Biology Contribution
Kinetics of Intratumoral Immune Cell Activation During Chemoradiation for Cervical Cancer

https://doi.org/10.1016/j.ijrobp.2018.06.404Get rights and content

Purpose

Radiation therapy has direct cytotoxic effects on tumor-infiltrating lymphocytes, but it also has immune stimulatory effects that increase immune cell infiltration. The dynamics of these competing effects on immune cells at the site of the tumor are poorly characterized during chemoradiation treatment (CRT) because of the difficulty of obtaining consecutive tumor biopsies. We used a minimally invasive cervical cytobrushing method to analyze the kinetics of intratumoral immune cell changes in patients with cervical cancer during CRT.

Methods and Materials

Cervical brushings were obtained from 20 patients with cervical cancer at baseline and during fractionated radiation therapy and cisplatin (weeks 1, 3, and 5). Matching peripheral blood mononuclear cells were obtained from 9 patients at the same time points. Cells were analyzed using multispectral flow cytometry to identify T cell and myeloid cell subsets and their activation status. Changes in immune cell subsets throughout treatment were calculated using matched-pair analysis with Wilcoxon rank sum test.

Results

We observed a significant decline in CD3+ total T cells, as well as CD8+ and CD4+ T-cell subsets in the first week of treatment from baseline, followed by variable expansion at weeks 3 and 5. This coincided with higher levels of proliferating CD8+ T cells expressing Ki67 at week 3 of treatment. The percentages of activated CD8+ T cells expressing CD69 continuously increased over the course of treatment, whereas the percentage of activated CD11c+CD11b– dendritic cells was highest during the first week. Many of these changes were not observed in the blood.

Conclusions

Our results identified immune dynamic changes during CRT, indicating that CRT may be immune activating at the site of the tumor. This study also suggests the importance of sequential analyses of the local tumor microenvironment in addition to peripheral blood.

Introduction

Cervical cancer is among the most common malignancies among female patients worldwide, with an annual incidence of more than 500,000 women and an annual death rate of more than 250,000 women. (1) Locally advanced cervical cancer can be treated effectively with chemoradiation therapy (CRT) over a 7-week course of treatment requiring daily external beam radiation followed by brachytherapy (2); however, the rate of response to treatment is highly variable. 3, 4 Rapid responders to CRT are more likely to remain free of disease in the long term, but the mechanisms that underlie this heterogeneity in response rates are not well understood. Historically, radiation therapy (RT) was considered to have immunosuppressive effects; lymphocytes are one of the most radiosensitive cell types, (5) and peripheral lymphocyte counts generally decrease through the course of pelvic radiation. (6) More recently, radiation has been shown to uncover tumor antigens through tumor cell death, which enhances antigen presentation and induces antitumor T-cell responses. (7) Furthermore, radiation increases the release of damage associated molecular patterns, which attract and activate cells of the innate and adaptive immune system. (8)

Because of the challenges of obtaining frequent biopsies during RT, studies of immune activation are based on animal models using hypofractionated regimens, clinical studies with evaluation of tumors at very limited time points, or a focus on peripheral immune responses. As a result, the kinetics of intratumoral immune activation during RT are poorly understood. To discern the sequential changes within the cervix tumor microenvironment during CRT, we adapted the cytobrush methodology, demonstrated to be a reliable and minimally invasive technique to characterize immune cells within the female genital tract of HIV-positive patients in a multicenter clinical trial. (9) We recently reported the utility of this methodology for monitoring CD4 T cells at multiple mucosal tissues after intranasal vaccination in rhesus macaques. (10) By using this minimally invasive method, we characterized the immune infiltrate with multispectral flow cytometry at baseline and at weeks 1, 3, and 5 of CRT. Because T-cell infiltration has been associated with better prognosis in patients with cervical cancer and myeloid cells are known to modulate this infiltration, we focused our analysis on these populations. (11)

Section snippets

Patient population

Between January 2016 and January 2018, 20 patients were enrolled in a prospective observational clinical trial at MD Anderson Cancer Center and Lyndon B. Johnson Hospital designed to evaluate immunologic and metagenomic changes in the cervical and intestinal microbiota during CRT. Institutional review board approval was obtained, and all participants provided informed consent before enrolling in the study. Inclusion criteria and treatment are available in Methods E1 (available online at 10.1016/j.ijrobp.2018.06.404

Patient and tumor characteristics

Twenty patients with locally advanced cervical cancer (stage IBI-IIIB) were enrolled (Table E1; available online at https://doi.org/10.1016/j.ijrobp.2018.06.404). All patients completed treatment with external beam radiation with concurrent weekly cisplatin followed by pulsed-dose-rate or high-dose-rate brachytherapy.

T cells are transiently decreased after CRT followed by expansion in the cervical tumor

Cervical brushes were collected from visible tumors in patients; for patients with tumors that were not visible at week 5, the treated tumor site was brushed. On average, the yield

Discussion

The kinetics of immune activation within the local tumor microenvironment during CRT has not been well investigated because of challenges in obtaining serial biopsies during treatment. To our knowledge, this is the first study to evaluate the kinetics of changes in intratumoral immune cell subsets and their activation status during CRT.

We found that CRT induces an early and transient decrease of T cells during the first week of treatment. The percentage of activated CD69+ T cells increased

References (24)

  • L.R. McKinnon et al.

    Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility

    J Immunol

    (2011)
  • S. Dorta-Estremera et al.

    Minimally invasive monitoring of CD4 T cells at multiple mucosal tissues after intranasal vaccination in rhesus macaques

    PLoS One

    (2017)
  • Cited by (36)

    • Chemotherapy to potentiate the radiation-induced immune response

      2023, International Review of Cell and Molecular Biology
    • Pre-treatment immune status predicts disease control in NSCLCs treated with chemoradiation and durvalumab

      2022, Radiotherapy and Oncology
      Citation Excerpt :

      Clinical studies have also found the drop in ALC due to RT to be more reflective of changes in peripheral B-cells rather than in T-cells [17]. Additionally, prospective evaluation of cervical cancer patients undergoing cCRT has found expansion of cytotoxic T-cells in the tumor microenvironment during cCRT despite a decreasing peripheral blood ALC [18]. Therefore, decreases in the peripheral ALC count do not necessarily equate functional immunosuppression, and the ALC may moreover be too crude of a measure.

    • Development of oncolytic viruses for cancer therapy

      2021, Translational Research
      Citation Excerpt :

      Radiation causes DNA damage and cell death primarily through apoptosis.153 Apoptosis of the cancer cells was found to release tumor antigens and DAMPs which led to the activation of the immune system.154 Combinatorial therapy between OVs and radiation was reported to have a synergistic antitumor role in preclinical models by improving apoptosis.155,156

    View all citing articles on Scopus

    J.K.S. and A.K. contributed equally to this study.

    This study was funded by The University of Texas MD Anderson Cancer Center HPV-related Cancers Moonshot. The Flow Cytometry Core at MD Anderson Cancer Center is supported by the Cancer Center Support Grant NCI# P30 CA16672.

    Conflict of interest: J.W. reports being a Healios cofounder, a MolecularMatch cofounder, a MolecularMatch officer, an OncoResponse cofounder/SAB, on the Reflexion Medical Scientific Advisor Board, on the Checkmate Pharmaceuticals Scientific Advisor Board, and on the Mavu Scientific Advisory Board. J.W. also reports BMS Clinical research support, Merck Clinical research support, Varian Laboratory research support, Incyte Laboratory research support, Merck Laboratory research support, Calithera Laboratory research support, Checkmate Pharmaceuticals Laboratory research support, and OncoResponse Laboratory research support. L.L.L reports an investigator-initiated study that is supported by AstraZeneca.

    View full text