Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 10.
Published in final edited form as: Br J Haematol. 2019 Dec 6;188(1):159–167. doi: 10.1111/bjh.16358

Checkpoint inhibitors in AML: are we there yet?

Arnab Ghosh 1,2, Pere Barba 3, Miguel-Angel Perales 1,2
PMCID: PMC9549702  NIHMSID: NIHMS1832010  PMID: 31808941

Summary

Immunotherapy is distinct from traditional chemotherapy in that it acts on immune cells rather than cancer cells themselves. Monoclonal antibodies targeting immune checkpoints on T cells – CTLA-4 and PD-1 – and PD-L1 on the cells of immune microenvironment are now approved for clinical use in several solid tumors and hematological malignancies. This article provides a general overview of the use of checkpoint inhibitors in hematologic malignancies with a special focus in acute myeloid leukemia.

Keywords: acute myeloid leukemia, checkpoint inhibitors, PD-1, PD-L1, CTLA-4


Immunotherapy is distinct from traditional chemotherapy in that it acts on immune cells rather than cancer cells themselves (Batlevi et al., 2016; Khalil et al., 2016; Weiss et al., 2019). Monoclonal antibodies targeting immune checkpoints on T cells – CTLA-4 and PD-1 - and PD-L1 on the cells of immune microenvironment are now approved by the Food and Drug Administration (FDA) and European Medical Agency (EMA) for the treatment of malignancies including melanoma (Hodi et al., 2010), non-small cell lung cancer (Brahmer et al., 2015), renal cancer (Motzer et al., 2018) and Hodgkin lymphoma (Ansell et al., 2015). Inhibition of checkpoints activates T cells and recognition of tumor-associated antigens leads to lysis of tumors and regression of cancer. Thus immune checkpoint inhibition enhances recognition of tumors as non-self and an expanding body of evidence in solid tumors identified several factor for successful applications of immune checkpoint blockers, including recognition of tumor-related antigens presented in the context of human leukocyte antigen (HLA) by T cells, presence of the checkpoints on T cells and the ligands of checkpoints on the antigen presenting cells or the tumor cells (Rizvi et al., 2015). These therapies have demonstrated antitumor responses in several malignancies either alone or in combination with other anti-cancer therapies (Ansell et al., 2015; Younes et al., 2016; Younes et al., 2019). This article provides a general overview of the use of checkpoint inhibitors in hematologic malignancies with a special focus in acute myeloid leukemia. We also discuss their treatment-related toxicity as well as the use of these agents in specific populations such as allogeneic hematopoietic stem cell transplantation (alloHCT) recipients.

Immune escape of AML after allogeneic hematopoietic stem cell transplantation

AlloHCT is a potentially curative option for patients with high-risk AML (Gooley et al., 2010; Jenq & van den Brink, 2010). Despite increases in overall survival due to improved patient selection, HLA-matching and supportive care, relapse remains a major cause of death in patients with AML who undergo alloHCT (Wayne et al., 2013). In addition to the effectiveness of high-intensity chemotherapy with or without radiation, alloHCT recipients benefit from the ability of donor-derived T cells to recognize host AML cells, which bear non-self HLA, as foreign and thus mount a graft versus leukemia (GVL) effect that eliminates any residual disease after alloHCT and maintains lasting immunosurveillance to prevent relapse (Copelan, 2006; van den Brink et al., 2010). However, host tissue recognition as foreign by donor T cells can also lead to a broader immune reaction causing potentially lethal graft-versus-host disease (GVHD) (Ferrara & Reddy, 2006).

In preventing relapse after AML, the relative contributions of the immunological effects of alloHCT, namely GVL effects and the cytotoxic effects of high-intensity conditioning have not been completely elucidated. A detailed analysis of leukemic cells that relapse after alloHCT may however provide useful insights into mechanisms of relapse. An early study of patients with myeloid leukemia that relapsed after alloHCT demonstrated decreased ability to stimulate donor T cell proliferation in 4 of 5 patients (Dermime et al., 1997). The relapsed cells also had decreased MHC-expression, and decreased susceptibility to T and NK cell lysis.

With the advent of high-dimensional analsysis, a comprehensive analysis of samples from 15 patients with AML who relapsed after alloHCT was performed. The study included whole exome sequencing, whole transcriptome analysis and multicolor flow cytometry (Christopher et al., 2018). Curiously, whole exome sequencing revealed relapse after alloHCT was not associated with the acquisition of new AML-specific mutations or structural variations in immune-related genes. Whole transcriptome studies by RNAseq and flow revealed involvement of immune-related pathways and most notably HLA class II regulation. In a second study, serial whole transcriptome and genome analysis was performed on leukemic blasts at different time points (Toffalori et al., 2014). A signature consistent with immune dysregulation was found that affected both antigen presentation and T cells. The AML blasts at relapse after alloHCT expressed checkpoints including PD-L1, and loss of HLA class II genes concurrently with down-regulation of its regulator CIITA.

An investigation into the donor T cell compartment in the bone marrow infiltrating CD4+ and CD8+ T cells from patients with AML after alloHCT demonstrated an exhaustion signature (Noviello et al., 2019). Bone marrow infiltrating T memory stem cells and central memory T cells, expressing the irreversibly exhausted PD-1+Eomes+T-bet—predicted relapse. Accumulation of regulatory T cells was also noted in patients with relapse. In HLA-matched alloHCT, CD8+ T cells infiltrating the bone marrow of patients expressing PD-1, CTLA-4, and/or TIM-3 was higher in patients who relapsed than in patients without disease.

Given the primacy of immune dysregulation associated with AML relapse after alloHCT, T cell-based therapies are a reasonable approach to treat or prevent relapse. Adoptive transfer of donor T cells in the form of donor lymphocyte infusion (DLI) is one such approach to overcome the exhausted T cells. Early success with DLI in chronic myelogenous leukemia (Kolb et al., 1995; Collins et al., 1997) led exploration of its use in AML and MDS with more limited success (Depil et al., 2004; Schmid et al., 2007). One key limitation of the use of DLI to treat a full-blown relapse is the high burden of disease. Donor T cells, driven by expansion of alloreactive T cells, are slower than chemotherapy and also dependent on the expression of HLA. Hence hypomethylating agents like azacytadine have also been used in conjunction with DLI (Schroeder et al., 2013; Tessoulin et al., 2014; Steinmann et al., 2015; Motabi et al., 2016). In one of the larger retrospective analyses of outcomes of intensive chemotherapy with that of hypomethylating agents in relapsed AML and MDS after alloHCT, 56% of patients in the chemotherapy group and 33% of patients in the hypomethylating agents group received at least one DLI after treatment. The use of chemotherapy followed by DLI offered better outcomes (ORR, 68%; CR, 59%, 1-year OS, 44%; and median OS, 9·8 months) than hypomethylating agents (Motabi et al., 2016).

Checkpoint inhibitors in lymphoid malignancies

After some early clinical trials of PD-1 blockade using the mAb pidilizumab (Berger et al., 2008; Westin et al., 2014) and the anti-CTLA-4 mAb ipilimumab (Ansell et al., 2009) suggested activity in lymphoid malignancies, recent studies have focused on two different PD-1 mAbs, nivolumab and pembrolizumab (Perales et al., 2016). The most promising results have been observed in Hodgkin lymphoma, which has frequent amplification of genetic material at 9p24, which results in the over-expression of the PD-1 ligands, PD-L1 and PD-L2 (Green et al., 2010). Due to this and additional mechanisms, the very high frequency of PD-L1/PD-L2 expression on the surface of the HL tumor cell surface suggested that HL is a uniquely vulnerable target for PD-1 blockade (Chen et al., 2013). The clinical results in Hodgkin lymphoma have lead to approval of both drugs in this disease. Initial studies in Hodgkin lymphoma reported ORR and CR rates of 65% and 16% with pembroluzimab (Armand et al., 2016), and 87% and 17% with nivolumab (Ansell et al., 2015), respectively (Table I). These results lead to two large studies testing each drug in several cohorts. The Checkmate 205 study included patients who had not received brentuximab vedotin (BV), had received it after an autologous stem cell transplant (ASCT), and had received BV both pre and post ASCT (Younes et al., 2016; Armand et al., 2018). The objective response rate was 69% overall and 65– 73% in each cohort (Armand et al., 2018). The median duration of response was 16·6 months (95% CI, 13·2– 20·3 months), and median progression-free survival was 14·7 months (95% CI, 11·3–18·5 months). The KEYNOTE-087 study examined 3 cohorts of patients treated with pembroluzimab, patients who progressed after ASCT and BV, patients who failed salvage chemotherapy and BV, were ineligible for ASCT, and those who failed ASCT but did not receive BV (Chen et al., 2017; Chen et al., 2019). The ORR was 69·0% (95% CI, 62·3–75·2%), and the CR rate was 22·4% (95% CI, 16·9–28·6%). ORRs were 73·9% for cohort 1, 64·2% for cohort 2, and 70·0% for cohort 3. More recently, nivolumab has been tested in the upfront setting in combination with AVD (Ramchandren et al., 2019). The ORR was 84% (71–93%), with 67% (52–79%) achieving CR. Additional studies are ongoing and nivolumab has also been combined with BV (Brockelmann & Engert, 2017; Herrera et al., 2018). The exact sequence of treatments in patients with Hodgkin lymphoma, including the role of ASCT and alloHCT remains an evolving picture as new data becomes available (Perales et al., 2015). BV has now moved upfront in selected patients, and is also being incorporated in salvage regimens pre ASCT. Checkpoint inhibitors are typically reserved for patients who have failed an ASCT, and are being used in BV naïve as well as BV refractory patients. While the prior standard of care in ASCT failures was to proceed with alloHCT in patients responding to salvage therapy, this paradigm has shifted with BV and checkpoint inhibitors, where many clinicians are deferring alloHCT until progression or in patients with incomplete responses.

Table I.

Select trials of anti-PD-1 antibody therapy in Hodgkin lymphoma.

Checkpoint inhibitor Additional agents Patients n Main outcomes Citation

Nivolumab Relapsed/refractory HL 23 ORR: 87%; CR: 17% Ansell et al., 2015
Pembrolizumab cHL after Brentuximab vedotin failure (KEYNOTE-013) 31 ORR: 65%; CR: 16% Armand et al., 2016
Nivolumab Treatment history: Cohort A: Brentuximab vedotin (BV)-naïve
Cohort B: BV received after auto-HSCT
Cohort C: BV received before and/or after auto-HSCT
Relapsed/refractory cHL after auto-HCT treatment failure (CheckMate 205) 243
Cohort A: 63
Cohort B: 80
Cohort C: 100
ORR: 69%; CR: 16% Cohort A: ORR:
65%; CR: 18%
Cohort B: ORR: 68%; CR: 10%
Cohort C: ORR: 73%;
CR: 12%
Armand et al., 2018
Pembrolizumab Relapsed or refractory classic Hodgkin lymphoma (KEYNOTE-087) 210 ORR: 719%; CR: 276% Chen et al., 2019
Nivolumab Doxorubicin, vinblastine, dacarbazine (N-AVD) Newly diagnosed cHL (CheckMate 205) Cohort D ORR: 84%; CR: 67% Ramchandren et al., 2019

In contrast to the excellent results in Hodgkin lymphoma, the activity of nivolumab in other hematologic malignancies has been quite variable, with response rates of 36–40% noted in follicular lymphoma, DLBCL, and T-cell lymphoma (Lesokhin et al., 2014; Armand et al., 2015).

Checkpoint inhibitors in AML

Studies have shown that polyclonally expanded T cells might recognize tumor cells that have a higher mutation burden (i.e., the total number of mutations per coding area of the tumor genome), which have higher odds of presenting antigens that evoke recognition as non-self (Yarchoan et al., 2017). In several solid tumors, a significant correlation exists between tumor mutation burden and objective response rates, and tumor mutation burden is therefore an important predictor of responses to immunotherapy (Snyder et al., 2014; Rizvi et al., 2015; McGranahan et al., 2016; Samstein et al., 2019).

AML and MDS express somatic mutations that could be recognized as non-self, but the tumor mutation burden is relatively lower than most other tumors (Lawrence et al., 2013). Immune checkpoint expression is generally low at baseline. Additionally, AML and MDS are associated with a dysfunctional and suppressive immune compartment (Kordasti et al., 2007; Wei et al., 2013; Arandi et al., 2018; Han et al., 2018). Simlar to the approach discussed above in patients receiving DLI, investigators have proposed combining agents that can enhance antigen presentation and/or increase immune checkpoint expression with immune checkpoint inhibitors (Table II).

Table II.

Immune checkpoint therapy for AML.

Checkpoint inhibitor Additional agents Patients N Main outcomes Citation

Pre alloHCT
 Nivolumab Cytarabine and idarubicin Newly diagnosed AML 44 Median EFS not reached (95% CI 7·93-NR).
Median OS and RFS in
responders 18·5 months
Ravandi et al., 2019
 Nivolumab Azacitidine Relapsed/refractory AML 70 ORR: 33%; CR/iCR: 22%.
Median OS 6·3 months
Daver et al., 2019
Post alloHCT
Ipilimumab Relapsed heme malignancies after conventional alloHCT 29 (including 2 AML) Bashey et al., 2009
Ipilimumab Relapsed heme malignancies after conventional alloHCT 28 (including 12 AML) CRs in 4 patients with extramedullary AML and 1 patient with MDS transformed to AML. Davids et al., 2016

One such approach has been to utilize chemotherapy to release antigens that might help prime T cells. In a phase 2 single-arm study reported from MD Anderson Cancer Center, nivolumab was used in combination with idarubicin and cytarabine as a frontline therapy for AML or high risk MDS (Ravandi et al., 2019). Induction included cytarabine 1·5 g/ m2 by continuous infusion daily on days 1–4 (3 days in patients >60 years) and idarubicin 12 mg/m2 daily on days 1–3. Nivolumab 3 mg/kg was started on about day 24 (±2 days) and continued every 2 weeks for up to a year in responders. Of the 44 patients enrolled in this study, the primary endpoint of median event free survival was not reached at a median follow up of 17·25 months. The median relapse free survival of the responders and overall survival were 18·54 months. Six patients had seven grade 3–4 immune-related adverse events which included rash, colitis, transaminitis, pancreatitis, and cholecystitis. Nineteen (43%) of 44 patients achieved a response and proceeded to allogeneic stem cell transplantation, with grade 3/4 graft-versus-host disease observed in five (26%).

Another promising strategy has been to use hypomethylating agents such as azacitadine, which are a standard antineoplastic therapy in AML/MDS. In a single arm, phase II study, a combination of nivolumab and azacytidine was tested in 70 patients with relapsed/refractory AML (Daver et al., 2019). Azacytidine 75 mg/m2 was administered on days 1–7 and nivolumab 3 mg/day on days 1 and 14 every 4–6 weeks. The results were encouraging with an ORR rate of 33%, including 4 (6%) complete remissions (CR), 11 (16%) CR with incomplete blood count recovery (CRi), 1 partial remission (PR) and 7 showing hematologic improvement. Six patients (9%) had stable disease. The combination was well tolerated with grade 2, and grade 3–4 immune-related adverse effects observed in eight (11%) patients respectively, which were mostly responsive to steroids or infliximab. Majority of the immune-related adverse effects happened in the first 8 weeks after initial treatment of nivolumab, and two patients died due to steroid and infliximab-resistant toxicities.

While the results of some of these early studies are encouraging, at the present time, checkpoint inhibitors are not recommended as part of standard of care in patients with AML. Ongoing studies should help to clarify the role of checkpoint inhibitors in myeloid malignancies.

Toxicities of checkpoint inhibitors

The significant benefits in clinical outcomes obtained with the use of checkpoint inhibitors in several indications have been hampered by their treatment-related toxicities. Most of these adverse events occur as a result of the dysregulation in the immune system balance produced by these drugs (Pardoll, 2012). Because these immune-related adverse events (irAERs) differ from classical chemotherapy-related side effects both in the clinical presentation and in their treatment, a multidisplinary approach to manage them has been proposed. In many centers, specific working groups where gastroenterologists, hepatologists, infectious disease specialists and neurologists (among others) work together with oncologists and hematologists, have been develop in order to improve patient care. Patient education is also important for early diagnosis and treatment of these complications, since it has been shown that early intervention of irAEs reduces both their severity and duration (Hodi et al., 2010).

The spectrum of irAEs is diverse and can involve almost any tissue or organ (Bair et al., 2019). These events include but are not limited to complications in skin (rash, vitiligo, toxic epidermal necrolysis) (Abdel-Rahman et al., 2015), the lungs (pneumonitis, pleural effusion) (Naidoo et al., 2017), thyroid gland (hypothyroidism) (Ryder et al., 2014), the gastrointestinal tract (colitis, pancreatitis, celiac disease) (Beck et al., 2006; Peyrony et al., 2019), the nervous system (encephalopathy, Guillain-Barre syndrome, myelitis) (Liao et al., 2014), the liver (hepatitis) (Lleo et al., 2019), the heart (myocarditis) (Quagliariello et al., 2019) and hematological cells (immune cytopenias) (Friedman et al., 2016). Most of these events can occur simultaneously or at different intervals and may involve different organs or tissues. Time to irAEs is also quite variable and can range from a few weeks to up to 2 years after the first treatment administration (Wolchok et al., 2010). Moreover, these events can occur even after discontinuation of the drug. Whereas most of these side effects are manageable, they can be potentially fatal in some cases (Johnson et al., 2016; Brahmer et al., 2018).

Patients receiving checkpoint inhibitors seem to be at a higher risk of developing infections, especially if they require corticosteroids or other immunosuppressant agents to treat immune-related adverse events (Redelman-Sidi et al., 2018). In a recent study, 7.3% of patients receiving checkpoint blockade for the treatment of melanoma developed severe infections (Del Castillo et al., 2016). In hematological patients, this risk might be higher due to previous immunosuppressive therapies administered to the patient and the nature of the malignancy (Ansell et al., 2015).

Risk factors for the development of irAEs events include the type of checkpoint inhibitor, combination therapy and previous autoimmune disease. In terms of the type of antibody, PD1/PDL1 axis inhibitors seem to have lower incidence of adverse events than CTLA-4 inhibitors whereas the combination of 2 inhibitors has a higher rate of immune-related events than either monotherapy (Friedman et al., 2016). Most of the trials evaluating the use of these drugs in cancer patients excluded patients with previous history of autoimmune diseases. There is limited data showing that checkpoint inhibitors can be safely administered to patients with autoimmune disease and the decision for the indication should be taken with caution (Kyi et al., 2014).

Role of checkpoint inhibitors in the context of alloHCT

While checkpoint inhibitors have a theoretical potential during peri-alloHCT to enhance donor-derived T cells, concerns for the development of uncontrolled GVHD and other fulminant immune adverse events have hampered wider use of these agents.

Given these concerns, the first trial to study Ipilimumab was attempted with very low doses to treat relapsed disease (Bashey et al., 2009). In this phase I trial, 29 patients with hematological malignancies including 2 with AML who relapsed after alloHCT were treated with 0·3–1 mg/kg in a single infusion. Responses were noted in only three of the patients with lymphoid malignancies. Importantly, dose-limiting toxicity was not encountered and no evidence of GVHD or graft rejection was found. This led to a phase I/II trial of 28 patients with post-alloHCT relapsed hematological malignancies that enrolled to receive ipilimumab at 3 or 10 mg/kg every 3 weeks for up to 4 doses (Davids et al., 2016). Additional doses were given every 12 weeks for up to 60 weeks in patients who showed clinical benefit. No responses or significant immune adverse effects were seen in the 3 mg/kg arm. In the 10 mg/kg arm, CRs occurred in 4 patients with extramedullary AML and 1 patient with the MDS transformed into AML. Four patients had a durable response that lasted more than 1 year. Responses were associated with infiltration of CD8+ T cells, decreased regulatory T cells, and expansion of effector T cells subsets.

In contrast to ipilimumab, the use of anti-PD1 antibodies in the context of alloHCT has been hampered with reports of significant GVHD and other irAEs. The effects of PD-1 when used to treat lymphoma prior to alloHCT resulted in grade II–IV and grade III–IV acute GVHD in 44% and 23% of the patients, respectively (Merryman et al., 2017). While the rates of II-IV aGVHD were expected in this patient population, the rates of grade III-IV were higher than predicted. Furthermore, a number of patients required steroids for treatment of a persistent febrile syndrome. Additionally Hodgkin lymphoma patients who relapsed after nivolumab and underwent alloHCT also had high rates of acute GVHD (Armand et al., 2018). In spite of the setbacks with anti-PD1 therapies, there are ongoing trials with nivolumab and pembrolizumab after alloHCT with encouraging early results that continue to accrue.

Future directions

The success and enthusiasm for immune checkpoint inhibitors have hitherto been less in AML compared to other cancers like solid tumors and Hodgkin lymphoma. However greater understanding of the immunological basis for relapse and differences between the mechanisms behind commonly used immune checkpoint inhibitors will likely see greater use of these and newer agents. Since the greatest concern in the use of immune checkpoints is development of uncontrollable GVHD, alloHCT platforms with low GVHD potential could be used. In vivo and in vitro T cell depleted alloHCT are associated with lower incidence of acute and chronic GVHD (Papadopoulos et al., 1998; Jakubowski et al., 2007; Jakubowski et al., 2011; Barba et al., 2017; Malard et al., 2018; Cho & Perales, 2019) (Papadopoulos et al., 1998; Jakubowski et al., 2011; Jakubowski et al., 2017). Further studies will be required to identify the optimal treatment combinations with checkpoint inhibitors in AML as well as their use in the context of alloHCT.

Financial support and sponsorship

This research was supported in part by National Institutes of Health award numbers P01 CA23766 and NIH/NCI Cancer Center Support Grant P30 CA008748. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. PB received funding from the Carlos III FIS16/01433 Health Institute and a PERIS 2018–2020 grant from the Generalitat de Catalunya (BDNS357800).

Footnotes

Conflict of interest

Dr. Perales reports honoraria from Abbvie, Bellicum, Bristol-Myers Squibb, Incyte, Merck, Novartis, Nektar Therapeutics, Omeros, and Takeda. He serves on DSMBs for Servier and Medigene, and the scientific advisory boards of MolMed and NexImmune. He has received research support for clinical trials from Incyte, Kite/Gilead and Miltenyi Biotec. He serves in a volunteer capacity as a member of the Board of Directors of American Society for Transplantation and Cellular Therapy (ASTCT) and Be The Match (National Marrow Donor Program, NMDP), as well as on the CIBMTR Cellular Immunotherapy Data Resource (CIDR) Committee. Dr. Barba declares have received honoraria from Amgen, Celgene, Gilead, Incyte, Jazz Pharmaceuticals, MSD, Novartis, Pfizer and Roche, not related with the present article. Dr. Ghosh declares no conflicting interests.

References

  1. Abdel-Rahman O, ElHalawani H & Fouad M. (2015) Risk of cutaneous toxicities in patients with solid tumors treated with immune checkpoint inhibitors: a meta-analysis. Future Oncol, 11, 2471–2484. [DOI] [PubMed] [Google Scholar]
  2. Ansell SM, Hurvitz SA, Koenig PA, LaPlant BR, Kabat BF, Fernando D, Habermann TM, Inwards DJ, Verma M, Yamada R, Erlichman C, Lowy I & Timmerman JM. (2009) Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clinical Cancer Research, 15, 6446–6453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, Rodig SJ, Chapuy B, Ligon AH, Zhu L, Grosso JF, Kim SY, Timmerman JM, Shipp MA & Armand P. (2015) PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. New England Journal of Medicine, 372, 311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Arandi N, Ramzi M, Safaei F & Monabati A. (2018) Overexpression of indoleamine 2,3-dioxygenase correlates with regulatory T cell phenotype in acute myeloid leukemia patients with normal karyotype. Blood Research, 53, 294–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Armand P, Timmerman JM, Lesokhin AM, Halwani A, Millenson MM, Schuster SJ, Gutierrez M, Scott EC, Cattry D, Freeman GJ, Chapuy B, Ligon AH, Rodig S, Zhu L, Grosso JF, Simon J, Shipp MA, Cohen AD, Lebovic D, Dhodapkar MV, Avigan D, Ansell S & Borrello I. (2015) Nivolumab in patients with relapsed or refractory lymphoid malignancies and classical hodgkin lymphoma: updated safety and efficacy results of a phase 1 study (ca209–039). European Hematology Association Meeting Abstracts, S808. [Google Scholar]
  6. Armand P, Shipp MA, Ribrag V, Michot JM, Zinzani PL, Kuruvilla J, Snyder ES, Ricart AD, Balakumaran A, Rose S & Moskowitz CH. (2016) Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. Journal of Clinical Oncology, 34, 3733–3739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Armand P, Engert A, Younes A, Fanale M, Santoro A, Zinzani PL, Timmerman JM, Collins GP, Ramchandren R, Cohen JB, De Boer JP, Kuruvilla J, Savage KJ, Trneny M, Shipp MA, Kato K, Sumbul A, Farsaci B & Ansell SM. (2018) Nivolumab for relapsed/refractory classic hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the multicohort singlearm phase II CheckMate 205 trial. Journal of Clinical Oncology, 36, 1428–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bair SM, Strelec LE, Feldman TA, Ahmed G, Armand P, Shah NN, Singavi AN, Reddy N, Khan N, Andreadis C, Vu K, Huntington SF, Giri S, Ujjani C, Howlett C, Faheem M, Youngman MR, Nasta SD, Landsburg DJ, Schuster SJ & Svoboda J. (2019) Outcomes and toxicities of programmed death-1 (PD-1) inhibitors in Hodgkin lymphoma patients in the United States: a real-world, multicenter retrospective analysis. The Oncologist, 24, 955–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Barba P, Hilden P, Devlin SM, Maloy M, Dierov D, Nieves J, Garrett MD, Sogani J, Cho C, Barker JN, Kernan NA, Castro-Malaspina H, Jakubowski AA, Koehne G, Papadopoulos EB, Prockop S, Sauter C, Tamari R, van den Brink MR, Avecilla ST, Meagher R, O’Reilly RJ, Goldberg JD, Young JW, Giralt S, Perales MA & Ponce DM. (2017) Ex vivo CD34(+)-selected T cell-depleted peripheral blood stem cell grafts for allogeneic hematopoietic stem cell transplantation in acute leukemia and myelodysplastic syndrome is associated with low incidence of acute and chronic graft-versus-host disease and high treatment response. Biology of Blood and Marrow Transplantation, 23, 452–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, Lowy I, Solomon SR, Morris LE, Holland HK, Mason JR, Alyea EP, Soiffer RJ & Ball ED. (2009) CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood, 113, 1581–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Batlevi CL, Matsuki E, Brentjens RJ & Younes A. (2016) Novel immunotherapies in lymphoid malignancies. Nature Reviews Clinical Oncology, 13, 25–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Beck KE, Blansfield JA, Tran KQ, Feldman AL, Hughes MS, Royal RE, Kammula US, Topalian SL, Sherry RM, Kleiner D, Quezado M, Lowy I, Yellin M, Rosenberg SA & Yang JC. (2006) Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. Journal of Clinical Oncology, 24, 2283–2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A & Nagler A. (2008) Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clinical Cancer Research, 14, 3044–3051. [DOI] [PubMed] [Google Scholar]
  14. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Aren Frontera O, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B & Spigel DR. (2015) Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. New England Journal of Medicine, 373, 123–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, Chau I, Ernstoff MS, Gardner JM, Ginex P, Hallmeyer S, Holter Chakrabarty J, Leighl NB, Mammen JS, McDermott DF, Naing A, Nastoupil LJ, Phillips T, Porter LD, Puzanov I, Reichner CA, Santomasso BD, Seigel C, Spira A, Suarez-Almazor ME, Wang Y, Weber JS, Wolchok JD, Thompson JA; National Comprehensive Cancer Network (2018) Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. Journal of Clinical Oncology, 36, 1714–1768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. van den Brink MR, Porter DL, Giralt S, Lu SX, Jenq RR, Hanash A & Bishop MR. (2010) Relapse after allogeneic hematopoietic cell therapy. Biology of Blood and Marrow Transplantation, 16, S138–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Brockelmann PJ & Engert A. (2017) Checkpoint inhibition in Hodgkin lymphoma – a review. Oncology Research and Treatment, 40, 654–660. [DOI] [PubMed] [Google Scholar]
  18. Chen BJ, Chapuy B, Ouyang J, Sun HH, Roemer MG, Xu ML, Yu H, Fletcher CD, Freeman GJ, Shipp MA & Rodig SJ. (2013) PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clinical Cancer Research, 19, 3462–3473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, Radford J, Ribrag V, Molin D, Vassilakopoulos TP, Tomita A, von Tresckow B, Shipp MA, Zhang Y, Ricart AD, Balakumaran A, Moskowitz CH; KEYNOTE-087. (2017) Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. Journal of Clinical Oncology, 35, 2125–2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen R, Zinzani PL, Lee HJ, Armand P, Johnson NA, Brice P, Radford J, Ribrag V, Molin D, Vassilakopoulos TP, Tomita A, von Tresckow B, Shipp MA, Lin J, Kim E, Nahar A, Balakumaran A & Moskowitz CH. (2019) Pembrolizumab in relapsed or refractory Hodgkin lymphoma: two-year follow-up of KEYNOTE-087. Blood, 134, 1144–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cho C & Perales MA. (2019) Expanding therapeutic opportunities for hematopoietic stem cell transplantation: T cell depletion as a model for the targeted allograft. Annual Review of Medicine, 70, 381–393. [DOI] [PubMed] [Google Scholar]
  22. Christopher MJ, Petti AA, Rettig MP, Miller CA, Chendamarai E, Duncavage EJ, Klco JM, Helton NM, O’Laughlin M, Fronick CC, Fulton RS, Wilson RK, Wartman LD, Welch JS, Heath SE, Baty JD, Payton JE, Graubert TA, Link DC, Walter MJ, Westervelt P, Ley TJ & DiPersio JF. (2018) Immune escape of relapsed AML cells after allogeneic transplantation. New England Journal of Medicine, 379, 2330–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Collins RH Jr, Shpilberg O, Drobyski WR, Porter DL, Giralt S, Champlin R, Goodman SA, Wolff SN, Hu W, Verfaillie C, List A, Dalton W, Ognoskie N, Chetrit A, Antin JH & Nemunaitis J. (1997) Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. Journal of Clinical Oncology, 15, 433–444. [DOI] [PubMed] [Google Scholar]
  24. Copelan EA. (2006) Hematopoietic stem-cell transplantation. New England Journal of Medicine, 354, 1813–1826. [DOI] [PubMed] [Google Scholar]
  25. Daver N, Garcia-Manero G, Basu S, Boddu PC, Alfayez M, Cortes JE, Konopleva M, Ravandi-Kashani F, Jabbour E, Kadia T, Nogueras-Gonzalez GM, Ning J, Pemmaraju N, DiNardo CD, Andreeff M, Pierce SA, Gordon T, Kornblau SM, Flores W, Alhamal Z, Bueso-Ramos C, Jorgensen JL, Patel KP, Blando J, Allison JP, Sharma P & Kantarjian H. (2019) Efficacy, safety, and biomarkers of response to azacitidine and nivolumab in relapsed/refractory acute myeloid leukemia: a nonrandomized, open-label, phase II study. Cancer Discovery, 9, 370–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, Lukez AP, Avigan D, Chen YB, McSweeney P, LeBoeuf NR, Rooney MS, Bowden M, Zhou CW, Granter SR, Hornick JL, Rodig SJ, Hirakawa M, Severgnini M, Hodi FS, Wu CJ, Ho VT, Cutler C, Koreth J, Alyea EP, Antin JH, Armand P, Streicher H, Ball ED, Ritz J, Bashey A, Soiffer RJ & Leukemia & Lymphoma Society Blood Cancer Research Partnership (2016) Ipilimumab for patients with relapse after allogeneic transplantation. The New England Journal of Medicine, 375, 143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Del Castillo M, Romero FA, Arguello E, Kyi C, Postow MA & Redelman-Sidi G. (2016) The spectrum of serious infections among patients receiving immune checkpoint blockade for the treatment of melanoma. Clinical Infectious Diseases, 63, 1490–1493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Depil S, Deconinck E, Milpied N, Sutton L, Witz F, Jouet JP, Damaj G, Yakoub-Agha I; Societe Francaise de Greffe de Moelle et Therapie cellulaire. (2004) Donor lymphocyte infusion to treat relapse after allogeneic bone marrow transplantation for myelodysplastic syndrome. Bone Marrow Transplantation, 33, 531–534. [DOI] [PubMed] [Google Scholar]
  29. Dermime S, Mavroudis D, Jiang YZ, Hensel N, Molldrem J & Barrett AJ. (1997) Immune escape from a graft-versus-leukemia effect may play a role in the relapse of myeloid leukemias following allogeneic bone marrow transplantation. Bone Marrow Transplantation, 19, 989–999. [DOI] [PubMed] [Google Scholar]
  30. Ferrara JL & Reddy P. (2006) Pathophysiology of graft-versus-host disease. Seminars in Hematology, 43, 3–10. [DOI] [PubMed] [Google Scholar]
  31. Friedman CF, Proverbs-Singh TA & Postow MA. (2016) Treatment of the immune-related adverse effects of immune checkpoint inhibitors: a review. JAMA Oncology, 2, 1346–1353. [DOI] [PubMed] [Google Scholar]
  32. Gooley TA, Chien JW, Pergam SA, Hingorani S, Sorror ML, Boeckh M, Martin PJ, Sandmaier BM, Marr KA, Appelbaum FR, Storb R & McDonald GB. (2010) Reduced mortality after allogeneic hematopoietic-cell transplantation. New England Journal of Medicine, 363, 2091–2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O’Donnell E, Chapuy B, Takeyama K, Neuberg D, Golub TR, Kutok JL & Shipp MA. (2010) Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood, 116, 3268–3277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Han Y, Dong Y, Yang Q, Xu W, Jiang S, Yu Z, Yu K & Zhang S. (2018) Acute myeloid leukemia cells express ICOS ligand to promote the expansion of regulatory T cells. Frontiers in Immunology, 9, 2227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Herrera AF, Moskowitz AJ, Bartlett NL, Vose JM, Ramchandren R, Feldman TA, LaCasce AS, Ansell SM, Moskowitz CH, Fenton K, Ogden CA, Taft D, Zhang Q, Kato K, Campbell M & Advani RH. (2018) Interim results of brentuximab vedotin in combination with nivolumab in patients with relapsed or refractory Hodgkin lymphoma. Blood, 131, 1183–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A & Urba WJ. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. New England Journal of Medicine, 363, 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Jakubowski AA, Petrlik E, Maloy M, Hilden P, Papadopoulos E, Young JW, Boulad F, Castro-Malaspina H, Tamari R, Dahi PB, Goldberg J, Koehne G, Perales MA, Sauter CS, O’Reilly RJ & Giralt S. (2017) T cell depletion as an alternative approach for patients 55 years or older undergoing allogeneic stem cell transplantation as curative therapy for hematologic malignancies. Biology of Blood and Marrow Transplantation, 23, 1685–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Jakubowski AA, Small TN, Young JW, Kernan NA, Castro-Malaspina H, Hsu KC, Perales MA, Collins N, Cisek C, Chiu M, van den Brink MR, O’Reilly RJ & Papadopoulos EB. (2007) T cell depleted stem-cell transplantation for adults with hematologic malignancies: sustained engraftment of HLA-matched related donor grafts without the use of antithymocyte globulin. Blood, 110, 4552–4559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Jakubowski AA, Small TN, Kernan NA, Castro-Malaspina H, Collins N, Koehne G, Hsu KC, Perales MA, Papanicolaou G, van den Brink MR, O’Reilly RJ, Young JW & Papadopoulos EB. (2011) T cell-depleted unrelated donor stem cell transplantation provides favorable disease-free survival for adults with hematologic malignancies. Biology of Blood and Marrow Transplantation, 17, 1335–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Jenq RR & van den Brink MR. (2010) Allogeneic haematopoietic stem cell transplantation: individualized stem cell and immune therapy of cancer. Nature Reviews Cancer, 10, 213–221. [DOI] [PubMed] [Google Scholar]
  41. Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, Hicks M, Puzanov I, Alexander MR, Bloomer TL, Becker JR, Slosky DA, Phillips EJ, Pilkinton MA, Craig-Owens L, Kola N, Plautz G, Reshef DS, Deutsch JS, Deering RP, Olenchock BA, Lichtman AH, Roden DM, Seidman BE, Koralnik IJ, Seidman JG, Hoffman RD, Taube JM, Diaz LA Jr, Anders RA, Sosman JA & Moslehi JJ. (2016) Fulminant myocarditis with combination immune checkpoint blockade. New England Journal of Medicine, 375, 1749–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Khalil DN, Smith EL, Brentjens RJ & Wolchok JD. (2016) The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nature Reviews Clinical Oncology, 13, 273–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B, Jacobsen N, Arcese W, Ljungman P, Ferrant A, Verdonck L, Niederwieser D, van Rhee F, Mittermueller J, de Witte T, Holler E & Ansari H. (1995) Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood, 86, 2041–2050. [PubMed] [Google Scholar]
  44. Kordasti SY, Ingram W, Hayden J, Darling D, Barber L, Afzali B, Lombardi G, Wlodarski MW, Maciejewski JP, Farzaneh F & Mufti GJ. (2007) CD4 + CD25high Foxp3 + regulatory T cells in myelodysplastic syndrome (MDS). Blood, 110, 847–850. [DOI] [PubMed] [Google Scholar]
  45. Kyi C, Carvajal RD, Wolchok JD & Postow MA. (2014) Ipilimumab in patients with melanoma and autoimmune disease. Journal for ImmunoTherapy of Cancer, 2, 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, Kiezun A, Hammerman PS, McKenna A, Drier Y, Zou L, Ramos AH, Pugh TJ, Stransky N, Helman E, Kim J, Sougnez C, Ambrogio L, Nickerson E, Shefler E, Cortes ML, Auclair D, Saksena G, Voet D, Noble M, DiCara D, Lin P, Lichtenstein L, Heiman DI, Fennell T, Imielinski M, Hernandez B, Hodis E, Baca S, Dulak AM, Lohr J, Landau DA, Wu CJ, Melendez-Zajgla J, Hidalgo-Miranda A, Koren A, McCarroll SA, Mora J, Crompton B, Onofrio R, Parkin M, Winckler W, Ardlie K, Gabriel SB, Roberts CWM, Biegel JA, Stegmaier K, Bass AJ, Garraway LA, Meyerson M, Golub TR, Gordenin DA, Sunyaev S, Lander ES & Getz G. (2013) Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature, 499, 214–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, Millenson MM, Cohen AD, Schuster SJ, Lebovic D, Dhodapkar MV, Avigan D, Chapuy B, Ligon AH, Rodig SJ, Cattry D, Zhu L, Grosso JF, Kim SY, Shipp MA, Borrello I & Timmerman J. (2014) Preliminary results of a phase I study of nivolumab (BMS-936558) in patients with relapsed or refractory lymphoid malignancies. Blood, 124, 291–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Liao B, Shroff S, Kamiya-Matsuoka C & Tummala S. (2014) Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma. Neuro-Oncology, 16, 589–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Lleo A, Rimassa L & Colombo M. (2019) Hepatotoxicity of immune check point inhibitors: Approach and management. Digestive and Liver Disease: Official Journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver, 51, 1074–1078. [DOI] [PubMed] [Google Scholar]
  50. Malard F, Labopin M, Cho C, Blaise D, Papadopoulos EB, Passweg J, O’Reilly R, Forcade E, Maloy M, Volin L, CastroMalaspina H, Hicheri Y, Jakubowski AA, Orvain C, Giralt S, Mohty M, Nagler A & Perales MA. (2018) Ex vivo and in vivo T cell-depleted allogeneic stem cell transplantation in patients with acute myeloid leukemia in first complete remission resulted in similar overall survival: on behalf of the ALWP of the EBMT and the MSKCC. Journal of Hematology & Oncology, 11, 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, Jamal-Hanjani M, Wilson GA, Birkbak NJ, Hiley CT, Watkins TB, Shafi S, Murugaesu N, Mitter R, Akarca AU, Linares J, Marafioti T, Henry JY, Van Allen EM, Miao D, Schilling B, Schadendorf D, Garraway LA, Makarov V, Rizvi NA, Snyder A, Hellmann MD, Merghoub T, Wolchok JD, Shukla SA, Wu CJ, Peggs KS, Chan TA, Hadrup SR, Quezada SA & Swanton C. (2016) Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science, 351, 1463–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Merryman RW, Kim HT, Zinzani PL, Carlo-Stella C, Ansell SM, Perales MA, Avigdor A, Halwani AS, Houot R, Marchand T, Dhedin N, Lescaut W, Thiebaut-Bertrand A, Francois S, Stamatoullas-Bastard A, Rohrlich PS, Labussiere Wallet H, Castagna L, Santoro A, Bachanova V, Bresler SC, Srivastava A, Kim H, Pesek E, Chammas M, Reynolds C, Ho VT, Antin JH, Ritz J, Soiffer RJ & Armand P. (2017) Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood, 129, 1380–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Motabi IH, Ghobadi A, Liu J, Schroeder M, Abboud CN, Cashen AF, Stockler-Goldstein KE, Uy GL, Vij R, Westervelt P & DiPersio JF. (2016) Chemotherapy versus hypomethylating agents for the treatment of relapsed acute myeloid leukemia and myelodysplastic syndrome after allogeneic stem cell transplant. Biology of Blood and Marrow Transplantation, 22, 1324–1329. [DOI] [PubMed] [Google Scholar]
  54. Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthelemy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm MO, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B & CheckMate I. (2018) Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. New England Journal of Medicine, 378, 1277–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J, Chaft JE, Segal NH, Callahan MK, Lesokhin AM, Rosenberg J, Voss MH, Rudin CM, Rizvi H, Hou X, Rodriguez K, Albano M, Gordon RA, Leduc C, Rekhtman N, Harris B, Menzies AM, Guminski AD, Carlino MS, Kong BY, Wolchok JD, Postow MA, Long GV & Hellmann MD. (2017) Pneumonitis in patients treated with anti-programmed death-1/ programmed death ligand 1 therapy. Journal of Clinical Oncology, 35, 709–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Noviello M, Manfredi F, Ruggiero E, Perini T, Oliveira G, Cortesi F, De Simone P, Toffalori C, Gambacorta V, Greco R, Peccatori J, Casucci M, Casorati G, Dellabona P, Onozawa M, Teshima T, Griffioen M, Halkes CJM, Falkenburg JHF, Stolzel F, Altmann H, Bornhauser M, Waterhouse M, Zeiser R, Finke J, Cieri N, Bondanza A, Vago L, Ciceri F & Bonini C. (2019) Bone marrow central memory and memory stem T-cell exhaustion in AML patients relapsing after HSCT. Nature Communications, 10, 1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Papadopoulos EB, Carabasi MH, Castro-Mala-spina H, Childs BH, Mackinnon S, Boulad F, Gillio AP, Kernan NA, Small TN, Szabolcs P, Taylor J, Yahalom J, Collins NH, Bleau SA, Black PM, Heller G, O’Reilly RJ & Young JW. (1998) T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease. Blood, 91, 1083–1090. [PubMed] [Google Scholar]
  58. Pardoll DM. (2012) The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer, 12, 252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Perales MA, Ceberio I, Armand P, Burns LJ, Chen R, Cole PD, Evens AM, Laport GG, Moskowitz CH, Popat U, Reddy NM, Shea TC, Vose JM, Schriber J, Savani BN & Carpenter PA. (2015) Role of cytotoxic therapy with hematopoietic cell transplantation in the treatment of Hodgkin lymphoma: guidelines from the American Society for Blood and Marrow Transplantation. Biology of Blood and Marrow Transplantation, 21, 971–983. [DOI] [PubMed] [Google Scholar]
  60. Perales MA, Sauter CS & Armand P. (2016) Fast cars and no brakes: autologous stem cell transplantation as a platform for novel immunotherapies. Biology of Blood and Marrow Transplantation, 22, 17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Peyrony O, Tieghem Y, Franchitti J, Ellouze S, Morra I, Madelaine-Chambrin I, Flicoteaux R, Baroudjian B, Azoulay E, Chevret S & Fontaine JP. (2019) Immune checkpoint blockade toxicity among patients with cancer presenting to the emergency department. Emergency Medical Journal, 36, 306–309. [DOI] [PubMed] [Google Scholar]
  62. Quagliariello V, Passariello M, Coppola C, Rea D, Barbieri A, Scherillo M, Monti MG, Iaffaioli RV, De Laurentiis M, Ascierto PA, Botti G, De Lorenzo C & Maurea N. (2019) Cardiotoxicity and pro-inflammatory effects of the immune checkpoint inhibitor pembrolizumab associated to trastuzumab. International Journal of Cardiology, 292, 171–179. [DOI] [PubMed] [Google Scholar]
  63. Ramchandren R, Domingo-Domenech E, Rueda A, Trneny M, Feldman TA, Lee HJ, Provencio M, Sillaber C, Cohen JB, Savage KJ, Willenbacher W, Ligon AH, Ouyang J, Redd R, Rodig SJ, Shipp MA, Sacchi M, Sumbul A, Armand P & Ansell SM. (2019) Nivolumab for newly diagnosed advanced-stage classic Hodgkin lymphoma: safety and efficacy in the phase II Checkmate 205 study. Journal of Clinical Oncology, 37, 1997–2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ravandi F, Assi R, Daver N, Benton CB, Kadia T, Thompson PA, Borthakur G, Alvarado Y, Jabbour EJ, Konopleva M, Takahashi K, Kornblau S, DiNardo CD, Estrov Z, Flores W, Basu S, Allison J, Sharma P, Pierce S, Pike A, Cortes JE, Garcia-Manero G & Kantarjian HM. (2019) Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study. The Lancet Haematology, 6, e480–e488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Redelman-Sidi G, Michielin O, Cervera C, Ribi C, Aguado JM, Fernandez-Ruiz M & Manuel O. (2018) ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors). Clinical Microbiology & Infection, 24 (Suppl. 2), S95–S107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, [Google Scholar]
  67. Gasmi B, Zappasodi R, Maeda Y, Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN & Chan TA. (2015) Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science, 348, 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Ryder M, Callahan M, Postow MA, Wolchok J & Fagin JA. (2014) Endocrine-related adverse events following ipilimumab in patients with advanced melanoma: a comprehensive retrospective review from a single institution. Endocrine-Related Cancer, 21, 371–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, Kaley TJ, Kendall SM, Motzer RJ, Hakimi AA, Voss MH, Russo P, Rosenberg J, Iyer G, Bochner BH, Bajorin DF, Al-Ahmadie HA, Chaft JE, Rudin CM, Riely GJ, Baxi S, Ho AL, Wong RJ, Pfister DG, Wolchok JD, Barker CA, Gutin PH, Brennan CW, Tabar V, Mellinghoff IK, DeAngelis LM, Ariyan CE, Lee N, Tap WD, Gounder MM, D’Angelo SP, Saltz L, Stadler ZK, Scher HI, Baselga J, Razavi P, Klebanoff CA, Yaeger R, Segal NH, Ku GY, DeMatteo RP, Ladanyi M, Rizvi NA, Berger MF, Riaz N, Solit DB, Chan TA & Morris LGT. (2019) Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nature Genetics, 51, 202–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Schmid C, Labopin M, Nagler A, Bornhauser M, Finke J, Fassas A, Volin L, Gurman G, Maertens J, Bordigoni P, Holler E, Ehninger G, Polge E, Gorin NC, Kolb HJ & Rocha V. (2007) Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. Journal of Clinical Oncology, 25, 4938–4945. [DOI] [PubMed] [Google Scholar]
  71. Schroeder T, Czibere A, Platzbecker U, Bug G, Uharek L, Luft T, Giagounidis A, Zohren F, Bruns I, Wolschke C, Rieger K, Fenk R, Germing U, Haas R, Kroger N & Kobbe G. (2013) Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia, 27, 1229–1235. [DOI] [PubMed] [Google Scholar]
  72. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, Hollmann TJ, Bruggeman C, Kannan K, Li Y, Elipenahli C, Liu C, Harbison CT, Wang L, Ribas A, Wolchok JD & Chan TA. (2014) Genetic basis for clinical response to CTLA-4 blockade in melanoma. New England Journal of Medicine, 371, 2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Steinmann J, Bertz H, Wasch R, Marks R, Zeiser R, Bogatyreva L, Finke J & Lubbert M. (2015) 5-Azacytidine and DLI can induce long-term remissions in AML patients relapsed after allograft. Bone Marrow Transplantation, 50, 690–695. [DOI] [PubMed] [Google Scholar]
  74. Tessoulin B, Delaunay J, Chevallier P, Loirat M, Ayari S, Peterlin P, Le Gouill S, Gastinne T, Moreau P, Mohty M & Guillaume T. (2014) Azacitidine salvage therapy for relapse of myeloid malignancies following allogeneic hematopoietic SCT. Bone Marrow Transplantation, 49, 567–571. [DOI] [PubMed] [Google Scholar]
  75. Toffalori C, Riba M, Zito L, Barcella M, Spinelli O, Crucitti L, Cieri N, Peccatori J, Bernardi M, Bonini C, Cittaro D, Lazarevic D, Rambaldi A, Barlassina C, Stupka E, Ciceri F, Fleischhauer K & Vago L. (2014) Acute myeloid leukemia relapses after allogenenic HSCT display a distinctive immune-related signature, with frequent and functionally relevant alterations in HLA class II antigen presentation and T cell costimulation. Blood, 124, 427. [Google Scholar]
  76. Wayne AS, Giralt S, Kroger N & Bishop MR. (2013) Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: introduction. Biology of Blood and Marrow Transplantation, 19, 1534–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Wei Y, Chen R, Dimicoli S, Bueso-Ramos C, Neuberg D, Pierce S, Wang H, Yang H, Jia Y, Zheng H, Fang Z, Nguyen M, GananGomez I, Ebert B, Levine R, Kantarjian H & Garcia-Manero G. (2013) Global H3K4me3 genome mapping reveals alterations of innate immunity signaling and overexpression of JMJD3 in human myelodysplastic syndrome CD34 + cells. Leukemia, 27, 2177–2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Weiss SA, Wolchok JD & Sznol M. (2019) Immunotherapy of melanoma: facts and hopes. Clinical Cancer Research, 25, 5191–5201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Westin JR, Chu F, Zhang M, Fayad LE, Kwak LW, Fowler N, Romaguera J, Hagemeister F, Fanale M, Samaniego F, Feng L, Baladandayuthapani V, Wang Z, Ma W, Gao Y, Wallace M, Vence LM, Radvanyi L, Muzzafar T, Rotem-Yehudar R, Davis RE & Neelapu SS. (2014) Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. The Lancet Oncology, 15, 69–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Wolchok JD, Neyns B, Linette G, Negrier S, Lutzky J, Thomas L, Waterfield W, Schadendorf D, Smylie M, Guthrie T Jr, Grob JJ, Chesney J, Chin K, Chen K, Hoos A, O’Day SJ & Lebbe C. (2010) Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. The Lancet Oncology, 11, 155–164. [DOI] [PubMed] [Google Scholar]
  81. Yarchoan M, Hopkins A & Jaffee EM. (2017) Tumor mutational burden and response rate to PD-1 inhibition. New England Journal of Medicine, 377, 2500–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Younes A, Santoro A, Shipp M, Zinzani PL, Timmerman JM, Ansell S, Armand P, Fanale M, Ratanatharathorn V, Kuruvilla J, Cohen JB, Collins G, Savage KJ, Trneny M, Kato K, Farsaci B, Parker SM, Rodig S, Roemer MG, Ligon AH & Engert A. (2016) 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. The Lancet Oncology, 17, 1283–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Younes A, Brody J, Carpio C, Lopez-Guillermo A, Ben-Yehuda D, Ferhanoglu B, Nagler A, Ozcan M, Avivi I, Bosch F, Caballero Barrigon MD, Hellmann A, Kuss B, Ma DDF, Demirkan F, Yagci M, Horowitz NA, Marlton P, Cordoba R, Wrobel T, Buglio D, Streit M, Hodkinson BP, Schaffer M, Alvarez J, Ceulemans R, Balasubramanian S, de Jong J, Wang SS, Fourneau N & Jurczak W. (2019) Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: a phase 1/2a study. The Lancet Haematology, 6, e67–e78. [DOI] [PubMed] [Google Scholar]

RESOURCES