You are on page 1of 3

Cancer Cell

Previews

Stressing Out about Cancer Immunotherapy


Xue-Yan He,1 David Ng,1 Linda Van Aelst,1 and Mikala Egeblad1,*
1Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY 11724, USA

*Correspondence: egeblad@cshl.edu
https://doi.org/10.1016/j.ccell.2019.10.013

Stress has long been suspected to negatively influence cancer mortality, yet the molecular mechanisms
responsible for this effect have only recently been identified. A new study identifies a stress-induced
response in dendritic cells—the activation of the glucocorticoid-inducible transcriptional regulator
TSC22D3—as a potent, immunosuppressive effect of stress on cancer.

Intrinsic (genetic and epigenetic) changes the use of synthetic glucocorticoids in can- stress-induced elevation in glucocorticoid
in cancer cells and extrinsic changes cer treatment. Here, we discuss another levels triggers an immunosuppressive
occurring to the microenvironment and newly identified mechanism by which state that dampens chemo- and immuno-
organism together drive cancer progres- glucocorticoid receptor signaling pro- therapeutic effects.
sion and resistance to therapy. One motes cancer and resistance to therapy, Cancer immunotherapy harnesses and
extrinsic factor long suspected to drive namely by affecting dendritic cells’ (DCs) enhances the immune system to fight can-
cancer progression is stress. Stress is a ability to suppress the immune response cer. Certain types of chemotherapy also
complex physiological process initiated against tumors (Yang et al., 2019). activate the immune system by inducing
by environmental or psychosocial factors, Yang and coworkers took advantage of so-called immunogenic cell death
including social isolation, that leads to a two mouse models that have been used (Figure 1A). This happens when chemo-
cascade of systemic effects, starting extensively in studies of stress-related therapy-induced cancer cell death causes
with the processing of information in the disorders—the social defeat and the the release of danger-associated molecu-
central nervous system (Moreno-Smith acute restraint models. They combined lar patterns (DAMPs), including calreticu-
et al., 2010). The stress response involves these stress models with models of non- lin, ATP, and HMGB1 from the malignant
secretion of corticotropin-releasing hor- small-cell lung cancer (NSCLC) (caused cells (Kroemer et al., 2013). This, in turn, at-
mone from the hypothalamus, which by the carcinogen urethane) and colon tracts DCs and facilitates the uptake of tu-
leads to the release of adrenocorticotro- cancer (caused by the carcinogen azoxy- mor antigens by such DCs, allowing them
phic hormone from the anterior pituitary, methane and the inflammatory agent to engage and stimulate tumor-antigen-
in turn resulting in secretion of glucocorti- dextran sodium sulfate) and found that specific, cytotoxic T lymphocytes (CTLs)
coids from the adrenal gland (McEwen, social defeat causes an increase in the through a process known as cross-pre-
2007). Cancer patients experience many number and size of tumors. In addition, sentation. These CTLs produce interferon
different types of stress, including the social defeat stress dampened the thera- (IFN)-g and mediate long-term effects of
acute stress of getting the cancer diag- peutic response to oxaliplatin, a type of immunogenic chemotherapy. Yang and
nosis and the chronic stress of worrying chemotherapy that acts in part by coworkers showed that stress causes a
about recurrence or undergoing weeks inducing immunogenic cell death to reduction of calreticulin expression in
of chemo- or radiotherapy. Epidemiolog- mount an immune response against the DCs and exclusion of IFN-g expressing
ical and clinical studies have provided ev- tumor. Additionally, social defeat and sus- CTLs in the tumors after chemotherapy,
idence for links between stress or social tained acute restraint stress also reduced likely causing the reduced anticancer
isolation and cancer development and responses to immunogenic chemo- response in stressed mice (Yang et al.,
survival (Moreno-Smith et al., 2010; Seph- therapy, prophylactic tumor vaccination, 2019). Moreover, reduced plasma levels
ton et al., 2000). Yet, very little is known and PD-1-targeted immunotherapy in of cytokines including IFN-b, interleukin
about the mechanisms by which stress two transplanted fibrosarcoma and (IL)-15, IL-23A, and C-X-C motif ligand 9
promotes cancer. NSCLC models. (CXCL9) and CXCL10 indicated that stress
Recent studies have indicated that Next, Yang and coworkers identified causes systemic immunosuppression.
stress hormones, particularly glucocorti- corticosterone, a glucocorticoid and major Specialized, cross-presenting DCs
coids, have a dark side that can negatively stress hormone in mice, as the main have emerged as a central cell type in
influence cancer treatment response neuroendocrine stress mediator linking the stimulation of anti-cancer immune re-
(Arbour et al., 2018; Obradovic  et al., stress and immunosuppression. A gluco- sponses. However, analyzing RNA
2019). Notably, Obradovic  and coworkers corticoid receptor antagonist, mifepris- expression profiles in tumor-infiltrating
reported earlier this year that activation of tone, efficiently abolished the immunosup- DCs after chemotherapy, Yang and co-
glucocorticoid receptor signaling in cancer pressive effect of stress on chemotherapy, workers found no evidence that stress
cells, via ROR1 kinase signaling, results in and conversely, synthetic glucocorticoids causes changes in lineage commitment
increased metastatic colonization and attenuated the therapeutic effects of tumor or differentiation of DCs. Instead,
resistance to chemotherapy (Obradovic  vaccination and chemotherapy in un- they found increased expression of the
et al., 2019), thus raising concerns about stressed mice. These data show that glucocorticoid-inducible transcriptional

468 Cancer Cell 36, November 11, 2019 ª 2019 Elsevier Inc.
Cancer Cell

Previews

A
Immunogenic
Chemotherapy

Stress Induced
Glucocorticoids

B Stress Induced
Glucocorticoids
Immunogenic
Chemotherapy
Tsc22d3

Type I IFN responses:


IFN-E
CXCL1/9/10
...

Tumor Glucocorticoid
Dendritic Cell Tumor Cell TCR Glucocorticoid
Antigen Receptor D

Cytotoxic T Dying Glucocorticoid


MHC I IFN-J HSP90
Lymphocyte Tumor Cell Response Elements

Figure 1. A Stress-Glucocorticoid-Dendritic Cell Axis Compromises Therapy-Induced Antitumor Immunity


(A) Immunogenic chemotherapy causes the release of tumor antigens and danger-associated molecular patterns. The antigens are taken up by dendritic cells
(DCs), which cross-represent them to cytotoxic T lymphocytes (CTLs), activating the CTLs so they can kill the tumor cells.
(B) Yang and coworkers elegantly show that stress-induced glucocorticoids inhibit therapy-induced anti-tumor immunity, including after immunogenic
chemotherapy, by upregulating the glucocorticoid-inducible transcriptional regulator Tsc22 domain family protein 3 (Tsc22d3) in DCs. The mechanisms involved
likely include reduced expression of IFN-b, CXCL1/9/10, and CTL suppression (Yang et al., 2019). CXCL, C-X-C motif ligand; HSP90, heat shock protein 90; IFN,
interferon; MHC I, major histocompatibility complex I; TCR, T cell receptor.

regulator Tsc22 domain family protein 3 sion remains to be determined, but the healthy volunteers and that elevated
(Tsc22d3; also known as glucocorticoid- authors noted that the expression of glucocorticoid levels correlated with a
induced leucine zipper, Gilz) in DCs from Calr, Ifnb1, Cxcl9, and Cxcl10 in the tu- high score for a negative mood (so-called
mice exposed to stress (Figure 1B). mor-infiltrating dendritic cells was ‘‘profile of mood states’’ score). Also,
Transgenic overexpression of Tsc22d3 in restored in mice lacking Tsc22d3 in DCs. TSC22D3 expression was higher in pe-
the DCs was sufficient to abolish the re- Modeling stress responses in animals ripheral blood mononuclear cells from
sponses to immunogenic chemotherapy has obvious limitations due to the higher cancer patients than from healthy volun-
and tumor vaccine in unstressed mice, awareness and emotional aptitude of hu- teers and correlated with elevated cortisol
whereas stress-exposed mice lacking mans, so do these findings from mice levels. Taken together, Yang et al. have
Tsc22d3 specifically in DCs showed an translate to humans? Yang and co- identified a new stress-glucocorticoid-
enhanced response to anti-PD-1 treat- workers found that glucocorticoid levels DC axis that compromises therapy-
ment and immunogenic chemotherapy. were higher in sera from patients with induced antitumor immunity (Yang
How Tsc22d3 caused immune suppres- colorectal cancer or NSCLC than in et al., 2019).

Cancer Cell 36, November 11, 2019 469


Cancer Cell

Previews

Prior work showed that glucocorticoid could be detrimental to treatment war- Death-1 and Programmed Death-Ligand 1
blockade in patients with non-small-cell lung can-
elevation induced by metabolic stress rants further investigation. cer. J. Clin. Oncol. 36, 2872–2878.
suppresses intratumoral adaptive immu- Future work will hopefully clarify the
nity, causing failure of cancer immuno- types and extent of stress that affect can- Flint, T.R., Janowitz, T., Connell, C.M., Roberts,
E.W., Denton, A.E., Coll, A.P., Jodrell, D.I., and
therapy (Flint et al., 2016). Thus, both psy- cer progression and therapy responses in Fearon, D.T. (2016). Tumor-induced IL-6 repro-
chological and metabolic stress, via people and will likely also identify addi- grams host metabolism to suppress anti-tumor im-
glucocorticoids, have major effects on tional mechanisms responsible for the ef- munity. Cell Metab. 24, 672–684.

cancer progression and treatment in fects of stress. Such knowledge may Gopalakrishnan, V., Helmink, B.A., Spencer, C.N.,
mice. Correspondingly, abnormal gluco- enable devising strategies to reduce the Reuben, A., and Wargo, J.A. (2018). The influence
of the gut microbiome on cancer, immunity, and
corticoid levels, consistent with stress, harmful effects of stress and allow syn- cancer immunotherapy. Cancer Cell 33, 570–580.
were reported to correlate with reduced thetic glucocorticoids to be used to con-
survival in breast cancer (Sephton et al., trol side effects and symptoms while pre- Kroemer, G., Galluzzi, L., Kepp, O., and Zitvogel, L.
(2013). Immunogenic cell death in cancer therapy.
2000). Intriguingly, stress also modulates venting them from promoting cancer Annu. Rev. Immunol. 31, 51–72.
the gastrointestinal microbiome (Rhee growth, metastasis, and therapy resis-
et al., 2009), which can affect the efficacy tance. The above-mentioned research McEwen, B.S. (2007). Physiology and neurobi-
ology of stress and adaptation: central role of the
of cancer immune checkpoint therapy also highlights that cancer is not only a brain. Physiol. Rev. 87, 873–904.
(Gopalakrishnan et al., 2018). Collectively, disease of genomic, cell-biological, or
Moreno-Smith, M., Lutgendorf, S.K., and Sood,
these studies suggest that psychological microenvironmental alterations but, in A.K. (2010). Impact of stress on cancer metastasis.
support for cancer patients should be a addition, cancer relies on altered body Future Oncol. 6, 1863–1881.
critical component of cancer treatment. physiology. The interface between cancer
Obradovic, M.M.S., Hamelin, B., Manevski, N.,
But the implications reach further: syn- biology and physiology must therefore be Couto, J.P., Sethi, A., Coissieux, M.-M., Mu€nst,
thetic glucocorticoids are widely used in understood to ultimately achieve better S., Okamoto, R., Kohler, H., Schmidt, A., and
Bentires-Alj, M. (2019). Glucocorticoids promote
cancer treatment to combat side effects survival of cancer patients.
breast cancer metastasis. Nature 567, 540–544.
of chemotherapy and to treat symptoms
related to advanced cancer. Multiple ACKNOWLEDGMENTS Rhee, S.H., Pothoulakis, C., and Mayer, E.A.
(2009). Principles and clinical implications of the
groups have now highlighted that in ani- brain-gut-enteric microbiota axis. Nat. Rev.
mal models, glucocorticoids have clear M.E. and L.V.A. were supported by a grant from the Gastroenterol. Hepatol. 6, 306–314.
Simons Foundation to Cold Spring Harbor Labora-
negative effects on the outcome of ther- tory. X.-Y.H. was supported by a fellowship from Sephton, S.E., Sapolsky, R.M., Kraemer, H.C., and
apy (Flint et al., 2016; Obradovic  et al., the State of New York (contract #C150158). Spiegel, D. (2000). Diurnal cortisol rhythm as a pre-
2019; Yang et al., 2019). In line with this, dictor of breast cancer survival. J. Natl. Cancer
Inst. 92, 994–1000.
glucocorticoid use was recently reported REFERENCES
to be associated with poorer outcome in Yang, H., Xia, L., Chen, J., Zhang, S., Martin, V., Li,
NSCLC patients treated with anti-PD-L1 Arbour, K.C., Mezquita, L., Long, N., Rizvi, H., Q., Lin, S., Chen, J., Calmette, J., Lu, M., et al.
Auclin, E., Ni, A., Martı́nez-Bernal, G., Ferrara, R., (2019). Stress-glucocorticoid-TSC22D3 axis com-
(Arbour et al., 2018). Thus, the possibility Lai, W.V., Hendriks, L.E.L., et al. (2018). Impact of promises therapy-induced antitumor immunity.
that synthetic glucocorticoid treatment baseline steroids on efficacy of Programmed Cell Nat. Med. 25, 1428–1441.

470 Cancer Cell 36, November 11, 2019

You might also like