Oxygen starvation during T cell priming boosts cancer-killing potential
Editorial

Oxygen starvation during T cell priming boosts cancer-killing potential

Špela Konjar*, Birte Blankenhaus*, Marc Veldhoen

Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal

*These authors contributed equally to this work.

Correspondence to: Špela Konjar; Birte Blankenhaus. Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisbon 1649-028, Portugal. Email: spelakonjar@medicina.ulisboa.pt; birte.blankenhaus@medicina.ulisboa.pt.

Comment on: Gropper Y, Feferman T, Shalit T, et al. Culturing CTLs under Hypoxic Conditions Enhances Their Cytolysis and Improves Their Antitumor Function. Cell Rep 2017;20:2547-55.


Submitted Nov 22, 2017. Accepted for publication Dec 04, 2017.

doi: 10.21037/tcr.2017.12.07


Immune-based tumor-therapy has seen substantial progress in recent years and has made important inroads in the fight against cancer. Checkpoint inhibitors are a clinical success and the use of cell-based therapy models is rapidly expanding. T cells, such as chimeric antigen receptor T cells (CARs) and tumor infiltrating lymphocytes (TILs), are now routinely cultured and activated in labs and subsequently adoptively transferred to patients with the aim of reducing and eradicating tumors. However, although positive results have been obtained in clinical trials, immune rejection of tumors is often not successful. This has focused attention on improving delivery and cytotoxic potential of transferred cells. Gropper et al. (1), now show that culturing CD8 T cells under hypoxic conditions, during in vitro priming and prior to adoptive transfer, enhances their cytolytic capacity resulting in more robust anti-tumor activity in a murine cancer model. These findings could have implications for the future of T cell-based tumor therapies and may improve the design of protocols aimed at potentiating cytotoxicity prior to adoptive transfer of T cells.

Immunotherapy with adoptive T cells has become one of the leading forces in the fight against cancer. Advanced clinical trials demonstrated positive results in treating a variety of cancers including melanoma, cervical cancer, lymphoma, leukemia and neuroblastoma (2). Although clinical trials show promising results, the efficiency of those therapies needs further improvement with a main goal to increase T cell survival and anti-tumor activity (3). The success of cell-based cancer immunotherapy is thought, at least in part, to be determined at the tumor site and by the tumor environment (4). Hypoxia, a state of low oxygen tension caused by a reduction in blood supply, is one of the environmental determinants that can influence the effectiveness of T cell mediated tumor rejection. Hypoxia is a particular characteristic of solid tumors due to their often limited vascularization. This is associated with poor prognosis in many types of cancers, including cervical carcinoma, head and neck cancer and sarcoma (5). Hypoxia is reported to promote tumor cell stemness, migration, metastasis, invasiveness, and resistance to radio- and chemotherapy (6).

The impact of hypoxia on T cells and their function in anti-tumor immunity is not fully understood (7). T cells migrate in the oxygen-rich bloodstream and are thought to be primed in oxygen sufficient lymph nodes (8), while encountering hypoxic conditions when migrating into the tissues. An early study of activated human T cells suggested a positive correlation between hypoxia, T cell viability and proliferation (9). However, a study by Gaber et al. (10) showed that hypoxia decreases the proliferation of mitogen stimulated human CD4+ T cells. Tumor-associated hypoxia has been reported to promote the recruitment of regulatory T (Treg) cells through induction of the CC-chemokine ligand 28, which could reduce tumor rejection (11). In addition, hypoxia can alter the differentiation of CD4+ T cells, thereby altering their functional capacities and reducing T cell migration toward tumors (12,13). Furthermore, increased use of anaerobic glycosylation in hypoxia and the subsequent secretion of lactate causes acidification of the tumor environment that can decrease T cell activation, proliferation and cytotoxicity (14). Collectively, the influence of hypoxia on T cells can be manifold with direct and indirect consequences on tumor rejection. This has hampered our understanding of the impact of hypoxia on T cell-mediated tumor cell cytotoxicity and apparent contradictory results have been reported, with both T cell inhibitory and boosting observations (15-17).

Gropper et al. took a reductionist approach to explore the direct impact of hypoxia during murine CD8 T cell priming (1). They used ovalbumin (OVA)-specific CD8+ T cells from OT-I T cell receptor (TCR) transgenic mice. OT-I T cells were activated in vitro using the SIINFEKL peptide and interleukin (IL)-2 under hypoxic (1% O2) or atmospheric (20% O2) conditions for 5 days. Of note, the concentration of oxygen within lymph nodes, where T cells are primed, are reported to be between 0.5% and 6% (8). T cells cultured under hypoxic conditions were viable, larger in size and contained more mitochondrial mass compared with T cells cultured under atmospheric oxygen pressure. T cell proliferation assayed with CFSE-dilution showed less proliferation of CD8 T cells primed under hypoxic condition compared with atmospheric oxygen pressure, which reduced the yield of T cells harvested under reduced oxygen pressure. This can be of concern for adoptive T cell therapies, which require substantial T cell numbers. However, although the maturation status of hypoxic cytotoxic T cells (CTLs) up to day 3 post-activation was indiscriminate to those cultured under atmospheric oxygen pressure, 5-day post-activation cells primed under hypoxic conditions harbored an increased percentage of highly activated CD44hiCD62Llo cells. Transcriptome analysis of CTLs generated under different oxygen pressures revealed increased glycolysis capacity of cells primed under hypoxia, such as higher levels of transcripts for glucose transporter (Glut) 1 and hypoxia-inducible factor (HIFs) family members. Together, the data showed that priming of CD8 T cells under hypoxic conditions results in reduced yield of CTLs, but with an increased mature phenotype such as previously reported for TILs in vivo (18).

To characterize the CTLs generated, markers associated with T cells exhaustion and inhibition, CTLA-4, PD-1, TIM3, LAG3 were compared between CD8 T cells primed under atmospheric and hypoxic conditions. Especially the co-expression of PD-1 with TIM3 and LAG3 on TILs has been used as a marker of CTL dysfunction, demonstrated by a reduced activation profile and cytokine production (19,20). Neutralizing monoclonal antibodies against PD-1 and CTLA-4 are approved for clinical use as checkpoint inhibitors and show very promising results in late stage clinical trials (21). The expression levels of PD-1 and CTLA-4 were, however, similar between CTLs generated under hypoxic or atmospheric oxygen conditions. Instead, the expression of TIM3 and LAG3 was increased on hypoxic CTLs. In contrast to PD-1, the increased expression of LAG3 and TIM3 on CTLs generated under hypoxic conditions is in common with TILs found in different tumors (19,20). These data indicated that CTLs generated under conditions of low or high oxygen pressure are not approaching senescence or exhaustion, but that priming under low oxygen pressure infers a phenotype more resembling TILs.

The increase in size and glycolytic capacity suggested an increased fitness of CTLs primed under hypoxia. This was strengthened by the observation that CTLs primed under hypoxic conditions contain higher granzyme B protein levels per granule. However, compared with CTLs primed under atmospheric oxygen levels, those generated under hypoxia do not contain more granules, perforin protein or degranulation capacity. In vitro cultures of CTLs and OVA-expressing B16 tumor cells revealed increased target killing of cells generated under hypoxic conditions compared with those stimulated under atmospheric oxygen levels. The increased level of granzyme B in CTLs generate under limited oxygen levels is in line with a previous report and suggests that these cells might also exhibit more efficient tumor killing in vivo (15). CTLs generated under high or low oxygen pressure adoptively transferred into B16-OVA tumor bearing mice were indistinguishable with respect to their migration pattern, speed and depth of tissue penetration. However, mice treated with CTLs primed under hypoxia showed increased regression of the tumors and prolonged survival compared with those treated with CTLs generated under high oxygen levels. Collectively the authors argue that these results may have beneficial implications for adoptive T cell therapies using either tumor isolated antigen-specific TILs or engineered antigen-specific CAR T cells. Priming or re-stimulation of CTLs under hypoxic conditions may be an effective way of enhancing anti-tumor efficiency.

Gropper et al., show that the priming of mouse CD8 T cells under reduced oxygen levels results in slower proliferation but enhanced maturation with respect to glycolysis potential, mitochondrial mass and storage of the cytolytic protein granzyme B. The data suggest that suboptimal T cell proliferation results in a qualitatively better T cell. This may be explained by the large energy levels required during clonal expansion, sustaining rapid proliferation as well as the anabolic processes of generating new cellular structures to be equally divided among daughter cells, such as mitochondria, and the synthesis of effector molecules such as granzymes. It remains to be determined if human CD8 T cells show a similar phenotype when cultured under different oxygen pressures. T cell therapy depends on the generation of sufficiently large amounts of T cells in often a short time and hence optimal T cell proliferation has been a prime focus. The results from Gropper et al. suggest a qualitative measure of T cell functionality is important. Although reducing T cell proliferation, such as achieved under limited oxygen conditions, comes at a cost of total T cell numbers, the efficacy of T cell therapy may be improved. Upon fast clonal expansion, resources or metabolic wiring may be insufficient to service both proliferation and T cell maturation optimally. If the enhanced quality of T cells is able to sustain the anti-tumor response for longer compared with those generated under atmospheric conditions remains to be determined. The CTLs transferred by Gropper et al., do not show signs of exhaustion or senescence prior to transfer. However, the cells might well acquire this phenotype after entering to the immunosuppressive tumor environment. The immunosuppressive tumor environment remains a significant hurdle to overcome for the success of adoptive transfer therapies resulting in tumor elimination and not only temporal control. Combinations of adoptive transfer of CTLs cultured under hypoxic conditions and checkpoint blockade, as suggested before (3), could be a potential strategy to further increase the success of tumor therapies.


Acknowledgments

Funding: This work in the MV laboratory is supported by the European Union H2020 ERA project (N°667824—EXCELLtoINNOV).


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Yan Li (Experimental Therapeutics Centre, Agency for Science, Technology and Research, Singapore).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2017.12.07). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Gropper Y, Feferman T, Shalit T, et al. Culturing CTLs under Hypoxic Conditions Enhances Their Cytolysis and Improves Their Anti-tumor Function. Cell Rep 2017;20:2547-55. [Crossref] [PubMed]
  2. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348:62-8. [Crossref] [PubMed]
  3. Houot R, Schultz LM, Marabelle A, et al. T-cell-based Immunotherapy: Adoptive Cell Transfer and Checkpoint Inhibition. Cancer Immunol Res. Cancer Immunol Res 2015;3:1115-22. [Crossref] [PubMed]
  4. Chen HHW, Kuo MT. Improving radiotherapy in cancer treatment: Promises and challenges. Oncotarget 2017;8:62742-58. [PubMed]
  5. Tatum JL, Kelloff GJ, Gillies RJ, et al. Hypoxia: importance in tumor biology, noninvasive measurement by imaging, and value of its measurement in the management of cancer therapy. Int J Radiat Biol 2006;82:699-757. [Crossref] [PubMed]
  6. Harris AL. Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer 2002;2:38-47. [Crossref] [PubMed]
  7. Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017;17:774-85. [Crossref] [PubMed]
  8. Huang JH, Cárdenas-Navia LI, Caldwell CC, et al. Requirements for T lymphocyte migration in explanted lymph nodes. J Immunol 2007;178:7747-55. [Crossref] [PubMed]
  9. Makino Y, Nakamura H, Ikeda E, et al. Hypoxia-inducible factor regulates survival of antigen receptor-driven T cells. J Immunol 2003;171:6534-40. [Crossref] [PubMed]
  10. Gaber T, Tran CL, Schellmann S, et al. Pathophysiological hypoxia affects the redox state and IL-2 signalling of human CD4+ T cells and concomitantly impairs survival and proliferation. Eur J Immunol 2013;43:1588-97. [Crossref] [PubMed]
  11. Facciabene A, Peng X, Hagemann IS, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011;475:226-30. [Crossref] [PubMed]
  12. Brucklacher-Waldert V, Ferreira C, Stebegg M, et al. Cellular Stress in the Context of an Inflammatory Environment Supports TGF-beta-Independent T Helper-17 Differentiation. Cell Rep 2017;19:2357-70. [Crossref] [PubMed]
  13. Hatfield SM, Kjaergaard J, Lukashev D, et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med 2015;7:277ra30 [Crossref] [PubMed]
  14. Hirschhaeuser F, Sattler UG, Mueller-Klieser W. Lactate: a metabolic key player in cancer. Cancer Res 2011;71:6921-5. [Crossref] [PubMed]
  15. Doedens AL, Phan AT, Stradner MH, et al. Hypoxia-inducible factors enhance the effector responses of CD8(+) T cells to persistent antigen. Nat Immunol 2013;14:1173-82. [Crossref] [PubMed]
  16. Caldwell CC, Kojima H, Lukashev D, et al. Differential effects of physiologically relevant hypoxic conditions on T lymphocyte development and effector functions. J Immunol 2001;167:6140-9. [Crossref] [PubMed]
  17. Nakagawa Y, Negishi Y, Shimizu M, et al. Effects of extracellular pH and hypoxia on the function and development of antigen-specific cytotoxic T lymphocytes. Immunol Lett 2015;167:72-86. [Crossref] [PubMed]
  18. Nguyen HH, Kim T, Song SY, et al. Naïve CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-β immunosuppression in the tumor microenvironment. Sci Rep 2016;6:28208. [Crossref] [PubMed]
  19. Anderson AC. Tim-3: an emerging target in the cancer immunotherapy landscape. Cancer Immunol Res 2014;2:393-8. [Crossref] [PubMed]
  20. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A 2010;107:7875-80. [Crossref] [PubMed]
  21. Homet Moreno B, Ribas A. Anti-programmed cell death protein-1/ligand-1 therapy in different cancers. Br J Cancer 2015;112:1421-7. [Crossref] [PubMed]
Cite this article as: Konjar Š, Blankenhaus B, Veldhoen M. Oxygen starvation during T cell priming boosts cancer-killing potential. Transl Cancer Res 2018;7(Suppl 1):S34-S37. doi: 10.21037/tcr.2017.12.07

Download Citation