Next Article in Journal
Targeting Receptor Kinases in Colorectal Cancer
Previous Article in Journal
Validation of miRNAs as Breast Cancer Biomarkers with a Machine Learning Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Validation of Neutrophil Count as An Algorithm-Based Predictive Factor of Progression-Free Survival in Patients with Metastatic Soft Tissue Sarcomas Treated with Trabectedin

by
Alexandre de Nonneville
1,*,
Dominique Barbolosi
2,
Maeva Andriantsoa
3,
Raouf El-Cheikh
2,
Florence Duffaud
3,
François Bertucci
1 and
Sebastien Salas
3
1
CRCM, Department of Medical Oncology, Institut Paoli-Calmettes, INSERM, CNRS, Aix-Marseille University, 13009 Marseille, France
2
SMARTc Unit, CRCM Inserm U1068, Aix-Marseille Université, 13005 Marseille, France
3
Department of Medical Oncology, Hôpital de la Timone, APHM, INSERM U910, Aix-Marseille University, 13005 Marseille, France
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(3), 432; https://doi.org/10.3390/cancers11030432
Submission received: 4 March 2019 / Revised: 22 March 2019 / Accepted: 25 March 2019 / Published: 26 March 2019

Abstract

:
Introduction: Based on a mathematical model of trabectedin-induced neutropenia, we assessed the predictive value of absolute neutrophil count (ANC) on progression-free survival (PFS) in an independent validation cohort of patients treated with trabectedin. Methods: We collected data from 87 patients in two expert centers who received at least two cycles of trabectedin for soft tissue sarcomas (STS) treatment. Correlations between ANC, patients’ characteristics, and survival were assessed, and a multivariate model including tumor grade, performance status, ANC, and hemoglobin level was developed. Results: Therapeutic ANC ≥ 7.5 G/L level was associated with shorter PFS: 3.22 months (95% confidence interval (CI), 1.57–4.87) in patients with ANC ≥ 7.5 G/L vs. 5.78 months (95% CI, 3.95–7.61) in patients with ANC < 7.5 G/L (p = 0.009). Age, primary localization, lung metastases, dose reduction, hemoglobin, and albumin rates were also associated with PFS. In multivariate analysis, ANC ≥ 7.5 G/L was independently associated with poor PFS and overall survival. Conclusion: We validated increased pre-therapeutic ANC as a predictive factor of short PFS in patients starting trabectedin for STS. ANC appears to have an impact on survival rates and may be used as a decision-making tool for personalizing second-line strategies in patients with metastatic STS.

1. Introduction

Soft tissue sarcomas (STS) are rare malignancies of mesenchymal origin, accounting for approximately 1% of adult cancers [1]. In patients with metastatic disease not amenable to curative-intent surgery, the first-line systemic treatment involves palliative doxorubicin-based chemotherapy, which has changed very little over the past three decades. After intolerance or failure, the second-line therapies include chemotherapies (trabectedin, ifosfamide, dacarbazine) and targeted therapies (pazopanib, olaparumab), but the results remain disappointing. Clearly, the improvement of systemic therapies is crucial, including the identification of predictive factors for response to specific drugs.
Trabectedin is a synthetic antineoplastic agent, derived from a sea squirt. It has demonstrated efficacy in several randomized phase II studies [2,3,4] in non-selected patients with advanced, unresectable disease and no previous exposure to chemotherapy [5], who are in compassionate programs [6,7,8]. Furthermore, a very recent, randomized phase III study showed a significant improvement in median progression-free survival (PFS) with trabectedin vs. Best Supportive Care, in patients with pre-treated, advanced STS of multiple histological types [9]. Liposarcoma-STS patients benefit the most from trabectedin therapy in terms of prolonged tumor control. Identification of the predictors of response to trabectedin is warranted, as well, because of its morbid and financial toxicity.
One of the main toxicities of trabectedin is neutropenia. The incidence of grade 3 and 4 neutropenia has been estimated at 26% and 24%, respectively. Although reversible, trabectedin-induced neutropenia represents a major concern for treatment continuation and optimal dose delivery. In order to develop an alternative administration schedule, in which similar doses would be less hematotoxic, we recently developed an original, mathematical pharmacokinetics/pharmacodynamics (PK/PD) model describing the time course of neutrophils, under trabectedin treatment, in a monocenter retrospective series of 39 patients with metastatic STS [10]. Model parameters were estimated using a nonlinear mixed effects analysis with MONOLIX software (Lixoft, Anrony, France). A visual inspection of the baseline absolute neutrophil count (ANC) revealed the existence of two subpopulations of patients: one with a baseline ANC ≥ 7.5 G/L and one with ANC < 7.5 G/L. To assess the validity of this hypothesis, a latent covariate structure was introduced at the individual parameter level, namely on ANC at baseline. This was accomplished using the between-subject mixture option in MONOLIX. Population analysis results confirmed the existence (p < 0.005 for the latent covariate) of two distinct distributions for the parameter representing ANC at baseline, thus confirming the existence of two subpopulations of patients, distinguishable by their baseline ANC. Importantly, progression-free survival (PFS) was significantly lower in the group with ANC ≥ 7.5 G/L (hazard ratio (HR) = 3.819; 95% confidence interval (CI) (0.44–32.96); p = 0.015).
The primary objective of the present study was to validate the impact of pre-therapeutic ANC on PFS in an independent external cohort of STS patients treated with trabectedin. Secondary objectives consisted of an exploratory analysis of the impact of pre-therapeutic ANC on overall survival (OS) and the identification of other predictive factors for PFS and OS.

2. Results

2.1. Characteristics of Study Population

A total of 87 patients who received trabectedin for metastatic STS were considered eligible for analysis. Baseline characteristics are reported in Table 1. The most prevalent histologic type was leiomyosarcomas, followed by liposarcomas, and undifferentiated sarcomas, with 17% of total cases corresponding to translocation-related sarcomas (myxoid liposarcoma: 40%, synovialosarcomas: 34%, rhabdomyosarcoma: 13%, clear cell sarcoma: 7%, and Ewing sarcoma: 6%). Primary tumors were located in a limb in 33% of cases. Other localizations were variable, including visceral localizations (uterine: 19%; digestive: 7%) and primitive serous sarcomas (5%). Lung was the most common site of metastatic disease, followed by liver and bone.
A median of five cycles of trabectedin were administered (range 2–73). Dose reduction occurred in 73% of patients either during the first cycle, due to initial fragility, or during treatment, due to trabectedin-related toxicities. Pre-therapeutic assessment found an initial ANC < 7.5 in 59 patients (80%) and ≥7.5 in 15 patients (20%). Baseline characteristics did not differ according to these two ANC-based subgroups (Table 1).

2.2. Impact of Pre-Therapeutic ANC on PFS

After a median follow-up of 9.26 months (range 1.31–52.86), 74 patients (85%) had progressive disease. Median PFS of the overall population was 4.17 months (95% CI, 3.10–5.24). In univariate analysis, a pre-therapeutic ANC ≥ 7.5 G/L was associated with a shorter median PFS of 3.22 months (95% CI, 1.57–4.87) vs. 5.78 months (95% CI, 3.95–7.61) in patients with ANC < 7.5 G/L (p = 0.009) (Figure 1A). Age, tumor grade (Figure 1B), primary tumor localization, lung metastases, trabectedin dose reduction, hemoglobin level (Figure 1C), and albumin level also impacted PFS (Table 2). The predictive negative effect of ANC ≥ 7.5 G/L on PFS was independently maintained in multivariate analysis (HR = 3.39) (95% CI, 1.35–8.53; p = 0.010) (Figure 2A), as well as high tumor grade and low hemoglobin level (Table 3).

2.3. Impact of Pre-Therapeutic ANC on Overall Survival: An Exploratory Analysis

Thirty-nine deaths (45%) occurred during the follow-up. The median OS of the overall population was 26.78 months (95% CI, 14.38–39.17). In univariate analysis, a pre-therapeutic ANC ≥ 7.5 G/L was associated with a shorter median OS of 4.89 months (95% CI, 0–11.81) vs. 26.78 months (95% CI, 13.47–40.08) in patients with ANC < 7.5 G/L (p = 0.011) (Figure 1D). Eastern Cooperative Oncology Group (ECOG) performance status, hemoglobin level (Figure 1F), and albumin level also impacted OS (Table 2). Despite a clear trend, tumor grade did not reach statistical significance (Figure 1E). The predictive negative effect of ANC ≥ 7.5 G/L on OS was independently maintained in multivariate analysis (HR = 4.90 (95% CI, 1.57–15.34); p = 0.006) (Figure 2B), as well aslow hemoglobin level (Table 3). As in univariate analysis, tumor grade failed to reach statistical significance (HR = 2.49 for grade 3 (95% CI, 0.92–6.73); p = 0.071).

3. Discussion

In this assessment of the predictive value of pre-therapeutic ANC on PFS, in an independent validation cohort of patients treated with trabectedin for metastatic STS, an ANC ≥ 7.5 G/L was strongly associated with poorer PFS. Our previous mathematical model of trabectedin-induced neutropenia had shown that pre-therapeutic ANC could discriminate two populations of patients with different survival rates. It revealed a predictive factor that we would not necessarily have intuitively identified, and allowed us to choose the right threshold for validation as a future decision-making parameter. Using mathematical models to improve cancer treatment is a strategy on the rise, encompassing model-driven dosing and scheduling, and leading to the development of algorithms for precision medicine [11]. Median PFS of the overall population in our cohort was very similar to the pivotal, multicenter phase III trial comparing trabectedin and dacarbazine (4.2 months in the trabectedin arm) [12]. In the T-SAR trial, the median PFS was 1.5 months (m) in the best supportive care (BSC) arm and 3.1 m in the trabectedin arm. In the liposarcoma-STS cohort, the median PFS was 1.4 m and 5.1 m in the BSC and trabectedin arms, respectively, whereas it was 1.5 m and 1.8 m in the non-liposarcoma-STS group. In our study, histologic type had no prognostic value, although liposarcomas represented only 18% of all cases.
Although high histological grade is widely recognized as a factor of poor prognosis [13,14,15,16,17], high pre-therapeutic ANC outperformed its prognostic value in multivariate analysis. The independent negative prognostic value of high ANC has already been described in other cancer types, regardless of trabectedin use, notably in non-small-cell lung carcinomas [18], squamous oropharyngeal carcinomas [19], and melanoma [20], with a range of optimal thresholds from 4.5 to 7.5 G/L. Recent data showed that trabectedin selectively targets tumor-associated macrophages (TAM), and decreases the production of pro-inflammatory mediators, thus modifying the tumor microenvironment. This mode of action may contribute to the antitumor and anti-angiogenic activities of trabectedin [21,22,23,24]. Although neutrophilia is often present at cancer diagnosis, the biological pathways that may explain this observation have not been clearly identified and the origin appears to be multifactorial. One of these could be the paraneoplastic production of hematopoietic growth factors by tumor cells. Several studies have shown the existence of autocrine loops in the production of granulocyte growth factors (G-CSF and GM-CSF) in these tumors [25,26,27,28,29]. Interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNFα), which are involved in tumor-related inflammatory processes, are known to induce neutrophilia independently [30,31,32]. Tumor inflammation could lead to an antitumor immunity deficiency by involving T-helpers and T-regulating lymphocytes, notably via immune checkpoints (PD1–PDL1, CTLA4) and cytokines [33,34], thus promoting tumor growth. Another hypothesis is that neutrophils directly inhibit cellular immunity and, therefore, have an impact on the host antitumor response. In vitro studies have shown that an increased ANC inhibits the cytolytic activity of lymphocytes and (natural killer) NK cells [35,36,37]. Our study validated ANC ≥ 7.5 G/L as a negative predictive factor for PFS (HR = 3.39 (95% CI, 1.35–8.53); p = 0.010).
High histological grade and Hb < 12 g/dL were also independently associated with poor PFS. Although high histological grade is already recognized as a negative prognostic factor, we found that this initial prognostic value is still maintained in heavily pre-treated metastatic STS. Other prognostic factors of STS have already been identified, including in the early stages, such as tumor size and depth, primary and metastasis sites, patient’s age at diagnosis, gender, ECOG performance status, and previous treatments for advanced STS level [38,39,40,41,42,43,44,45,46,47,48]. Hemoglobin level has a prognostic value in metastatic renal cell carcinoma, as part of the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) score [49], and tends to be associated with shorter OS in lung cancer patients [50]. Nevertheless, there is no data on the prognostic value of baseline Hb levels and outcomes in sarcomas. The increasingly widespread use of molecular biology is now promoting a better understanding of the complexity of STS and making it possible to identify predictive signatures of the risk of metastatic recurrence in localized STS [51,52]. In vitro studies have suggested that cells with a deficient homologous recombination DNA repair pathway have a higher sensitivity to trabectedin [53], and retrospective studies have shown that BRCA1 (breast cancer gene 1) status may be predictive of trabectedin efficacy [54,55,56]. This observation was very recently assessed in a prospective EORTC (European Organisation for Research and Treatment of Cancer) study comparing the efficacy of trabectedin to doxorubicin, as a first line for advanced/metastatic soft tissue sarcoma [57]. It failed to confirm the predictive value of BRCA1 haplotype for trabectedin efficacy, but the number of informative patients was limited and there was no assessment of BRCA1 expression levels.
The six-month, progression-free rate with trabectedin is about 35–40% [12]. Thus, it is essential to establish predictive biomarkers of the clinical benefit of trabectedin to identify potential responders. This would offer a long-term tumor stabilization strategy for such patients and avoid treating those who would, potentially, only have side-effects with it. In our study, the PFS of patients with high ANC was equivalent to the PFS of liposarcoma patients in the trabectedin arm of the T-SAR trial, yet our cohort included only a minor proportion of liposarcomas. Baseline ANC may help in selecting candidates who would benefit from trabectedin treatment for advanced STS, leaving a gemcitabine-docetaxel regimen for those with higher ANC levels. Pazopanib, eribulin, aldoxorubicin, and regorafenib are other therapeutic options for use in advanced STS [58]. In our exploratory analysis of OS, we demonstrated here that high ANC could even be predictive of poorer OS. As discussed, high ANC could reflect tumor-related inflammatory processes involving T-helpers and T-regulating lymphocytes, or those directly inhibiting cellular immunity such as the paraneoplastic production of hematopoietic growth factors. This might reflect individual variations with regard to disease progression and patients’ general condition.
Our study is limited by its retrospective design due to a potential selection bias and lack of standardization in previous treatment strategies. However, the results concern a multicenter cohort of real-life patients treated in two expert centers and are, therefore, more likely to reflect the true diversity of patients treated by trabectedin in routine clinical practice.

4. Materials and Methods

4.1. Study Design and Data Source

We conducted a retrospective analysis of patients treated with trabectedin between January, 2010 and February, 2018 in two French, academic cancer centers. Because of the observational, non-interventional nature of this work, and since all patients were treated according to current guidelines such that their care was not affected or modified by the study, no ethical committee approval or clinical trial number was necessary under French law. All patients had metastatic STS and received at least two cycles of trabectedin, at the recommended dose of 1.5 mg/m² in 24-hour infusion every 21 days, as a standard therapy or in a clinical trial. Patient and tumor characteristics (i.e., age, sex, ECOG performance status (PS), body mass index (BMI), histological type, FNCLCC tumor grade (Fédération Nationale des Centres de Lutte Contre le Cancer), primary tumor location, existence of pulmonary, bone and/or hepatic metastases, pre-trabectedin ANC, hemoglobin and albumin levels, and progression and death, if any), were retrieved from medical files. Systemic treatment data (number of cycles, initial dose of trabectedin and eventual dose reduction, and previous and/or following treatment line, if any) were extracted from common computerized prescription software in our institutions (Chimio®, Computer Engineering, Paris, France).

4.2. Statistical Analysis

Categorical variables were described using counts and frequencies, and quantitative variables were reported using median and range. Patient characteristics were compared with the χ2 test for categorical variables and with the rank-Wilcoxon test for quantitative variables. Some variables were categorized according to cut-off values: age ≥ or <65 years, PS ECOG ≤ or >1, BMI < 19, between 19 and 25 or >25, number of prior chemotherapy lines > or ≤2, PFS under previous chemotherapy regimen, if any > or ≤4 months, ANC ≥ or <7.5 G/L, Hb level ≥ or <12 g/dL, and albumin level ≥ or <35 g/L, but also histological type (leiomyosarcoma, liposarcoma, undifferentiated sarcoma, and others), FNCLCC grade (1–2 or 3), and location of primary tumor (limb or other). PFS was defined as the time from the first course of trabectedin, to clinical and/or radiological progression, or to death from any cause, dependent on whichever occurred first. OS was defined as the time from the first course of trabectedin to death from any cause. Survival curves for PFS and OS rates were generated using the Kaplan-Meier method and compared with log-rank tests. The independent prognostic impact of the different clinico-biological factors identified in univariate analysis (p < 0.05) was tested by a multivariate Cox regression model including the following covariates categorized as mentioned above: performance status, tumor grade, ANC, and hemoglobin level.
The level of statistical significance was set at α = 0.05. Statistical analyses were carried out with SPSS® software version 24 (IBM, Armonk, New York, USA) and R software version 3.2.4. (R Foundation for Statistical Computing, Vienna, Austria). We followed the reporting recommendations specified in the REMARK (Reporting recommendations for tumor Marker prognostic studies) Statement [59].

5. Conclusions

We have validated high (i.e., >7.5) pre-therapeutic ANC as a predictive factor of poor PFS in patients starting trabectedin for STS. ANC could thus serve as a simple and inexpensive pre-therapeutic marker for decision-making in patients with metastatic STS.

Author Contributions

Conceptualization, A.d.N., M.A., F.B. and S.S.; methodology, A.d.N., M.A., F.B. and S.S.; software, A.d.N., M.A.; validation, A.d.N., M.A., F.B. and S.S.; formal analysis, A.d.N., M.A., F.B. and S.S.; investigation, A.d.N., M.A., F.B. and S.S.; resources, A.d.N., M.A., F.B. and S.S.; data curation, A.d.N., M.A., F.B. and S.S.; writing—original draft preparation, A.d.N. and M.A.; writing—review and editing, A.d.N., D.B., M.A., R.E.-C., F.D., F.B. and S.S.; visualization, A.d.N., D.B., M.A., R.E.-C., F.D., F.B. and S.S.; supervision, S.S.; project administration, F.B. and S.S.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef]
  2. Le Cesne, A.; Blay, J.Y.; Judson, I.; Van Oosterom, A.; Verweij, J.; Radford, J.; Lorigan, P.; Rodenhuis, S.; Ray-Coquard, I.; Bonvalot, S.; et al. Phase II Study of ET-743 in Advanced Soft Tissue Sarcomas: A European Organisation for the Research and Treatment of Cancer (EORTC) Soft Tissue and Bone Sarcoma Group Trial. J. Clin. Oncol. 2005, 23, 576–584. [Google Scholar] [CrossRef]
  3. Yovine, A.; Riofrio, M.; Blay, J.Y.; Brain, E.; Alexandre, J.; Kahatt, C.; Taamma, A.; Jimeno, J.; Martin, C.; Salhi, Y.; et al. Phase II study of ecteinascidin-743 in advanced pretreated soft tissue sarcoma patients. J. Clin. Oncol. 2004, 22, 890–899. [Google Scholar] [CrossRef] [PubMed]
  4. Garcia-Carbonero, R.; Supko, J.G.; Manola, J.; Seiden, M.V.; Harmon, D.; Ryan, D.P.; Quigley, M.T.; Merriam, P.; Canniff, J.; Goss, G.; et al. Phase II and pharmacokinetic study of ecteinascidin 743 in patients with progressive sarcomas of soft tissues refractory to chemotherapy. J. Clin. Oncol. 2004, 22, 1480–1490. [Google Scholar] [CrossRef] [PubMed]
  5. Garcia-Carbonero, R.; Supko, J.G.; Maki, R.G.; Manola, J.; Ryan, D.P.; Harmon, D.; Puchalski, T.A.; Goss, G.; Seiden, M.V.; Waxman, A.; et al. Ecteinascidin-743 (ET-743) for chemotherapy-naive patients with advanced soft tissue sarcomas: Multicenter phase II and pharmacokinetic study. J. Clin. Oncol. 2005, 23, 5484–5492. [Google Scholar] [CrossRef]
  6. Blay, J.-Y.; Italiano, A.; Ray-Coquard, I.; Le Cesne, A.; Duffaud, F.; Rios, M.; Collard, O.; Bertucci, F.; Bompas, E.; Isambert, N.; et al. Long-term outcome and effect of maintenance therapy in patients with advanced sarcoma treated with trabectedin: An analysis of 181 patients of the French ATU compassionate use program. BMC Cancer 2013, 13, 64. [Google Scholar] [CrossRef]
  7. Samuels, B.L.; Chawla, S.; Patel, S.; von Mehren, M.; Hamm, J.; Kaiser, P.E.; Schuetze, S.; Li, J.; Aymes, A.; Demetri, G.D. Clinical outcomes and safety with trabectedin therapy in patients with advanced soft tissue sarcomas following failure of prior chemotherapy: Results of a worldwide expanded access program study. Ann. Oncol. 2013, 24, 1703–1709. [Google Scholar] [CrossRef]
  8. Fayette, J.; Boyle, H.; Chabaud, S.; Favier, B.; Engel, C.; Cassier, P.; Thiesse, P.; Méeus, P.; Sunyach, M.-P.; Vaz, G.; et al. Efficacy of trabectedin for advanced sarcomas in clinical trials versus compassionate use programs: Analysis of 92 patients treated in a single institution. Anticancer. Drugs 2010, 21, 113–119. [Google Scholar] [CrossRef]
  9. Le Cesne, A.; Blay, J.-Y.; Cupissol, D.; Italiano, A.; Delcambre, C.; Penel, N.; Isambert, N.; Chevreau, C.; Bompas, E.; Bertucci, F.; et al. Results of a prospective randomized phase III T-SAR trial comparing trabectedin vs best supportive care (BSC) in patients with pretreated advanced soft tissue sarcoma (ASTS). Ann. Oncol. 2016, 27. [Google Scholar] [CrossRef] [Green Version]
  10. Andriantsoa, M.; El Cheikh, R.; Imbs, D.C.; Serre, R.; Duffaud, F.; Ciccolini, J.; Barbolosi, D.; Salas, S. Optimization of trabectedin temporal protocol using mathematical PK/PD modeling. J. Clin. Oncol. 2018, 36, e23553. [Google Scholar] [CrossRef]
  11. Barbolosi, D.; Ciccolini, J.; Lacarelle, B.; Barlési, F.; André, N. Computational oncology—Mathematical modelling of drug regimens for precision medicine. Nat. Rev. Clin. Oncol. 2016, 13, 242–254. [Google Scholar] [CrossRef] [PubMed]
  12. Demetri, G.D.; von Mehren, M.; Jones, R.L.; Hensley, M.L.; Schuetze, S.M.; Staddon, A.; Milhem, M.; Elias, A.; Ganjoo, K.; Tawbi, H.; et al. Efficacy and Safety of Trabectedin or Dacarbazine for Metastatic Liposarcoma or Leiomyosarcoma After Failure of Conventional Chemotherapy: Results of a Phase III Randomized Multicenter Clinical Trial. J. Clin. Oncol. 2016, 34, 786–793. [Google Scholar] [CrossRef]
  13. Trojani, M.; Contesso, G.; Coindre, J.M.; Rouesse, J.; Bui, N.B.; de Mascarel, A.; Goussot, J.F.; David, M.; Bonichon, F.; Lagarde, C. Soft-tissue sarcomas of adults; study of pathological prognostic variables and definition of a histopathological grading system. Int. J. Cancer 1984, 33, 37–42. [Google Scholar] [CrossRef] [PubMed]
  14. Costa, J.; Wesley, R.A.; Glatstein, E.; Rosenberg, S.A. The grading of soft tissue sarcomas. Results of a clinicohistopathologic correlation in a series of 163 cases. Cancer 1984, 53, 530–541. [Google Scholar] [CrossRef]
  15. van Unnik, J.A.; Coindre, J.M.; Contesso, C.; Albus-Lutter, C.E.; Schiodt, T.; Sylvester, R.; Thomas, D.; Bramwell, V.; Mouridsen, H.T. Grading of soft tissue sarcomas: Experience of the EORTC Soft Tissue and Bone Sarcoma Group. Eur. J. Cancer Oxf. Engl. 1993, 29A, 2089–2093. [Google Scholar] [CrossRef]
  16. Myhre-Jensen, O.; Kaae, S.; Madsen, E.H.; Sneppen, O. Histopathological grading in soft-tissue tumours. Relation to survival in 261 surgically treated patients. Acta Pathol. Microbiol. Immunol. Scand. 1983, 91, 145–150. [Google Scholar]
  17. Russell, W.O.; Cohen, J.; Enzinger, F.; Hajdu, S.I.; Heise, H.; Martin, R.G.; Meissner, W.; Miller, W.T.; Schmitz, R.L.; Suit, H.D. A clinical and pathological staging system for soft tissue sarcomas. Cancer 1977, 40, 1562–1570. [Google Scholar] [CrossRef] [Green Version]
  18. Teramukai, S.; Kitano, T.; Kishida, Y.; Kawahara, M.; Kubota, K.; Komuta, K.; Minato, K.; Mio, T.; Fujita, Y.; Yonei, T.; et al. Pretreatment neutrophil count as an independent prognostic factor in advanced non-small-cell lung cancer: An analysis of Japan Multinational Trial Organisation LC00-03. Eur. J. Cancer 2009, 45, 1950–1958. [Google Scholar] [CrossRef] [Green Version]
  19. Huang, S.H.; Waldron, J.N.; Milosevic, M.; Shen, X.; Ringash, J.; Su, J.; Tong, L.; Perez-Ordonez, B.; Weinreb, I.; Bayley, A.J.; et al. Prognostic value of pretreatment circulating neutrophils, monocytes, and lymphocytes in oropharyngeal cancer stratified by human papillomavirus status. Cancer 2015, 121, 545–555. [Google Scholar] [CrossRef]
  20. Schmidt, H.; Suciu, S.; Punt, C.J.; Gore, M.; Kruit, W.; Patel, P.; Lienard, D.; von der Maase, H.; Eggermont, A.M.; Keilholz, U. 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. [Google Scholar] [CrossRef] [PubMed]
  21. D’Incalci, M.; Galmarini, C.M. A review of trabectedin (ET-743): A unique mechanism of action. Mol. Cancer Ther. 2010, 9, 2157–2163. [Google Scholar] [CrossRef]
  22. Allavena, P.; Signorelli, M.; Chieppa, M.; Erba, E.; Bianchi, G.; Marchesi, F.; Olimpio, C.O.; Bonardi, C.; Garbi, A.; Lissoni, A.; et al. Anti-inflammatory properties of the novel antitumor agent yondelis (trabectedin): Inhibition of macrophage differentiation and cytokine production. Cancer Res. 2005, 65, 2964–2971. [Google Scholar] [CrossRef]
  23. Germano, G.; Frapolli, R.; Simone, M.; Tavecchio, M.; Erba, E.; Pesce, S.; Pasqualini, F.; Grosso, F.; Sanfilippo, R.; Casali, P.G.; et al. Antitumor and Anti-inflammatory Effects of Trabectedin on Human Myxoid Liposarcoma Cells. Cancer Res. 2010, 70, 2235–2244. [Google Scholar] [CrossRef]
  24. Germano, G.; Frapolli, R.; Belgiovine, C.; Anselmo, A.; Pesce, S.; Liguori, M.; Erba, E.; Uboldi, S.; Zucchetti, M.; Pasqualini, F.; et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013, 23, 249–262. [Google Scholar] [CrossRef]
  25. Nagata, S.; Tsuchiya, M.; Asano, S.; Kaziro, Y.; Yamazaki, T.; Yamamoto, O.; Hirata, Y.; Kubota, N.; Oheda, M.; Nomura, H. Molecular cloning and expression of cDNA for human granulocyte colony-stimulating factor. Nature 1986, 319, 415–418. [Google Scholar] [CrossRef]
  26. Asahi, Y.; Kubonishi, I.; Imamura, J.; Kamioka, M.; Matsushita, H.; Furihata, M.; Ohtsuki, Y.; Miyoshi, I. Establishment of a clonal cell line producing granulocyte colony-stimulating factor and parathyroid hormone-related protein from a lung cancer patient with leukocytosis and hypercalcemia. Jpn. J. Cancer Res. Gann 1996, 87, 451–458. [Google Scholar] [CrossRef]
  27. Lilly, M.B.; Devlin, P.E.; Devlin, J.J.; Rado, T.A. Production of granulocyte colony-stimulating factor by a human melanoma cell line. Exp. Hematol. 1987, 15, 966–971. [Google Scholar]
  28. Tachibana, M.; Miyakawa, A.; Tazaki, H.; Nakamura, K.; Kubo, A.; Hata, J.; Nishi, T.; Amano, Y. Autocrine growth of transitional cell carcinoma of the bladder induced by granulocyte-colony stimulating factor. Cancer Res. 1995, 55, 3438–3443. [Google Scholar]
  29. Oshika, Y.; Nakamura, M.; Abe, Y.; Fukuchi, Y.; Yoshimura, M.; Itoh, M.; Ohnishi, Y.; Tokunaga, T.; Fukushima, Y.; Hatanaka, H.; et al. Growth stimulation of non-small cell lung cancer xenografts by granulocyte-macrophage colony-stimulating factor (GM-CSF). Eur. J. Cancer Oxf. Engl. 1998, 34, 1958–1961. [Google Scholar] [CrossRef]
  30. Ulich, T.R.; del Castillo, J.; Keys, M.; Granger, G.A.; Ni, R.X. Kinetics and mechanisms of recombinant human interleukin 1 and tumor necrosis factor-alpha-induced changes in circulating numbers of neutrophils and lymphocytes. J. Immunol. 1987, 139, 3406–3415. [Google Scholar]
  31. Ulich, T.R.; del Castillo, J.; Guo, K.; Souza, L. The hematologic effects of chronic administration of the monokines tumor necrosis factor, interleukin-1, and granulocyte-colony stimulating factor on bone marrow and circulation. Am. J. Pathol. 1989, 134, 149–159. [Google Scholar] [PubMed]
  32. Ulich, T.R.; del Castillo, J.; Guo, K.Z. In vivo hematologic effects of recombinant interleukin-6 on hematopoiesis and circulating numbers of RBCs and WBCs. Blood 1989, 73, 108–110. [Google Scholar] [PubMed]
  33. Domingues, D.; Turner, A.; Silva, M.D.; Marques, D.S.; Mellidez, J.C.; Wannesson, L.; Mountzios, G.; de Mello, R.A. Immunotherapy and lung cancer: Current developments and novel targeted therapies. Immunotherapy 2014, 6, 1221–1235. [Google Scholar] [CrossRef] [PubMed]
  34. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer Immunoediting: Integrating Immunity’s Roles in Cancer Suppression and Promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef]
  35. Petrie, H.T.; Klassen, L.W.; Kay, H.D. Inhibition of human cytotoxic T lymphocyte activity in vitro by autologous peripheral blood granulocytes. J. Immunol. 1985, 134, 230–234. [Google Scholar] [PubMed]
  36. el-Hag, A.; Clark, R.A. Immunosuppression by activated human neutrophils. Dependence on the myeloperoxidase system. J. Immunol. 1987, 139, 2406–2413. [Google Scholar]
  37. Shau, H.Y.; Kim, A. Suppression of lymphokine-activated killer induction by neutrophils. J. Immunol. 1988, 141, 4395–4402. [Google Scholar]
  38. Coindre, J.M.; Terrier, P.; Bui, N.B.; Bonichon, F.; Collin, F.; Le Doussal, V.; Mandard, A.M.; Vilain, M.O.; Jacquemier, J.; Duplay, H.; et al. Prognostic factors in adult patients with locally controlled soft tissue sarcoma. A study of 546 patients from the French Federation of Cancer Centers Sarcoma Group. J. Clin. Oncol. 1996, 14, 869–877. [Google Scholar] [CrossRef]
  39. Pisters, P.W.; Leung, D.H.; Woodruff, J.; Shi, W.; Brennan, M.F. Analysis of prognostic factors in 1,041 patients with localized soft tissue sarcomas of the extremities. J. Clin. Oncol. 1996, 14, 1679–1689. [Google Scholar] [CrossRef] [PubMed]
  40. Bentz, B.G.; Singh, B.; Woodruff, J.; Brennan, M.; Shah, J.P.; Kraus, D. Head and Neck Soft Tissue Sarcomas: A Multivariate Analysis of Outcomes. Ann. Surg. Oncol. 2004, 11, 619–628. [Google Scholar] [CrossRef] [PubMed]
  41. Mattavelli, D.; Miceli, R.; Radaelli, S.; Mattavelli, F.; Cantù, G.; Barisella, M.; Quattrone, P.; Stacchiotti, S.; Sangalli, C.; Casali, P.G.; et al. Head and neck soft tissue sarcomas: Prognostic factors and outcome in a series of patients treated at a single institution. Ann. Oncol. 2013, 24, 2181–2189. [Google Scholar] [CrossRef] [PubMed]
  42. de Nonneville, A.; Toullec, C.; Blay, J.Y.; Ranchere, D.; Stoeckle, P.E.; Italiano, A.; Bonvalot, S.; Terrier, A.P.; Duffaud, F.; Bertucci, F. Patients with primary localized high-grade sarcomas of the digestive tract excluding GIST: A retrospective study from the French sarcoma group. Acta Gastro-Enterol. Belg. 2017, 80, 481–486. [Google Scholar]
  43. Le, Q.-T.X.; Fu, K.K.; Kroll, S.; Fitts, L.; Massullo, V.; Ferrell, L.; Kaplan, M.J.; Phillips, T.L. Prognostic factors in adult soft-tissue sarcomas of the head and neck. Int. J. Radiat. Oncol. 1997, 37, 975–984. [Google Scholar] [CrossRef]
  44. Kowalski, L.P.; San, C.I. Prognostic factors in head and neck soft tissue sarcomas: Analysis of 128 cases. J. Surg. Oncol. 1994, 56, 83–88. [Google Scholar] [CrossRef] [PubMed]
  45. Levine, E.A. Prognostic factors in soft tissue sarcoma. Semin. Surg. Oncol. 1999, 17, 23–32. [Google Scholar] [CrossRef]
  46. Levay, J.; O’sullivan, B.; Catton, C.; Bell, R.; Fornasier, V.; Cummings, B.; Hao, D.; Warr, D.; Quirt, I. Outcome and prognostic factors in soft tissue sarcoma in the adult. Int. J. Radiat. Oncol. 1993, 27, 1091–1099. [Google Scholar] [CrossRef]
  47. Ueda, T.; Aozasa, K.; Tsujimoto, M.; Hamada, H.; Hayashi, H.; Ono, K.; Matsumoto, K. Multivariate analysis for clinical prognostic factors in 163 patients with soft tissue sarcoma. Cancer 1988, 62, 1444–1450. [Google Scholar] [CrossRef] [Green Version]
  48. Van Glabbeke, M.; van Oosterom, A.T.; Oosterhuis, J.W.; Mouridsen, H.; Crowther, D.; Somers, R.; Verweij, J.; Santoro, A.; Buesa, J.; Tursz, T. Prognostic Factors for the Outcome of Chemotherapy in Advanced Soft Tissue Sarcoma: An Analysis of 2,185 Patients Treated with Anthracycline-Containing First-Line Regimens—A European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group Study. J. Clin. Oncol. 1999, 17, 150–157. [Google Scholar]
  49. Ko, J.J.; Xie, W.; Kroeger, N.; Lee, J.-L.; Rini, B.I.; Knox, J.J.; Bjarnason, G.A.; Srinivas, S.; Pal, S.K.; Yuasa, T.; et al. The International Metastatic Renal Cell Carcinoma Database Consortium model as a prognostic tool in patients with metastatic renal cell carcinoma previously treated with first-line targeted therapy: A population-based study. Lancet Oncol. 2015, 16, 293–300. [Google Scholar] [CrossRef]
  50. Gauthier, I.; Ding, K.; Winton, T.; Shepherd, F.A.; Livingston, R.; Johnson, D.H.; Rigas, J.R.; Whitehead, M.; Graham, B.; Seymour, L. Impact of hemoglobin levels on outcomes of adjuvant chemotherapy in resected non-small cell lung cancer: The JBR.10 trial experience. Lung Cancer Amst. Neth. 2007, 55, 357–363. [Google Scholar] [CrossRef]
  51. Chibon, F.; Lagarde, P.; Salas, S.; Pérot, G.; Brouste, V.; Tirode, F.; Lucchesi, C.; de Reynies, A.; Kauffmann, A.; Bui, B.; et al. Validated prediction of clinical outcome in sarcomas and multiple types of cancer on the basis of a gene expression signature related to genome complexity. Nat. Med. 2010, 16, 781–787. [Google Scholar] [CrossRef] [PubMed]
  52. Bertucci, F.; De Nonneville, A.; Finetti, P.; Perrot, D.; Nilbert, M.; Italiano, A.; Le Cesne, A.; Skubitz, K.M.; Blay, J.Y.; Birnbaum, D. The Genomic Grade Index predicts postoperative clinical outcome in patients with soft-tissue sarcoma. Ann. Oncol. 2018, 29, 459–465. [Google Scholar] [CrossRef]
  53. Soares, D.G.; Escargueil, A.E.; Poindessous, V.; Sarasin, A.; de Gramont, A.; Bonatto, D.; Henriques, J.A.P.; Larsen, A.K. Replication and homologous recombination repair regulate DNA double-strand break formation by the antitumor alkylator ecteinascidin 743. Proc. Natl. Acad. Sci. USA 2007, 104, 13062–13067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Schöffski, P.; Taron, M.; Jimeno, J.; Grosso, F.; Sanfilipio, R.; Casali, P.G.; Cesne, A.L.; Jones, R.L.; Blay, J.-Y.; Poveda, A.; et al. Predictive impact of DNA repair functionality on clinical outcome of advanced sarcoma patients treated with trabectedin: A retrospective multicentric study. Eur. J. Cancer 2011, 47, 1006–1012. [Google Scholar] [CrossRef]
  55. Italiano, A.; Laurand, A.; Laroche, A.; Casali, P.; Sanfilippo, R.; Le Cesne, A.; Judson, I.; Blay, J.-Y.; Ray-Coquard, I.; Bui, B.; et al. ERCC5/XPG, ERCC1, and BRCA1 gene status and clinical benefit of trabectedin in patients with soft tissue sarcoma. Cancer 2011, 117, 3445–3456. [Google Scholar] [CrossRef] [Green Version]
  56. Laroche-Clary, A.; Chaire, V.; Le Morvan, V.; Neuville, A.; Bertucci, F.; Salas, S.; Sanfilippo, R.; Pourquier, P.; Italiano, A. BRCA1 haplotype and clinical benefit of trabectedin in soft-tissue sarcoma patients. Br. J. Cancer 2015, 112, 688–692. [Google Scholar] [CrossRef] [Green Version]
  57. Italiano, A.; Touati, N.; Litière, S.; Collin, F.; Pourquier, P.; Gronchi, A. Prospective assessment of the predictive value of the BRCA1 gene status in sarcoma patients treated with trabectedin: An updated analysis of the EORTC 62091 trial. Cancer Med. 2018, 7, 1575–1577. [Google Scholar] [CrossRef]
  58. Blay, J.-Y. Getting up-to-date in the management of soft tissue sarcoma. Future Oncol. Lond. Engl. 2018, 14, 3–13. [Google Scholar] [CrossRef]
  59. McShane, L.M.; Altman, D.G.; Sauerbrei, W.; Taube, S.E.; Gion, M.; Clark, G.M. REporting recommendations for tumour MARKer prognostic studies (REMARK). Br. J. Cancer 2005, 93, 387–391. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Kaplan-Meier estimates of progression-free survival (left panels) and overall survival (right panels), respectively, in patients with absolute neutrophil count (ANC) < or ≥ 7.5 G/L (A,D) patients with grade 1, 2, or 3 tumors (B,E) and patients with hemoglobin level (Hb) < or ≥ 12 g/L (C,F).
Figure 1. Kaplan-Meier estimates of progression-free survival (left panels) and overall survival (right panels), respectively, in patients with absolute neutrophil count (ANC) < or ≥ 7.5 G/L (A,D) patients with grade 1, 2, or 3 tumors (B,E) and patients with hemoglobin level (Hb) < or ≥ 12 g/L (C,F).
Cancers 11 00432 g001
Figure 2. Cox-adjusted curves of progression-free survival (A) and overall survival (B) in patients with ANC < or ≥ 7.5 G/L.
Figure 2. Cox-adjusted curves of progression-free survival (A) and overall survival (B) in patients with ANC < or ≥ 7.5 G/L.
Cancers 11 00432 g002
Table 1. Patient and tumor characteristics. Percentages are calculated in relation to the number of available data.
Table 1. Patient and tumor characteristics. Percentages are calculated in relation to the number of available data.
VariablesTotal PopulationPatients According to Pre-Therapeutic ANC
ANC < 7.5 G/LANC ≥ 7.5 G/Lp-Value
n%n%n%(Khi2 or Wilcoxon)
Gender
Female4956%3763%640%0.111
Male3844%2237%960%
Age (years)
Median (Min–Max)59.75(19.96–83.58)59.75(22.03–83.58)55.32(19.96–70.13)0.480
<65 y6271%4271%1067%0.732
≥65 y2529%1729%533%
ECOG status
<26881%1119%429%0.446
≥21619%4681%1071%
MD3
Histological type
LMS3844%2644%427%0.672
LPS1618%1119%427%
UPS45%35%17%
OTHER2933%1932%640%
Translocation-related sarcoma
No7283%4983%1173%0.391
Yes1517%1017%427%
Grade (FNCLCC)
G146%24%218%0.164
G22844%2351%327%
G33250%2044%655%
MD23
Location of the primary site
Limb2933%2034%533%0.967
Other5867%3966%1067%
Lung metastases
No2327%1628%320%0.551
Yes6373%4272%1280%
MD1
Liver metastases
No5463%3662%1280%0.192
Yes3237%2238%320%
MD1
Bone metastases
No6375%4173%1173%0.993
Yes2125%1527%427%
MD3
BMI
<191416%916%320%0.914
19–254148%2848%747%
>253136%2136%533%
MD1
Number of prior lines of chemotherapy
Median (Min–Max)2(0–7)2(0–5)3(0–7)0.269
≤25464%3967%643%0.091
>23136%1933%857%
MD2
PFS under previous chemotherapy regimen
Median (Min–Max) (months)5.09(0.29–20.53)5.06(1.61–19.81)3.43(0.76–20.53)0.103
≤4 months3348%2349%758%0.561
>4 months3652%2451%542%
MD18
Trabectedin dose reduction
No2327%1627%429%0.913
Yes6373%4373%1071%
MD1
Absolute neutrophil count at the 1st cycle of Trabectedin
<7.55980%
≥7.51520%
MD13
Hemoglobin at the 1st cycle of Trabectedin
<123447%2646%542%0.803
≥123853%3154%758%
MD15
Albumin at the 1st cycle of Trabectedin
<351741%932%660%0.122
≥352459%1968%440%
MD46
Abbreviation: LMS, Leiomyosarcoma; LPS, Liposarcoma; UPS, Undifferentiated pleomorphic sarcoma; BMI, Body Mass Index; PFS, Progression-free survival; MD, Missing data.
Table 2. Univariate analysis of factors associated with progression-free survival (PFS) and overall survival (OS).
Table 2. Univariate analysis of factors associated with progression-free survival (PFS) and overall survival (OS).
VariablesClassesMedian PFS (CI 95%)p-Value Log-RankMedian OS (CI 95%)p-Value Log-Rank
SexMale4.07 (2.51–5.65)0.35517.18 (5.08–29.28)0.787
Female4.83 (3.44–6.22) 26.78 (12.70–40.85)
Age≥655.78 (0.15–11.41)0.03827.50 (27.10–27.90)0.135
<654.07 (2.56–5.58) 14.88 (8.21–21.55)
Performance Status ≤14.17 (2.27–6.07)0.9927.30 (13.63–40.97)0.015
>13.94 (2.41–5.48) 6.96 (0.88–13.05)
Histological typeLMS5.42 (2.89–7.95)0.1527.49 (25.39–29.60)0.126
LPS4.07 (0.49–7.66) 6.97 (3.65–10.28)
UPS1.54 (0.84–2.25) 5.91 (0.405–11.42)
OTHER3.88 (2.32–5.43) 17.18 (10.06–24.31)
Translocation-related sarcomaYes3.71 (1.46–5.97)0.85514.88 (9.76–29.73)0.959
No4.60 (3.31–5.89) 26.78 (14.93–38.63)
Grade (FNCLCC)G14.07 (0–8.29)0.0084.89 (0–21.94)0.052
G28.31 (2.15–14.47) 28.78 (26.28–31.28)
G33.22 (0.76–5.68) 14.160 (7.77–20.55)
Location of the primary siteLimb5.78 (2.11–9.45)0.02833.12 (9.05–57.18)0.184
Other3.94 (2.45–5.43) 17.18 (9.8–24.56)
Lung metastasesYes3.88 (2.55–5.19)0.02926.78 (5.78–47.77)0.090
No11.89 (2.43–21.35) 45.68 (8.54–59.42)
Liver metastasesYes4.60 (2.60–6.60)0.66728.78 (19.92–37.64)0.145
No3.88 (2.25–5.51) 14.88 (6.38–23.39)
Bone metastasesYes3.22 (1.16–5.28)0.29214.88 (3.60–26.16)0.297
No4.34 (3.07–5.60) 26.78 (15.44–38.10)
BMI<192.04 (1.43–2.64)0.58911.37 (3.08–19.66)0.388
19–254.17 (2.85–5.49) 27.30 (11.05–43.56)
>255.16 (3.44–6.87) 26.78 (4.69–48.86)
Number of prior lines of chemotherapy>23.71 (2.00–5.42)0.25417.18 (3.38–30.99)0.814
≤24.83 (3.07–6.59) 27.30 (13.95–40.65)
Trabectedin dose reductionYes5.42 (3.48–7.36)0.01827.30 (16.25–38.35)0.484
No2.89 (1.91–3.87) 14.88 (8.59–21.18)
Absolute neutrophil count at the 1st cycle of Trabectedin>7.53.22 (1.60–4.87)0.0094.89 (0–11.82)0.011
≤7.55.78 (3.95–7.61) 26.78 (13.47–40.08)
Hemoglobin at the 1st cycle of Trabectedin≥126.70 (2.99–10.42)0.00743.14 (17.87–68.40)<0.001
<121.40 (0–4.81) 8.41 (0.66–16.16)
Albumin at the 1st cycle of Trabectedin≥354.83 (3.89–5.77)0.04326.78 (10.06–43.49)0.036
<352.07 (1.37–2.77) 4.89 (1.39–8.40)
Table 3. Multivariate analysis of factors associated with progression-free survival (PFS) and overall survival (OS).
Table 3. Multivariate analysis of factors associated with progression-free survival (PFS) and overall survival (OS).
VariablesPFSOS
HR(95% CI)p-ValueHR(95% CI)p-Value
minmaxminmax
Performance Status
1–2Reference category
3–41.520.703.300.2891.000.372.740.998
Grade (FNCLCC)
1–2Reference category
32.751.405.400.0032.490.926.730.071
Absolute neutrophil count
<7.5 G/LReference category
≥7.5 G/L3.391.358.530.0104.901.5715.340.006
Haemoglobin
<12 g/dLReference category
≥12 g/dL0.350.170.720.0050.170.060.500.001
FNCLCC: Fédération Nationale des Centres de Lutte Contre le Cancer; HR: hazard ratio.

Share and Cite

MDPI and ACS Style

de Nonneville, A.; Barbolosi, D.; Andriantsoa, M.; El-Cheikh, R.; Duffaud, F.; Bertucci, F.; Salas, S. Validation of Neutrophil Count as An Algorithm-Based Predictive Factor of Progression-Free Survival in Patients with Metastatic Soft Tissue Sarcomas Treated with Trabectedin. Cancers 2019, 11, 432. https://doi.org/10.3390/cancers11030432

AMA Style

de Nonneville A, Barbolosi D, Andriantsoa M, El-Cheikh R, Duffaud F, Bertucci F, Salas S. Validation of Neutrophil Count as An Algorithm-Based Predictive Factor of Progression-Free Survival in Patients with Metastatic Soft Tissue Sarcomas Treated with Trabectedin. Cancers. 2019; 11(3):432. https://doi.org/10.3390/cancers11030432

Chicago/Turabian Style

de Nonneville, Alexandre, Dominique Barbolosi, Maeva Andriantsoa, Raouf El-Cheikh, Florence Duffaud, François Bertucci, and Sebastien Salas. 2019. "Validation of Neutrophil Count as An Algorithm-Based Predictive Factor of Progression-Free Survival in Patients with Metastatic Soft Tissue Sarcomas Treated with Trabectedin" Cancers 11, no. 3: 432. https://doi.org/10.3390/cancers11030432

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop