8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Geospatial Cellular Distribution of Cancer-Associated Fibroblasts Significantly Impacts Clinical Outcomes in Metastatic Clear Cell 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

          Simple Summary

          Cancer-associated fibroblasts (CAFs) are highly prevalent cells in the clear cell renal cell carcinoma (ccRCC) tumor immune microenvironment. CAFs are thought to potentiate tumor proliferation primarily through paracrine interactions, as evidenced by laboratory-based studies. We sought to corroborate these findings using surgically removed tissue samples from 96 patients with metastatic ccRCC and associate geospatial relationships between CAFs and rapidly proliferating tumor cells with survival outcomes. We found that CAFs exhibited more geospatial clustering with proliferating tumor cells than with dying tumor cells, and patients whose samples exhibited higher tumor cell proliferation had worse overall survival and were more likely to be resistant to systemic tyrosine-kinase-inhibiting targeted therapies. Immunotherapy resistance was not associated with the geospatial metrics measured in this analysis. Overall, these findings suggest that close proximity to CAFs potentiates tumor cell proliferation, worsening survival and conferring resistance to targeted therapies.

          Abstract

          Cancer-associated fibroblasts (CAF) are highly prevalent cells in the tumor microenvironment in clear cell renal cell carcinoma (ccRCC). CAFs exhibit a pro-tumor effect in vitro and have been implicated in tumor cell proliferation, metastasis, and treatment resistance. Our objective is to analyze the geospatial distribution of CAFs with proliferating and apoptotic tumor cells in the ccRCC tumor microenvironment and determine associations with survival and systemic treatment. Pre-treatment primary tumor samples were collected from 96 patients with metastatic ccRCC. Three adjacent slices were obtained from 2 tumor-core regions of interest (ROI) per patient, and immunohistochemistry (IHC) staining was performed for αSMA, Ki-67, and caspase-3 to detect CAFs, proliferating cells, and apoptotic cells, respectively. H-scores and cellular density were generated for each marker. ROIs were aligned, and spatial point patterns were generated, which were then used to perform spatial analyses using a normalized Ripley’s K function at a radius of 25 μm (nK(25)). The survival analyses used an optimal cut-point method, maximizing the log-rank statistic, to stratify the IHC-derived metrics into high and low groups. Multivariable Cox regression analyses were performed accounting for age and International Metastatic RCC Database Consortium (IMDC) risk category. Survival outcomes included overall survival (OS) from the date of diagnosis, OS from the date of immunotherapy initiation (OS-IT), and OS from the date of targeted therapy initiation (OS-TT). Therapy resistance was defined as progression-free survival (PFS) <6 months, and therapy response was defined as PFS >9 months. CAFs exhibited higher cellular clustering with Ki-67 + cells than with caspase-3 + cells (nK(25): Ki-67 1.19; caspase-3 1.05; p = 0.04). The median nearest neighbor (NN) distance from CAFs to Ki-67 + cells was shorter compared to caspase-3 + cells (15 μm vs. 37 μm, respectively; p < 0.001). Multivariable Cox regression analyses demonstrated that both high Ki-67 + density and H-score were associated with worse OS, OS-IT, and OS-TT. Regarding αSMA+CAFs, only a high H-score was associated with worse OS, OS-IT, and OS-TT. For caspase-3 +, high H-score and density were associated with worse OS and OS-TT. Patients whose tumors were resistant to targeted therapy (TT) had higher Ki-67 density and H-scores than those who had TT responses. Overall, this ex vivo geospatial analysis of CAF distribution suggests that close proximity clustering of tumor cells and CAFs potentiates tumor cell proliferation, resulting in worse OS and resistance to TT in metastatic ccRCC.

          Related collections

          Most cited references30

          • Record: found
          • Abstract: found
          • Article: not found

          The biology and function of fibroblasts in cancer.

          Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Turning foes to friends: targeting cancer-associated fibroblasts

            Current paradigms of cancer-centric therapeutics are usually not sufficient to eradicate the malignancy, as the cancer stroma may prompt tumour relapse and therapeutic resistance. Among all the stromal cells that populate the tumour microenvironment, cancer-associated fibroblasts (CAFs) are the most abundant and are critically involved in cancer progression. CAFs regulate the biology of tumour cells and other stromal cells via cell-cell contact, releasing numerous regulatory factors and synthesizing and remodelling the extracellular matrix, and thus these cells affect cancer initiation and development. The recent characterization of CAFs based on specific cell surface markers not only deepens our insight into their phenotypic heterogeneity and functional diversity but also brings CAF-targeting therapies for cancer treatment onto the agenda. In this Review, we discuss the current knowledge of biological hallmarks, cellular origins, phenotypical plasticity and functional heterogeneity of CAFs and underscore their contribution to cancer progression. Moreover, we highlight relevant translational advances and potential therapeutic strategies that target CAFs for cancer treatment.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The Ki-67 protein: from the known and the unknown.

              The expression of the human Ki-67 protein is strictly associated with cell proliferation. During interphase, the antigen can be exclusively detected within the nucleus, whereas in mitosis most of the protein is relocated to the surface of the chromosomes. The fact that the Ki-67 protein is present during all active phases of the cell cycle (G(1), S, G(2), and mitosis), but is absent from resting cells (G(0)), makes it an excellent marker for determining the so-called growth fraction of a given cell population. In the first part of this study, the term proliferation marker is discussed and examples of the applications of anti-Ki-67 protein antibodies in diagnostics of human tumors are given. The fraction of Ki-67-positive tumor cells (the Ki-67 labeling index) is often correlated with the clinical course of the disease. The best-studied examples in this context are carcinomas of the prostate and the breast. For these types of tumors, the prognostic value for survival and tumor recurrence has repeatedly been proven in uni- and multivariate analysis. The preparation of new monoclonal antibodies that react with the Ki-67 equivalent protein from rodents now extends the use of the Ki-67 protein as a proliferation marker to laboratory animals that are routinely used in basic research. The second part of this review focuses on the biology of the Ki-67 protein. Our current knowledge of the Ki-67 gene and protein structure, mRNA splicing, expression, and cellular localization during the cell-division cycle is summarized and discussed. Although the Ki-67 protein is well characterized on the molecular level and extensively used as a proliferation marker, the functional significance still remains unclear. There are indications, however, that Ki-67 protein expression is an absolute requirement for progression through the cell-division cycle. Copyright 2000 Wiley-Liss, Inc.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                26 July 2021
                August 2021
                : 13
                : 15
                : 3743
                Affiliations
                [1 ]Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Ali.Hajiran@ 123456moffitt.org (A.H.); andrew.chang@ 123456duke.edu (A.C.); ahmetmurataydin@ 123456gmail.com (A.M.A.); logan.zemp@ 123456moffitt.org (L.Z.); Esther.Katende@ 123456moffitt.org (E.K.); Jad.Chahoud@ 123456moffitt.org (J.C.); philippe.spiess@ 123456moffitt.org (P.E.S.); Natasha.Francis@ 123456moffitt.org (N.F.); michelle.fournier@ 123456moffitt.org (M.F.); brandon.manley@ 123456moffitt.org (B.J.M.)
                [2 ]Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; gregory.kimmel@ 123456moffitt.org (G.J.K.); mferrall.fairbanks@ 123456bme.ufl.edu (M.C.F.-F.); philipp.altrock@ 123456moffitt.org (P.M.A.)
                [3 ]Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; youngchul.kim@ 123456moffitt.org
                [4 ]Analytic Microcopy Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Joseph.Johnson@ 123456moffitt.org
                [5 ]Tissue Core Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; noel.clark@ 123456moffitt.org
                [6 ]Department of Pathology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; Jasreman.Dhillon@ 123456moffitt.org
                [7 ]Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA; Jong.Park@ 123456moffitt.org
                [8 ]Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Liang.Wang@ 123456moffitt.org
                [9 ]Immunology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; james.mule@ 123456moffitt.org
                Author notes
                [* ]Correspondence: nicholas.chakiryan@ 123456moffitt.org ; Tel.: +1-813-745-3208; Fax: +1-813-745-8494
                Author information
                https://orcid.org/0000-0001-9263-8957
                https://orcid.org/0000-0002-6047-8431
                https://orcid.org/0000-0002-3380-6989
                https://orcid.org/0000-0002-5723-1972
                https://orcid.org/0000-0002-6384-6447
                https://orcid.org/0000-0002-9364-8572
                https://orcid.org/0000-0002-7927-1510
                Article
                cancers-13-03743
                10.3390/cancers13153743
                8345222
                34359645
                c53d43a3-7ec5-4701-a8f2-befa7691058c
                © 2021 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( https://creativecommons.org/licenses/by/4.0/).

                History
                : 24 May 2021
                : 22 July 2021
                Categories
                Article

                metastatic clear cell renal cell carcinoma,cancer associated fibroblasts,ki-67,spatial analysis,immunohistochemistry

                Comments

                Comment on this article