Skip to main content
Top
Published in: Current Hematologic Malignancy Reports 1/2017

01-02-2017 | Stem Cell Transplantation (R Maziarz, Section Editor)

Immune Checkpoint Blockade and Hematopoietic Stem Cell Transplant

Authors: Reid W. Merryman, Philippe Armand

Published in: Current Hematologic Malignancy Reports | Issue 1/2017

Login to get access

Abstract

Allogeneic hematopoietic stem cell transplant (HSCT) relies primarily upon graft-versus-tumor activity for cancer eradication. Relapse remains the principal cause of treatment failure after HSCT, implying frequent immune escape, which in at least some cases, appears to be mediated by increased expression of inhibitory immune checkpoints. In an attempt to restore anti-tumor immunity, checkpoint blockade therapy (CBT) targeting PD-1 and CLTA-4 has been used in conjunction with both allogeneic and autologous HSCT. Clinical experience in this setting is limited to several small clinical trials and case series, but together they suggest that treatment with CBT can effectively amplify anti-tumor immune responses. However, intrinsic to its mechanism is also the risk that CBT in the HSCT setting may also cause significant immune toxicity. Fatal immune-related adverse events and graft-versus-host disease have been observed, but in most cases, immune side effects appear to be reversible with steroids and CBT discontinuation. As clinical investigation continues, improved understanding of immune checkpoint biology will be critical to optimize safe and efficacious treatment strategies.
Literature
1.
go back to reference Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.PubMed Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.PubMed
2.
go back to reference Norde WJ, Maas F, Hobo W, et al. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. Cancer Res. 2011;71(15):5111–22.CrossRefPubMed Norde WJ, Maas F, Hobo W, et al. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. Cancer Res. 2011;71(15):5111–22.CrossRefPubMed
3.
go back to reference Iwai Y, Ishida M, Tanaka Y, et al. 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 U S A. 2002;99(19):12293–7.CrossRefPubMedPubMedCentral Iwai Y, Ishida M, Tanaka Y, et al. 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 U S A. 2002;99(19):12293–7.CrossRefPubMedPubMedCentral
5.
go back to reference Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17.CrossRefPubMed Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17.CrossRefPubMed
6.
go back to reference Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.CrossRefPubMed Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.CrossRefPubMed
7.
go back to reference Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.CrossRefPubMed Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.CrossRefPubMed
8.
go back to reference Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13.CrossRefPubMed Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13.CrossRefPubMed
9.
go back to reference Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet (London, England). 2016;387(10031):1909–20.CrossRef Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet (London, England). 2016;387(10031):1909–20.CrossRef
10.
go back to reference Ferris RL, Blumenschein G, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016. Ferris RL, Blumenschein G, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016.
11.
go back to reference Ansell SM, Hurvitz SA, Koenig PA, et al. Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009;15(20):6446–53.CrossRefPubMedPubMedCentral Ansell SM, Hurvitz SA, Koenig PA, et al. Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009;15(20):6446–53.CrossRefPubMedPubMedCentral
12.
go back to reference Roemer MGM, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol. 2016;34(23):2690–7.CrossRefPubMed Roemer MGM, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol. 2016;34(23):2690–7.CrossRefPubMed
13.
go back to reference • Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2014;372(4):311–9. PD-1 blockade in Hodgkin Lymphoma is well-tolerated with a high overall response rate and durable remissions.CrossRefPubMedPubMedCentral • Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2014;372(4):311–9. PD-1 blockade in Hodgkin Lymphoma is well-tolerated with a high overall response rate and durable remissions.CrossRefPubMedPubMedCentral
14.
go back to reference Armand P, Shipp MA, Ribrag V, et al. Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. J Clin Oncol. 2016. Armand P, Shipp MA, Ribrag V, et al. Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. J Clin Oncol. 2016.
15.
go back to reference Younes A, Santoro A, Shipp M, et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016;17(9):1283–94.CrossRefPubMed Younes A, Santoro A, Shipp M, et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016;17(9):1283–94.CrossRefPubMed
16.
go back to reference Lesokhin AM, Ansell SM, Armand P, Scott ECHA. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol. 2016;34(23):2698–704.CrossRefPubMed Lesokhin AM, Ansell SM, Armand P, Scott ECHA. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol. 2016;34(23):2698–704.CrossRefPubMed
17.
18.
go back to reference Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39(1):74–88.CrossRefPubMed Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39(1):74–88.CrossRefPubMed
19.
go back to reference Porrata LF, Litzow MR, Markovic SN. Immune reconstitution after autologous hematopoietic stem cell transplantation. Mayo Clin Proc. 2001;76(4):407–12.CrossRefPubMed Porrata LF, Litzow MR, Markovic SN. Immune reconstitution after autologous hematopoietic stem cell transplantation. Mayo Clin Proc. 2001;76(4):407–12.CrossRefPubMed
20.
go back to reference Armand P, Nagler A, Weller EA, et al. Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol. 2013;31(33):4199–206.CrossRefPubMedPubMedCentral Armand P, Nagler A, Weller EA, et al. Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol. 2013;31(33):4199–206.CrossRefPubMedPubMedCentral
21.
go back to reference • Merryman RW, Kim HT, Zinzani PL, et al. Safety and efficacy of allogeneic hematopoetic stem cell transplant (HSCT) after treatment with programmed cell death 1 (PD-1) inhibitors. Blood. 2015;126(23). Allo-HSCT after PD-1 blockade is feasible but may be associated with increased early immune toxicities. • Merryman RW, Kim HT, Zinzani PL, et al. Safety and efficacy of allogeneic hematopoetic stem cell transplant (HSCT) after treatment with programmed cell death 1 (PD-1) inhibitors. Blood. 2015;126(23). Allo-HSCT after PD-1 blockade is feasible but may be associated with increased early immune toxicities.
22.
go back to reference Armand P, Kim HT, Sainvil M-M, et al. The addition of sirolimus to the graft-versus-host disease prophylaxis regimen in reduced intensity allogeneic stem cell transplantation for lymphoma: a multicentre randomized trial. Br J Haematol. 2016;173(1):96–104.CrossRefPubMed Armand P, Kim HT, Sainvil M-M, et al. The addition of sirolimus to the graft-versus-host disease prophylaxis regimen in reduced intensity allogeneic stem cell transplantation for lymphoma: a multicentre randomized trial. Br J Haematol. 2016;173(1):96–104.CrossRefPubMed
23.
go back to reference Kanate AS, Mussetti A, Kharfan-Dabaja MA, et al. Reduced-intensity transplantation for lymphomas using haploidentical related donors vs HLA-matched unrelated donors. Blood. 2016;127(7):938–47.CrossRefPubMedPubMedCentral Kanate AS, Mussetti A, Kharfan-Dabaja MA, et al. Reduced-intensity transplantation for lymphomas using haploidentical related donors vs HLA-matched unrelated donors. Blood. 2016;127(7):938–47.CrossRefPubMedPubMedCentral
24.
go back to reference Armand P, Kim HT, Logan BR, et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014;123(23):3664–71.CrossRefPubMedPubMedCentral Armand P, Kim HT, Logan BR, et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014;123(23):3664–71.CrossRefPubMedPubMedCentral
25.
go back to reference Alyea EP, Kim HT, Ho V, et al. Comparative outcome of nonmyeloablative and myeloablative allogeneic hematopoietic cell transplantation for patients older than 50 years of age. Blood. 2005;105(4):1810–4.CrossRefPubMed Alyea EP, Kim HT, Ho V, et al. Comparative outcome of nonmyeloablative and myeloablative allogeneic hematopoietic cell transplantation for patients older than 50 years of age. Blood. 2005;105(4):1810–4.CrossRefPubMed
26.
go back to reference Antin JH. Graft-versus-leukemia: no longer an epiphenomenon. Blood. 1993;82(8):2273–7.PubMed Antin JH. Graft-versus-leukemia: no longer an epiphenomenon. Blood. 1993;82(8):2273–7.PubMed
27.
go back to reference • Kong Y, Zhang J, Claxton DF, et al. PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J. 2015;5:e330. Inhibitory checkpoint receptors are upregulated on T cells in patients who relapse after allo-HSCT and may predict relapse several months before clinically apparent recurrence.CrossRefPubMedPubMedCentral • Kong Y, Zhang J, Claxton DF, et al. PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J. 2015;5:e330. Inhibitory checkpoint receptors are upregulated on T cells in patients who relapse after allo-HSCT and may predict relapse several months before clinically apparent recurrence.CrossRefPubMedPubMedCentral
28.
go back to reference Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol. 2015;8:93.CrossRefPubMedPubMedCentral Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol. 2015;8:93.CrossRefPubMedPubMedCentral
29.
go back to reference Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Sharpe AH, Vallera DA. Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. J Immunol. 1999;162(11):6368–77.PubMed Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Sharpe AH, Vallera DA. Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. J Immunol. 1999;162(11):6368–77.PubMed
30.
go back to reference Fevery S, Billiau AD, Sprangers B, et al. CTLA-4 blockade in murine bone marrow chimeras induces a host-derived antileukemic effect without graft-versus-host disease. Leukemia. 2007;21(7):1451–9.CrossRefPubMed Fevery S, Billiau AD, Sprangers B, et al. CTLA-4 blockade in murine bone marrow chimeras induces a host-derived antileukemic effect without graft-versus-host disease. Leukemia. 2007;21(7):1451–9.CrossRefPubMed
31.
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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
32.
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. Blockade of CTLA-4 with ipilimumab resulted in durable responses in patients with various hematologic malignancies who had relapsed after allogeneic HSCT. It is the largest prospective trial to date showing that immune checkpoint blockade is a viable strategy for relapse after allo-HSCT.CrossRefPubMedPubMedCentral •• 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. Blockade of CTLA-4 with ipilimumab resulted in durable responses in patients with various hematologic malignancies who had relapsed after allogeneic HSCT. It is the largest prospective trial to date showing that immune checkpoint blockade is a viable strategy for relapse after allo-HSCT.CrossRefPubMedPubMedCentral
33.
go back to reference Schade H, Sen S, Neff CP, et al. Programmed death 1 expression on CD4(+) T cells predicts mortality after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2016. Schade H, Sen S, Neff CP, et al. Programmed death 1 expression on CD4(+) T cells predicts mortality after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2016.
34.
go back to reference • Krönig H, Kremmler L, Haller B, et al. Interferon-induced programmed death-ligand 1 (PD-L1/B7-H1) expression increases on human acute myeloid leukemia blast cells during treatment. Eur J Haematol. 2014;92(3):195–203. Alterations in expression of PD-1 and its ligands appears to be an important mechanism of tumor immune escape.CrossRefPubMed • Krönig H, Kremmler L, Haller B, et al. Interferon-induced programmed death-ligand 1 (PD-L1/B7-H1) expression increases on human acute myeloid leukemia blast cells during treatment. Eur J Haematol. 2014;92(3):195–203. Alterations in expression of PD-1 and its ligands appears to be an important mechanism of tumor immune escape.CrossRefPubMed
35.
go back to reference • Michonneau D, Sagoo P, Breart B, et al. The PD-1 axis enforces an anatomical segregation of CTL activity that creates tumor niches after allogeneic hematopoietic stem cell transplantation. Immunity. 2016;44(1):143–54. In a mouse model, differential upregulation of PD-1 ligand resulted in a compartmentalized GVT response and the emergence of niches for tumor immune escape. PD-1 blockade restored the efficacy of GVT across these anatomic niches.CrossRefPubMed • Michonneau D, Sagoo P, Breart B, et al. The PD-1 axis enforces an anatomical segregation of CTL activity that creates tumor niches after allogeneic hematopoietic stem cell transplantation. Immunity. 2016;44(1):143–54. In a mouse model, differential upregulation of PD-1 ligand resulted in a compartmentalized GVT response and the emergence of niches for tumor immune escape. PD-1 blockade restored the efficacy of GVT across these anatomic niches.CrossRefPubMed
36.
go back to reference Koestner W, Hapke M, Herbst J, et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood. 2011;117(3):1030–41.CrossRefPubMedPubMedCentral Koestner W, Hapke M, Herbst J, et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood. 2011;117(3):1030–41.CrossRefPubMedPubMedCentral
37.
go back to reference Blazar BR, Carreno BM, Panoskaltsis-Mortari A, et al. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171(3):1272–7.CrossRefPubMed Blazar BR, Carreno BM, Panoskaltsis-Mortari A, et al. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171(3):1272–7.CrossRefPubMed
38.
go back to reference Fujiwara H, Maeda Y, Kobayashi K, et al. Programmed death-1 pathway in host tissues ameliorates Th17/Th1-mediated experimental chronic graft-versus-host disease. J Immunol. 2014;193(5):2565–73.CrossRefPubMed Fujiwara H, Maeda Y, Kobayashi K, et al. Programmed death-1 pathway in host tissues ameliorates Th17/Th1-mediated experimental chronic graft-versus-host disease. J Immunol. 2014;193(5):2565–73.CrossRefPubMed
39.
go back to reference Amarnath S, Mangus CW, Wang JCM, et al. The PDL1-PD1 axis converts human TH1 cells into regulatory T cells. Sci Transl Med. 2011;3(111):111–20.CrossRef Amarnath S, Mangus CW, Wang JCM, et al. The PDL1-PD1 axis converts human TH1 cells into regulatory T cells. Sci Transl Med. 2011;3(111):111–20.CrossRef
40.
go back to reference Asano T, Kishi Y, Meguri Y, et al. PD-1 signaling has a critical role in maintaining regulatory T cell homeostasis; implication for Treg depletion therapy by PD-1 blockade. Blood. 2015;126(23):848. Asano T, Kishi Y, Meguri Y, et al. PD-1 signaling has a critical role in maintaining regulatory T cell homeostasis; implication for Treg depletion therapy by PD-1 blockade. Blood. 2015;126(23):848.
41.
go back to reference Khan AR, Hams E, Floudas A, et al. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun. 2015;6:5997.CrossRefPubMed Khan AR, Hams E, Floudas A, et al. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun. 2015;6:5997.CrossRefPubMed
42.
go back to reference Saha A, Aoyama K, Taylor PA, et al. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood. 2013;122(17):3062–73.CrossRefPubMedPubMedCentral Saha A, Aoyama K, Taylor PA, et al. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood. 2013;122(17):3062–73.CrossRefPubMedPubMedCentral
43.
go back to reference Saha A, O’Connor RS, Thangavelu G, et al. Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality. J Clin Invest. 2016;126(7):2642–60.CrossRefPubMedPubMedCentral Saha A, O’Connor RS, Thangavelu G, et al. Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality. J Clin Invest. 2016;126(7):2642–60.CrossRefPubMedPubMedCentral
44.
go back to reference Villasboas JC, Ansell SM, Witzig TE. Targeting the PD-1 pathway in patients with relapsed classic Hodgkin lymphoma following allogeneic stem cell transplant is safe and effective. Oncotarget. 2016;7(11):13260–4.PubMedPubMedCentral Villasboas JC, Ansell SM, Witzig TE. Targeting the PD-1 pathway in patients with relapsed classic Hodgkin lymphoma following allogeneic stem cell transplant is safe and effective. Oncotarget. 2016;7(11):13260–4.PubMedPubMedCentral
45.
go back to reference Kwong Y-L. Safety of pembrolizumab after allogeneic haematopoietic stem cell transplantation. Ann Hematol. 2016;95(7):1191–2.CrossRefPubMed Kwong Y-L. Safety of pembrolizumab after allogeneic haematopoietic stem cell transplantation. Ann Hematol. 2016;95(7):1191–2.CrossRefPubMed
46.
go back to reference Chan TSY, Khong P-L, Kwong Y-L. Pembrolizumab for relapsed anaplastic large cell lymphoma after allogeneic haematopoietic stem cell transplantation: efficacy and safety. Ann Hematol. 2016. Chan TSY, Khong P-L, Kwong Y-L. Pembrolizumab for relapsed anaplastic large cell lymphoma after allogeneic haematopoietic stem cell transplantation: efficacy and safety. Ann Hematol. 2016.
47.
go back to reference Singh AK, Porrata LF, Aljitawi O, et al. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin’s lymphoma. Bone Marrow Transplant. 2016;51(9):1268–70.CrossRefPubMed Singh AK, Porrata LF, Aljitawi O, et al. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin’s lymphoma. Bone Marrow Transplant. 2016;51(9):1268–70.CrossRefPubMed
48.
go back to reference Herbaux C, Gauthier J, Brice P, et al. Nivolumab is effective and reasonably safe in relapsed or refractory Hodgkin’s lymphoma after allogeneic hematopoietic cell transplantation: a study from the Lysa and SFGM-TC. Blood. 2015;126(23):3979. Herbaux C, Gauthier J, Brice P, et al. Nivolumab is effective and reasonably safe in relapsed or refractory Hodgkin’s lymphoma after allogeneic hematopoietic cell transplantation: a study from the Lysa and SFGM-TC. Blood. 2015;126(23):3979.
49.
go back to reference Yared JA, Hardy N, Singh Z, et al. Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51(6):850–2.CrossRefPubMed Yared JA, Hardy N, Singh Z, et al. Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51(6):850–2.CrossRefPubMed
50.
go back to reference Angenendt L, Schliemann C, Lutz M, et al. Nivolumab in a patient with refractory Hodgkin’s lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51(3):443–5.CrossRefPubMed Angenendt L, Schliemann C, Lutz M, et al. Nivolumab in a patient with refractory Hodgkin’s lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51(3):443–5.CrossRefPubMed
Metadata
Title
Immune Checkpoint Blockade and Hematopoietic Stem Cell Transplant
Authors
Reid W. Merryman
Philippe Armand
Publication date
01-02-2017
Publisher
Springer US
Published in
Current Hematologic Malignancy Reports / Issue 1/2017
Print ISSN: 1558-8211
Electronic ISSN: 1558-822X
DOI
https://doi.org/10.1007/s11899-017-0362-5

Other articles of this Issue 1/2017

Current Hematologic Malignancy Reports 1/2017 Go to the issue

Stem Cell Transplantation (R Maziarz, Section Editor)

Clinical Studies in Hematologic Microtransplantation

Chronic Lymphocytic Leukemias (N Jain, Section Editor)

A Concise Review of Autoimmune Cytopenias in Chronic Lymphocytic Leukemia

Chronic Lymphocytic Leukemias (N Jain, Section Editor)

Targeted Therapy in Chronic Lymphocytic Leukemia (CLL)