Skip to main content

EDITORIAL article

Front. Oncol., 07 May 2019
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Cancer Plasticity and The Microenvironment: Implications for Immunity and Therapy Response View all 16 articles

Editorial: Cancer Plasticity and the Microenvironment: Implications for Immunity and Therapy Response

  • 1School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
  • 2Cancer Immunobiology Program, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
  • 3Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, and The Translational Research Institute, Brisbane, QLD, Australia
  • 4Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
  • 5Clinical Pathology and Surgery, The University of Melbourne, Melbourne, VIC, Australia

Over the last decade, our understanding of how cancer cells interact with their microenvironment has grown exponentially. It has become evident that a complex interplay exists between malignant cells and benign host cells. The cancer cells and the “normal” cells, especially the immune compartment, undergo constant co-evolution to dynamically “shape” each other. Early attempts to utilize immune cells to favor anti-tumor responses date back more than a hundred years, when William Coley used bacteria to evoke immune responses in cancer patients, however, the results were controversial. Now, in the twenty-first century, “evading the immune system” has been recognized as a key hallmark of cancer (1).

The importance of the immune system in the recognition and clearance of cancer has been illustrated by the unprecedented success of immune checkpoint therapies in the treatment of multiple cancers and the durable responses observed in patients with late-stage disease (2, 3). Presently, clinical success has outpaced scientific understanding despite an ever increasing number of publications on a multitude of factors and pathways contributing to therapy response and resistance. What is clear, however, is that cancer cell plasticity—the ability of cancer cells to alter their phenotype or function—is linked to tumor cell dormancy, tumor progression, metastatic processes and treatment resistance (4). This can affect clonal selection and is implicated in immune evasion, thus influencing response to immunotherapy and patient outcome (5, 6). Cancer plasticity can be induced by a multitude of factors, most notably the tumor microenvironment (TME) (7). In this research topic, we have focused on plasticity within the tumor in the host TME, and various selective pressures that can dictate plasticity, or be influenced by it, particularly treatment responses.

Various features that are components of the TME have been recognized as potential prognostic and predictive biomarkers. The presence of tumor-infiltrating lymphocytes has been shown to correlate with improved survival (8), and this has led to the establishment of the biopsy-based immunoscore for colorectal cancer risk assessment (9). While the main focus of these studies was on the presence of T cells, the impact of several other immune cell types within the tumor microenvironment has also been reported (10). Using a retrospective correlative study of 269 triple-negative breast cancers (TNBCs), Yeong et al. reported that the density of plasma cells within TNBC tumors had a significant association with disease-free survival rates, and high expression of IgG genes was associated with improved survival outcomes. Through analysis of publicly available datasets for patients with hepatocellular carcinoma (HCC) and known outcomes, Shrestha et al. assessed immune modulators as potential biomarkers, highlighting that PD-L1 expression was closely associated with epithelial-to-mesenchymal transition (EMT) marker expression and acted as prognostic factor for poor survival in the high-risk patient group.

Tumor cells are known to exert an immune-suppressive or immune-evasive phenotype through various mechanisms, and the crosstalk between TME and tumor is now a major focus of research for understanding and exploiting the critical role of the immune system. Weidenfeld and Barkan highlight the role of EMT and its reverse process, mesenchymal-to-epithelial transition (MET) in tumor dissemination and dormancy. They discuss how EMT may lead to the acquisition of cancer stem cell-like traits in tumors, which can change the immune-regulating properties of these cancer cells, although the effect on immunotherapy outcomes is unclear. Poltavets et al. review how the extracellular matrix (ECM) can influence cancer cell plasticity, discussing the implications for immunotherapy and the potential to exploit targeting ECM regulators as novel therapeutic strategies. Ham et al. present data indicating that exosomes secreted by breast cancer cells can skew macrophage polarization toward a pro-tumoral M2 phenotype partially via gp130/STAT3 signaling, suggesting that the exosomes can enhance the immune-suppressive activity of macrophages. Hamilton et al. describe how down-regulation of cyclin-dependent kinase (CDK) inhibitor 1 (p21CIP1) in cancer cells by the transcription factor brachyury leads to less stable CDK1 and renders the tumor cells more resistant to chemotherapy and immune-mediated cytotoxicity.

The authors also speculate that the same mechanism may drive EMT, which in turn may influence tumor immunogenicity.

Heterogeneity and plasticity in the tumor, as well as within the TME, can affect treatment outcomes. This has been exemplified recently by tumors that are responsive/“hot” or resistant/“cold” to immunotherapies. A similar concept regarding therapeutic resistance in melanoma was reviewed by Ahmed and Haass. They highlight that specific gene expression patterns known to dictate cell phenotype and function could also be influenced by the microenvironmental conditions that are selective for subpopulations differing in proliferation rates, invasiveness and drug responsiveness. Tissue-specific TMEs at different anatomical locations can regulate tumor growth, determine metastatic progression and impact on the outcome of therapy responses as reviewed by Oliver et al. suggesting that the organ site of metastasis can also influence immunotherapy outcomes due to differences in the local TMEs.

One of the main mediators of immune stimulation following immunogenic cell death (ICD) is the type I interferons. Budhwani et al. review our current knowledge about how type 1 interferons assist in mounting an immune response, but also note that long term, chronic exposure to type I interferons may diminish the efficacy of radiotherapy and chemotherapy. Conversely, a loss of interferon signaling can lead to resistance to immunotherapy, similar to defects in the IFNγ signaling cascade, which can lead to resistance to anti-PD-1 therapies (11). Alavi et al. demonstrated that only 33% of a large panel of melanoma cell lines with diverse mutational drivers had a strong INFγ response, and displayed induction of all measured targets. The importance of these signaling pathways and the influence of the TME on signaling and gene expression patterns may, at least partially, explain why response to immunotherapy is restricted to subsets of patients.

More recently, additional tumor extrinsic features including the patient's innate immunity, extent and duration of inflammation, balance of the microbiome and even stress levels, are thought to influence patient outcomes. Andrews et al. review various factors that influence the “visibility” of the tumor to the immune system and how they together determine the susceptibility of the tumor to immune attack. They highlight the central role of the gut microbiome in influencing the overall immune set-point (12), through diverse effects on local and systemic inflammatory processes, to influence disease and treatment outcomes. As immune checkpoint blockade treatments are effective only in subsets of patients and can lead to severe immune-related adverse events, it is important to identify which patients are most likely to benefit from these treatments, and also those at risk of developing complications. In order to predict which patients will develop adverse effects to treatment before toxicity becomes clinically evident, Da Gama Duarte et al. reported the potential use of a protein array to capture auto-antibodies from a cohort of melanoma patients treated with immunotherapy.

As a high proportion of patients do not respond to current immunotherapies, novel targets that can enhance immunogenicity could be clinically beneficial. Fan et al. review the potential of retinoic acid receptor-related orphan receptors to directly affect tumor cell behavior, including their ability to directly influence immune cells, given their expression by regulatory T cells and other immune cell subsets. Effective immunotherapy requires cancer cells to be recognized as foreign by the immune system, triggering the initiation of a directed immune-response (13). Many current treatment approaches, either as mono-therapy or combination therapy, aim to initiate a strong and broad immune response that results in long-lasting anti-cancer immunity. Cruickshank et al. review the data showing that immunogenic cell death (ICD) leads to immune stimulation that is epigenetically regulated, and propose that epigenetic drugs like HDAC inhibitors could be used to modify ICD. Poh and Ernst review the role of macrophages, a key component of the TME that orchestrate various aspects of immunity to regulate tumor progression. They discuss the targeting of tumor-associated macrophages (TAMs) as anti-cancer treatment strategies, evaluating the contribution of macrophages in moderating the effectiveness of current therapies and the challenges for successfully incorporating these strategies in cancer treatment regimens.

We currently face several challenges for the clinical implementation of combination therapies as effective treatment strategies for cancer. This second edition of our Research Topic on Cancer Plasticity and the Microenvironment builds upon our first edition, Cellular and Phenotypic Plasticity in Cancer, by expanding the focus to aspects of immunity and therapy responses across multiple cancer types. Our topic emphasizes various aspects of cancer plasticity that are highly pertinent to the incorporation and effective use of immune-modulating drugs in conventional cancer treatment regimens. As we embark on an exciting and promising era of multi-modal precision therapy, we have highlighted here some relevant points for consideration in our efforts to improve cancer treatment strategies for better patient outcomes.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

PF is supported by the National Health and Medical Research Council (NHMRC) of Australia (1126048). AB is the recipient of a Fellowship from the Victorian Government Department of Health and Human Services acting through the Victorian Cancer Agency. The Translational Research Institute is supported by a grant from the Australian Government.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The Olivia Newton-John Cancer Research Institute acknowledges the support of the Operational Infrastructure Program of Victorian Government.

References

1 . Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. (2011) 144:646–74. doi: 10.1016/j.cell.2011.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. (2012) 366:2443–54. doi: 10.1056/NEJMoa1200690

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Stambrook PJ, Maher J, Farzaneh F. Cancer immunotherapy: whence and whither. Mol Cancer Res. (2017) 15:635–50. doi: 10.1158/1541-7786.MCR-16-0427

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in cancer. Nat Rev Cancer. (2018) 18:128–34. doi: 10.1038/nrc.2017.118

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Chouaib S, Janji B, Tittarelli A, Eggermont A, Thiery JP. Tumor plasticity interferes with anti-tumor immunity. Crit Rev Immunol. (2014)34:91–102. doi: 10.1615/CritRevImmunol.2014010183

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery JP, et al. New insights into the role of EMT in tumor immune escape. Mol Oncol. (2017) 11:824–46. doi: 10.1002/1878-0261.12093

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. (2019) 20:69–84. doi: 10.1038/s41580-018-0080-4

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al., Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. (2006) 313:1960–4. doi: 10.1126/science.1129139

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Mlecnik B, Tosolini M, Kirilovsky A, Berger A, Bindea G, Meatchi T, et al., Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Onol. (2011) 29:610–8. doi: 10.1200/JCO.2010.30.5425

CrossRef Full Text | Google Scholar

10. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. (2012) 12:298–306. doi: 10.1038/nrc3245

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Zaretasky JM, Garcia-Diaz A, Shin SS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. (2016) 375:819–29. doi: 10.1056/NEJMoa1604958

CrossRef Full Text | Google Scholar

12. Chen DS, Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. (2017) 541:321–30. doi: 10.1038/nature21349

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Galluzzi L, Chan TA, Kroemer G, Wolchok JD, López-Soto A. The hallmarks of successful anticancer immunotherapy. Sci Transl Med. (2018) 10:eaat7807. doi: 10.1126/scitranslmed.aat7807

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immune evasion, therapy resistance, epithelial-to-mesenchymal transition (EMT), immunotherapy, tumor microenvironment (TME), heterogeneity, cancer plasticity, immune checkpoint inhibitors

Citation: Behren A, Thompson EW, Anderson RL and Ferrao PT (2019) Editorial: Cancer Plasticity and the Microenvironment: Implications for Immunity and Therapy Response. Front. Oncol. 9:276. doi: 10.3389/fonc.2019.00276

Received: 19 March 2019; Accepted: 26 March 2019;
Published: 07 May 2019.

Edited and reviewed by: Catherine Sautes-Fridman, INSERM U1138 Centre de Recherche des Cordeliers, France

Copyright © 2019 Behren, Thompson, Anderson and Ferrao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Petranel T. Ferrao, petranelferrao@gmail.com

Present Address: Petranel T. Ferrao, South Australian Health and Medical Research Institute, Adelaide, SA, Australia

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.