannot ignore the angiogenesis aspect. Previous studies have indeed suggested that the plasma levels of CAFs might be used to identify prognostic and predictive markers in solid tumors [4449] and are indicative of the response to antineoplastic therapy [2932].
Seven CAFs of the nine evaluated (ANG-2, FGF-2, HGF, IL-8, PDGF-BB, TNF-α, and VEGF) initially emerged as being related to disease activity, but further testing showed that only FGF-2 and VEGF were significantly associated with PFS and OS and were therefore evaluated for prognostic stratification of patients. We also assessed whether determination of the studied CAFs could add prognostic information to D&S [50] and International Staging Systems (ISS) [40] or whether these systems were, in fact, predictive of benefit. D&S and ISS staging were associated with prognosis based on PFS in both groups; however, they were not strong prognostic parameters as FGF-2 and/or VEGF. Thus, the plasma levels of the two angiogenic cytokines provided prognostic and, more importantly, predictive value beyond that of standard clinical staging.
Our analyses of survival were based on assignment at initial randomization. High FGF-2 and VEGF plasma levels were negative prognostic markers and were associated with lower relative OS in both harms. The associations reported here with FGF-2 and VEGF were akin to previous studies [2932], in which patients with the highest reduction of circulating CAF concentrations obtained the greatest benefit from anticancer therapy. However, other studies assessing the use of VEGF as a predictive marker of benefit from VEGF-targeted therapies in renal-cell carcinoma have yielded inconsistent results [51].
Previous studies have shown that groups of related angiogenic or inflammatory factors are often correlated [44, 46, 52]. Hierarchical clustering of six circulating CAFs showed a strong correlation among many of them, including osteopontin and VEGF [46, 52]. Patients defined by high concentrations of these CAF and inflammatory or immunomodulatory factors had a significantly worse prognosis, but derived a greater relative OS benefit from therapy. The circulating CAFs identified in these studies might themselves have important biological roles or might be markers for alternative pathways or mechanisms affecting treatment responses (e.g., hypoxia-induced factor-1α or NF-κB pathways). This association between factors suggests that common mechanisms might regulate their production. Studying a cohort of patients with metastatic renal-cell carcinoma, a classification of the disease based on the higher expression of angiogenic versus inflammatory circulating CAFs defined by a six-cytokine signature was established [46]. In that study, the angiogenic group derived greater benefit from sorafenib alone, whereas the other group benefited from the combination of sorafenib and IFN-α. In the present study, a similar correlation of treatment benefit with circulating CAF signature was observed in MM patients.
All the evidences provided by the studies on the role of microenvironment [3, 17], and particularly of the angiogenic process [23, 37] in myeloma progression as well as in cancer cell protection mediated by microenvironment components [37], indicated that the response to therapy is also related to the activity of anticancer drugs on tumor microenvironment [42, 43]. Therefore, the inhibition of cytokine production which mediates the interaction between cancer cells and their microenvironment represents one of the major goals of the modern therapeutic approaches. On these bases, the evaluation of CAF levels is indicative of the response to therapy and may represent a good indicator of refractoriness in cancer patients.

Conclusion

Overall, our findings support the use of circulating CAF profiling to define biologically distinct subgroups of MM patients whose tumors have a greater angiogenic drive. As such, these patients might have a more aggressive disease course but are likely to derive relative benefit from inhibition of angiogenic pathways. Circulating CAF profiling might be particularly well suited for angiogenesis inhibitors and other drugs targeting the tumor microenvironment, in which both circulating host-derived and tumor-derived factors could affect response. Such an approach may have important advantages, including straightforward and relatively non-invasive sample collection, availability of robust analytical platforms, and the ability to monitor changes during treatment or disease progression, which can help identify markers of resistance.
The limitation of this study is that the results have been obtained in older patients not-eligible for ASCT, whereas the major strength of the study is the homogeneous stratification and longer follow-up of patients. Further studies on a greater cohort of subjects, including young patients eligible for ASCT before and after treatment, will be needed to evaluate the variation of CAFs after therapy, to study their power as an indicator of minimal residual disease and, then, of risk of relapse, and to evaluate the value of the risk stratification based on CAFs in the new drugs era, with the aim of definitively establish the value of this approach in the application of precision, personalized therapy for patients with MM.

Acknowledgements

The authors would like to thank the Italian Association for Cancer Research (AIRC, Milan), Investigator Grant 2013 (no. 14095) and the 5 per thousand Molecular Clinical Oncology Special Program (Grant no. 9965 to Angelo Vacca).

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request and with permission of Local IRB.
The Local IRB approved this study and all patients gave their informed consent before the entry into the clinical trial in agreement with institutional guidelines.
Not applicable.

Competing interests

Lorenzo De Paoli: honorary from Jansen, Abbvie, Celgene, Gilead, AMGEN, and Roche. Massimo Offidani: honorary and advisory from Janssen. Maria Teresa Petrucci: Honoraria and Advisory Board: Celgene, Janssen-Cilag, BMS, Takeda, Amgen. Roberto Ria: Honoraria and Advisory Board: Celgene, Janssen-Cilag, BMS, Italfarmaco, Amgen. The other authors declare no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Noone AM, Howlader N, Krapcho M, Miller D, Brest A, Yu M, et al. SEER Cancer Statistics Review, 1975–2015, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2015/, based on November 2017 SEER data submission, posted to the SEER web site. April 2018. Noone AM, Howlader N, Krapcho M, Miller D, Brest A, Yu M, et al. SEER Cancer Statistics Review, 1975–2015, National Cancer Institute. Bethesda, MD, https://​seer.​cancer.​gov/​csr/​1975_​2015/​, based on November 2017 SEER data submission, posted to the SEER web site. April 2018.
2.
Zurück zum Zitat Alexanian R, Haut A, Khan AU, Lane M, McKelvey EM, Migliore PJ, et al. Treatment for multiple myeloma: combination chemotherapy with different melphalan dose regimens. JAMA. 1969;208:1680–5.CrossRef Alexanian R, Haut A, Khan AU, Lane M, McKelvey EM, Migliore PJ, et al. Treatment for multiple myeloma: combination chemotherapy with different melphalan dose regimens. JAMA. 1969;208:1680–5.CrossRef
3.
Zurück zum Zitat Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med. 2004;351:1860–73.CrossRef Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med. 2004;351:1860–73.CrossRef
4.
Zurück zum Zitat Myeloma Trialists’ Collaborative Group. Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma. An overview of 6,633 patients from 27 randomized trials. J Clin Oncol. 1998;16:3832–42. Myeloma Trialists’ Collaborative Group. Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma. An overview of 6,633 patients from 27 randomized trials. J Clin Oncol. 1998;16:3832–42.
5.
Zurück zum Zitat Ria R, Reale A, Vacca A. Novel agents and new therapeutic approaches for treatment of multiple myeloma. World J Methodol. 2014;4:73–90.CrossRef Ria R, Reale A, Vacca A. Novel agents and new therapeutic approaches for treatment of multiple myeloma. World J Methodol. 2014;4:73–90.CrossRef
6.
Zurück zum Zitat Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med. 2005;352:2487–98.CrossRef Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med. 2005;352:2487–98.CrossRef
7.
Zurück zum Zitat San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008;359:906–17.CrossRef San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008;359:906–17.CrossRef
8.
Zurück zum Zitat Palumbo A, Bringhen S, Caravita T, Merla E, Capparella V, Callea V, et al. Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial. Lancet. 2006;367:825–31.CrossRef Palumbo A, Bringhen S, Caravita T, Merla E, Capparella V, Callea V, et al. Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial. Lancet. 2006;367:825–31.CrossRef
9.
Zurück zum Zitat Facon T, Mary JY, Hulin C, Benboubker L, Attal M, Pegourie B, et al. Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial. Lancet. 2007;370:1209–18.CrossRef Facon T, Mary JY, Hulin C, Benboubker L, Attal M, Pegourie B, et al. Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial. Lancet. 2007;370:1209–18.CrossRef
10.
Zurück zum Zitat Hulin C, Facon T, Rodon P, Pegourie B, Benboubker L, Doyen C, et al. Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial. J Clin Oncol. 2009;27:3664–70.CrossRef Hulin C, Facon T, Rodon P, Pegourie B, Benboubker L, Doyen C, et al. Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial. J Clin Oncol. 2009;27:3664–70.CrossRef
11.
Zurück zum Zitat Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, Dmoszynska A, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med. 2007;357:2123–32.CrossRef Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, Dmoszynska A, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med. 2007;357:2123–32.CrossRef
12.
Zurück zum Zitat Weber DM, Chen C, Niesvizky R. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007;357:2133–42.CrossRef Weber DM, Chen C, Niesvizky R. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007;357:2133–42.CrossRef
13.
Zurück zum Zitat Palumbo A, Falco P, Corradini P, Falcone A, Di Raimondo F, Giuliani N, et al. Melphalan, prednisone, and lenalidomide treatment for newly diagnosed myeloma: a report from the GIMEMA—Italian Multiple Myeloma Network. J Clin Oncol. 2007;25:4459–65.CrossRef Palumbo A, Falco P, Corradini P, Falcone A, Di Raimondo F, Giuliani N, et al. Melphalan, prednisone, and lenalidomide treatment for newly diagnosed myeloma: a report from the GIMEMA—Italian Multiple Myeloma Network. J Clin Oncol. 2007;25:4459–65.CrossRef
14.
Zurück zum Zitat Biran N, Jagannath S, Chari A. Risk stratification in multiple myeloma, part 1: characterization of high-risk disease. Clin Adv Hematol Oncol. 2013;11:489–503.PubMed Biran N, Jagannath S, Chari A. Risk stratification in multiple myeloma, part 1: characterization of high-risk disease. Clin Adv Hematol Oncol. 2013;11:489–503.PubMed
15.
Zurück zum Zitat Fonseca R, Monge J, Dimopoulos MA. Staging and prognostication of multiple myeloma. Expert Rev Hematol. 2014;7:21–31.CrossRef Fonseca R, Monge J, Dimopoulos MA. Staging and prognostication of multiple myeloma. Expert Rev Hematol. 2014;7:21–31.CrossRef
16.
Zurück zum Zitat Mihailovic J, Goldsmith SJ. Multiple myeloma: 18F-FDG-PET/CT and diagnostic imaging. Semin Nucl Med. 2015;45:16–31.CrossRef Mihailovic J, Goldsmith SJ. Multiple myeloma: 18F-FDG-PET/CT and diagnostic imaging. Semin Nucl Med. 2015;45:16–31.CrossRef
17.
Zurück zum Zitat Landgren O, Morgan GJ. Biologic frontiers in multiple myeloma: from biomarker identification to clinical practice. Clin Cancer Res. 2014;20:804–13.CrossRef Landgren O, Morgan GJ. Biologic frontiers in multiple myeloma: from biomarker identification to clinical practice. Clin Cancer Res. 2014;20:804–13.CrossRef
18.
Zurück zum Zitat Hart AJ, Jagasia MH, Kim AS, Mosse CA, Savani BN, Kassim A. Minimal residual disease in myeloma: are we there yet? Biol Blood Marrow Transplant. 2012;18:1790–9.CrossRef Hart AJ, Jagasia MH, Kim AS, Mosse CA, Savani BN, Kassim A. Minimal residual disease in myeloma: are we there yet? Biol Blood Marrow Transplant. 2012;18:1790–9.CrossRef
19.
Zurück zum Zitat Shaughnessy JD, Zhou Y, Haessler J, van Rhee F, Anaissie E, Nair B, et al. TP53 deletion is not an adverse feature in multiple myeloma treated with total therapy 3. Br J Haematol. 2009;147:347–51.CrossRef Shaughnessy JD, Zhou Y, Haessler J, van Rhee F, Anaissie E, Nair B, et al. TP53 deletion is not an adverse feature in multiple myeloma treated with total therapy 3. Br J Haematol. 2009;147:347–51.CrossRef
20.
Zurück zum Zitat Neben K, Lokhorst HM, Jauch A, Bertsch U, Hielscher T, van der Holt B, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 2012;119:940–8.CrossRef Neben K, Lokhorst HM, Jauch A, Bertsch U, Hielscher T, van der Holt B, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 2012;119:940–8.CrossRef
21.
Zurück zum Zitat Stella F, Pedrazzini E, Agazzoni M, Ballester O, Slavutsky I. Cytogenetic alterations in multiple myeloma: prognostic significance and the choice of frontline therapy. Cancer Investig. 2015;33:496–504.CrossRef Stella F, Pedrazzini E, Agazzoni M, Ballester O, Slavutsky I. Cytogenetic alterations in multiple myeloma: prognostic significance and the choice of frontline therapy. Cancer Investig. 2015;33:496–504.CrossRef
22.
Zurück zum Zitat Chng WJ, Dispenzieri A, Chim CS, Fonseca R, Goldschmidt H, Lentzsch S, et al. International Myeloma Working Group. IMWG consensus on risk stratification in multiple myeloma. Leukemia. 2014;28:269–77.CrossRef Chng WJ, Dispenzieri A, Chim CS, Fonseca R, Goldschmidt H, Lentzsch S, et al. International Myeloma Working Group. IMWG consensus on risk stratification in multiple myeloma. Leukemia. 2014;28:269–77.CrossRef
23.
Zurück zum Zitat Vacca A, Ria R, Reale A, Ribatti D. Angiogenesis in multiple myeloma. Chem Immunol Allergy. 2014;99:180–96.CrossRef Vacca A, Ria R, Reale A, Ribatti D. Angiogenesis in multiple myeloma. Chem Immunol Allergy. 2014;99:180–96.CrossRef
24.
Zurück zum Zitat Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–24.CrossRef Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–24.CrossRef
25.
Zurück zum Zitat Hutson TE, Davis ID, Machiels JP, De Souza PL, Rottey S, Hong BF, et al. Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma. J Clin Oncol. 2010;28:475–80.CrossRef Hutson TE, Davis ID, Machiels JP, De Souza PL, Rottey S, Hong BF, et al. Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma. J Clin Oncol. 2010;28:475–80.CrossRef
26.
Zurück zum Zitat Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–34.CrossRef Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–34.CrossRef
27.
Zurück zum Zitat Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370:2103–11.CrossRef Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370:2103–11.CrossRef
28.
Zurück zum Zitat Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–8.CrossRef Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–8.CrossRef
29.
Zurück zum Zitat Ria R, Cirulli T, Giannini T, Bambace S, Serio G, Portaluri M, et al. Serum levels of angiogenic cytokines decrease after radiotherapy in non-Hodgkin lymphomas. Clin Exp Med. 2008;8:141–5.CrossRef Ria R, Cirulli T, Giannini T, Bambace S, Serio G, Portaluri M, et al. Serum levels of angiogenic cytokines decrease after radiotherapy in non-Hodgkin lymphomas. Clin Exp Med. 2008;8:141–5.CrossRef
30.
Zurück zum Zitat Ria R, Portaluri M, Russo F, Cirulli T, Di Pietro G, Bambace S, et al. Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Lett. 2004;216:103–7.CrossRef Ria R, Portaluri M, Russo F, Cirulli T, Di Pietro G, Bambace S, et al. Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Lett. 2004;216:103–7.CrossRef
31.
Zurück zum Zitat Dietz A, Rudat V, Conradt C, Weidauer H, Ho A, Moehler T. Prognostic relevance of serum levels of the angiogenic peptide bFGF in advanced carcinoma of the head and neck treated by primary radiochemotherapy. Head Neck. 2000;22:666–73.CrossRef Dietz A, Rudat V, Conradt C, Weidauer H, Ho A, Moehler T. Prognostic relevance of serum levels of the angiogenic peptide bFGF in advanced carcinoma of the head and neck treated by primary radiochemotherapy. Head Neck. 2000;22:666–73.CrossRef
32.
Zurück zum Zitat Chala E, Manes C, Iliades H, Skaragkas G, Mouratidou D, Kapantais E. Insulin resistance, growth factors and cytokine levels in overweight women with breast cancer before and after chemotherapy. Hormones. 2006;5:137–46.CrossRef Chala E, Manes C, Iliades H, Skaragkas G, Mouratidou D, Kapantais E. Insulin resistance, growth factors and cytokine levels in overweight women with breast cancer before and after chemotherapy. Hormones. 2006;5:137–46.CrossRef
33.
Zurück zum Zitat Palumbo A, Bringhen S, Rossi D, Cavalli M, Larocca A, Ria R, et al. Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: a randomized controlled trial. J Clin Oncol. 2010;28:5101–9.CrossRef Palumbo A, Bringhen S, Rossi D, Cavalli M, Larocca A, Ria R, et al. Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: a randomized controlled trial. J Clin Oncol. 2010;28:5101–9.CrossRef
34.
Zurück zum Zitat Morabito F, Gentile M, Mazzone C, Rossi D, Di Raimondo F, Bringhen S, et al. Safety and efficacy of bortezomib-melphalan-prednisone-thalidomide followed by bortezomib-thalidomide maintenance (VMPT-VT) versus bortezomib-melphalan-prednisone (VMP) in untreated multiple myeloma patients with renal impairment. Blood. 2011;118:5759–66.CrossRef Morabito F, Gentile M, Mazzone C, Rossi D, Di Raimondo F, Bringhen S, et al. Safety and efficacy of bortezomib-melphalan-prednisone-thalidomide followed by bortezomib-thalidomide maintenance (VMPT-VT) versus bortezomib-melphalan-prednisone (VMP) in untreated multiple myeloma patients with renal impairment. Blood. 2011;118:5759–66.CrossRef
35.
Zurück zum Zitat Palumbo A, Bringhen S, Larocca A, Rossi D, Di Raimondo F, Magarotto V, et al. Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: updated follow-up and improved survival. J Clin Oncol. 2014;32:634–40.CrossRef Palumbo A, Bringhen S, Larocca A, Rossi D, Di Raimondo F, Magarotto V, et al. Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: updated follow-up and improved survival. J Clin Oncol. 2014;32:634–40.CrossRef
36.
Zurück zum Zitat Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–57.CrossRef Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–57.CrossRef
37.
Zurück zum Zitat Ria R, Reale A, De Luisi A, Ferrucci A, Moschetta M, Vacca A. Bone marrow angiogenesis and progression in multiple myeloma. Am J Blood Res. 2011;1:76–89.PubMedPubMedCentral Ria R, Reale A, De Luisi A, Ferrucci A, Moschetta M, Vacca A. Bone marrow angiogenesis and progression in multiple myeloma. Am J Blood Res. 2011;1:76–89.PubMedPubMedCentral
38.
Zurück zum Zitat Cosemans C, Oben B, Arijs I, Daniëls A, Declercq J, Vanhees K, et al. Prognostic biomarkers in the progression from MGUS to multiple myeloma: a systematic review. Clin Lymphoma Myeloma Leuk. 2018;18:235–48.CrossRef Cosemans C, Oben B, Arijs I, Daniëls A, Declercq J, Vanhees K, et al. Prognostic biomarkers in the progression from MGUS to multiple myeloma: a systematic review. Clin Lymphoma Myeloma Leuk. 2018;18:235–48.CrossRef
39.
Zurück zum Zitat Hanbali A, Hassanein M, Rasheed W, Aljurf M, Alsharif F. The evolution of prognostic factors in multiple myeloma. Adv Hematol. 2017;2017:4812637.CrossRef Hanbali A, Hassanein M, Rasheed W, Aljurf M, Alsharif F. The evolution of prognostic factors in multiple myeloma. Adv Hematol. 2017;2017:4812637.CrossRef
40.
Zurück zum Zitat Greipp PR, San Miguel J, Durie BG, Crowley JJ, Barlogie B, Bladé J, et al. International staging system for multiple myeloma. J Clin Oncol. 2005;23:3412–20.CrossRef Greipp PR, San Miguel J, Durie BG, Crowley JJ, Barlogie B, Bladé J, et al. International staging system for multiple myeloma. J Clin Oncol. 2005;23:3412–20.CrossRef
41.
Zurück zum Zitat Roccaro AM, Hideshima T, Raje N, Kumar S, Ishitsuka K, Yasui H, et al. Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells. Cancer Res. 2006;66:184–91.CrossRef Roccaro AM, Hideshima T, Raje N, Kumar S, Ishitsuka K, Yasui H, et al. Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells. Cancer Res. 2006;66:184–91.CrossRef
42.
Zurück zum Zitat Moschetta M, Di Pietro G, Ria R, Gnoni A, Mangialardi G, Guarini A, et al. Bortezomib and zoledronic acid on angiogenic and vasculogenic activities of bone marrow macrophages in patients with multiple myeloma. Eur J Cancer. 2010;46:420–9.CrossRef Moschetta M, Di Pietro G, Ria R, Gnoni A, Mangialardi G, Guarini A, et al. Bortezomib and zoledronic acid on angiogenic and vasculogenic activities of bone marrow macrophages in patients with multiple myeloma. Eur J Cancer. 2010;46:420–9.CrossRef
43.
Zurück zum Zitat De Luisi A, Ferrucci A, Coluccia AM, Ria R, Moschetta M, de Luca E, et al. Lenalidomide restrains motility and overangiogenic potential of bone marrow endothelial cells in patients with active multiple myeloma. Clin Cancer Res. 2011;17:1935–46.CrossRef De Luisi A, Ferrucci A, Coluccia AM, Ria R, Moschetta M, de Luca E, et al. Lenalidomide restrains motility and overangiogenic potential of bone marrow endothelial cells in patients with active multiple myeloma. Clin Cancer Res. 2011;17:1935–46.CrossRef
44.
Zurück zum Zitat Nikolinakos PG, Altorki N, Yankelevitz D, Tran HT, Yan S, Rajagopalan D, et al. Plasma cytokine and angiogenic factor profiling identifies markers associated with tumor shrinkage in early-stage non-small cell lung cancer patients treated with pazopanib. Cancer Res. 2010;70:2171–9.CrossRef Nikolinakos PG, Altorki N, Yankelevitz D, Tran HT, Yan S, Rajagopalan D, et al. Plasma cytokine and angiogenic factor profiling identifies markers associated with tumor shrinkage in early-stage non-small cell lung cancer patients treated with pazopanib. Cancer Res. 2010;70:2171–9.CrossRef
45.
Zurück zum Zitat Hanrahan EO, Lin HY, Kim ES, Yan S, Du DZ, McKee KS, et al. Distinct patterns of cytokine and angiogenic factor modulation and markers of benefit for vandetanib and/or chemotherapy in patients with non-small-cell lung cancer. J Clin Oncol. 2010;28:193–201.CrossRef Hanrahan EO, Lin HY, Kim ES, Yan S, Du DZ, McKee KS, et al. Distinct patterns of cytokine and angiogenic factor modulation and markers of benefit for vandetanib and/or chemotherapy in patients with non-small-cell lung cancer. J Clin Oncol. 2010;28:193–201.CrossRef
46.
Zurück zum Zitat Zurita AJ, Jonasch E, Wang X, Khajavi M, Yan S, Du DZ, et al. A cytokine and angiogenic factor (CAF) analysis in plasma for selection of sorafenib therapy in patients with metastatic renal cell carcinoma. Ann Oncol. 2012;23:46–52.CrossRef Zurita AJ, Jonasch E, Wang X, Khajavi M, Yan S, Du DZ, et al. A cytokine and angiogenic factor (CAF) analysis in plasma for selection of sorafenib therapy in patients with metastatic renal cell carcinoma. Ann Oncol. 2012;23:46–52.CrossRef
47.
Zurück zum Zitat Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol. 2009;27:5794–9.CrossRef Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol. 2009;27:5794–9.CrossRef
48.
Zurück zum Zitat Peña C, Lathia C, Shan M, Escudier B, Bukowski RM. Biomarkers predicting outcome in patients with advanced renal cell carcinoma: results from sorafenib phase III treatment approaches in Renal Cancer Global Evaluation Trial. Clin Cancer Res. 2010;16:4853–63.CrossRef Peña C, Lathia C, Shan M, Escudier B, Bukowski RM. Biomarkers predicting outcome in patients with advanced renal cell carcinoma: results from sorafenib phase III treatment approaches in Renal Cancer Global Evaluation Trial. Clin Cancer Res. 2010;16:4853–63.CrossRef
49.
Zurück zum Zitat Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 2012;13:827–37.CrossRef Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 2012;13:827–37.CrossRef
50.
Zurück zum Zitat Durie BG, Salmon SE. A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer. 1975;36:842–54.CrossRef Durie BG, Salmon SE. A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer. 1975;36:842–54.CrossRef
51.
Zurück zum Zitat Melichar B, Procházková-Študentová H, Vitásková D. Bevacizumab in combination with IFN-α in metastatic renal cell carcinoma: the AVOREN trial. Expert Rev Anticancer Ther. 2012;12:1253–61.CrossRef Melichar B, Procházková-Študentová H, Vitásková D. Bevacizumab in combination with IFN-α in metastatic renal cell carcinoma: the AVOREN trial. Expert Rev Anticancer Ther. 2012;12:1253–61.CrossRef
52.
Zurück zum Zitat Heymach JV, Shackleford TJ, Tran HT, Yoo SY, Do KA, Wergin M, et al. Effect of low-fat diets on plasma levels of NFκB-regulated inflammatory cytokines and angiogenic factors in men with prostate cancer. Cancer Prev Res (Phila). 2011;4:1590–8.CrossRef Heymach JV, Shackleford TJ, Tran HT, Yoo SY, Do KA, Wergin M, et al. Effect of low-fat diets on plasma levels of NFκB-regulated inflammatory cytokines and angiogenic factors in men with prostate cancer. Cancer Prev Res (Phila). 2011;4:1590–8.CrossRef
Metadaten
Titel
Prognostic or predictive value of circulating cytokines and angiogenic factors for initial treatment of multiple myeloma in the GIMEMA MM0305 randomized controlled trial
verfasst von
Ilaria Saltarella
Fortunato Morabito
Nicola Giuliani
Carolina Terragna
Paola Omedè
Antonio Palumbo
Sara Bringhen
Lorenzo De Paoli
Enrica Martino
Alessandra Larocca
Massimo Offidani
Francesca Patriarca
Chiara Nozzoli
Tommasina Guglielmelli
Giulia Benevolo
Vincenzo Callea
Luca Baldini
Mariella Grasso
Giovanna Leonardi
Manuela Rizzo
Antonietta Pia Falcone
Daniela Gottardi
Vittorio Montefusco
Pellegrino Musto
Maria Teresa Petrucci
Franco Dammacco
Mario Boccadoro
Angelo Vacca
Roberto Ria
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2019
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-018-0691-4

Weitere Artikel der Ausgabe 1/2019

Journal of Hematology & Oncology 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.