Skip to main content
Top
Published in: Medical Oncology 11/2019

Open Access 01-11-2019 | Liver Transplantation | Review Article

Liver graft rejection following immune checkpoint inhibitors treatment: a review

Authors: Bo Hu, Xiao-Bo Yang, Xin-Ting Sang

Published in: Medical Oncology | Issue 11/2019

Login to get access

Abstract

Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a variety of solid tumors; nonetheless, they have not been well investigated and are still recognized as a relative contraindication for patients with a liver transplantation (LT) history, since ICIs treatment might potentially lead to graft rejection. The program death-1 (PD-1) and the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways are implicated in the tolerance of transplanted organ, as well as blockade of the pathways, which contribute to eliminating tumors and may inadvertently lead to peripheral transplant rejection. Currently, no guidelines are available regarding the treatment for ICIs patients with a prior LT history. Therefore, this study was carried out to review the recent studies, attempting to introduce the ICIs-related graft rejection after LT from various aspects. We believed that ICIs could be given for the well-informed patients receiving LT and developed recurrence in a controlled setting. Typically, these patients should be treated according to a clinical care path or a prospective clinical trial, so as obtain a persistent anti-tumor immune response in the meantime of avoiding graft rejection, adjust the immunosuppression, reduce the possibility of graft loss following rejection, and have the opportunity to develop biomarkers for tumor response and transplant rejection.
Literature
1.
go back to reference Kuo JC, Lilly LB, Hogg D. Immune checkpoint inhibitor therapy in a liver transplant recipient with a rare subtype of melanoma: a case report and literature review. Melanoma Res. 2018;28(1):61–4.PubMed Kuo JC, Lilly LB, Hogg D. Immune checkpoint inhibitor therapy in a liver transplant recipient with a rare subtype of melanoma: a case report and literature review. Melanoma Res. 2018;28(1):61–4.PubMed
2.
go back to reference Munker S, De Toni EN. Use of checkpoint inhibitors in liver transplant recipients. United Eur Gastroenterol J. 2018;6(7):970–3. Munker S, De Toni EN. Use of checkpoint inhibitors in liver transplant recipients. United Eur Gastroenterol J. 2018;6(7):970–3.
3.
go back to reference Jackson C, Ruzevick J, Phallen J, et al. Challenges in immunotherapy presented by the glioblastoma multiforme microenvironment. Clin Dev Immunol. 2011;1:732413. Jackson C, Ruzevick J, Phallen J, et al. Challenges in immunotherapy presented by the glioblastoma multiforme microenvironment. Clin Dev Immunol. 2011;1:732413.
4.
go back to reference Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer. 2019;145:639–48.PubMed Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer. 2019;145:639–48.PubMed
5.
go back to reference Chen C, Zhang F, Zhou N, et al. Efficacy and safety of immune checkpoint inhibitors in advanced gastric or gastroesophageal junction cancer: a systematic review and meta-analysis. OncoImmunology. 2019;8(5):e1581547.PubMed Chen C, Zhang F, Zhou N, et al. Efficacy and safety of immune checkpoint inhibitors in advanced gastric or gastroesophageal junction cancer: a systematic review and meta-analysis. OncoImmunology. 2019;8(5):e1581547.PubMed
6.
go back to reference Pundole X, Abdel-Wahab N, Suarez-Almazor ME. Arthritis risk with immune checkpoint inhibitor therapy for cancer. Curr Opinion Rheumatol. 2019;31(3):293–9. Pundole X, Abdel-Wahab N, Suarez-Almazor ME. Arthritis risk with immune checkpoint inhibitor therapy for cancer. Curr Opinion Rheumatol. 2019;31(3):293–9.
7.
go back to reference Som A, Mandaliya R, Alsaadi D, et al. Immune checkpoint inhibitor-induced colitis: a comprehensive review. World J Clin Cases. 2019;7(4):405–18.PubMedPubMedCentral Som A, Mandaliya R, Alsaadi D, et al. Immune checkpoint inhibitor-induced colitis: a comprehensive review. World J Clin Cases. 2019;7(4):405–18.PubMedPubMedCentral
8.
go back to reference Jennings JJ, Mandaliya R, Nakshabandi A, et al. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opinion Drug Metab Toxicol. 2019;15:231–44. Jennings JJ, Mandaliya R, Nakshabandi A, et al. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opinion Drug Metab Toxicol. 2019;15:231–44.
9.
go back to reference Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer. 2018;118(1):1–8. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer. 2018;118(1):1–8.
10.
go back to reference Temel JS, Gainor JF, Sullivan RJ, et al. Keeping expectations in check with immune checkpoint inhibitors. J Clin Oncol. 2018;36(17):1654–7.PubMed Temel JS, Gainor JF, Sullivan RJ, et al. Keeping expectations in check with immune checkpoint inhibitors. J Clin Oncol. 2018;36(17):1654–7.PubMed
11.
go back to reference Thallinger C, Thorsten F, Preusser M, et al. Review of cancer treatment with immune checkpoint inhibitors: current concepts expectations limitations and pitfalls. Wien Klin Wochenschr. 2017;130(164):85–91.PubMedPubMedCentral Thallinger C, Thorsten F, Preusser M, et al. Review of cancer treatment with immune checkpoint inhibitors: current concepts expectations limitations and pitfalls. Wien Klin Wochenschr. 2017;130(164):85–91.PubMedPubMedCentral
12.
go back to reference Yu Y. Molecular classification and precision therapy of cancer: immune checkpoint inhibitors. Front Med. 2018;12(2):229–35.PubMed Yu Y. Molecular classification and precision therapy of cancer: immune checkpoint inhibitors. Front Med. 2018;12(2):229–35.PubMed
13.
go back to reference Krenzien F, Schmelzle M, Struecker B, et al. Liver transplantation and liver resection for cirrhotic patients with hepatocellular carcinoma: comparison of long-term survivals. J Gastrointest Surg. 2018;22:840–8.PubMed Krenzien F, Schmelzle M, Struecker B, et al. Liver transplantation and liver resection for cirrhotic patients with hepatocellular carcinoma: comparison of long-term survivals. J Gastrointest Surg. 2018;22:840–8.PubMed
14.
go back to reference Nasralla D, Coussios CC, Mergental H, et al. A randomized trial of normothermic preservation in liver transplantation. Nature. 2018;557:50.PubMed Nasralla D, Coussios CC, Mergental H, et al. A randomized trial of normothermic preservation in liver transplantation. Nature. 2018;557:50.PubMed
15.
go back to reference She WH, Chan ACY, Cheung TT, et al. Survival outcomes of liver transplantation for hepatocellular carcinoma in patients with normal, high and very high preoperative alpha-fetoprotein levels. World J Hepatol. 2018;10(2):308.PubMedPubMedCentral She WH, Chan ACY, Cheung TT, et al. Survival outcomes of liver transplantation for hepatocellular carcinoma in patients with normal, high and very high preoperative alpha-fetoprotein levels. World J Hepatol. 2018;10(2):308.PubMedPubMedCentral
16.
go back to reference Mogul DB, Luo X, Garonzik-Wang J, Bowring MG, Massie AB, Schwarz KB, et al. Expansion of the liver donor supply through greater use of split-liver transplantation: identifying optimal recipients. Liver Transplant. 2019;25(1):119–27. Mogul DB, Luo X, Garonzik-Wang J, Bowring MG, Massie AB, Schwarz KB, et al. Expansion of the liver donor supply through greater use of split-liver transplantation: identifying optimal recipients. Liver Transplant. 2019;25(1):119–27.
17.
go back to reference Sapisochin G, Bruix J. Liver transplantation for hepatocellular carcinoma: outcomes and novel surgical approaches. Nat Rev Gastroenterol Hepatol. 2017;14(4):203–17.PubMed Sapisochin G, Bruix J. Liver transplantation for hepatocellular carcinoma: outcomes and novel surgical approaches. Nat Rev Gastroenterol Hepatol. 2017;14(4):203–17.PubMed
18.
go back to reference Kotlyar DS, Campbell MS, Reddy KR. Recurrence of diseases following orthotopic liver transplantation. Am J Gastroenterol. 2006;101(6):1370.PubMed Kotlyar DS, Campbell MS, Reddy KR. Recurrence of diseases following orthotopic liver transplantation. Am J Gastroenterol. 2006;101(6):1370.PubMed
19.
go back to reference Carrión JA, Navasa M, García-Retortillo M, et al. Efficacy of antiviral therapy on hepatitis C recurrence after liver transplantation: a randomized controlled study. Gastroenterology. 2007;132(5):1746–56.PubMed Carrión JA, Navasa M, García-Retortillo M, et al. Efficacy of antiviral therapy on hepatitis C recurrence after liver transplantation: a randomized controlled study. Gastroenterology. 2007;132(5):1746–56.PubMed
20.
go back to reference Blasco A, Forns X, Carrión JA, García-Pagán JC, Gilabert R, Rimola A, et al. Hepatic venous pressure gradient identifies patients at risk of severe hepatitis C recurrence after liver transplantation. Hepatology. 2006;43(3):492–9.PubMed Blasco A, Forns X, Carrión JA, García-Pagán JC, Gilabert R, Rimola A, et al. Hepatic venous pressure gradient identifies patients at risk of severe hepatitis C recurrence after liver transplantation. Hepatology. 2006;43(3):492–9.PubMed
21.
go back to reference Lai MC, Yang Z, Zhou L, et al. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 2012;29(3):1810–6.PubMed Lai MC, Yang Z, Zhou L, et al. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 2012;29(3):1810–6.PubMed
22.
go back to reference Testa G, Crippin JS, Netto GJ, et al. Liver transplantation for hepatitis C: Recurrence and disease progression in 300 patients. Liver Transpl. 2000;6(5):553–61.PubMed Testa G, Crippin JS, Netto GJ, et al. Liver transplantation for hepatitis C: Recurrence and disease progression in 300 patients. Liver Transpl. 2000;6(5):553–61.PubMed
23.
go back to reference Filgueira NA. Hepatocellular carcinoma recurrence after liver transplantation: risk factors, screening and clinical presentation. World J Hepatol. 2019;11(3):261–72.PubMedPubMedCentral Filgueira NA. Hepatocellular carcinoma recurrence after liver transplantation: risk factors, screening and clinical presentation. World J Hepatol. 2019;11(3):261–72.PubMedPubMedCentral
24.
go back to reference Samuel R, Bilal M, Nawgiri R, et al. Recurrence of hepatocellular carcinoma at the porta-hepatis following liver transplantation diagnosed on EUS-FNA. Clin J Gastroenterol. 2019;12:336–40.PubMed Samuel R, Bilal M, Nawgiri R, et al. Recurrence of hepatocellular carcinoma at the porta-hepatis following liver transplantation diagnosed on EUS-FNA. Clin J Gastroenterol. 2019;12:336–40.PubMed
25.
go back to reference Li DB, Si XY, Wang SJ, et al. Long-term outcomes of combined hepatocellular-cholangiocarcinoma after hepatectomy or liver transplantation: a systematic review and meta-analysis. Hepatobiliary Pancreat Dis Int. 2018;18:12–8.PubMed Li DB, Si XY, Wang SJ, et al. Long-term outcomes of combined hepatocellular-cholangiocarcinoma after hepatectomy or liver transplantation: a systematic review and meta-analysis. Hepatobiliary Pancreat Dis Int. 2018;18:12–8.PubMed
26.
go back to reference Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.PubMed Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.PubMed
27.
go back to reference Riella LV, Paterson AM, Sharpe AH, et al. Role of the PD-1 pathway in the immune response. Am J Transplant. 2012;12(10):2575–87.PubMedPubMedCentral Riella LV, Paterson AM, Sharpe AH, et al. Role of the PD-1 pathway in the immune response. Am J Transplant. 2012;12(10):2575–87.PubMedPubMedCentral
28.
go back to reference Dong Y, Sun Q, Zhang X. PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget. 2017;8(2):2171.PubMed Dong Y, Sun Q, Zhang X. PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget. 2017;8(2):2171.PubMed
29.
go back to reference Ito T, Ueno T, Clarkson MR, et al. Analysis of the role of negative T cell costimulatory pathways in CD4 and CD8 T cell-mediated alloimmune responses in vivo. J Immunol. 2005;174(11):6648–56.PubMed Ito T, Ueno T, Clarkson MR, et al. Analysis of the role of negative T cell costimulatory pathways in CD4 and CD8 T cell-mediated alloimmune responses in vivo. J Immunol. 2005;174(11):6648–56.PubMed
30.
go back to reference Tanaka K, Albin MJ, Yuan X, et al. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J Immunol. 2007;179(8):5204–10.PubMedPubMedCentral Tanaka K, Albin MJ, Yuan X, et al. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J Immunol. 2007;179(8):5204–10.PubMedPubMedCentral
31.
go back to reference Morita M, Fujino M, Jiang G, et al. PD-1/B7-H1 interaction contribute to the spontaneous acceptance of mouse liver allograft. Am J Transplant. 2010;10(1):40–6.PubMed Morita M, Fujino M, Jiang G, et al. PD-1/B7-H1 interaction contribute to the spontaneous acceptance of mouse liver allograft. Am J Transplant. 2010;10(1):40–6.PubMed
32.
go back to reference Wang L, Han R, Hancock WW. Programmed cell death 1 (PD-1) and its ligand PD-L1 are required for allograft tolerance. Eur J Immunol. 2007;37(10):2983–90.PubMed Wang L, Han R, Hancock WW. Programmed cell death 1 (PD-1) and its ligand PD-L1 are required for allograft tolerance. Eur J Immunol. 2007;37(10):2983–90.PubMed
33.
go back to reference Riella LV, Watanabe T, Sage PT, et al. Essential role of PDL1 expression on nonhematopoietic donor cells in acquired tolerance to vascularized cardiac allografts. Am J Transplant. 2011;11(4):832–40.PubMed Riella LV, Watanabe T, Sage PT, et al. Essential role of PDL1 expression on nonhematopoietic donor cells in acquired tolerance to vascularized cardiac allografts. Am J Transplant. 2011;11(4):832–40.PubMed
34.
go back to reference Starke A, Lindenmeyer MT, Segerer S, et al. Renal tubular PD-L1 (CD274) suppresses alloreactive human T-cell responses. Kidney Int. 2010;78(1):38–47.PubMed Starke A, Lindenmeyer MT, Segerer S, et al. Renal tubular PD-L1 (CD274) suppresses alloreactive human T-cell responses. Kidney Int. 2010;78(1):38–47.PubMed
35.
go back to reference Judge TA, Wu Z, Zheng XG, et al. The role of CD80, CD86, and CTLA4 in alloimmune responses and the induction of long-term allograft survival. J Immunol. 1999;162(4):1947–51.PubMed Judge TA, Wu Z, Zheng XG, et al. The role of CD80, CD86, and CTLA4 in alloimmune responses and the induction of long-term allograft survival. J Immunol. 1999;162(4):1947–51.PubMed
36.
go back to reference Zhang T, Fresnay S, Welty E, et al. Selective CD28 blockade attenuates acute and chronic rejection of murine cardiac allografts in a CTLA-4-dependent manner. Am J Transplant. 2011;11(8):1599–609.PubMedPubMedCentral Zhang T, Fresnay S, Welty E, et al. Selective CD28 blockade attenuates acute and chronic rejection of murine cardiac allografts in a CTLA-4-dependent manner. Am J Transplant. 2011;11(8):1599–609.PubMedPubMedCentral
37.
go back to reference Lin H, Rathmell JC, Gray GS, et al. Cytotoxic T lymphocyte antigen 4 (CTLA4) blockade accelerates the acute rejection of cardiac allografts in CD28-deficient mice: CTLA4 can function independently of CD28. J Exp Med. 1998;188(1):199–204.PubMedPubMedCentral Lin H, Rathmell JC, Gray GS, et al. Cytotoxic T lymphocyte antigen 4 (CTLA4) blockade accelerates the acute rejection of cardiac allografts in CD28-deficient mice: CTLA4 can function independently of CD28. J Exp Med. 1998;188(1):199–204.PubMedPubMedCentral
38.
go back to reference Li W, Zheng XX, Kuhr CS, et al. CTLA4 engagement is required for induction of murine liver transplant spontaneous tolerance. Am J Transplant. 2005;5(5):978–86.PubMed Li W, Zheng XX, Kuhr CS, et al. CTLA4 engagement is required for induction of murine liver transplant spontaneous tolerance. Am J Transplant. 2005;5(5):978–86.PubMed
39.
go back to reference Wanchoo R, Riella LV, Uppal NN, et al. Immune checkpoint inhibitors in the cancer patient with an organ transplant. J Onco-Nephrol. 2017;1(1):42–8. Wanchoo R, Riella LV, Uppal NN, et al. Immune checkpoint inhibitors in the cancer patient with an organ transplant. J Onco-Nephrol. 2017;1(1):42–8.
40.
go back to reference Ijaz A, Khan AY, Malik SU, et al. Significant risk of graft-versus-host disease with exposure to checkpoint inhibitors before and after allogeneic transplantation. Biol Blood Marrow Transplant. 2019;25(1):94–9.PubMed Ijaz A, Khan AY, Malik SU, et al. Significant risk of graft-versus-host disease with exposure to checkpoint inhibitors before and after allogeneic transplantation. Biol Blood Marrow Transplant. 2019;25(1):94–9.PubMed
41.
go back to reference Gassmann D, Weiler S, Mertens JC, et al. Liver allograft failure after nivolumab treatment—a case report with systematic literature research. Transplant Direct. 2018;4(8):e376.PubMedPubMedCentral Gassmann D, Weiler S, Mertens JC, et al. Liver allograft failure after nivolumab treatment—a case report with systematic literature research. Transplant Direct. 2018;4(8):e376.PubMedPubMedCentral
42.
go back to reference Friend BD, Venick RS, McDiarmid SV, et al. Fatal orthotopic liver transplant organ rejection induced by a checkpoint inhibitor in two patients with refractory, metastatic hepatocellular carcinoma. Pediatr Blood Cancer. 2017;64(12):e26682. Friend BD, Venick RS, McDiarmid SV, et al. Fatal orthotopic liver transplant organ rejection induced by a checkpoint inhibitor in two patients with refractory, metastatic hepatocellular carcinoma. Pediatr Blood Cancer. 2017;64(12):e26682.
43.
go back to reference Boils CL, Aljadir DN, Cantafio AW. Use of the PD-1 pathway inhibitor nivolumab in a renal transplant patient with malignancy. Am J Transplant. 2016;16(8):2496–7.PubMed Boils CL, Aljadir DN, Cantafio AW. Use of the PD-1 pathway inhibitor nivolumab in a renal transplant patient with malignancy. Am J Transplant. 2016;16(8):2496–7.PubMed
44.
go back to reference Lipson EJ, Bagnasco SM, Moore J Jr, et al. Tumor regression and allograft rejection after administration of anti-PD-1. N Engl J Med. 2016;374(9):896–8.PubMedPubMedCentral Lipson EJ, Bagnasco SM, Moore J Jr, et al. Tumor regression and allograft rejection after administration of anti-PD-1. N Engl J Med. 2016;374(9):896–8.PubMedPubMedCentral
45.
go back to reference Ong M, Ibrahim AM, Bourassa-Blanchette S, et al. Antitumor activity of nivolumab on hemodialysis after renal allograft rejection. J Immunother Cancer. 2016;4(1):64.PubMedPubMedCentral Ong M, Ibrahim AM, Bourassa-Blanchette S, et al. Antitumor activity of nivolumab on hemodialysis after renal allograft rejection. J Immunother Cancer. 2016;4(1):64.PubMedPubMedCentral
46.
go back to reference Owonikoko TK, Kumar M, Yang S, et al. Cardiac allograft rejection as a complication of PD-1 checkpoint blockade for cancer immunotherapy: a case report. Cancer Immunol Immunother. 2017;66(1):45–50.PubMed Owonikoko TK, Kumar M, Yang S, et al. Cardiac allograft rejection as a complication of PD-1 checkpoint blockade for cancer immunotherapy: a case report. Cancer Immunol Immunother. 2017;66(1):45–50.PubMed
47.
go back to reference Spain L, Higgins R, Gopalakrishnan K, et al. Acute renal allograft rejection after immune checkpoint inhibitor therapy for metastatic melanoma. Ann Oncol. 2016;27(6):1135–7.PubMed Spain L, Higgins R, Gopalakrishnan K, et al. Acute renal allograft rejection after immune checkpoint inhibitor therapy for metastatic melanoma. Ann Oncol. 2016;27(6):1135–7.PubMed
48.
go back to reference Tio M, Rai R, Ezeoke OM, et al. Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection. Eur J Cancer. 2018;104:137–44.PubMed Tio M, Rai R, Ezeoke OM, et al. Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection. Eur J Cancer. 2018;104:137–44.PubMed
49.
go back to reference Kittai AS, Oldham H, Cetnar J, et al. Immune checkpoint inhibitors in organ transplant patients. J Immunother. 2017;40(7):277–81.PubMed Kittai AS, Oldham H, Cetnar J, et al. Immune checkpoint inhibitors in organ transplant patients. J Immunother. 2017;40(7):277–81.PubMed
50.
go back to reference DeLeon TT, Salomao MA, Aqel BA, et al. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: the Mayo Clinic experience. J Gastrointest Oncol. 2018;9(6):1054.PubMedPubMedCentral DeLeon TT, Salomao MA, Aqel BA, et al. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: the Mayo Clinic experience. J Gastrointest Oncol. 2018;9(6):1054.PubMedPubMedCentral
51.
go back to reference Ranganath HA, Panella TJ. Administration of ipilimumab to a liver transplant recipient with unresectable metastatic melanoma. J Immunother. 2015;38(5):211.PubMed Ranganath HA, Panella TJ. Administration of ipilimumab to a liver transplant recipient with unresectable metastatic melanoma. J Immunother. 2015;38(5):211.PubMed
52.
go back to reference De Bruyn P, Van Gestel D, Ost P, et al. Immune checkpoint blockade for organ transplant patients with advanced cancer: how far can we go? Curr Opin Oncol. 2019;31(2):54–64.PubMed De Bruyn P, Van Gestel D, Ost P, et al. Immune checkpoint blockade for organ transplant patients with advanced cancer: how far can we go? Curr Opin Oncol. 2019;31(2):54–64.PubMed
53.
go back to reference Morales RE, Shoushtari AN, Walsh MM, et al. Safety and efficacy of ipilimumab to treat advanced melanoma in the setting of liver transplantation. J Immunother Cancer. 2015;3(1):22.PubMedPubMedCentral Morales RE, Shoushtari AN, Walsh MM, et al. Safety and efficacy of ipilimumab to treat advanced melanoma in the setting of liver transplantation. J Immunother Cancer. 2015;3(1):22.PubMedPubMedCentral
54.
go back to reference Schvartsman G, Perez K, Sood G, et al. Immune checkpoint inhibitor therapy in a liver transplant recipient with melanoma. Ann Intern Med. 2017;167(5):361–2.PubMed Schvartsman G, Perez K, Sood G, et al. Immune checkpoint inhibitor therapy in a liver transplant recipient with melanoma. Ann Intern Med. 2017;167(5):361–2.PubMed
55.
go back to reference Biondani P, De Martin E, Samuel D. Safety of an anti-PD-1 immune checkpoint inhibitor in a liver transplant recipient. Ann Oncol. 2018;29:286–7.PubMed Biondani P, De Martin E, Samuel D. Safety of an anti-PD-1 immune checkpoint inhibitor in a liver transplant recipient. Ann Oncol. 2018;29:286–7.PubMed
56.
go back to reference Rammohan A, Reddy MS, Farouk M, et al. Pembrolizumab for metastatic hepatocellular carcinoma following live donor liver transplantation: the silver bullet? Hepatology. 2018;67(3):1166–8.PubMed Rammohan A, Reddy MS, Farouk M, et al. Pembrolizumab for metastatic hepatocellular carcinoma following live donor liver transplantation: the silver bullet? Hepatology. 2018;67(3):1166–8.PubMed
57.
go back to reference Davids MS, Kim HT, Costello C, et al. Optimizing checkpoint blockade as a treatment for relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. Blood. 2017;130:275. Davids MS, Kim HT, Costello C, et al. Optimizing checkpoint blockade as a treatment for relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. Blood. 2017;130:275.
58.
go back to reference Bashey A, Medina B, Corringham S, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113(7):1581–8.PubMedPubMedCentral Bashey A, Medina B, Corringham S, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113(7):1581–8.PubMedPubMedCentral
59.
go back to reference Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for patients with relapse after allogeneic transplantation. N Engl J Med. 2016;375(2):143–53.PubMedPubMedCentral Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for patients with relapse after allogeneic transplantation. N Engl J Med. 2016;375(2):143–53.PubMedPubMedCentral
60.
go back to reference Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood. 2017;129(18):2471–8.PubMed Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood. 2017;129(18):2471–8.PubMed
61.
go back to reference Dueland S, Guren TK, Boberg KM, et al. Acute liver graft rejection after ipilimumab therapy. Ann Oncol. 2017;28(10):2619–20.PubMed Dueland S, Guren TK, Boberg KM, et al. Acute liver graft rejection after ipilimumab therapy. Ann Oncol. 2017;28(10):2619–20.PubMed
62.
go back to reference Varkaris A, Lewis DW, Nugent FW. Preserved liver transplant after PD-1 pathway inhibitor for hepatocellular carcinoma. Am J Gastroenterol. 2017;112(12):1895.PubMed Varkaris A, Lewis DW, Nugent FW. Preserved liver transplant after PD-1 pathway inhibitor for hepatocellular carcinoma. Am J Gastroenterol. 2017;112(12):1895.PubMed
63.
go back to reference De Toni EN, Gerbes AL. Tapering of immunosuppression and sustained treatment with nivolumab in a liver transplant recipient. Gastroenterology. 2017;152(6):1631–3.PubMed De Toni EN, Gerbes AL. Tapering of immunosuppression and sustained treatment with nivolumab in a liver transplant recipient. Gastroenterology. 2017;152(6):1631–3.PubMed
65.
go back to reference Haverkos BM, Abbott D, Hamadani M, et al. PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. Blood. 2017;130(2):221–8.PubMedPubMedCentral Haverkos BM, Abbott D, Hamadani M, et al. PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. Blood. 2017;130(2):221–8.PubMedPubMedCentral
66.
go back to reference Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors: a review. JAMA Oncol. 2016;2(10):1346–53.PubMed Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors: a review. JAMA Oncol. 2016;2(10):1346–53.PubMed
67.
go back to reference Jennings JJ, Mandaliya R, Nakshabandi A, et al. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opinion Drug Metab Toxicol. 2019;15(3):231–44. Jennings JJ, Mandaliya R, Nakshabandi A, et al. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opinion Drug Metab Toxicol. 2019;15(3):231–44.
68.
go back to reference Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4(5):560.PubMedPubMedCentral Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4(5):560.PubMedPubMedCentral
69.
go back to reference Schmitt TM, Phillips M, Sawyer RG, et al. Anti-thymocyte globulin for the treatment of acute cellular rejection following liver transplantation. Dig Dis Sci. 2010;55(11):3224–34.PubMed Schmitt TM, Phillips M, Sawyer RG, et al. Anti-thymocyte globulin for the treatment of acute cellular rejection following liver transplantation. Dig Dis Sci. 2010;55(11):3224–34.PubMed
70.
go back to reference Kamar N, Lavayssiere L, Muscari F, et al. Early plasmapheresis and rituximab for acute humoral rejection after ABO-compatible liver transplantation. World J Gastroenterol. 2009;15(27):3426.PubMedPubMedCentral Kamar N, Lavayssiere L, Muscari F, et al. Early plasmapheresis and rituximab for acute humoral rejection after ABO-compatible liver transplantation. World J Gastroenterol. 2009;15(27):3426.PubMedPubMedCentral
71.
go back to reference Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275.PubMedPubMedCentral Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275.PubMedPubMedCentral
72.
go back to reference Garraway LA. Genomics-driven oncology: framework for an emerging paradigm. J Clin Oncol. 2013;31(15):1806–14.PubMed Garraway LA. Genomics-driven oncology: framework for an emerging paradigm. J Clin Oncol. 2013;31(15):1806–14.PubMed
73.
go back to reference Sumimoto H, Imabayashi F, Iwata T, et al. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med. 2006;203(7):1651–6.PubMedPubMedCentral Sumimoto H, Imabayashi F, Iwata T, et al. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med. 2006;203(7):1651–6.PubMedPubMedCentral
74.
go back to reference Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74.PubMed Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74.PubMed
75.
go back to reference Ngiow SF, Von Scheidt B, Akiba H, et al. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Can Res. 2011;71(10):3540–51. Ngiow SF, Von Scheidt B, Akiba H, et al. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Can Res. 2011;71(10):3540–51.
77.
go back to reference Deaglio S, Dwyer KM, Gao W, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–65.PubMedPubMedCentral Deaglio S, Dwyer KM, Gao W, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–65.PubMedPubMedCentral
78.
go back to reference Zarek PE, Huang CT, Lutz ER, et al. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood. 2008;111(1):251–9.PubMedPubMedCentral Zarek PE, Huang CT, Lutz ER, et al. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood. 2008;111(1):251–9.PubMedPubMedCentral
Metadata
Title
Liver graft rejection following immune checkpoint inhibitors treatment: a review
Authors
Bo Hu
Xiao-Bo Yang
Xin-Ting Sang
Publication date
01-11-2019

Other articles of this Issue 11/2019

Medical Oncology 11/2019 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.