Immune checkpoint-targeted therapy: cancer and autoimmune diseases represent two sides of the same coin

Noah Isakov*

The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and the Cancer Research Center, Ben Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel

*Corresponding Author:
Isakov N
The Shraga Segal Department of Microbiology
Immunology and Genetics, Ben-Gurion University
Beer-Sheva, Israel
Tel: 972-8-6477267
Fax: 972-8-6477626
E-mail: noah@bgu.ac.il

Received date: April 11, 2016; Accepted date: April 28, 2016; Published date: May 02, 2016

Citation: Isakov N. Immune Checkpoint-Targeted Therapy: Cancer and Autoimmune Diseases Represent Two Sides of the Same Coin. J Autoimmun Disod. 2016, 2:2. DOI:10.21767/2471-8513.100017

Copyright: © 2016 Isakov N. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Visit for more related articles at Journal of Autoimmune Disorders

Abstract

The immune system plays a critical role in tumor surveillance and cancer prevention. However, some cancer cells can evade immune destruction by acquiring the ability to inhibit immune checkpoint regulatory pathways and suppress anti-cancer immune responses. In recent years, the immune checkpoints took center stage in cancer immunotherapy and several promising strategies, based on the intervention in immune checkpoint-regulated pathways, have been designed in order to overcome mechanisms of immunosuppression. Clinical studies, using anti-inhibitory immune checkpoint-receptor antibodies, demonstrated durable clinical responses even in patients with advanced cancer. However, the clinical benefit was limited to a subset of cancer patients. Furthermore, the immune checkpoint therapy, which unleashes the immune system in order to augment the anti-cancer immune response, also increases the incidence of autoimmune diseases and induces an array of immune-related adverse effects. Here we briefly discuss some of the pros and cons of immune checkpoint-directed immunotherapy.

Keywords

Immune checkpoint; Immunotherapy; Cancer; Autoimmune diseases; T lymphocytes

Description

One of the most promising approaches for cancer therapy involves the manipulation of immune checkpoint receptors, which consist of a number of stimulatory and inhibitory cell surface proteins that are critical for the regulation of immune cell functions.

Under normal physiological conditions, immune checkpoints are implicated in the initiation of immune responses and determination of the intensity and duration of the response. As such, they operate to limit collateral tissue damage during anti-microbial immune responses. More important, they maintain self-tolerance and are critical for the prevention of autoimmune diseases.

Recent studies indicated that cancer cells could evade immune destruction by acquiring abilities to inhibit immune checkpoint pathways and suppression of anti-cancer T cell responses. More specifically, immune checkpoint receptorexpressing tumor infiltrating lymphocytes were found to interact with their cognate ligands on the surface of the tumor cell thereby undergoing local, selective immunosuppression at the tumor microenvironment [1]. Based on these observations, antibodies (Abs) were designed to downregulate the inhibitory immune-checkpoint receptors, and there in vivo administration was found to unleash effective anti-tumor immunity. Clinical studies demonstrated that such Abs could induce durable clinical responses even in patients with advanced cancer [2,3].

The cytotoxic T lymphocyte associated antigen-4 (CTLA-4; CD152) is the most studied immune-checkpoint receptor in the context of cancer immunotherapy and the first immunecheckpoint receptor to be clinically targeted [4-6]. It is a potent negative regulator of T cell responses which counteracts the activity of the costimulatory receptor CD28. CTLA-4 and CD28 share the B7.1 (CD80) and B7.2 (CD86) ligands, but a higher affinity of CTLA-4 allows it outcompete CD28 in ligand binding and delivery of inhibitory signals to the T cells [7,8]. Thus, while cytotoxic T lymphocytes (Tc) can identify and destroy a large variety of cancer cells, signal delivery via the CTLA-4 surface receptor downregulates Tc functions and their ability to eradicate cancer. Nevertheless, a monoclonal antibody (mAb), which blocks CTLA-4, termed Ipilimumab, was found to turn off this inhibitory signal and allow the cytotoxic T cells to destroy cancer cells. Ipilimumab (trade name Yervoy, a human IgG1 mAb) was the first to demonstrate survival benefit for patients with metastatic melanoma [6]. It received U.S. Food and Drug Administration (FDA) approval for melanoma treatment in 2011, and is currently under clinical trials in various types of tumors, including lung carcinoma, bladder cancer and metastatic hormone-refractory prostate cancer.

Programmed death-1 (PD-1; CD279) is the second immunecheckpoint receptor that emerged as a promising target for immunotherapy. PD-1 is expressed on T cells and a variety of other types of immunocytes and plays a major role in down regulating inflammatory immune responses to infection and inhibition of autoimmune responses [4]. High expression of PD-1 was noted on tumor infiltrating lymphocytes [9] and regulatory T cells (Tregs) [10], and PD-1 ligands, including PDL1 (B7-H1; CD274) and PD-L2 (B7-DC; CD273), were found on the surface of multiple cell types, including dendritic cells, monocytes and macrophages. The expression of PD-1 ligands on T-, B-, NK-, and dendritic-cells, as well as, epithelial cells and vascular endothelial cells is upregulated during inflammatory responses, predominantly by the effect of interferon [11,12]. The high expression of PD-1 ligands, in general, and in the tumor microenvironment in particular, suppresses Tc functions, and allows cancer cells to evade immune surveillance [13-15].

To test whether blocking of the PD-1/PD-L pathway will impact on anti-tumor immunity, PD-1-specific Abs were administered to patients with advanced melanoma. A significant overall survival improvement was observed is several independent studies. Two of the anti-PD-1 mAbs, pembrolizumab (Keytruda, a humanized mAb) and nivolumab (Opdivo a human IgG4 mAb), received FDA approval in 2014, and studies are in progress in order to test their effectiveness in different cancer diseases.

A similar strategy to target PD-L1 was tested in squamous and non-squamous non-small-cell lung cancer (NSCLC) and bladder cancer patients, and initial results of clinical trials, using durvalumab (anti-PD-L1 IgG1; MEDI4736) and atezolizumab (anti-PD-L1 IgG1; MPDL3280A) demonstrated a limited durable benefit [16,17].

Immunocytes express additional types of inhibitory immune-checkpoint receptors, such as the B- and Tlymphocyte attenuator (BTLA; CD272), lymphocyte activation gene-3 (LAG-3; CD223), T-cell immunoglobulin (Ig) domain and mucin domain 3 (TIM-3), and the killer-cell immunoglobulinlike receptor (KIR) [18,19]. Some of the inhibitory immune checkpoint receptor-ligands were found to be selectively upregulated in certain types of cancer cells. Attempts to block these receptors and their cognate ligands in order to augment anticancer immune responses are in progress.

The clinical use of Abs against negative checkpoint regulators represents a new and novel approach in cancer immunotherapy since the targets of the Abs are receptors on T cells that regulate their biological activity, rather than tumor cell-specific target molecules. In addition, the anti-immune checkpoint receptor Abs do not activate T cell responses against specific tumor antigens, but remove inhibitory pathways that block effective T cell responses against multiple types of tumors. While the immune checkpoint therapy represents an important weapon against cancer, unfortunately, it elicits long-term remission of tumors only in a fraction of the cancer patients. This is likely to reflect the large genetic and phenotypic heterogeneity existing between tumors and among cells within individual tumors, as well as the enormous genetic polymorphism prevailing among people, which determines not only the susceptibility to various diseases, but also the ability to respond to drugs and recover. In an attempt to overcome some of these difficulties, ongoing studies will be aimed to identify predictive biomarkers that will help select optimal therapies to individual cancer patients. Additional information on regulatory pathways that control T cell functions will help identify new drug targets, while a combination therapy which targets distinct immune cell types and intervenes at different stages of the immune response is likely to provide an overall survival benefit for a greater number of patients.

Despite the proven effectiveness of immune checkpointbased immunotherapy in a range of cancer diseases, these new treatment modalities came with a price. The intervention in the normal physiological regulation of T cells has led to immune dysfunction and an increase in the incidence of autoimmune diseases.

As indicated earlier, CTLA-4 is an important attenuator of T cell activation and an essential component of the regulatory system that controls peripheral tolerance [20,21]. Direct linkage of CTLA-4 to autoimmunity was clearly demonstrated in germline Ctla4-deficient mice, which developed a T cellmediated autoimmune lymphoproliferative disorder associated with splenomegaly, lymphadenopathy, growth retardation, and early death [22,23]. In addition, a blockade of CTLA-4 in various mouse models of autoimmunity led to exacerbate autoimmune responses [24-26], while genetic evidence provided a further link between Ctla4 and an array of human autoimmune diseases [26,27].

Mice with a restricted germline deletion of Ctla4 in Tregs suffered from T cell-mediated autoimmune symptoms, which resembled those observed in Ctla4-deficient mice [28]. Although some studies support a role for CTLA-4 in Treg suppressor activity [29,30], others demonstrated that Ctla4- deficient Tregs are capable of suppressing certain autoimmune diseases [31,32] suggesting that CTLA-4 might be involved in selective Treg functions that predispose hosts to autoimmune diseases [33].

The entire spectrum of side effects induced by anti-CTLA-4 Abs, as well as by Abs directed against other co-inhibitory immune checkpoint receptors, is referred to as ‘immunerelated adverse events’ (irAEs). The frequency and type of irAE symptoms varied in clinical trials, depending on the nature of the target molecule and the patient cohort, the type and amount of Ab, as well as the particular protocol, and involved a range of tissues and organs. These symptoms led to dermatitis, enterocolitis, hepatitis, and wide spread endocrinopathies (hypophysitis, thyroiditis, adrenal insufficiency), with less frequent symptoms of uveitis, nephritis, arthritis, and inflammatory myopathy [6,34,35].

These autoimmune-like side effects are likely to reflect the immune tolerance breaking by the CTLA-4 (or other coinhibitory receptor) blockade, since these symptoms tend to resolve upon cessation of Ab treatment. Unfortunately, the development of autoimmune symptoms did not correlate with health improvement and/or tumor regression in anti-CTLA-4/ anti-PD-1-treated cancer patients, indicating that they cannot serve as valid predictors of treatment outcome.

These autoimmune-like side effects are likely to reflect the immune tolerance breaking by the CTLA-4 (or other coinhibitory receptor) blockade, since these symptoms tend to resolve upon cessation of Ab treatment. Unfortunately, the development of autoimmune symptoms did not correlate with health improvement and/or tumor regression in anti-CTLA-4/ anti-PD-1-treated cancer patients, indicating that they cannot serve as valid predictors of treatment outcome.

Immune checkpoint therapy has also been considered in autoimmune diseases and one of the major approaches is based on the inhibition of costimulatory immune checkpoint receptors.

It is well established that T cells require two independent signals in order to become fully activated. The first signal is obtained by TCR interaction with a peptide antigen presented on a major histocompatibility complex (MHC) receptor on the surface of an antigen-presenting cell (APC), while the second signal is provided by interaction of the CD28 costimulatory receptor with the APC-expressed CD80 or CD86 ligand. TCRmediated signaling in the absence of costimulation allows T cells to interpret signals as "self" and acquire a state of anergy. In contrast, simultaneous triggering of the TCR and the CD28 costimulatory receptor promotes the differentiation into potent effector T cells.

An autoregulatory mechanism that prevents the constitutive activation of the T cells is mediated by the TCR/CD28 activation-induced expression of CTLA-4, which can then interact with CD80/CD86 on the surface of APC, and deliver an inhibitory signal that terminate the response. This inhibitory signal is delivered to the fully differentiated effector T cells that have already synthesized and secreted a multitude of proinflammatory cytokines which amplified the immune response. Therefore, intervention with CTLA-4-dependent signals in autoimmune patients cannot alleviate autoimmune response.

In order to downregulate the activity of autoreactive T cells at a very early stage, scientists designed a soluble protein mimetic molecule, which binds CD80/86 and blocks T cell activation. This drug, termed abatacept (Orencia) consists of an extracellular domain of CTLA-4 fused to the Fc portion of a human IgG1 (CTLA4Ig), and since CTLA-4 exhibits a higher binding affinity to CD80/CD86, compared to CD28, the drug competes with CD28, occupies CD80/CD86, prevents the delivery of the costimulatory signal, and dampens-down autoimmune and inflammatory responses [36]. Indeed, abatacept was able to prevent antigen-presenting cells (APCs) from delivering CD28-dependent costimulatory signals to T cells [37]. Furthermore, abatacept has demonstrated longterm efficacy in various forms of rheumatoid arthritis [38,39], as well as other disease conditions [40,41].

It should be noted that besides, CD28, T cells express a number of immune-checkpoint receptors with costimulatory activity, including the CD28-family member, inducible T-cell costimulator (ICOS), members of the tumor necrosis factor (TNF) receptor superfamily, such as CD27, CD40, CD134 (OX40), CD137 (4-1BB), and the glucocorticoid-induced TNFR family related gene (GITR) [18,19,42,43]. Engagement of these receptors can potentially contribute to the T cell activation response, and therefore these costimulatory receptors and their cognate ligands can serve as putative targets for immunotherapy in various autoimmune diseases.

The progress made in immunotherapy, using immune checkpoint blocking Abs, has significantly improved the outlook for patients with a variety of malignancies. Further discoveries of novel immune-checkpoint receptors and characterization of their mechanism of action will increase the spectrum of target molecules for immunotherapy. In addition, determination of the most effective combinatorial approaches for particular diseases will enhance the rate of recovery. Since the current treatments are effective only in a fraction of cancer patients, the identification of predictive biomarkers will help define optimal therapies for individuals and maximize the treatment benefit. The intense future studies to optimize immune checkpoint-targeted therapy must take into account the drug-induced immune-related adverse events and develop highly effective strategies to counteract the common iatrogenic effects.

Funding

This work was funded in part by the USA-Israel Binational Science Foundation, the Israel Science Foundation administered by the Israel Academy of Science and Humanities, and a donation by Mr. Martin Kolinsky. N.I. holds the Joseph H. Krupp Chair in Cancer Immunobiology.

References

  1. Topalian SL, Drake CG, Pardoll DM (2012) Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. CurrOpinImmunol 24: 207-212.
  2. Bhatia S, Thompson JA2 (2014) Melanoma: immune checkpoint blockade story gets better. Lancet 384: 1078-1079.
  3. Korman AJ, Peggs KS, Allison JP (2006) Checkpoint blockade in cancer immunotherapy. AdvImmunol 90: 297-339.
  4. Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, et al. (2005) CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 25: 9543-9553.
  5. Hodi FS, MC Mihm, RJ Soiffer, FG Haluska, M Butler, et al. (2003) Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. ProcNatlAcadSci USA 100: 4712-4717.
  6. Hodi FS, SJ O'Day, DF McDermott, RW Weber, JA Sosman, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363: 711-723.
  7. Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, et al. (1994) Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1: 793-801.
  8. Schneider H, Downey J, Smith A, Zinselmeyer BH, Rush C, et al. (2006) Reversal of the TCR stop signal by CTLA-4. Science 313: 1972-1975.
  9. Ahmadzadeh M, LA Johnson, B Heemskerk, JR Wunderlich, ME Dudley, et al.(2009)Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114: 1537-1544.
  10. Wang W, Lau R, Yu D, Zhu W, Korman A, et al. (2009) PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. IntImmunol 21: 1065-1077.
  11. Flies DB, Chen L (2007) The new B7s: playing a pivotal role in tumor immunity. J Immunother 30: 251-260.
  12. Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, et al. (2002) Expression of programmed death 1 ligands by murine T cells and APC. J Immunol 169: 5538-5545.
  13. Shi L, Chen S, Yang L, Li Y1 (2013) The role of PD-1 and PD-L1 in T-cell immune suppression in patients with hematological malignancies. J HematolOncol 6: 74.
  14. Chinai JM, Janakiram M, Chen F, Chen W, Kaplan M, et al. (2015) New immunotherapies targeting the PD-1 pathway. Trends PharmacolSci 36: 587-595.
  15. Baptista MZ, Sarian LO, Derchain SF, Pinto GA, Vassallo J (2016) Prognostic significance of PD-L1 and PD-L2 in breast cancer. Hum Pathol 47: 78-84.
  16. Fehrenbacher L, A Spira, M Ballinger, M Kowanetz, J Vansteenkiste, et al. (2016)Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet.
  17. Antonia S, Goldberg SB, Balmanoukian A, Chaft JE, Sanborn RE, et al. (2016) Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol 17: 299-308.
  18. Baitsch L, A Legat, L Barba, SA Fuertes Marraco, JP Rivals, et al. (2012) Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization. PLoS One 7: e30852.
  19. Le Mercier I, Lines JL, Noelle RJ (2015) Beyond CTLA-4 and PD-1, the Generation Z of Negative Checkpoint Regulators. Front Immunol 6: 418.
  20. Krummel MF, Allison JP (1996) CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med 183: 2533-2540.
  21. Walunas TL, DJ Lenschow, CY Bakker, PS Linsley, GJ Freeman, et al.(1994) CTLA-4 can function as a negative regulator of T cell activation. Immunity 1: 405-413.
  22. Tivol EA, F Borriello, AN Schweitzer, WP Lynch, JA Bluestone, et al. (1995) Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3: 541-547.
  23. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, et al. (1995) Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270: 985-988.
  24. Karandikar NJ, Vanderlugt CL, Walunas TL, Miller SD, Bluestone JA (1996) CTLA-4: a negative regulator of autoimmune disease. J Exp Med 184: 783-788.
  25. Lühder F, Höglund P, Allison JP, Benoist C, Mathis D (1998) Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) regulates the unfolding of autoimmune diabetes. J Exp Med 187: 427-432.
  26. Scalapino KJ, Daikh DI (2008) CTLA-4: a key regulatory point in the control of autoimmune disease. Immunol Rev 223: 143-155.
  27. Ueda H, JM Howson, L Esposito, J Heward, H Snook, et al. (2003) Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 423: 506-511.
  28. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, et al. (2008) CTLA-4 control over Foxp3+ regulatory T cell function. Science 322: 271-275.
  29. Takahashi T, Tagami T, Yamazaki S,Uede T, Shimizu J, et al. (2000) Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 192: 303-310.
  30. Read S, Malmström V, Powrie F (2000) Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med 192: 295-302.
  31. Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, et al. (2006) Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J Immunol 177: 4376-4383.
  32. Verhagen J, Gabrysová L, Minaee S, Sabatos CA, Anderson G, et al. (2009) Enhanced selection of FoxP3+ T-regulatory cells protects CTLA-4-deficient mice from CNS autoimmune disease. ProcNatlAcadSci U S A 106: 3306-3311.
  33. Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, et al. (2015) Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med 212: 1603-1621.
  34. Di Giacomo AM, Biagioli M, Maio M (2010) The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. SeminOncol 37: 499-507.
  35. Kong YC, Flynn JC (2014) Opportunistic Autoimmune Disorders Potentiated by Immune-Checkpoint Inhibitors Anti-CTLA-4 and Anti-PD-1. Front Immunol 5: 206.
  36. Iannone F, Lapadula G (2012) The inhibitor of costimulation of T cells: abatacept. J RheumatolSuppl 89: 100-102.
  37. Dall'Era M, Davis J (2004) CTLA4Ig: a novel inhibitor of costimulation. Lupus 13: 372-376.
  38. Keystone E, Alkhalaf A, Makkawy M (2015) Subcutaneous abatacept in rheumatoid arthritis: current update. Expert OpinBiolTher 15: 1221-1230.
  39. Pieper J, Herrath J, Raghavan S, Muhammad K, VollenhovenRv, et al. (2013) CTLA4-Ig (abatacept) therapy modulates T cell effector functions in autoantibody-positive rheumatoid arthritis patients. BMC Immunol 14: 34.
  40. Grasland A, Sterpu R, Boussoukaya S, Mahe I (2012) Autoimmune hepatitis induced by adalimumab with successful switch to abatacept. Eur J ClinPharmacol 68: 895-898.
  41. Gupta NK, Yilmaz O, Fisher M, Yajnik V (2014) Abatacept: a new treatment option for refractory adult autoimmune enteropathy. J ClinGastroenterol 48: 55-58.
  42. Inman BA, Frigola X, Dong H, Kwon ED (2007) Costimulation, coinhibition and cancer. Curr Cancer Drug Targets 7: 15-30.
  43. Watts TH (2005) TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol 23: 23-68.
Select your language of interest to view the total content in your interested language

Viewing options

Flyer image

Share This Article

paper.io

agar io

wowcappadocia.com
cappadocia-hotels.com
caruscappadocia.com
brothersballoon.com
balloon-rides.net

wormax io