GET THE APP

Immune Checkpoint Blockade Therapy: Merits and Demerits
Journal of Clinical & Experimental Dermatology Research

Journal of Clinical & Experimental Dermatology Research
Open Access

ISSN: 2155-9554

+44 1478 350008

Review Article - (2016) Volume 7, Issue 5

Immune Checkpoint Blockade Therapy: Merits and Demerits

Keiji Sugiura* and Mariko Sugiura
Department of Environmental Dermatology & Allergology, Daiichi Clinic, Japan
*Corresponding Author: Keiji Sugiura, Department of Environmental Dermatology & Allergology, Daiichi Clinic, Nittochi Nagoya Bld., 2F, 1-1 Sakae 2, Nakaku, Nagoya, 468-0008, Japan, Tel: +81-52-204-0834 Email:

Abstract

The up regulation of anti-tumor responses by immune-checkpoint blockade is a new approach for the treatment of advanced solid malignant tumors. There are some demerits to this therapy that require an evaluation of eligibility and efficacy, balanced against adverse effects and cost. Treatment for patients without immune checkpoint receptors or ligands remains an issue of concern. The cost of this therapy is huge and is further increased when treatment is required for adverse effects. The appropriate use of immune checkpoint therapy requires careful consideration.

<

Keywords: Immune checkpoint; PD-1; PD-L1 (CD274); PD-L2 (CD273); Melanoma; Cost

Introduction

The up regulation of anti-tumor responses by immune-checkpoint blockade is a new approach for the treatment of advanced solid malignant tumors. The checkpoints are important immune interactions between T cells and tumor cells. One key molecule in these immune interactions is programmed cell-death 1 (PD-1). There are two ligands of PD-1 and PD-L2. The blockade of PD-1 and PD-L1 binding using these antibodies upregulates the anti-tumor response. There are some demerits to this therapy that require an evaluation of eligibility and efficacy, balanced against adverse effects and cost. In cases with a low positive or negative rate of these receptors or ligands, immune checkpoint therapy is not effective.

Treatment for patients without immune checkpoint receptors or ligands remains an issue of concern. The cost of this therapy is huge and is further increased when treatment is required for adverse effects. The appropriate use of immune checkpoint therapy requires careful consideration.

Merits of Immune Checkpoint Therapy

Immune checkpoint blockade therapy

Immune checkpoint blockade therapy using immune checkpointtargeted antibodies is a new treatment option for advanced malignant melanoma, and this therapy is a new approaches in solid other malignant tumors.

In this treatment modality, the checkpoints are important immune interactions between T cells and antigen-presenting cells (APCs) or tumor cells (Figure 1). There are two key molecules in these immune interactions: programmed cell-death 1 (PD-1) and cytotoxic Tlymphocyte antigen 4 (CTLA-4) expressed on T cells [1].

clinical-experimental-dermatology-research-Interaction-between-T-cells

Figure 1: Interaction between T cells and tumor cells or APCs.

PD-1 is well-known as a T cell inhibitory receptor (antitumor immunity) expressed on activated T and B cells [2-5]. PD-1 belongs to the CD28 family [3,4], and is a marker of T cell exhaustion [6]. The functions of PD-1 in the immune response stop on-going immune activity in tissues [7]. There are two ligands of PD-1: PD-L1 (B7-H1, CD274) [8-11] and PD-L2 (B7-DC, CD273) [9-11]. These ligands are induced in inflammatory tissues [12]. Certain types of solid tumors and APCs express PD-L1 and PD-L2 on the cell surface [11-13]. PDL1 expressed on tumor cells is a key ligand in evading anti-tumor immunity [13], and can be induced by tumor-infiltrating CD4+ and CD8+ T lymphocytes [14-16]. When PD-1 on T cells binds with PDL1 on tumor cells, these T cells receive signals of inhibited T cell immunity against tumors [17,18], and tumor cells then grow and proliferate (Figure 1) [6]. The role of PD-L2 is not clear, but it is known that PD-L2 is restricted to and expressed on macrophages and dendritic cells [19,20]. PD-L1 and PD-L2 expressed on APCs induce T cell anergy or apoptosis via PD-1 on T cells [21]. The ligands of PD on tumor cells regulate adaptive Tregs, resulting in tumor-induced immune suppression [22]. Many PD-1 molecules are found on circulating T cells and tumor-infiltrating T cells in melanoma patients [23]. The blockade of PD-1 and PD-L1 binding using these antibodies up regulates the anti-tumor response (PD-L1 and PD-L2 reactivate the anti-tumor immune response (Figure 2) [18]), enhances the functional capacity of cytotoxic T cells against human melanoma antigen [24], decreases tumors, and prolongs patient survival.

clinical-experimental-dermatology-research-immune-checkpoint-agents

Figure 2: Action of immune checkpoint agents.

The agents of immune checkpoint therapy

Antibodies against PD-1 include nivolumab (BMS-936558, MDX-1106 and ONO-4538) and pembrolizumab (MK-3475): Nivolumab is an IgG4 monoclonal antibody that binds the PD-1 receptor to block its interaction with PD-L1 [23,25,26]. PD-L1 expression on tumor cells is a required biomarker of an effective response to nivolumab therapy [26]. Cases with PD-L1-negative tumors show no objective response [26,27]. Previous reports indicate that PD-L1 is expressed in 38-50% of melanomas (including metastatic melanoma) [28-31] and these PD-L1-positive cases all exhibited an objective response.

Nivolumab was approved in July 2014 in Japan and in December 2014 in the United States for unresectable or metastatic melanoma cases, and has already shown impressive and interesting results in clinical trials [2,27,32,33]. Nivolumab therapy is thus expected to make a substantial contribution to the treatment of advanced malignant melanoma.

Pembrolizumab is an IgG4 monoclonal antibody that binds to PD-1, resulting in T cell immunity against tumors. This agent has been approved by the Food and Drug Administration (FDA) in the US [26]. Iannone [34] reported that pembrolizumab achieved a high objective response rate (47%) in cases of advanced melanoma. In phase 1 trials using MK-3475 in 135 advanced melanoma patients, the overall objective response was 38%, and a 52% objective response was shown when using the maximum dose [35].

Antibodies against the ligand of PD-1 include BMS-936559 and MPDL3280A: BMS-936559 is an IgG4 monoclonal antibody that inhibits binding of PD-L1 to PD-1 and CD80 [20,36]. A phase 1 trial using this agent showed a 17% (9 of 52 melanoma patients) rate of complete or partial response [36].

MPDL3280A is a monoclonal antibody to PD-L1 that has achieved an objective response in clinical trials [35]. The objective response in advanced melanoma cases using this agent was reported to be 29% (10/35) at the 2013 annual conference of the American Society of Clinical Oncology [35].

Antibody against CTLA-4: Ipilimumab is an IgG1 monoclonal antibody that binds to CTLA-4. CTLA-4 reduces the T cell response against self-antigens and prevents autoimmunity [3]. This antibody prevents cytotoxic T cell down regulation [37]. Clinical trials using ipilimumab in melanoma cases have shown improved survival rates [38], which have been estimated to be 49-60% [39,40].

Demerits of Immune Checkpoint Therapy

In cases with a low positive or negative rate of these receptors or ligands, immune checkpoint therapy is not effective. It is important to study positive rates of immune checkpoint molecules before therapy using these agents, while keeping in mind that treatment for patients without immune checkpoint receptors or ligands remains an issue of concern.

The most frequent adverse effects of immune checkpoint therapy are skin rashes, followed by gastrointestinal symptoms; other adverse effects include endocrine, liver, pulmonary and general symptoms [18,20,37]. Select adverse events in clinical trials occurred in 40-60% of patients, and 3-20% of these cases were grade 3 or 4 [37].

Combination therapy using immune checkpoint agents could produce more effective results, but adverse effects are more common than when using a single agent [41]. Using a concurrent regimen, Jedd [41] reported grade 3 or 4 adverse effects in 53% of patients. The rates of adverse effects when using only ipilimumab or nivolumab are reported to be 20% and 15%, respectively [38]. Kourie [42] reports that combination therapy using many checkpoint inhibitors was associated with high response rates (>50%) in melanomas, but that 30% of patients had to discontinue combination therapy using nivolumab and ipilimumab because of high toxicity.

In the US, the average annual number of adults treated for skin cancer and the annual cost for skin cancer increased from 2002 to 2011 [43], and these numbers are likewise increasing in other countries as well. The costs of immune checkpoint therapy are huge, and those of combination therapies are even higher. Therefore, eligibility for this therapy should be determined based on disease severity, and the kinds and rates of positive immune checkpoints. The cost of using just one immune checkpoint therapy, nivolumab, is at least a few million yen per month in Japan, and almost all costs (about 97-98%) are covered by insurance for high medical expenses and by the national budget. Therefore, both in Japan and other countries, national health budgets and medical insurance companies could go bankrupt if all candidate cases are treated with immune checkpoint therapies. Moreover, the cost of immune checkpoint therapies is further increased when treatment is required for adverse effects.

References

  1. Sullivan RJ, Lorusso PM, Flaherty KT (2013) The intersection of immune-directed and molecularly targeted therapy in advanced melanoma: where we have been, are, and will be. Clin Cancer Res 19: 5283-5291.
  2. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, et al. (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab J Clin Oncol  32: 1020-1030.
  3. Fife BT, Bluestone JA (2008) Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev 224: 166-182.
  4. Zou W, Chen L (2008) Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 8: 467-477.
  5. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, et al. (2005) Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res 65: 1089-1096.
  6. Davies M (2014) New modalities of cancer treatment for NSCLC: focus on immunotherapy. Cancer Manag Res 6: 63-75.
  7. Dong H, Zhu G, Tamada K, Chen L (1999) B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med 5: 1365-1369.
  8. Tseng SY, Otsuji M, Gorski K, Huang X, Slansky JE, et al. (2001) B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J Exp Med 193: 839-846.
  9. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, et al. (2001) PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2: 261-268.
  10. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, et al. (2000) Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192: 1027-1034.
  11. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, et al. (2002) Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8: 793-800.
  12. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, et al. (2002) Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci 99: 12293-12297.
  13. Flies DB, Sandler BJ, Sznol M, Chen L (2011) Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy. Yale J Biol Med 84: 409-421.
  14. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, et al. (2003) Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 9: 562-567.
  15. Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, et al. (2010) Tumor cell expression of programmed celldeath-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116: 1757-1766.
  16. Menzies AM, Long GV (2013) New combinations and immunotherapies for melanoma: latest evidence and clinical utility. Ther Adv Med Oncol 5: 278-285.
  17. Brahmer JR, Hammers H, Lipson EJ (2015) Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol 11: 1307-1326.
  18. Ascierto PA, Kalos M, Schaer DA, Callahan MK, Wolchok JD (2013) Biomarkers for immunostimulatory monoclonal antibodies in combination strategies for melanoma and other tumor types. Clin Cancer Res 19: 1009-1020.
  19. Kirkwood JM (2012) Immunotherapy of cancer in 2012. CA Cancer J Clin 2012
  20. Okazaki T, Honjo T (2006) The PD-1-PD-L pathway in immunological tolerance. Trends Immunol 27: 195-201.
  21. Wang L, Pino-Lagos K, de Vries VC, Guleria I, Sayegh MH, et al. (2008) Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc Natl Acad Sci 105: 9331-9336.
  22. Weber JS, Kudchadkar RR, Yu B, Gallenstein D, Horak CE, et al. (2013) Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol 31: 4311-4318.
  23. Greenwald RJ, Freeman GJ, Sharpe AH (2005) The B7 family revisited. Annu Rev Immunol 23: 515-548.
  24. Butte MJ, Peña-Cruz V, Kim MJ, Freeman GJ, Sharpe AH (2008) Interaction of human PD-L1 and B7-1. Mol Immunol 45: 3567-3572.
  25. Johnson DB, Peng C, Sosman JA (2015) Nivolumab in melanoma: latest evidence and clinical potential. Ther Adv Med Oncol 7: 97-106.
  26. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med  366: 2443-2454.
  27. Petroff MG, Chen L, Phillips TA, Azzola D, Sedlmayr P, et al. (2003) B7 family molecules are favourably positioned at the human maternal-fetal interface. Biol Reprod 68: 1496-1504.
  28. Kim J, Myers AC, Chen L, Pardoll DM, Truong-Tran QA, et al. (2005) Constitutive and inducible expression of b7 family of ligands by human airway epithelial cells. Am J Respir Cell Mol Biol 33: 280-289.
  29. Taube JM, Anders RA, Young GD, Xu H, Sharma R, et al. (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med  4: 127ra37.
  30. Gordon MS, Hamid O, Powderly J et al. (2013) A phase 1 study of MPDL3280A, anengineered PD-L1 natibody in patients with locally advanced or metastatic tumors. Presented at the Annual Meeting of the American Association of Cancer Research, Washington, DC, April 6-10, 2013.
  31. Stephen Hodi F, Sznol M, Kluger HM, McDermott DF, Carvajal RD, et al. (2014) Long-term survival of ipilimumab-naive patients (pts) with advanced melanoma (MEL) treated with nivolmab (anti-PD-1; BMS-936558; ONO-4538) in Phase 1 trial. J Clin Oncol  32.
  32. Kiyohara Y, Tahara H, Uhara H, Moroi Y, Yamazaki N, et al. (2014) Long-term survival of patients with advanced melanoma in Phase 2 study of nivolumab (anti-PD-!, ONO-4538/BMS-936558). Ann Oncol  25: iv379-iv380.
  33. Iannone R, Gergich K, Cong C, et al. (2012) Efficacy and safety of MK-3475 in patients with advanced melanoma. Pigmented cell Melanoma Res 25: 864.
  34. Hamid O, Sosman JA, Lawrence DP, Sullivan RJ, Ibrahim N, et al. (2013) Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). J Clin Oncol : 31.
  35. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366: 2455-2465.
  36. Davies M (2014) New modalities of cancer treatment for NSCLC: focus on immunotherapy. Cancer Manag Res 6: 63-75.
  37. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010; 363(8):711-723.
  38. Margolin KA, Wong SL, Penrod JR, et al. (2013) Effectiveness and safety of first-line ipilimumab 3 mg/kg therapy for advanced melanoma: evidence from a US multisite retrospective chart review. Poster 3742 presented at the European Cancer Congress, September 27 to October 1, 2013, Amsterdam, The Netherlands.
  39. Patt D, Juday T, Prnrod JR, Chen C, Wong SL (2013) A community-based, real-word, study of treatment-naïve advanced melanoma (AM) patients treated with 3 mg/kg ipilimumab (IPI) in the United States. Poster 3751 presented at the European Cancer Congress, Amsterdam, The Netherlands.
  40. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, et al. (2013) Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013; 369(2):122-133.
  41. Kourie HR, Klastersky JA (2016) Side-effects of checkpoint inhibitor-based combination therapy. Curr Opin Oncol 28: 306-313.
  42. Guy GP Jr, Machlin SR, Ekwueme DU, Yabroff KR (2015) Prevalence and costs of skin cancer treatment in the U.S., 2002-2006 and 2007-2011. Am J Prev Med 48: 183-187.
Citation: Sugiura K, Sugiura M (2016) Immune Checkpoint Blockade Therapy: Merits and Demerits. J Clin Exp Dermatol Res 7:372.

Copyright: © 2016 Sugiura K, et al. 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.
Top
https://www.olimpbase.org/1937/