Chapter Eight - Control of CD8 T-Cell Infiltration into Tumors by Vasculature and Microenvironment

https://doi.org/10.1016/bs.acr.2015.05.001Get rights and content

Abstract

CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.

Section snippets

Prognostic Significance of Immune Cell Representation in Tumors

A role for the immune system in cancer regression was suggested in the late-nineteenth century by William Coley, who observed that spontaneous remission of tumors sometimes occurred in patients who contracted acute bacterial infections. He subsequently developed a mixture of bacterial toxins that he believed activated the immune system and reported they were effective and even curative for some patients (Coley, 1893). Still, his method was controversial, and with the advent of chemo- and

CD8 T-Cell Representation in Tumors as a Predictive Marker of Responsiveness to Therapy

The emergence of clinically evident cancers reveals that despite the presence and activity of CD8 T-cells, tumors can escape their control (Vesely et al., 2011). Numerous strategies to harness and/or enhance the antitumor properties of CD8 T-cells have been developed, and in recent years, have led to encouraging successes. Melanoma has historically been the most studied tumor for immunotherapies, in part, because it is also the most responsive to a wide spectrum of such therapies. However,

Determinants of CD8 T-Cell Representation in Tumors and Other Tissues

The overall representation of T-cells in tissues, including tumors, is determined by the balance of several fundamental processes: cells entering tissues from the blood vasculature, cells leaving through draining lymphatics, and cells proliferating and dying in situ. To accumulate in a tumor at a peripheral site, however, typically effector T-cells are first activated by specific antigen in the draining LN (see Fig. 1). Tumors that are poorly infiltrated by effector T-cells may be poorly

Tumors Develop HEV-Like Vasculature

Classically, naïve CD8 T-cells are thought to primarily recirculate through the blood and secondary lymphoid organs to scan antigen-presenting cells for their cognate antigen (Von Andrian & Mackay, 2000). This tissue selectivity is based upon interactions of L-selectin and CCR7 with peripheral node addressin (PNAd) and CCL19/CCL21, respectively, which are normally selectively expressed on the specialized high endothelial venules (HEV) found in LN but not the vessels of peripheral tissues (

Rationale for Modifying Tumor-Associated Vasculature

As we have reviewed, characteristics of the tumor vasculature determine whether or not tumors are permissive to the entry of both effector and naïve CD8 T-cells. Because the presence of CD8 T-cells in tumors is such a strong prognostic factor and predictor of responsiveness to immunotherapies, strategies that can alter the tumor vasculature to support the enhanced entry of T-cells hold the potential to extend the effectiveness of these immunotherapies to a much broader cross section of patients

References (257)

  • M. De Palma et al.

    Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors

    Cancer Cell

    (2005)
  • S. Demaria et al.

    Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated

    International Journal of Radiation Oncology, Biology, Physics

    (2004)
  • L. Deng et al.

    STING-dependent cytosolic DNA sensing promotes radiation-induced type i interferon-dependent antitumor immunity in immunogenic tumors

    Immunity

    (2014)
  • M.-C. Dieu-Nosjean et al.

    Tertiary lymphoid structures in cancer and beyond

    Trends in Immunology

    (2014)
  • K. Enarsson et al.

    Differential mechanisms for T lymphocyte recruitment in normal and neoplastic human gastric mucosa

    Clinical Immunology

    (2006)
  • A. Fütterer et al.

    The lymphotoxin β receptor controls organogenesis and affinity maturation in peripheral lymphoid tissues

    Immunity

    (1998)
  • J. Galon et al.

    The continuum of cancer immunosurveillance: Prognostic, predictive, and mechanistic signatures

    Immunity

    (2013)
  • S.E. Goelz et al.

    ELFT: A gene that directs the expression of an ELAM-1 ligand

    Cell

    (1990)
  • S. Goelz et al.

    Differential expression of an E-selectin ligand (SLex) by two Chinese hamster ovary cell lines transfected with the same alpha (1,3)-fucosyltransferase gene (ELFT)

    The Journal of Biological Chemistry

    (1994)
  • R. Abdi et al.

    The role of CC chemokine receptor 5 (CCR5) in islet allograft rejection

    Diabetes

    (2002)
  • M.B. Alimzhanov et al.

    Abnormal development of secondary lymphoid tissues in lymphotoxin β-deficient mice

    Proceedings of the National Academy of Sciences of the United States of America

    (1997)
  • F. Aloisi et al.

    Lymphoid neogenesis in chronic inflammatory diseases

    Nature Reviews. Immunology

    (2006)
  • R. Alon et al.

    The integrin VLA-4 supports tethering and rolling in flow on VCAM-1

    The Journal of Cell Biology

    (1995)
  • T. Ando et al.

    MAdCAM-1 expression and regulation in murine colonic endothelial cells in vitro

    Inflammatory Bowel Diseases

    (2005)
  • D.P. Andrew et al.

    C-C chemokine receptor 4 expression defines a major subset of circulating nonintestinal memory T cells of both Th1 and Th2 potential

    Journal of Immunology

    (2001)
  • F. Austrup et al.

    P- and E-selectin mediate recruitment of T-helper-1 but not T-helper-2 cells into inflammed tissues

    Nature

    (1997)
  • G. Avram et al.

    The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma

    Histopathology

    (2013)
  • S.R. Bailey et al.

    Th17 cells in cancer: The ultimate identity crisis

    Frontiers in Immunology

    (2014)
  • P. Balermpas et al.

    Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer

    British Journal of Cancer

    (2014)
  • E. Barbera-Guillem et al.

    B lymphocyte pathology in human colorectal cancer. Experimental and clinical therapeutic effects of partial B cell depletion

    Cancer Immunology, Immunotherapy

    (2000)
  • J.L. Baron et al.

    Surface expression of alpha 4 integrin by CD4 T cells is required for their entry into brain parenchyma

    The Journal of Experimental Medicine

    (1993)
  • S.R. Barthel et al.

    Targeting selectins and selectin ligands in inflammation and cancer

    Expert Opinion on Therapeutic Targets

    (2007)
  • G.L. Beatty et al.

    Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce antitumor activity in solid malignancies

    Cancer Immunology Research

    (2014)
  • E.L. Berg et al.

    The cutaneous lymphocyte antigen is a skin lymphocyte homing receptor for the vascular lectin endothelial cell-leukocyte adhesion molecule 1

    The Journal of Experimental Medicine

    (1991)
  • H. Bernhard et al.

    Adoptive transfer of autologous, HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing breast cancer

    Cancer Immunology, Immunotherapy

    (2008)
  • B. Beverly et al.

    Reversal of in vitro T cell clonal anergy by IL-2 stimulation

    International Immunology

    (1992)
  • C. Blank et al.

    ICAM-1 contributes to but is not essential for tumor antigen cross-priming and CD8 + T cell-mediated tumor rejection in vivo

    Journal of Immunology

    (2005)
  • C. Blank et al.

    PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8 + T cells

    Cancer Research

    (2004)
  • A. Boissonnas et al.

    In vivo imaging of cytotoxic T cell infiltration and elimination of a solid tumor

    The Journal of Experimental Medicine

    (2007)
  • E. Borges et al.

    P-selectin glycoprotein ligand-1 (PSGL-1) on T helper 1 but not on T helper 2 cells binds to P-selectin and supports migration into inflamed skin

    The Journal of Experimental Medicine

    (1997)
  • C. Bourquin et al.

    Efficient eradication of subcutaneous but not of autochthonous gastric tumors by adoptive T cell transfer in an SV40 T antigen mouse model

    Journal of Immunology

    (2010)
  • C.C. Brinkman et al.

    Peripheral tissue homing receptors enable T cell entry into lymph nodes and affect the anatomical distribution of memory cells

    Journal of Immunology

    (2013)
  • C.C. Brinkman et al.

    Activated CD8 T cells redistribute to antigen-free lymph nodes and exhibit effector and memory characteristics

    Journal of Immunology

    (2008)
  • R.J. Buckanovich et al.

    Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy

    Nature Medicine

    (2008)
  • Y. Bulliard et al.

    Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies

    The Journal of Experimental Medicine

    (2013)
  • D.J. Campbell et al.

    Rapid acquisition of tissue-specific homing phenotypes by CD4(+) T cells activated in cutaneous or mucosal lymphoid tissues

    The Journal of Experimental Medicine

    (2002)
  • J.J. Campbell et al.

    The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells

    Nature

    (1999)
  • N. Casares et al.

    Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death

    The Journal of Experimental Medicine

    (2005)
  • E. Cha et al.

    Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients

    Science Translational Medicine

    (2014)
  • S. Champiat et al.

    Exomics and immunogenics: Bridging mutational load and immune checkpoints efficacy

    OncoImmunology

    (2014)
  • Cited by (125)

    • PAK4 inhibition improves PD1 blockade immunotherapy in prostate cancer by increasing immune infiltration

      2023, Cancer Letters
      Citation Excerpt :

      Our RNASeq results showed that Icam1 increased in KD and KD αPD1 tumors (Fig. 5C), and Vcam1 increased in KD αPD1 tumors (Fig. 5D). Icam1 and Vcam1 are integrin ligands that regulate leukocyte-endothelial interaction and play an important role in the structure of the tumor vasculature and T-cell trafficking into tumor.11 12 Vcam1 binds to VLA4 integrin to initiate T-cell slow rolling, whereas Icam1 binds to LFA1 to mediate cell arrest [31]. To confirm their role in CD8+ cell infiltration, we blocked CD18 and CD49D, which are the binding integrin partners to Icam1 and Vcam1, respectively.

    • A comparison of cancer vaccine adjuvants in clinical trials

      2023, Cancer Treatment and Research Communications
    View all citing articles on Scopus
    View full text