561
Views
15
CrossRef citations to date
0
Altmetric
Editorial

Inhibiting the inhibitors

Checkpoints blockade in solid tumors

&
Article: e26535 | Received 04 Sep 2013, Accepted 04 Sep 2013, Published online: 26 Sep 2013

It has been known for many years that the immune system can actively respond to (pre)neoplastic cells and prevent oncogenesis (a concept known as cancer immunosurveillance).Citation1 However, in most cases, antitumor immune responses are inefficient and neoplasms are allowed to establish. Indeed, effector T cells often infiltrate developing malignant lesions and may significantly influence the prognosis of the disease,Citation2,Citation3 but are frequently functionally impaired.Citation4 As compared with circulating T cells or T cells infiltrating healthy tissues, these cells (1) secrete reduced amounts of effector cytokines, (2) exhibit limited proliferative capacity, (3) express low levels of pro-survival receptors, and (4) display a terminally differentiated phenotype. Multiple mechanisms have been associated such a functional impairment, most of which can be reconducted to the immunosuppressive nature of the tumor microenvironment.Citation5 Malignant cells actively secrete cytokines, metabolites and other molecules that directly inhibit the function of antitumor T lymphocytes. In addition, cancer cells can recruit immune cell populations that are capable of hindering effector responses. These include, among others, CD4+CD25+FOXP3+ regulatory T cells and myeloid-derived suppressor cells, which mediate immunosuppressive effects via contact-dependent as well as contact-independent mechanisms.Citation5 Thus, neoplastic cells as well as immunosuppressive cells of the immune system express multiple inhibitory ligands on their surface that engage cognate receptors on T lymphocytes.

The CD28 family of co-receptors plays a major role in the regulation of immune responses, mostly as they deliver co-stimulatory or co-inhibitory signals to T-cells that have recognized a peptide/MHC complex on the surface of an antigen-presenting cell (APC).Citation6 Besides CD28, which operates as a major co-stimulatory receptor upon binding to CD80 (also known as B7-H1) or CD86 (also known as B7-H2) on APCs, this family include cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, commonly known as PD-1). CTLA-4, which also interacts with CD80 and CD86, is expressed on the surface of activated T cells (including Tregs) and delivers co-inhibitory signals. Along similar lines, upon interaction with its main ligands (CD274, also known as PD-L1, and PDCD1LG2), PD-1 negatively regulates multiple T-cell functions, including their activation, proliferative potential and ability to secrete effector cytokines. The most prominent physiological role of these molecules is to limit T-cell responses and avoid autoimmune diseases.Citation6 However, the elevated expression levels of CTLA4 and PD-1 on cancer cells led to the hypothesis that these factors could underlie a major mechanism of immune escape mediated by the inhibition of effector T-cell responses.Citation7

Chronic antigenic or inflammatory stimulation induces the expression of CTLA4 and PD-1 on the surface of T cells, as demonstrated in individuals chronically infected with HIV-1 and the hepatitis C virus (HCV).Citation8 Similarly, these receptors are highly upregulated by tumor antigen-specific T cells, resulting in the robust impairment of effector functions. In line with this notion, the blockade of CTLA4 and PD-1 with specific monoclonal antibodies has been shown to promote tumor regression in multiple preclinical models as it stimulates the function of tumor-specific CD4+ and CD8+ T cells.Citation7 CTLA4-targeting interventions also boost immune responses by simultaneously interfering with the inhibitory activity of Tregs.Citation7 These data strongly suggested that the blockade of CTLA4 and/or PD1 could offer a valid approach for the treatment of a wide variety of cancers.Citation9

Monoclonal antibodies are nowadays widely employed in anticancer immunotherapy.Citation10,Citation11 In 2 clinical trials published in June 2012, 2 different monoclonal antibodies targeting PD-1 or PD-L1 were given to patients affected by different solid tumors, including neoplasms that are generally refractory to immunotherapy such as lung carcinoma.Citation12,Citation13 Both antibodies exhibited significant clinical activity as they induced tumor regression (objective response rate of 18–27% and 6–17% for PD-1- and PD-L1-targeting antibodies, respectively) or stabilized disease progression. Importantly, none of the patients with PD-L1- tumors receiving anti-PD-1 antibodies manifested an objective response, suggesting that the expression of PD-L1 on the surface of cancer cells constitutes a robust predictive biomarker that may allow clinicians to implement personalized therapeutic approaches.Citation13

In a recent Phase I clinical trial, melanoma patients were treated with the hitherto experimental anti-PD-1 antibody nivolumab plus ipilimumab, a CTLA4-blocking antibody approved by US Food and Drug Administration (FDA) for use in this oncological indication in 2011.Citation14 The authors reported an objective response rate of 40% and evidence of clinical activity in 65% of the patients. In spite of the fact that up to 53% of patients experienced Grade III-IV adverse events (although reversible, for the vast majority), some degree of tumor regression could be observed in more than 80% of cases.

These results lend strong support to the notion that the dormant immune system can be awaken by the simultaneous inhibition of distinct immunological checkpoints. High-content single-cell analysis of the phenotype and functionality of the immune cell subpopulations involved in such a reactivation will provide further insights into the effector mechanisms that mediate cancer immunosurveillance in humans. Given the large number of functions regulated by CTLA4 and PD-1, these mechanisms are likely to involve multiple cell types. Currently, a number of biological agents including immunostimulatory cytokines such as interleukin-15,Citation15 are being tested to break the immunological tolerance of cancer patients and boost antitumor T-cell immunity. As it has already been demonstrated in preclinical settings,Citation16 the combination of immunostimulatory agents with checkpoint inhibitors may boost even further anticancer immune responses and hence constitute an optimal immunotherapeutic approach against (at least some types of) cancer.

Abbreviations:
APC=

antigen-presenting cell

CTLA4=

cytotoxic T lymphocyte-associated protein 4

FDA=

US Food and Drug Administration

HCV=

hepatitis C virus

PD-1=

programmed cell death 1

Treg=

regulatory T cell

Citation: Mavilio D, Lugli E. Inhibiting the inhibitors: Checkpoints blockade in solid tumors. OncoImmunology 2013; 2:e26535; 10.4161/onci.26535

References

  • Senovilla L, Galluzzi L, Zitvogel L, Kroemer G. Immunosurveillance as a regulator of tissue homeostasis. Trends Immunol 2013; http://dx.doi.org/10.1016/j.it.2013.06.005; PMID: 23891238
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323 - 43; http://dx.doi.org/10.4161/onci.22009; PMID: 23243596
  • Lança T, Silva-Santos B. The split nature of tumor-infiltrating leukocytes: Implications for cancer surveillance and immunotherapy. Oncoimmunology 2012; 1:717 - 25; http://dx.doi.org/10.4161/onci.20068; PMID: 22934263
  • Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 2012; 12:269 - 81; http://dx.doi.org/10.1038/nri3191; PMID: 22437939
  • Becker JC, Andersen MH, Schrama D, Thor Straten P. Immune-suppressive properties of the tumor microenvironment. Cancer Immunol Immunother 2013; 62:1137 - 48; http://dx.doi.org/10.1007/s00262-013-1434-6; PMID: 23666510
  • Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008; 26:677 - 704; http://dx.doi.org/10.1146/annurev.immunol.26.021607.090331; PMID: 18173375
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12:252 - 64; http://dx.doi.org/10.1038/nrc3239; PMID: 22437870
  • Wherry EJ. T cell exhaustion. Nat Immunol 2011; 12:492 - 9; http://dx.doi.org/10.1038/ni.2035; PMID: 21739672
  • Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology 2012; 1:1223 - 5; http://dx.doi.org/10.4161/onci.21335; PMID: 23243584
  • Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2:e22789; http://dx.doi.org/10.4161/onci.22789; PMID: 23482847
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2012; 1:28 - 37; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455 - 65; http://dx.doi.org/10.1056/NEJMoa1200694; PMID: 22658128
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366:2443 - 54; http://dx.doi.org/10.1056/NEJMoa1200690; PMID: 22658127
  • Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369:122 - 33; http://dx.doi.org/10.1056/NEJMoa1302369; PMID: 23724867
  • Lugli E, Goldman CK, Perera LP, Smedley J, Pung R, Yovandich JL, Creekmore SP, Waldmann TA, Roederer M. Transient and persistent effects of IL-15 on lymphocyte homeostasis in nonhuman primates. Blood 2010; 116:3238 - 48; http://dx.doi.org/10.1182/blood-2010-03-275438; PMID: 20631381
  • Yu P, Steel JC, Zhang M, Morris JC, Waldmann TA. Simultaneous blockade of multiple immune system inhibitory checkpoints enhances antitumor activity mediated by interleukin-15 in a murine metastatic colon carcinoma model. Clin Cancer Res 2010; 16:6019 - 28; http://dx.doi.org/10.1158/1078-0432.CCR-10-1966; PMID: 20924130