Weekly Digests
‹ Back to July

Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma

July 12, 2017

Despite extensive CD8+ T cell infiltration, clear cell renal cell carcinoma (ccRCC) responds only moderately to checkpoint blockade therapy, suggesting other immunosuppressive mechanisms. One mechanism by which tumors can avoid effector T cell function is by metabolically inhibiting the tumor-infiltrating lymphocytes (TIL). Siska et al. used a combination of mass and flow cytometry, microscopy, and metabolic perturbations to examine the metabolic pathways and mitochondrial state in TIL from 54 patients with ccRCC and revealed how much energy and the environment matter.

The ccRCC CD8+ TIL exhibited the phenotype of an effector memory T cell and uniformly expressed high levels of PD-1, indicative of their suppressed or exhausted state. Isolated CD8+ TIL were limited in activation and proliferation compared to CD8+ T cells from the peripheral blood. Probing the metabolic machinery revealed that despite normal expression of hexokinase and the glucose transporter Glut1, the glucose uptake process in CD8+ TIL was defective and comparable to resting CD8+ T cells. Further analysis showed that Glut1 in this cell population was not phosphorylated, possibly indicating that the glucose transporter was unable to import extracellular glucose into the cell, and that downstream GAPDH levels, key to efficient glycolysis, were reduced. Interestingly, ccRCC CD4+ TIL were only slightly affected, suggesting a differential response to the environment by the two T cell types.

Stimulating the CD8+ TIL did not result in increased glucose uptake, nor did exposure to low glucose result in further suppression, indicating a metabolic adaptation that is not dependent on exogenous glucose. It became clear that this adaptation was not due to low exogenous glucose levels, as the levels of interstitial glucose in the tumor tissue were similar to adjacent normal tissue. Such observation was surprising, as tumor cell metabolism in general, and the von Hippel-Lindau (VHL) tumor suppressor gene mutations characteristic of ccRCC in particular, often lead to increased glycolysis in cancer cells. However, the lactate levels in ccRCC interstitial fluid were elevated, pointing toward the role of other metabolic pathways, including PD-1 signaling.

In addition to the metabolic impairments, changes were also observed in the mitochondria, which play a key role in energy production and generation of reactive oxygen species (ROS). Although the mitochondrial mass and expression of electron transport components and transcription factors were normal, the ccRCC CD8+ TIL had small, dispersed mitochondria that were highly fragmented and in a condensed configuration, with an elevated mitochondrial membrane potential. High levels of ROS were found inside the mitochondria, likely due to the hyperpolarization as well as the decreased expression of SOD2, which helps neutralize mitochondrial ROS. However, the total cellular ROS was not elevated, as the expression of SOD1 (which plays a role in neutralizing cytoplasmic ROS) remained normal. In addition, CD8+ TIL utilized an adaptive cytosolic mechanism (cystine uptake and subsequent increased glutathione production) to neutralize ROS. The upregulation of cytosolic ROS neutralization may have contributed to defective T cell activation, as cytosolic ROS is important to calcium signaling in activated T cells.

As the CD8+ TIL adapted to decreased dependence on glucose and high mitochondrial ROS, the metabolic defects became persistent, even in the presence of additional nutrients and treatments, which only partially increased the activation of impaired TIL. Thus, metabolic adaptations within the tumor microenvironment appear to be imprinted upon the existing infiltrating T cells and have limited the activation and effectiveness of CD8+ TIL in ccRCC. Finally, CD8+ T cells demonstrating high membrane polarization can be found in the peripheral blood of ccRCC patients, potentially identifying tumor-experienced T cells.

by Anna Scherer

References:

Siska P.J., Beckermann K.E., Mason F.M., Andrejeva G., Greenplate A.R., Sendor A.B., Chiang Y.J., Corona A.L., Gemta L.F., Vincent B.G., Wang R.C., Kim B., Hong J., Chen C.L., Bullock T.N., Irish J.M., Rathmell W.K., Rathmell J.C. Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma. JCI Insight. 2017 Jun 15.

In the Spotlight...

Combining Local Immunotoxins Targeting Mesothelin with CTLA-4 Blockade Synergistically Eradicates Murine Cancer by Promoting Anti-Cancer Immunity

Leshem et al. tested locally-delivered, recombinant immunotoxins SS1P or LMB-100 with anti-CTLA-4 checkpoint blockade in mice with one or two tumors of BALB/c breast cancer expressing human mesothelin. The complete regression rate was 86% for injected tumors and 53% for uninjected tumors, indicating efficacy against metastatic tumor growth.

Monitoring immune-checkpoint blockade: response evaluation and biomarker development.

The ever-increasing use of immune-checkpoint blockade has led to the need to re-address the strategies to monitor and classify disease response (including long-term, stable disease), especially in light of “pseudoprogression”, and to identify biomarkers, standardized assays, and guidelines for predicting potential treatment responders, all comprehensively reviewed by Nishino et al.

Targeting vascular endothelial-cadherin in tumor-associated blood vessels promotes T cell-mediated immunotherapy.

An oligonucleotide-based microRNA inhibitor, CD5-2, upregulates the expression of the endothelial cell-specific junctional protein VE-cadherin, which normalizes blood vessel structure and function in the tumor microenvironment, alleviates some immunosuppressive effects (hypoxia, PD-L1 expression), supports tumor infiltration by CD8+ T cells, and enhances tumor control.

IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade.

Using an iterative approach coupling gene expression data with clinical outcomes, Ayers et al. identified an 18 member gene panel for prediction of response to pembrolizumab, which outperformed PD-L1 immunohistochemistry analysis. A highly T cell-infiltrated tumor is predicted, based on the genes induced, to correlate with positive outcomes.

Concurrent OX40 and CD30 Ligand Blockade Abrogates the CD4-Driven Autoimmunity Associated with CTLA4 and PD1 Blockade while Preserving Excellent Anti-CD8 Tumor Immunity.

CTLA-4 and PD-1 blockade have shown great anti-tumor efficacy, but significant autoimmune side-effects, likely driven by CD4+ cells, have limited their use. Nawaf et al. show that in mice depleted of intratumoral Tregs via anti-CTLA-4, an additional blockade of the OX40 and CD30 costimulatory receptors ameliorated liver autoimmune side effects, while retaining CD8+ T cell anti-tumor function.

De Novo Epigenetic Programs Inhibit PD-1 Blockade-Mediated T Cell Rejuvenation.

DNA methylation of key genomic regions in murine T cells during chronic viral infection and in tumor models establishes stable gene-silencing that leads to terminal exhaustion of CD8+ T cells, restricting response to PD-L1 blockade. Administration of demethylating agents prior to PD-L1 blockade in Tramp-C2 mice increased proliferation of CD8+ TIL and improved tumor control.

Critical biological parameters modulate affinity as a determinant of function in T-cell receptor gene-modified T-cells.

To better understand how to optimally select which TCRs to use for adoptive cell therapy, Spear et al. examined the contributions of TCR affinity, CD8 co-receptor dependence, antigen density, and TCR density to T cell functional readouts. Importantly, TCR affinity was not the best predictor of IFNγ release or degranulation, and the other factors such as antigen or TCR density are equally or more relevant.

GITR ligand fusion protein agonist enhances the tumor antigen-specific CD8 T-cell response and leads to long-lasting memory.

Durham et al. tested the effectiveness of GITR Ligand Fusion Protein (GITRL-FP) monotherapy in the immunogenic, self-priming CT26 tumor model and found that it promotes expansion of antigen-specific T cells, depletion of Treg cells, tumor clearance, and generation of a lasting CD8+ T cell memory response. In the non-self-priming TC-1 model, no GITRL-FP anti-tumor effects were observed unless accompanied with an E7-targeting vaccine.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.