Introduction
3.3’-diindolylmethane (DIM) is one of the best characterized bioactive compounds found in Cruciferae.
1- Thomson C.A.
- Ho E.
- Strom M.B.
Chemopreventive properties of 3,3’-diindolylmethane in breast cancer: Evidence from experimental and human studies.
The acid-catalyzed dimer of the bioactive indole has long been proposed for use as a cancer prevention agent.
2- Acharya A.
- Das I.
- Singh S.
- et al.
Chemopreventive properties of indole-3-carbinol, diindolylmethane and other constituents of cardamom against carcinogenesis.
DIM has been found to regulate cancer cell proliferation by acting as an AhR ligand, reducing oxidative stress, activating interferon-γ, and modulating estrogen signaling.
1- Thomson C.A.
- Ho E.
- Strom M.B.
Chemopreventive properties of 3,3’-diindolylmethane in breast cancer: Evidence from experimental and human studies.
Studies also suggest that DIM inhibits radiation therapy and chemotherapy-induced toxicity in normal cells.
3- Hajra S.
- Basu A.
- Singha Roy S.
- et al.
Attenuation of doxorubicin-induced cardiotoxicity and genotoxicity by an indole-based natural compound 3,3’-diindolylmethane (DIM) through activation of Nrf2/ARE signaling pathways and inhibiting apoptosis.
DIM was found to protect cultured cells against ionizing radiation in clonogenic survival assays by activating ataxia-telangiectasia mutated (ATM) signaling.
4- Fan S.
- Meng Q.
- Xu J.
- et al.
DIM (3,3’-diindolylmethane) confers protection against ionizing radiation by a unique mechanism.
DIM also protects mice against lethal doses of total body irradiation (TBI) and ameliorates TBI-induced hematopoietic injury by inhibiting oxidative stress responses and hematopoietic cell apoptosis.
5- Lu L.
- Dong J.
- Li D.
- et al.
3,3’-diindolylmethane mitigates total body irradiation-induced hematopoietic injury in mice.
However, DIM does not protect MDA-MB-231 human breast cancer xenografts in nude mice against fractionated radiation therapy (RT).
4- Fan S.
- Meng Q.
- Xu J.
- et al.
DIM (3,3’-diindolylmethane) confers protection against ionizing radiation by a unique mechanism.
This preferential protection of normal cells makes DIM an attractive adjuvant to radiation therapy, particularly when high-dose RT results in delayed tissue and organ toxicity.
Recently, the use of RT has been explored to enhance immunotherapy.
6- Demaria S.
- Coleman C.N.
- Formenti S.C.
Radiotherapy: Changing the game in immunotherapy.
,7- Formenti S.C.
- Rudqvist N.P.
- Golden E.
- et al.
Radiotherapy induces responses of lung cancer to CTLA-4 blockade.
Radiation enhances major histocompatibility complex (MHC) class I expression in tumor cells and increases cytotoxic T lymphocyte recognition of irradiated cells.
8- Reits E.A.
- Hodge J.W.
- Herberts C.A.
- et al.
Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy.
On the other hand, radiation exhibits immunosuppressive effects on the tumor microenvironment. RT induces expression of the immunosuppressive cytokine TGF-β
9- Hardee M.E.
- Marciscano A.E.
- Medina-Ramirez C.M.
- et al.
Resistance of glioblastoma-initiating cells to radiation mediated by the tumor microenvironment can be abolished by inhibiting transforming growth factor-beta.
,10- Vanpouille-Box C.
- Diamond J.M.
- Pilones K.A.
- et al.
TGFbeta is a master regulator of radiation therapy-induced antitumor immunity.
and increases infiltration of both regulatory T cells
11- Bos P.D.
- Plitas G.
- Rudra D.
- et al.
Transient regulatory T cell ablation deters oncogene-driven breast cancer and enhances radiotherapy.
and immunosuppressive myeloid cells.
12- Kozin S.V.
- Kamoun W.S.
- Huang Y.
- et al.
Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation.
Radiation also leads to higher expression of immune checkpoint ligands, including PD-L1, in tumor cells,
13- Deng L.
- Liang H.
- Burnette B.
- et al.
Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice.
as well as PD-1 in T cells.
14- Li D.
- Chen R.
- Wang Y.W.
- et al.
Prior irradiation results in elevated programmed cell death protein 1 (PD-1) in T cells.
The importance of tumor immunity warrants an investigation of DIM’s effects on the tumor-immune microenvironment (TIME) in response to radiation. In this study, we used a clinically relevant, syngeneic breast cancer model in immunocompetent mice
6- Demaria S.
- Coleman C.N.
- Formenti S.C.
Radiotherapy: Changing the game in immunotherapy.
to characterize how DIM influences the TIME and tumor regression after RT.
Methods and Materials
Cell cultures and chemical and in vitro treatment
Human breast cancer cell lines MDA-MB-231 and HCC1937 were obtained from the Tissue Culture Shared Resource. Murine breast cancer cell line E0771 and hTERT-immortalized human mammary epithelial cell line (HMEC-hTERT) were kindly provided by our collaborators. HMEC-hTERT cells were cultured in keratinocyte serum-free media (Gibco, Waltham, MA) containing 25 mg/mL of bovine pituitary extract and 5 ng/mL of human recombinant epidermal growth factor. The other cells were cultured in DMEM supplemented with 10% fetal bovine serum. Cells were maintained at 37°C in a 5% CO2 atmosphere. BR9001, a bioavailability enhancing, self-emulsifying DIM formulation (BioResponse, Boulder, CO) was a kind gift from Dr Michael Zeligs. Subconfluent HMEC-hTERT cells were exposed to ionizing radiation using a Precision X-RAD 320 irradiator (320 kV, 25 mA) at a dose rate of 0.864 Gy/min. For DIM treatment, the drug was administered 24 hours before irradiation.
Tumor models and treatments
Immune-competent wild-type C57BL/6J female mice were used as the syngeneic breast tumor model. Athymic nude (Crl:NU(NCr)-Foxn1nu) and NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice were purchased from Charles River and the Jackson Laboratory, respectively, and used as immunodeficient models. Tumor cells were injected subcutaneously in the right dorsal flank. Mice were randomized to 4 groups (n = 5-8): (1) vehicle (SV); (2) DIM (SD); (3) radiation + vehicle (RV); and (4) radiation + DIM (RD). Anesthetized mice were subjected to tumor site targeted radiation using narrow-beamed radiation with a shield (Precision X-Ray Inc, North Branford, CT). DIM was administered intraperitoneally at the concentration of 75 mg/kg. Mice were given 6 Gy per day at 0.864 Gy/min on 4 consecutive days. DIM was administered daily from 1 day before irradiation to 1 day after the last fraction. These experiments have been repeated 3 times using biological replicates. All animal experiments were performed in accordance with the Institutional Animal Care and Use Committee.
Immunohistochemistry (IHC)
Mice were sacrificed at 12 days after the date when the last radiation fraction was delivered, and tumor tissues were harvested. Harvested tumor tissues were subjected to IHC. For each tumor section, 12 different random fields of view were imaged at 400× magnification. Average integrated density for each treatment group (n = 8) was measured using ImageJ software installed with the plugin including color deconvolution function.
NanoString gene expression analysis
Total RNA was extracted from flash-frozen tumor tissues. Tumor samples from 5 mice in each group were used. RNA quality was assessed using an Agilent 2100 Bioanalyzer (Agilent, Santa Clara, CA). One hundred nanograms of tumor tissue RNA was subjected to gene expression profiling using the nCounter PanCancer Immune Profiling Panel (NanoString Tech, Seattle, WA). Sample labeling, hybridization, and scanning were performed using the nCounter MAX Analysis System. Quality control metrics were reported using the nSolver Analysis Software v4.0. Raw read counts normalization, differential expression, cell type profiling, and pathway score analysis were performed using nCounter Advanced Analysis Software v2.0.
Microarray and data analysis
Microarray analysis was performed on 3 replicates of each treatment. Detailed methods can be found in the
supplementary materials. Identification of differentially expressed genes was performed by filtering the data set using
P < .05 and a treatment-to-control ratio greater than 1.7, as well as by 1-way analysis of variance (ANOVA) statistical analysis followed by Benjamin-Hochberg multiple testing. Differential genes that met these statistical criteria in any of the 3 analyses (sham vs 3 Gy, sham vs DIM, and sham + DIM vs 3 Gy + DIM) were identified. In total, there were 1885 genes. These genes were subjected to 2-dimensional clustering visualized using Genesis (genome.tugraz.at) and the Ingenuity Pathway Analysis (Qiagen, Redwood City, CA).
Statistical analysis
The data are expressed as mean ± standard error of the mean (SEM). Significance was analyzed by 2-way ANOVA and 1-way ANOVA. Multiple comparisons were performed using GraphPad Prism 6 Software (GraphPad Software, San Diego, CA). Values were considered significant at P < .05, except in the NanoString cell type analysis, where the significant cutoff was set at P < .1.
Discussion
In this study, we demonstrate that DIM, when combined with RT, enhances the TIME by increasing intratumoral immune cells and promoting expression of genes involved in key immune responses in tumors. As a result, the efficacy of radiation treatment was enhanced for tumors engrafted in immunocompetent but not immunodeficient mice. The results indicate that DIM treatment alone does not affect tumor growth, regardless of the immune conditions, suggesting that DIM’s radioprotective effect on normal cells plays a critical role in promoting a TIME that favors antitumor immunity.
DIM has been shown to attenuate radiation-induced injury by inhibiting oxidative stress and cell apoptosis.
5- Lu L.
- Dong J.
- Li D.
- et al.
3,3’-diindolylmethane mitigates total body irradiation-induced hematopoietic injury in mice.
In this study, DIM treatment was found to precondition normal cells to become substantially less responsive to radiation-induced gene expression modulation. On the other hand, DIM treatment did not show radioprotection of tumor cells as, in these cells, canonical DNA damage response pathways are often malfunctional and/or the disordered activation of survival pathways and constitutive proliferation signaling override DNA-damage-induced cell cycle arrest and apoptosis.
Lymphocytes are one of the most sensitive cell types to radiation-induced cell death. In this study, lymphocyte levels in the peripheral blood of mice that received 3 Gy TBI dropped to less than 10% of corresponding levels in the sham control 1 week after radiation. The relative lymphocyte abundance increased around 50% in mice that received TBI and DIM treatment compared with radiation with the vehicle control, a finding that is consistent with previous work.
5- Lu L.
- Dong J.
- Li D.
- et al.
3,3’-diindolylmethane mitigates total body irradiation-induced hematopoietic injury in mice.
,17- Thekkekkara D.
- Basavan D.
- Chandna S.
- et al.
A combination of resveratrol and 3,3’-diindolylmethane, a potent radioprotector.
In addition to alleviating the cytotoxicity of circulating lymphocytes, protection of locoregional normal cells may also contribute to a better TIME. Radiation is known to induce apoptosis of endothelial cells through persistent p53 signaling and ceramide-mediated pathway activation.
18- Rashi-Elkeles S.
- Elkon R.
- Shavit S.
- et al.
Transcriptional modulation induced by ionizing radiation: p53 remains a central player.
,19Radiation and ceramide-induced apoptosis.
In our study, vascular density was found to be reduced in irradiated tumors. Although there was no statistically significant difference, vascular density was slightly higher in mice that received RD treatment compared with RV treatment (
Fig 4A, 4B), suggesting less endothelial injury. Endothelial activation can result in upregulation of adhesion molecules such as Vcam1 and cytokines, which attract immune cells to the site. Although the role of endothelial cells in tumor responses to radiation therapy remains controversial,
20- Guipaud O.
- Jaillet C.
- Clément-Colmou K.
- et al.
The importance of the vascular endothelial barrier in the immune-inflammatory response induced by radiotherapy.
it is possible that radioprotection of endothelial cells and other normal cells facilitates the recruitment and infiltration of immune cells in irradiated tumors.
Our results show enrichment of T cells, neutrophils, and macrophages in irradiated tumors after RD treatment. Previous studies strongly suggest that a higher density of tumor-infiltrating lymphocytes (TILs) is a favorable prognostic marker for a heightened response to radiochemotherapy and better survival rates.
21- Dieci M.V.
- Mathieu M.C.
- Guarneri V.
- et al.
Prognostic and predictive value of tumor-infiltrating lymphocytes in two phase III randomized adjuvant breast cancer trials.
, 22- Pagès F.
- Galon J.
- Dieu-Nosjean M.C.
- et al.
Immune infiltration in human tumors: A prognostic factor that should not be ignored.
, 23- Bayraktar S.
- Batoo S.
- Okuno S.
- et al.
Immunotherapy in breast cancer.
In some patients, radiochemotherapy turns “cold” tumors, which contain few TILs, to “hot” ones, with rich infiltration of T cells. Patients with tumors that remain or become “hot” after treatment show lower risk of relapse.
21- Dieci M.V.
- Mathieu M.C.
- Guarneri V.
- et al.
Prognostic and predictive value of tumor-infiltrating lymphocytes in two phase III randomized adjuvant breast cancer trials.
In the tumor model used in this study, RT alone did not significantly increase the density of infiltrated T cells, suggesting that RT by itself was not capable of effectively converting the “cold” TIME. In the presence of DIM, however, TIL density was significantly elevated after radiation (
Fig 2B), and tumor regression was significantly improved (
Fig 1A).
At this point, the roles of tumor-associated neutrophils (TANs) and macrophages (TAMs) remain controversial. Both pro- and antitumor roles have been reported for TANs. For example, TANs have been suggested to play a role in supporting tumor vascularization by promoting highly angiogenic MMP-9.
24- Deryugina E.I.
- Zajac E.
- Juncker-Jensen A.
- et al.
Tissue-infiltrating neutrophils constitute the major in vivo source of angiogenesis-inducing MMP-9 in the tumor microenvironment.
On the other hand, increased levels of TANs have been shown to promote higher levels of proinflammatory cytokines that are cytotoxic to tumor cells.
25- Fridlender Z.G.
- Sun J.
- Kim S.
- et al.
Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN.
Similarly, although M1-like TAMs generally exhibit antitumor activity, high infiltration of TAMs has been associated with poor clinical prognosis in patients with breast cancer.
26Tumor-associated macrophages as potential diagnostic and prognostic biomarkers in breast cancer.
, 27Targeting macrophages in cancer: from bench to bedside.
, 28- Qiu S.Q.
- Waaijer S.J.H.
- Zwager M.C.
- et al.
Tumor-associated macrophages in breast cancer: Innocent bystander or important player.
In this study, IHC staining with CD68, a pan-macrophage marker, indicated that RD treatment increased TAMs but not SD treatment (
Fig 2C), suggesting that these macrophages were recruited in tumors after radiation. It has been shown that the TIME usually tends to skew recruited monocytes toward the M2-like phenotype.
29- Xu M.
- Liu M.
- Du X.
- et al.
Intratumoral delivery of IL-21 overcomes anti-Her2/Neu resistance through shifting tumor-associated macrophages from M2 to M1 phenotype.
Our previous work demonstrated that differentiation of helper T cells to Th1 is impaired after radiation, leading to imbalanced Th1/Th2 function.
30- Chen R.
- Wang Y.W.
- Fornace A.J.
- et al.
Impairment of the intrinsic capability of Th1 polarization in irradiated mice: A close look at the imbalanced Th1/Th2 response after irradiation.
This imbalance may also contribute to macrophage polarization toward the M2-like phenotype. Considering the potential adverse effects of TAMs on the TIME, concomitant pharmacologic depletion of TAMs such as CSF1R signaling blockade
31- Ries C.H.
- Cannarile M.A.
- Hoves S.
- et al.
Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy.
,32- Dammeijer F.
- Lievense L.A.
- Kaijen-Lambers M.E.
- et al.
Depletion of tumor-associated macrophages with a CSF-1R kinase inhibitor enhances antitumor immunity and survival induced by DC immunotherapy.
may boost DIM’s effects in TIME modulation.
Radiation therapy is an effective treatment for cancer. Radiation eliminates cancer cells mostly by direct cytotoxicity. Preclinical and clinical data have suggested that RT acts as an immune modifier,
33- Demaria S.
- Golden E.B.
- Formenti S.C.
Role of local radiation therapy in cancer immunotherapy.
having both immunostimulatory and immunosuppressive effects. Radiation causes immunogenic cell death and releases autologous neoantigens to the immune system.
34- Nesseler J.P.
- Schaue D.
- McBride W.H.
- et al.
Irradiation to improve the response to immunotherapeutic agents in glioblastomas.
,35- Wennerberg E.
- Vanpouille-Box C.
- Bornstein S.
- et al.
Immune recognition of irradiated cancer cells.
It also generates danger signals that activate stimulator of interferon genes-mediated
36- Deng L.
- Liang H.
- Xu M.
- et al.
STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors.
or Toll-like receptor
37- Haikerwal S.J.
- Hagekyriakou J.
- MacManus M.
- et al.
Building immunity to cancer with radiation therapy.
,38- Wang H.T.
- Lee H.I.
- Guo J.H.
- et al.
Calreticulin promotes tumor lymphocyte infiltration and enhances the antitumor effects of immunotherapy by up-regulating the endothelial expression of adhesion molecules.
pathways. Given many lines of evidence supporting the notion that radiation may enrich tumor-specific effector lymphocytes and elicit priming of systemic antitumor T cell responses, it is expected that radiation treatment can be used to enhance both local and systemic antitumor immunity. It is of interest to note that abscopal effects did emerge more frequently in patients who received radiation combined with immunotherapy.
39- Golden E.B.
- Chhabra A.
- Chachoua A.
- et al.
Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumors: A proof-of-principle trial.
However, these effects are often unpredictable and not seen in most patients. These puzzling findings are likely the consequence of the complex immunosuppressive effects of radiation on the TIME.
Article info
Publication history
Published online: October 23, 2020
Accepted:
October 13,
2020
Received in revised form:
September 6,
2020
Received:
April 12,
2020
Footnotes
Sources of support: Research in this publication was supported by NCI of the NIH under award number R01CA184168.
Disclosures: None to report.
Research data are stored in an institutional repository and will be shared upon request to the corresponding author.
Copyright
© 2020 The Authors. Published by Elsevier Inc. on behalf of American Society for Radiation Oncology.