36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Change in Neutrophil-to-lymphocyte ratio (NLR) in response to immune checkpoint blockade for metastatic renal cell carcinoma

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          An elevated Neutrophil-to-lymphocyte ratio (NLR) is associated with worse outcomes in several malignancies. However, its role with contemporary immune checkpoint blockade (ICB) is unknown. We investigated the utility of NLR in metastatic renal cell carcinoma (mRCC) patients treated with PD-1/PD-L1 ICB.

          Methods

          We examined NLR at baseline and 6 (±2) weeks later in 142 patients treated between 2009 and 2017 at Dana-Farber Cancer Institute (Boston, USA). Landmark analysis at 6 weeks was conducted to explore the prognostic value of relative NLR change on overall survival (OS), progression-free survival (PFS), and objective response rate (ORR). Cox and logistic regression models allowed for adjustment of line of therapy, number of IMDC risk factors, histology and baseline NLR.

          Results

          Median follow up was 16.6 months (range: 0.7–67.8). Median duration on therapy was 5.1 months (<1–61.4). IMDC risk groups were: 18% favorable, 60% intermediate, 23% poor-risk. Forty-four percent were on first-line ICB and 56% on 2nd line or more. Median NLR was 3.9 (1.3–42.4) at baseline and 4.1 (1.1–96.4) at week 6. Patients with a higher baseline NLR showed a trend toward lower ORR, shorter PFS, and shorter OS. Higher NLR at 6 weeks was a significantly stronger predictor of all three outcomes than baseline NLR. Relative NLR change by ≥25% from baseline to 6 weeks after ICB therapy was associated with reduced ORR and an independent prognostic factor for PFS ( p < 0.001) and OS ( p = 0.004), whereas a decrease in NLR by ≥25% was associated with improved outcomes.

          Conclusions

          Early decline and NLR at 6 weeks are associated with significantly improved outcomes in mRCC patients treated with ICB. The prognostic value of the readily-available NLR warrants larger, prospective validation.

          Related collections

          Most cited references29

          • Record: found
          • Abstract: found
          • Article: found

          Hallmarks of Cancer: The Next Generation

          The hallmarks of cancer comprise six biological capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metabolism and evading immune destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment." Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer. Copyright © 2011 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).

            Assessment of the change in tumour burden is an important feature of the clinical evaluation of cancer therapeutics: both tumour shrinkage (objective response) and disease progression are useful endpoints in clinical trials. Since RECIST was published in 2000, many investigators, cooperative groups, industry and government authorities have adopted these criteria in the assessment of treatment outcomes. However, a number of questions and issues have arisen which have led to the development of a revised RECIST guideline (version 1.1). Evidence for changes, summarised in separate papers in this special issue, has come from assessment of a large data warehouse (>6500 patients), simulation studies and literature reviews. HIGHLIGHTS OF REVISED RECIST 1.1: Major changes include: Number of lesions to be assessed: based on evidence from numerous trial databases merged into a data warehouse for analysis purposes, the number of lesions required to assess tumour burden for response determination has been reduced from a maximum of 10 to a maximum of five total (and from five to two per organ, maximum). Assessment of pathological lymph nodes is now incorporated: nodes with a short axis of 15 mm are considered measurable and assessable as target lesions. The short axis measurement should be included in the sum of lesions in calculation of tumour response. Nodes that shrink to <10mm short axis are considered normal. Confirmation of response is required for trials with response primary endpoint but is no longer required in randomised studies since the control arm serves as appropriate means of interpretation of data. Disease progression is clarified in several aspects: in addition to the previous definition of progression in target disease of 20% increase in sum, a 5mm absolute increase is now required as well to guard against over calling PD when the total sum is very small. Furthermore, there is guidance offered on what constitutes 'unequivocal progression' of non-measurable/non-target disease, a source of confusion in the original RECIST guideline. Finally, a section on detection of new lesions, including the interpretation of FDG-PET scan assessment is included. Imaging guidance: the revised RECIST includes a new imaging appendix with updated recommendations on the optimal anatomical assessment of lesions. A key question considered by the RECIST Working Group in developing RECIST 1.1 was whether it was appropriate to move from anatomic unidimensional assessment of tumour burden to either volumetric anatomical assessment or to functional assessment with PET or MRI. It was concluded that, at present, there is not sufficient standardisation or evidence to abandon anatomical assessment of tumour burden. The only exception to this is in the use of FDG-PET imaging as an adjunct to determination of progression. As is detailed in the final paper in this special issue, the use of these promising newer approaches requires appropriate clinical validation studies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cancer-related inflammation.

              The mediators and cellular effectors of inflammation are important constituents of the local environment of tumours. In some types of cancer, inflammatory conditions are present before a malignant change occurs. Conversely, in other types of cancer, an oncogenic change induces an inflammatory microenvironment that promotes the development of tumours. Regardless of its origin, 'smouldering' inflammation in the tumour microenvironment has many tumour-promoting effects. It aids in the proliferation and survival of malignant cells, promotes angiogenesis and metastasis, subverts adaptive immune responses, and alters responses to hormones and chemotherapeutic agents. The molecular pathways of this cancer-related inflammation are now being unravelled, resulting in the identification of new target molecules that could lead to improved diagnosis and treatment.
                Bookmark

                Author and article information

                Contributors
                Aly-Khan_Lalani@DFCI.HARVARD.EDU
                wxie@jimmy.harvard.edu
                dylan.martini@emory.edu
                JohnA_Steinharter@DFCI.HARVARD.EDU
                craig.norton@umassmed.edu
                kmkrajewski@bwh.harvard.edu
                Audrey_Duquette@DFCI.HARVARD.EDU
                Dominick_Bosse@DFCI.HARVARD.EDU
                Joaquim_Bellmunt@DFCI.HARVARD.EDU
                EliezerM_VanAllen@DFCI.HARVARD.EDU
                Bradley_McGregor@DFCI.HARVARD.EDU
                creighto@bcm.edu
                LaurenC_Harshman@DFCI.HARVARD.EDU
                (617) 632-5456 , Toni_Choueiri@dfci.harvard.edu
                Journal
                J Immunother Cancer
                J Immunother Cancer
                Journal for Immunotherapy of Cancer
                BioMed Central (London )
                2051-1426
                22 January 2018
                22 January 2018
                2018
                : 6
                : 5
                Affiliations
                [1 ]ISNI 0000 0001 2106 9910, GRID grid.65499.37, Lank Center for Genitourinary Oncology, , Dana-Farber Cancer Institute, ; 450 Brookline Avenue, Dana 1230, Boston, MA 02215 USA
                [2 ]ISNI 0000 0001 2106 9910, GRID grid.65499.37, Department of Biostatistics and Computational Biology, , Dana-Farber Cancer Institute, ; 450 Brookline Avenue, Boston, MA 02215 USA
                [3 ]ISNI 0000 0001 0941 6502, GRID grid.189967.8, Emory University School of Medicine, ; 100 Woodruff Circle, Atlanta, GA 30322 USA
                [4 ]ISNI 0000 0004 0378 8294, GRID grid.62560.37, Department of Imaging, Dana-Farber Cancer Institute & Department of Radiology, , Brigham and Women’s Hospital, ; 450 Brookline Avenue, Boston, MA 02215 USA
                [5 ]GRID grid.66859.34, The Eli and Edythe L. Broad Institute of MIT and Harvard, ; 415 Main St, Cambridge, MA 02142 USA
                [6 ]ISNI 0000 0001 2160 926X, GRID grid.39382.33, Department of Medicine, , Dan L. Duncan Comprehensive Cancer Center, and Human Genome Sequencing Center, Baylor College of Medicine, ; One Baylor Plaza MS 305, Houston, TX 77030 USA
                Article
                315
                10.1186/s40425-018-0315-0
                5776777
                29353553
                566bbf48-30ab-41e3-aebb-3aaca508f4f7
                © The Author(s). 2018

                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.

                History
                : 2 November 2017
                : 3 January 2018
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000009, Foundation for the National Institutes of Health;
                Award ID: Kidney Spore
                Award Recipient :
                Categories
                Research Article
                Custom metadata
                © The Author(s) 2018

                neutrophil-to-lymphocyte ratio,pd-1/pd-l1 pd-l1,immunotherapy,renal cell carcinoma,prognostic biomarker

                Comments

                Comment on this article