Open access peer-reviewed chapter

Immune Checkpoint Inhibitors in Hodgkin Lymphoma and Non-Hodgkin Lymphoma

Written By

Jiawen Huang and Juan Huang

Submitted: 25 July 2022 Reviewed: 29 August 2022 Published: 31 October 2022

DOI: 10.5772/intechopen.107435

From the Edited Volume

Immune Checkpoint Inhibitors - New Insights and Recent Progress

Edited by Afsheen Raza

Chapter metrics overview

138 Chapter Downloads

View Full Metrics

Abstract

Lymphoma, which mainly includes Hodgkin lymphoma (HL) and Non-Hodgkin lymphoma (NHL), is the most common hematological malignance of the lymphoid tissues with significantly heterogeneous characteristics. Tumor immune disequilibrium is involved in tumor development and progression, evading tumor immunosurveillance and suppressing anti-tumor immune responses. The tumor microenvironment (TME) is a complex network that comprises stromal cells and extracellular matrix, playing important roles in the pathogenesis, progression, and drug resistance of lymphoma. Therefore, a promising therapeutic strategy for lymphoma is by targeting the TME to stimulate anticancer immunity either by enhancing the release of immunostimulatory molecules or by mediating immune cell populations. Notably, immune checkpoint therapy (ICT) can provide durable clinical responses and improve overall survival in HL and NHL. However, different subsets of patients with lymphoma have different responses to ICT. Thus, significant challenges remain, including understanding pathways of resistance, optimizing patient selection, improving the management of immune-related adverse events, and identifying rational therapeutic combinations. This will allow a better understanding of the potential applications of ICT in lymphoma, guiding decisions to develop novel combination strategies with maximum efficacy and minimal toxicities for patients.

Keywords

  • tumor microenvironment (TME)
  • immune checkpoint therapy (ICT)
  • lymphoma
  • Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL)

1. Introduction

1.1 Biology of immune checkpoints inhibitors (ICIs)

T-cell activation is central to the immune response [1]. However, uncontrolled T cell activation leads to T cell exhaustion and autoimmune diseases [2, 3]. Therefore, it is crucial to maintain immune homeostasis and the balance of both co-stimulatory and co-inhibitory signals. These signals are thus referred as immune checkpoints. The major co-inhibitory receptors expressed on activated T cells are programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Here, we will briefly discuss their mechanisms of action.

1.1.1 Programmed cell death 1

PD-1 is mainly expressed on mature effector T cells within the peripheral and tumor microenvironment [4], responsible for immune tolerance. Besides T cells, PD-1 expression is also found on B cells, natural killer (NK) cells, dendritic cells (DCs), macrophages, and monocytes [5]. Therefore, it is an inhibitor of both innate and adaptive immunity. In cancers, numerous pathways are responsible for the upregulation of PD-1/PD-L1 signaling; and these major pathways include phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, mitogen-activated protein kinase (MAPK) pathway, Jak-Stat pathway, Wnt pathway, NF-κB pathway, and Hedgehog (Hh) pathway [6]. Upon interaction with its ligands, programmed cell death-ligand 1 or 2 (PD-L1 or PD-L2) expressed on cancer cells or antigen-presenting cells (APCs) of the tumor microenvironments [7, 8, 9], PD-1 signaling leads to T cell dysfunction, reduced cytokine production and anergy, thus protecting cancer cells from immune attack [10].

However, the detailed underlying mechanism of PD-1 signaling requires further elucidation. The inhibitory signal transduction of PD-1 needs both the interaction of PD-1/PD-L1 and peptide/MHC class I complex (MHC-I) from the same cells [11]. Src homology region 2 domain-containing phosphatase-2 (SHP-2) is a major downstream mediator of PD-1 and is capable of inhibiting key molecules and pathways such as ZAP70, PI3K/Akt pathway, and Ras pathway. Ultimately, PD-1 signaling counters the T-cell receptor (TCR) cascade and co-stimulatory receptor CD28 signaling in T cells, leading to reduced T cell activation and proliferation [11, 12]. Moreover, PD-1 can directly exhaust T cells by upregulating the basic leucine transcription factor, ATF-like (BATF) [13]. Interestingly, PD-L1 may protect cancer cells in a PD-1 independent manner, leading to inhibition of autophagy and activation of mammalian target of rapamycin (mTOR; Figure 1) [14]. In addition, PD-1 is also highly expressed on regulatory T cells (Treg), enhancing its proliferation and immunosuppressive effects [12].

Figure 1.

Major immune checkpoints on T cells. PD-1 and CTLA-4 are the major co-inhibitory receptors expressed on activated T cells. Through ZAP70, PD-1 signaling is able to inhibit ZAP70, PI3K/Akt pathway, and Ras pathway, resulting in reduced T-cell activation. PD-1 can also directly induce T cell exhaustion by upregulating BATF. Furthermore, PD-L1 protects cancer cells in a PD-1-independent manner. CTLA-4 is a competitive inhibitor of co-stimulatory receptor CD28. It also inhibits T cell function via inhibition of ZAP70, PI3K/Akt pathway, cell-cycle progression, and trans-endocytosis of CD80/CD86.

1.1.2 Cytotoxic T-lymphocyte-associated protein 4

In contrast to PD-1, CTLA-4 is mainly expressed in the endocytic vesicles of naïve T cells, and it translocates to the cell surface upon TCR activation. CTLA-4 shares the same ligands (CD80 and CD86) with co-stimulatory receptor CD28 (as competitive binding with higher affinity). Therefore, it can suppress T-cell activation [15, 16]. In addition, like PD-1, CTLA-4 is also able to directly inhibit ZAP70 to suppress TCR signaling and reverse T cell activation [17, 18]. Moreover, CTLA-4 exerts its immunosuppressive function via inhibition of PI3K/Akt pathway, cell-cycle progression, and removal of CD80/CD86 from the APCs via trans-endocytosis (Figure 1) [19, 20, 21, 22]. Similar to PD-1, CTLA-4 is constitutively expressed in Tregs for immunosuppression and ligand (CD80/CD86) masking [4].

1.1.3 Blockade of immune checkpoints for cancer therapy

In cancers, the suppressive immune checkpoints introduced above are likely dysregulated, allowing them to escape from immune surveillance [23]. Therefore, blocking such immune checkpoints by antibodies is able to reverse the immune suppression for the treatment of cancers [24]. Preclinical studies have indicated that inhibition of immune checkpoints is able to enhance anti-tumor immunity. In the 1990s, initial research already indicated that the blockade of CTLA-4 by antibodies is able to reduce tumor burden in murine models [25, 26]. Since then, enormous advancements have been achieved in the use of immune checkpoint inhibition in cancer treatment, and the monoclonal antibodies targeting CTLA-4 and PD-1 have been approved by US Food and Drug Administration (FDA) for different cancers [27, 28]. In the following part of the chapter, we will summarize the current applications of immune checkpoint inhibitors (ICIs) for Hodgkin lymphomas (HLs) and non-Hodgkin lymphomas (NHLs).

Advertisement

2. Immune checkpoint inhibitors in Hodgkin lymphoma

2.1 Anti-PD-1 checkpoint inhibitors

In classical HL (cHL), malignant Reed-Sternberg cells harbor a recurrent chromosome 9p24.1 amplification. Such genetic abnormality encodes PD-L1 and PD-L2, as well as JAK2, which further upregulates PD-1 ligand via the JAK-STAT pathway [29]. This upregulated PD-1 signaling allows cHL to suppress surrounding immune cells and survive from immune surveillance. Therefore, blocking PD-1 is likely to restore anti-tumor immunity and eradicate HL cells.

2.1.1 Nivolumab and pembrolizumab

Nivolumab and pembrolizumab are among the first fully human anti-PD-1 IgG4 monoclonal antibodies approved by the US FDA (May 2016 for nivolumab and March 2017 for pembrolizumab) for the treatment of relapsed or progressed cHL after autologous hematopoietic stem cell transplantation (auto-HSCT) and brentuximab vedotin (therefore referred as relapsed/refractory, r/r) [30, 31]. Since then, numerous clinical trials and real-world experiences have demonstrated the efficacy and safety profiles of nivolumab and pembrolizumab against HL, which is mainly (but not limited to) r/r cHL. Nivolumab and pembrolizumab are the most common ICIs used for r/r cHL patients, and many groups use both drugs in the same clinical trials (refers to them both as PD-1 ICIs). The clinical data of pembrolizumab are summarized in Table 1 together with nivolumab.

GroupsStudy statusResponsesSurvivalAEs
DiseaseDrugNumberFollow-upORRCRPRPFSOSTreatment-related AEs>Grade 3
Liput et al. [32]cHLNivo10NA70%60%10%80%20%
Davis et al. [33]r/r cHL (young patients)Nivo1030 d30%10%20%Not reported for cHL separately
Kasamon et al. [34]r/r cHLNivo956 mo65%7%58%Lack overall summary
Bair et al. [35]r/r cHLNivo, Pembro5313 mo68%45%23%12 mo: 75%
Median: 29 mo
12 mo: 89%Lack overall summary
Armand et al. [36]r/r cHL after auto-HSCTNivo24318 mo69%16%53%Median: 14.7%12 mo: 92%Lack overall summary
Falchi et al. [37]r/r cHLNivo, Pembro99.9 mo89%78%11%100%67%
Armand et al. [38]r/r cHLPembro3117 mo65%16%48%24 wk.: 69%
52 wk.: 46%
24 wk.: 100%97%16%
Ansell et al. [39]r/r cHLNivo2340 week87%17%70%24 wk.: 86%78%22%
Chen et al. [40]r/r cHLPembro21027.6 mo72%28%44%Median: 13.7 mo24 mo: 100%73%12%
Younes et al. [41]r/r cHLNivo808.9 mo66%9%57%6 mo: 76.9%6 mo: 98.7%99%40%
Bekoz et al. [42, 43]r/r cHLNivo8729 mo70%36%34%24 mo: 58.5%24 mo: 78.7%58%12% of AEs
Geoerger et al. [44]r/r HL pediatric patientsPembro158.6 mo60%13%47%Median: 12.2 mo
6 mo: 72.7%
12 mo: 51.9%
6 and 12 mo: 100%Not reported for cHL separately
Maruyama et al. [45]r/r cHLNivo1638.8 mo87.5%31.3%56.3%Median: 11.7 mo3 yr.: 80.4%100%50%
Ramchandren et al. [46]untreated, advanced-stage cHLNivo519.4 mo84%67%17%9 mo: 92%96%59%
Chan et al. [47]r/r cHLPembro (low dose)11100%73%27%Median: 35 mo27.20%
Chan et al. [47]r/r cHLNivo
(low dose)
6100%67%17%Median: 33 mo67%0%
Dada et al. [48]r/r cHLNivo1012.3 mo80%70%10%40%0%
Kuruvilla et al. [49]r/r cHLPembro15125.7 mo65.6%25%41%Median: 13.2 mo75%20%
Momotow et al. [50]r/r HLNivo, Pembro6020.4 mo65%18%47%2 yr.: 42.3%2 yr.: 78.4%
3 yr.: 65.8%
32%
Hur et al. [51]Pretreated cHLNivo, Pembro2014 mo75%45%30%Median: 18 moMedian: 36 moLack overall summary
Lepik et al. [52]r/r cHLNivo9921 mo64%31%33%Median: 19.4 mo88%17%
Armand et al. [53]cHL after BV failurePembro3152.8 mo58%19%39%Median: 11.4 mo
24 mo: 30%
24 mo: 87%
36 mo: 81%
71%19%

Table 1.

Overview of clinical efficiency and toxicity results of PD-1 inhibitors, nivolumab and pembrolizumab, in Hodgkin’s lymphoma.

Numbers: number of patients; Follow-up: median or minimum follow-up; ORR: overall response rate; CR: complete response; PR: partial response; PFS: progression free survival; OS: overall survival; AEs: adverse events; r/r cHL: relapsed/refractory classical Hodgkin’s lymphoma; d: days; mo: months; yr.: years; Nivo: nivolumab; Pembro: pembrolizumab; BV: brentuximab vedotin.

As shown in Table 1, the objective response rate (ORR) for PD-1 ICIs is generally high (usually over 70%). However, the CR is rarely achieved, with a CR rate of around 30%–40%. Notably, some preconditions or previous treatments the patients experienced significantly enhance the outcomes of anti-PD-1 therapy. For example, 5 of 5 r/r cHL patients who have been given hypomethylating agents 5-azacitidine all achieved CR after ICI treatment [37]. This may suggest some potential combination therapies and numerous groups are assessing the efficacy of different treatment combinations.

2.1.1.1 Combination of PD-1 inhibitors and HSCT

The poor CR rate of anti-PD-1 antibodies suggests that monotherapy of PD-1 blockade alone may be not sufficient to cure r/r HL. Therefore, combined therapy of PD-1 blockade with other additional canonical treatments is necessary. It has been demonstrated that administration of PD-1 inhibitors before/after allogenic (allo-) or autologous (auto-) HSCT significantly enhances response rate and prolongs patient survival. Manson et al. [54] reported that none of the 13 r/r HL patients who underwent consolidation treatment of allo-HSCT together with nivolumab suffered from disease relapse. On contrary, 62.2% of those (n = 37) who did not undergo subsequent allo-HSCT relapsed. In another similar study, Merryman et al. [55] studied the 209 cHL patients who underwent subsequent allo-HSCT after PD-1 inhibition. With a median follow-up of 24 months, they reported that the 2-year progression-free survival (PFS) and overall survival (OS) were 69% and 82%, respectively. Merryman et al. also suggested that a shorter interval between PD-1 inhibition and allo-HSCT can significantly boost the graft-versus-lymphoma (GVL) effect of allo-HSCT. The real-life experience of 74 patients who underwent allo-HSCT after nivolumab treatment in Spain provided a similar conclusion (i.e., improved PFS and OS) as well [56].

Similarly, consolidation after auto-HSCT by PD-1 blockade also improves the treatment outcomes [57]. In this clinical trial (NCT02362997), the expected PFS after auto-HSCT increased from 60 to 82% upon pembrolizumab administration. Casadei et al. [58] also reported that auto-HSCT after PD-1 blockade further improved patient survival, with an estimated 5-year PFS of 73.4% and 4.8-year OS of 92.3%.

Although the combination of PD-1 inhibition and allo-HSCT seems to be a promising strategy against HL, increased graft-versus-host disease (GVHD) upon anti-PD-1 administration is a major concern of this treatment option [55, 5659]. Such GVHD can be severe and may cause multi-organ failure and even death [555659, 60, 61, 62, 63, 64]. Therefore, multiple studies indicated that post-transplant cyclophosphamide (PTCy)-based GVHD prophylaxis is required for improved PFS and therefore strongly suggested [55, 59].

2.1.1.2 Other combined therapy

The regimen AVD regimen (doxorubicin, vinblastine, and dacarbazine) is the backbone of the well-established chemotherapy regimen ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) for HL [65]. Therefore, the efficacy of nivolumab and AVD combination in early-stage cHL was assessed [66]. In total, 109 patients were given two different treatment strategies (of dosing and sequencing), and both groups displayed promising outcomes, with over 90% CR and nearly 100% 12-month PFS. Another multicenter, single-arm, phase II trial proved that pembrolizumab followed by AVD was both effective and safe in patients with untreated early unfavorable and advanced-stage cHL, with all patients (n = 30) achieving complete metabolic response (CMR) [67]. At the median follow-up of 22.5 months, the PFS and OS are 100%, indicating the superior efficacy of the strategy.

Brentuximab vedotin (BV) is a CD30-based antibody-drug conjugate. When used alone, it can lead to an ORR of 72% and CR rate of 33% in r/r HL patients [68]. Advani et al. [69] reported that BV combined with nivolumab can be the first salvage therapy in patients with r/r cHL, with an ORR of 85% and CR rate of 67%. In a median follow-up of 34.3 months, the estimated 3-year PFS and OS were 77% and 93%, respectively. Such combination treatment can be applied as a first-line option for older or chemotherapy-ineligible cHL patients, as demonstrated by Cheson et al. [70]. With a total of 46 patients and a median follow-up of 21.2 months, 48% of patients achieved CR and 13% achieved PR, with an ORR of 61%. Due to the high efficacy of this combination, it was considered as a salvage option after PD-1 blockade failure. In 21 r/r cHL patients who failed nivolumab monotherapy previously, BV combined with nivolumab resulted in an ORR of 57% [71]. Twenty-four-month PFS and OS were 31% and 80%, respectively. In total, 63% of patients suffered from adverse effects (AEs), but AEs of grade 3 or 4 were only observed in 10% of patients.

The potential synergistic effect of radiotherapy and ICIs has been proposed as well. In a cohort of 12 patients with r/r cHL, patients were given a combined treatment of radiotherapy and nivolumab/pembrolizumab, with an ORR of 100% and a CR rate of 58% [72]. With a median follow-up of 18 months, 92% of patients remained in CR (9 of 12 patients underwent HSCT consolidation). Forceville et al. [73] presented two case reports supporting that radiotherapy combined with nivolumab can lead to excellent outcomes.

Gemcitabine, vinorelbine, and liposomal doxorubicin (GVD) are traditional second-line treatment options for r/r cHL, with a CR rate of around 50% [74]. In comparison, the combination of GVD and pembrolizumab resulted in an ORR of 100% and a CR rate of 95%, with a total of 39 enrolled r/r cHL patients [75]. In total, 36 of these 39 patients underwent subsequent auto-HSCT, and they all remained in CR at a median post-transplant follow-up of 13.5 months. In a similar trial consisting of 103 patients (27 for GVD + PD-1 blockade, 76 for GVD), the combination group had a higher CR rate of 85.2% (65.8% for the GVD group) and an extended PFS (1-year PFS of 82.2% vs. 67.9% for GVD group) [76].

2.1.2 Camrelizumab

Camrelizumab (SHR-1210), which was developed in China, is a humanized high-affinity anti-PD-1 IgG4 monoclonal antibody. It has shown promising efficacy against numerous advanced solid tumors including nasopharyngeal carcinoma, esophageal carcinoma, gastric and gastroesophageal junction cancer [77, 78, 79, 80, 81]. In a single-arm, multicenter, phase II study (NCT03155425), a total of 75 patients with r/r cHL were given Camrelizumab 200 mg every 2 weeks intravenously. In a median follow-up of 12.9 months, 21 (28.0%) and 36 (48.0%) patients achieved complete or partial remission, respectively (i.e., objective response rate is 76.0%). Treatment-related adverse events (AE) were observed in all patients enrolled, with 20 (26.7%) of them exhibiting grade 3 or 4 treatment-related AEs [82]. The group further extended the follow-up of this clinical trial till 2020, with a median follow-up duration of 36.2 months. The objective response rate remained almost unchanged. The median PFS was 22.5 months and 3-year OS was 82.7%.

2.1.2.1 Combined therapy

Like other checkpoint inhibitors, although camrelizumab exhibits a high objective response rate in patients with r/r cHL, the CR rate remains low (as shown above). It has been proven that inhibition of de novo DNA methylation can boost T-cell function upon PD-1 blockade [83, 84]. Decitabine is a DNA demethylating agent [85]; therefore, clinical trial combining a low dose of decitabine with camrelizumab against r/r cHL was conducted (NCT02961101, NCT03250962). Indeed, when compared with camrelizumab monotherapy, r/r cHL patients receiving decitabine plus camrelizumab exhibited a higher CR rate (79% vs. 32%) and longer median PFS (35.0 vs. 15.5 months) [86, 87]. In addition, the administration of decitabine plus camrelizumab showed promising efficacy against HL with resistance to anti-PD-1 [86, 87]. Similarly, combination treatment of anti-angiogenic agent apatinib and camrelizumab might be a salvage option for r/r cHL patients who failed PD-1/PD-L1 inhibitor therapy, as demonstrated in the case reports presented by Yan et al. [88]. Out of seven enrolled patients, two achieved CR, and four achieved PR. The median PFS was 10 months, and no unexpected side effects were observed.

2.1.3 Sintilimab

Sintilimab is an anti-PD-1 antibody developed by Innovent Biologics, Suzhou, China. Shi et al. reported it exhibits comparable activity to nivolumab and pembrolizumab in patients with r/r cHL [89]. In their single-arm, multicenter, phase II trial (NCT03114683), 6-months PFS was 77.6%, and 74 of 92 fully analyzed patients (80.4%) achieved an objective response. Among those with objective responses, 31 (34%) had CR and 43 (47%) had PR. As for AE, 89 (93%) of 96 patients demonstrated treatment-related AE, including 17 (18%) with grade 3 or 4 and 11 (11%) with serious treatment-related AE (all expected).

2.1.4 Tislelizumab

Tislelizumab is a specially engineered humanized anti-PD-1 IgG4 monoclonal antibody. In contrast to other conventional PD-1 inhibitors, the Fcγ receptor (FcɤR) fragment of tislelizumab was modified to minimize the binding of macrophages and the subsequent antibody-dependent phagocytosis. The antibody-dependent phagocytosis by macrophages could potentially lead to T-cell clearance and greatly affect the efficacy of anti-PD-1 therapy [90]. Therefore, the FcɤR modification allows tislelizumab to exhibit improved anti-tumor function. In the single-arm, multicenter, phase II trial of tislelizumab in patients with r/r cHL (NCT03209973) [91], 61 of 70 (87.1%) patients achieved an objective response, including a high CR rate of 62.9% (44 of 70). The estimated median 9-month PFS was 74.5%. AEs were observed in 65 of 70 (92.9%) patients, with 15 (21.4%) experiencing grade 3 or 4 AEs.

2.1.4.1 Combined therapy.

Similar to other anti-PD-1 antibodies, co-administration of low-dose decitabine and tislelizumab for the treatment of r/r cHL has been reported. A 27-year-old male r/r cHL patient who failed eight lines of therapy (including PD-1 inhibition) achieved partial remission upon receiving decitabine plus tislelizumab treatment. No disease progression was observed during the entire 11.5 months of follow-up [92].

2.1.5 Zimberelimab

Zimberelimab (GLS-010) is the first fully human anti-PD-1 monoclonal antibody produced in a transgenic rat platform. While sharing the same heavy chain constant region as nivolumab and pembrolizumab, zimberelimab has two different modifications, namely S228P and N95S, in IgG4 core-hinge area and CDR3 area of the light chain, respectively. The S228P mutation prevents Fab-arm exchange, and the N95S mutation prevents the glycosylation of the antigen-binding domain [93]. Phase I studies for advanced solid tumors [94, 95] or preliminary studies for r/r cHL [96] have suggested high efficacy and acceptable safety. In a phase II trial for patients with r/r cHL (NCT03655483), 77 of 85 (90.6%) patients had objective responses, with a CR rate of 32.9% (28 patients). Twelve-month PFS and OS were 78% and 99%, respectively. Treatment-related AEs were found in 79 of 85 (92.9%) patients, with 24 (28.2%) of them demonstrated grade 3 or 4 and 1 exhibited grade 5 treatment-related AE (gastrointestinal infection) [93].

2.1.6 Penpulimab

Penpulimab is a humanized anti-PD-1 monoclonal antibody co-developed by Akeso Biopharma and Chia Tai Tianqing for the treatment of solid tumors. Similar to tislelizumab, the FcɤR fragment region of penpulimab is engineered, through which the FcɤR bindings of effectors (such as macrophages) are eliminated. As the results, T cells are protected from antibody-dependent cell-mediated cytotoxicity (ADCC), and the efficacy of tislelizumab is expected to be enhanced. In the open-label, multicenter, single-arm, phase I/II study (NCT03722147), the objective response rate was 89.4% (76 of 85 patients), with 40 (47.1%) patients achieving CR. Twelve-month PFS was 72.1%. Treatment-related AEs were observed in 97.9% (92 of 94) patients, with 25 (26.6%) experienced grade 3 or above treatment-related AEs [97, 98].

2.2 Anti-PD-L1 checkpoint inhibitors

Besides PD-1 blockade, targeting PD-L1 is an alternative strategy to avoid PD-1/PD-L1 immune checkpoints. However, it should be noted that PD-L1 and PD-L2, the two ligands to PD-1, are differentially expressed in the tumor microenvironment of cHL [29, 99]. Therefore, anti-PD-L1 monotherapy may be not sufficient to completely inhibit the PD-1 pathway, its efficacy may be lower than PD-1 inhibition alone. The use of PD-L1 inhibitors in HL should be carefully evaluated.

2.2.1 Avelumab

Avelumab (MSB0010718C) is a human anti-PD-L1 IgG1 monoclonal antibody. Besides blocking PD-1/PD-L1 interactions, the binding of avelumab on tumor cells induces ADCC via the FcɤR binding [100, 101]. Unlike the ADCC induced by anti-PD-1 antibodies that impair T-cell function and dampen the efficacy of treatment, the ADCC induced by anti-PD-L1 antibodies provides another mechanism of tumor clearance and further enhances treatment efficacy. In a phase Ib trial of avelumab against r/r cHL [102], 13 of 31 (41.2%) patients showed an objective response, with six (19.4%) achieving CR and seven (22.6%) achieved PR. Twelve-month PFS was 18.2%, and the median PFS was 5.7 months. Treatment-related AEs were observed in 26 (86.7%) patients and 13 (43.3%) of them are grade 3 or 4.

2.2.2 Sugemalimab

Sugemalimab is a fully human, full-length, anti-PD-L1 IgG4 monoclonal antibody developed by CStone Pharmaceuticals for advanced solid tumors and lymphoma. In 2021, it has been approved in China for the first-line treatment of various forms of non-small-cell lung cancer in combination with different treatments. Phase Ia and Ib studies have been finished for sugemalimab against advanced malignancies (including 5 cHL patients in phase Ia study) [103]. They have demonstrated the safety and anti-tumor efficacy of sugemalimab. Currently, a single-arm, phase 2 trial of sugemalimab against r/r cHL (as monotherapy) is underway (NCT03505996) and has enrolled 80 patients [104].

2.2.3 Durvalumab

Durvalumab is another human anti-PD-L1 monoclonal antibody and has been approved by US FDA for urothelial carcinoma and stage III non-small-cell lung cancer [105]. Ogasawara et al. have conducted a pharmacokinetic analysis of durvalumab in 267 patients with hematological malignancies (including HL) [105]. They suggested the dosing regimen (1500 mg every 4 weeks) for hematologic malignancies can be the same as other solid tumors. This suggests a potential application of durvalumab against HL.

2.2.4 Atezolizumab

Atezolizumab is an inhibitor of PD-L1, and it has been approved by the US FDA and the European Medicines Agency for certain forms of solid tumors (such as triple-negative breast cancer or non-small-cell lung carcinoma, as monotherapy or used in combination) [106]. iMATRIX was a multicenter, open-label, phase I/II trial of young patients (<30 years old) with solid tumors or lymphomas (including nine HL patients, NCT02541604) [106]. Unfortunately, only two patients demonstrated objective response (PR). For the rest of the HL patients, two of them remained with stable disease, and five suffered from disease progression. Another phase II clinical trial of atezolizumab in r/r HL (NCT03120676) was also terminated due to lack of accrual.

2.3 Anti-CTLA-4 checkpoint inhibitors

2.3.1 Ipilimumab

In contrast to the wild application of anti-PD-1/PD-L1 inhibitor for the treatment of HL, very few studies have been conducted to assess the efficacy of anti-CTLA-4 inhibitor against HL. Ipilimumab is a fully humanized anti-CTLA-4 IgG1κ monoclonal antibody. Although several clinical trials of ipilimumab have been conducted for numerous solid tumors [107, 108, 109, 110], and there are some ongoing clinical trials assessing the possibility of using ipilimumab in r/r cHL (like NCT04938232); currently very few reports have demonstrated the efficacy of ipilimumab as monotherapy in the treatment of HL. Bashey et al. reported that two out of 14 relapsed HL patients after allo-HSCT achieved CR upon ipilimumab treatment [111]. In comparison, the possibility of co-administrating ipilimumab with other agents against HL has been evaluated.

2.3.1.1 Combined therapy

In an open-label, multicenter, phase I trial assessing the efficacy of combination therapy in 61 patients with r/r HL (NCT01896999), patients were divided into three groups: combinations of brentuximab vedotin with ipilimumab (ipi-group) or nivolumab (nivo-group) or both (triplet-group) [112]. Although the overall response rates were similar for all three groups (76% for ipi-group, 89% for nivo-group, and 82% for triplet-group), triplet groups demonstrated a higher CR rate (73%), as compared with ipi- (57%) and nivo-groups (61%). These are also higher than the expected individual monotherapies. However, the inclusion of ipilimumab in the combination therapy significantly increased the chance of severe (grade 3 or 4) treatment-related AEs, with 43% in the ipi-group and 50% in the triplet-group. On contrary, this number is only 16% in the nivo-group. This may raise concerns for the possible higher toxicity of ipilimumab in treating Hodgkin lymphoma.

Lenalidomide is an FDA-approved drug for the treatment of multiple myeloma, with the ability of modulating cellular and humoral immunity and antiangiogenesis [113]. In a phase I dose-escalation study of ipilimumab and lenalidomide including seven refractory HL patients (NCT01750983) [114], PR was observed in one patient, and three patients experienced tumor shrinkage (less than PR).

2.4 Other potential immune checkpoint inhibitors

Besides the well-known immune checkpoints PD-1 and CTLA-4, novel immune checkpoints may be used as therapeutic targets for the treatment of HL. Halabi et al. found that lymphocyte-activation gene 3 (LAG-3) and T-cell immunoglobulin and mucin-domain containing 3 (TIM-3) are almost constitutively expressed in cHL [115]. Therefore, clinical trials targeting LAG-3 (relatlimab, NCT02061761) or TIM-3 (BMS-986258, NCT03446040) alone or in combination with nivolumab in the treatment of r/r HL are completed, and results will be released soon. In addition, T-cell Ig and ITIM domains (TIGIT) are another immune checkpoint receptor that is found to be highly co-expressed with PD-1 in r/r cHL patients [116]. Therefore, co-inhibition of PD-1 and TIGIT could be a novel strategy for treating r/r cHL.

Immune checkpoints are expressed in immune cells other than T cells, which could be targeted as well. In a phase Ib study, Armand et al. evaluated the efficacy and safety of dual inhibition of PD-1 and CTLA-4 (65 patients) or killer immunoglobulin-like receptors (KIRs) (72 patients) for r/r cHL [117]. KIR is expressed on NK cells and inhibits their function by interacting with MHC I [118]. However, the authors reported that the combination failed to further improve the efficacy, as compared with nivolumab monotherapy.

Advertisement

3. Immune checkpoints inhibitors in non-Hodgkin lymphoma

Non-Hodgkin lymphoma (NHL) is a mostly common and heterogeneous group of lymphomas derived from B and T lymphocytes, natural killer (NK), cells or precursors of these cells. Its pathology remains largely unexplained. Recent studies identified that tumor microenvironment (TME) in NHL is now playing a significant role in immune suppression and propagating tumor growth [119, 120]. Therefore, immunotherapies have been widely used and investigated in NHL to enhance or manipulate host anti-tumor immunity. In recent years, interference of PD-1/PD-L1 signaling, the immune checkpoint (therefore also known as checkpoint blockade), has been used in these kinds of lymphomas for its clinical efficacy by enhancing anti-tumor immune response. More importantly, therapeutic interference of checkpoint blockade has enjoyed significant success in cHL, but clinical response greatly varied in NHLs [121].

PD-1 and its ligands (PD-Ls), PD-L1 (also known as CD274 or B7-H1) and PD-L2 (as known as CD273 or B7-DC), form a signaling network that serves as a checkpoint to limit T-cell immunity, causing T-cell exhaustion [6, 122, 123]. Targeting PD-1 signaling to block T-cell activity with immune inhibitory antibodies can promote the activation, maturation, and proliferation of T-cells, eventually regulating anti-tumor activity, which has been investigated in NHL. Recent studies suggested that ICIs have been considered a promising and effective treatment strategy for some types of NHLs. Thus, PD-1 antibodies have been approved by the US FDA including nivolumab and pembrolizumab. Now, let us review the effectiveness of ICIs in NHL.

3.1 Immune checkpoints inhibitors in B-non-Hodgkin lymphoma

3.1.1 Diffuse large B-cell lymphoma (DLBCL)

Diffuse large B-cell lymphoma (DLBCL) represents 30–40% of all non-Hodgkin lymphomas (NHL) with a 60–70% curable rate in Rituximab Era [120]. However, about one-third of these patients are refractory or resistant to standard treatment. In addition, there are several subtypes of DLBCL in the 2016 World Health Organization (WHO) classification of lymphoid malignancies according to unique clinical and pathological features, including primary DLBCL of the central nervous system (PCNSL), primary cutaneous DLBCL, leg type, T-cell/histiocyte-rich large cell lymphoma, and EBV positive DLBCL of the elderly [124]. Nevertheless, most cases of DLBCL fall into the “not otherwise specified” (NOS) category [125]. As we know, immune evasion plays an important pathogenetic mechanism in DLBCL evolution, and immune checkpoint blockade therapy was explored in all kinds of lymphomas. But the outcome of immunotherapy remained controversial.

PD-L1 expression in DLBCL, with an incidence of ~25%, is associated with inferior outcomes, involving in DLBCL pathogenesis, which is considered a potential target [126, 127]. Importantly, chromosome 9p24.1 copy number alteration observed in DLBCL, in addition to cHL, is also involved in negative T cell regulation and NF-κb signaling pathway, which is associated with responsiveness to ICIs in relapsed/refractory DLBCL (r/r DLBCL) [126]. However, the results of ICIs in r/r DLBCL are disappointing [128, 129]. A phase I study to evaluate the safety and efficacy of nivolumab enrolled 81 r/r lymphoma patients (11 DLBCL) and showed an ORR of 36% in DLBCL. A recent phase II study (NCT02038933) showed that nivolumab monotherapy had good safety profiles but low ORR in DLBCL patients [130]. However, clinical trials of nivolumab combined with other immunochemotherapies are still in progress. Pembrolizumab (Keytruda), a humanized anti-PD-1 MoAb with excellent anti-tumor activity, was explored in DLBCL. This study including 30 DLBCL patients, evaluated the efficacy of pembrolizumab (200 mg) with R-CHOP, and showed a 90% ORR, 77% CR, and 83% 2y-PFS at a median follow-up of 25.5 months, suggesting that this combination may be a promising treatment strategy [131]. All in all, the results of anti-PD-1 antibody in DLBCL patients are not promising in the current clinical trials, and anti-PD-1 antibody combination therapy is also under investigation [126].

It is a worthy note that anti-PD-L1 antibody atezolizumab (MPDL-3280A) combined chemotherapy seems a promising approach in DLBCL. In a phase I/II study, atezolizumab-R-CHOP for DLBCL demonstrated high efficacy (ORR of 87.5%) and durable responses (24 months for 80% of patients) for the combinational group [132]. Fifty-eight DLBCL patients enrolled in the study to assess the anti-tumor activity of atezolizumab associated with Venetolax (a BCL-2 inhibitor) and Obinutuzumab with 23.6% ORR (NCT03276468). In another phase 1/2 study, atezolizumab in combination with rituximab and polatuzumab in 21 participants with r/r DLBCL showed 57.14% ORR and 33.33% CR. In additional, atezolizumab with mosunetuzumab (a bispecific CD20-CD3 monoclonal antibody) was evaluated (NCT02500407). Certainly, anti-PD-L1 antibodies have been extensively investigated in combination with new-generation CD20 antibodies (NCT03533283), Chimeric antigen receptor (CAR)-T (NCT02926833), and ASCT (NCT02362997).

Durvalumab, another humanized IgG1-kappa monoclonal antibody against PD-L1, showed markedly anti-tumor activity in vivo. Thus, like atezolizumab, numbers of clinical trials are ongoing to investigate the value of durvalumab as a single agent or in combination with other treatment approaches or CAR T-cells in B-NHL patients. Encouraging results were commonly seen in patients treated with durvalumab in combination therapy in early studies. Durvalumab with Ibrutinib in DLBCL has 25% ORR and 4.6 months PFS [133]. Also, durvalumab combined with R-CHOP showed 54.10% CR but 51% serious AEs [134]. More interesting, remarkable results were found when combined with durvalumab and CAR T-cells in B-NHL including 12 DLBCLs, 2 high-grade B-cell lymphomas, and 1 PMBL (NCT03310619 (PLATFORM) and NCT02706405), which reported an ORR of 91%, including 64% CR [135, 136]. From these clinical results, AEs were frequently seen in combination therapy, which needed to be noted [137, 138].

Another inhibitor signaling of CTLA-4 including ipilimumab was not explored for its efficacy and safety. Recently, the combination of ipilimumab and nivolumab in patients with high-risk DLBCL after Allo-SCT has been opened (NCT02681302) [139]. More results should be worthy of expectation.

3.1.2 Primary mediastinal large B-cell lymphoma (PMBCL)

Importantly, anti-PD-1 MoAb has promising results in some special DLBCL. Primary mediastinal large B-cell lymphoma (PMBCL) comprises approximately 10% of DLBCL with different clinicopathologic and molecular signature, which have a good prognosis with R-CHOP/R-DAEPOCH combined with radiotherapy, with a 5-year event-free survival rate of 93% and OS rate of 97% [140]. However, more than 10% of patients still suffered relapsed or refractory, and the outcomes in r/r PMBCL remain poor. Studies have elucidated that PMBCL shared many similar biological features with cHL, including the importance of JAK-STAT and NF-κB signaling pathways as well as immune evasion [141]. Aberration expressions of PD-L1 and PD-L2 were found in PMBCL tumors, and the efficacy of anti-PD-1 antibody (pembrolizumab) in r/r PMBCL was confirmed in phase 1 KEYNOTE-013 study [142]. Subsequently, a phase 2 study (KEYNOTE-170) has evaluated the efficacy of pembrolizumab in r/r PMBCL, and similar results were observed, with 45% ORR, and 13% CR, and median duration of response (DOR) not yet reached [143]. Thus, pembrolizumab has been approved in r/r PMBCL by FDA. After that, ICIs combined with other therapeutic agents for r/r PMBCL have been widely studied all over the world. Nivolumab combined with the anti-CD30 antibody-drug conjugate (ADC) brentuximab vedotin (BV) has been studied for r/r PMBCL in the CheckMate 436 study with an ORR was 73% and CR 37% [144]. These studies have identified the efficacy of PD-1 in PMBCL, especially combined with other agents. Numerous clinical trials assessing combination therapies with immune checkpoint inhibitors are ongoing [145].

3.1.3 Epstein-Barr virus (EBV)+ diffuse large B-cell lymphoma (DLBCL) and primary DLBCL of the central nervous system (PCNSL)

Epstein-Barr virus (EBV) is detected in a variety of B-cell lymphomas (BCLs) and lymphoproliferative disorders (B-LPD) with poor prognosis, associated with immunodeficiency, a key factor of lymphomagenesis. EBV+ DLBCL-NOS was first described as age-related EBV-associated LPD in 2003 with poor outcomes compared with EBV-negative DLBCL patients [146]. Unfortunately, the biology of EBV+ DLBCL-NOS remains unsure, and no standard approaches for these kinds of patients. Researchers have identified that 100% PD-L1 expression was seen in EBV+ DLBCL in a larger cohort study (n = 1100), which was significantly associated with EBV+ status [147, 148]. Liu et al. have identified that anti-PD-1 antibodies can restore and active function of T cells in EBV+ DLBCL [149]. Thus, the PD-L1/PD-1 pathway may be a potential therapeutic target for EBV+ DLBCL. Many studies are ongoing to assess the application of ICIs combined with chemotherapies in EBV+ DLBCL (e.g., NCT03212807, NCT04181489, NCT04705129, and so on).

Also, primary central nervous lymphoma (PCNSL) is a rare extra-nodal lymphoma with a high refractory/relapse rate using high-dose MTX-based treatment [150, 151]. For relapsed/refractory PCNSL (r/r PCNSL), new strategies have been explored including immunotherapy. PD-1 antibodies in r/r PCNSL have been reported with good efficacy in some case reports [152, 153]. Thus, some retrospective and prospective studies discussed the efficacy of anti-PD-1 antibody as monotherapy and in combination with other drugs [153, 154]. Unluckily, the results of ICIs in r/r PCNSL varied. Especially, we have seen promising efficacy in PCNSL with Bruton’s Tyrosine Kinase and Immune Modulatory Small Molecules. Some explorations of ICIs in primary and r/r PCNSL are under study, especially for those who have higher PD-L1 expression and no chance to do MTX-based chemotherapy (NCT04899427, NCT05425654, and NCT04831658).

In other B-NHL entities, the rates of PD-L1 expression on neoplastic B cells are low: ∼5% in FL, ∼10% in high-grade MZL, and 0% in MCL. Therefore, slightly rare studies have been explored in these types of B-NHL.

3.2 Immune checkpoints inhibitors in T-non-Hodgkin lymphoma

PD-1 or PD-L1 has been used in various kinds of NHLs, either alone or in combination with other agents, which have promising results in some Lymphoma. For T-cell lymphomas, this strategy has been challenging because these markers may be expressed on the tumor cells themselves resulting in inadvertent tumor growth.

Peripheral T-cell lymphoma (PTCL) is a group of lymphoproliferative disorders, originating from mature T/NK cells with highly heterogeneous, aggressive characteristics and poor prognosis. There are 27 subtypes of PTCL in the World Health Organization 2016 classification of lymphoid neoplasm, including extranodal NK/T-cell lymphoma, nasal-type (ENKTL), nonspecific (PTCL-NOS), angioimmunoblastic T-cell lymphoma (AITL), and anaplastic lymphoma kinase (ALK)+/− anaplastic large cell lymphoma (ALCL) [155, 156]. CHOP-based regimens are the first-line treatments for PTCL other than NK/T-cell lymphoma, but the efficacy is limited [157, 158]. Thus, effective treatments for relapsed or refractory (r/r) PTCL are urgently needed. PD-1 and PD-L1 expression is commonly observed in PTCL cells, and PD-1 or PD-L1 is considered a prognostic biomarker and target. A phase I study (five patients with r/r PTCL) and phase 2 study (12 patients with r/r PTCL) have identified the clinical value of Nivolumab in r/r PTCL patients with 33% ORR [159, 160]. Also, the promising results using pembrolizumab have been seen in seven relapsed ENKT lymphoma with 100% overall response rates after a median of 7 weeks of treatment, either EBV DNA-positive or negative. The remission has been maintained at a median follow-up of 6 months. Similar results were reported for nivolumab. Thus, several studies are ongoing to explore the efficacy of PD-1 inhibitors in the treatment of ENKL. Studies showed that the response is correlated with the level of PD-1 expression, especially in EBV DNA positive patients. Although, studies were halted early due to the short duration of response and concern for hyperprogression. Encouraging results were also seen with pembrolizumab in patients with r/r cutaneous T-cell lymphoma with 38% ORR in a phase 2 study [142]. There was an alarming report of hyperprogression in three patients with ATLL that were enrolled in the nivolumab trial. Clinical progression was also accompanied by an increase in the viral load [143]. In these cases, PD-1 tumor suppressor function may have been lifted by PD-1 blockade. The use of PD-1 and PD-L1 antibodies in ATLL has to be viewed with caution. Therefore, more clinical trials should be done to evaluate the efficacy and safety of ICIs in different subtypes of PTCL.

Ipilimumab, an inhibitor of CTLA-4 (also known as CD152), provides both positive and negative feedback for T-cell activation when combined with its costimulatory receptor CD28. But the effect of CTLA-4 inhibitors in PTCL is not well characterized. In general, immunotherapy for PTCL is promising. For other immune checkpoint proteins, such as TIGIT, TIM-3, and LAG-3, their evaluation in PTCL is still at the preclinical stage and needed to be further explored via relevant clinical trials. Some studies have shown that the combined blockade of the TIM-3 and PD-1 pathways has significant efficacy in hematological tumors [161]. More importantly, the combination of PD-1/PD-L1 inhibitors and CAR-T cell therapy are worthy of exploring [162]. These ICIs combined therapies may be the best strategy for tumor therapy and promote the prognosis in near future.

Advertisement

4. Immune checkpoints inhibitors toxicity

As reported by the current clinical trials, treatment-related AEs were very common in patients undergoing anti-PD-1 treatment (Table 1). However, grade 3 or above treatment-related AEs were generally only observed in less than 30% of patients. Here, we use the studies with most patients enrolled as examples (i.e., n = 243 for Armand et al. [36] and n = 210 for Chen et al. [40]). As reported by Armand et al. [36], the most common treatment-related AEs of any grade were fatigue (23%), diarrhea (15%), and infusion-related reactions (14%). However, none of them were severe (grade 3 or above). On contrary, the most common grade 3 or 4 treatment-related AEs were lipase increases (5%), neutropenia (3%), and ALT increases. The most common treatment-related AEs that led to treatment discontinuation were pneumonitis (2%) and autoimmune hepatitis (1%). Other serious treatment-relate AEs included infusion-related reactions (2%), pneumonia (1%), pleural effusion (1%), and pyrexia (1%). For the study conducted by Chen et al. [40], the most common treatment-related AEs were hypothyroidism (14.3%), pyrexia (11.4%), rash (11.0%), and fatigue (11.0%). The most common grade 3 or 4 treatment-relate AEs were neutropenia (2.4%) and diarrhea (1.4%). Fourteen patients discontinued treatment due to treatment-related AEs, and the most common causes were pneumonitis in seven (3.3%) and infusion-related reactions in two (1.0%). Here, we will briefly discuss some cases that deserve special attention.

4.1 Thyroid dysfunction

Thyroid dysfunction is one of the most common AEs observed during PD-1 inhibition and is heterogeneous in nature. In a study of 73 patients who underwent nivolumab therapy, Peiro et al. [163] reported that 23.3% of patients developed thyroid dysfunction. Among them, seven patients showed thyrotoxicosis and 10 patients showed primary hypothyroidism (four required levothyroxine treatment). They concluded that thyrotoxicosis occurred earlier than hypothyroidism. Before the onset of hypothyroidism, 33% of patients exhibited transient thyroiditis and five patients had hyperthyroid, which became hypothyroid later. In cases of thyroiditis, patients can be treated with beta-blockers, and thyroid hormone replacement may be required for hypothyroidism. For hyperthyroidism, beta-blockers and corticosteroids are very effective [164].

4.2 Treatment-related pneumonitis

In a meta-analysis of 11 clinical trials in patients treated with ICI (PD-1 or CTLA-4 blockade), the use of ICIs led to an increased risk of pneumonitis of all grades [165]. Younger age (<60 years old) may be a major risk factor [166]. Corticosteroids can be used for the treatment of pneumonitis, and those refractory cases should be treated with steroid-free immunosuppressants. For cases of grade 3 or above pneumonitis, potential infections should be considered. In cases of severe pneumonitis, the use of ICI should be stopped [167].

4.3 Treatment-related colitis/diarrhea

Gastrointestinal AEs are another most common treatment-related AEs during ICI therapy. Physicians should carefully distinguish colitis from diarrhea; and when colitis symptoms emerge, hospitalization and discontinuation of ICIs should be considered. In cases of mild symptoms, administration of corticosteroids or antidiarrheals could be applied [168], and additional infliximab may be needed [169].

4.4 Treatment-related cardiovascular disease

The incidences of treatment-related cardiovascular diseases are frequently underestimated, as reported by Jain et al. [170]. They identified 16,574 patients who received ICIs from a total of 2,687,301 patients and 1:1 matched to 2875 patients who received chemotherapy or 4611 patients who received targeted agents. They observed the onsets of treatment-related cardiovascular diseases included stroke (4.6%), heart failure (3.5%), atrial fibrillation (2.1%), conduction disorders (1.5%), myocardial infarction (0.9%), myocarditis (0.05%), vasculitis (0.05%), and pericarditis (0.2%). In addition, anti-CTLA-4 therapy was more commonly related to treatment-related cardiovascular diseases. Moreover, another retrospective analysis indicated that inhibition of PD-1/PD-L1 was significantly associated with the risk of myocarditis, and males may have an increased risk of certain cardiovascular AEs [171]. In another meta-analysis including 2576 trials/studies and 20,244 patients, combined therapy of PD-1 blockade and chemotherapy may increase the risk of myocardial disease of all grades; although there was no significant increase in the risk of other cardiovascular diseases [172].

4.5 Other autoimmune diseases

As PD-1 blockade non-specifically activates the immune system, the induction of autoimmune-like diseases is the major concern of the toxicity incurred. Examples of symptoms during the treatment of HL include autoimmune type I diabetes [173, 174, 175, 176], autoimmune encephalitis [177, 178, 179], autoimmune hepatitis [36], autoimmune nephritis [36, 38], and autoimmune hemolytic anemia [180, 181]. In cases of autoimmune diseases, the use of immunosuppressive treatment or delay of ICI therapy should be seriously considered.

4.6 Association between toxicity and efficacy

Although treatment-related AEs severely affect the treatment outcomes of ICIs, the onset of AEs that are immune-related may be directly associated with the efficacy of ICIs. In a study of 106 patients who underwent PD-1 blockade monotherapy, Rogado et al. [182] observed that patients with immune-related AEs have a higher ORR of 82.5% (vs 16.6%) and longer PFS of 10 months (vs 3 months), as compared with those without immune-related AEs. Although the detailed underlying mechanisms remain to be elucidated, concerns about the effect of corticosteroids and other immunosuppressants administration on ICI efficacy have been raised. However, some studies suggested that the use of corticosteroids and other immunosuppressants may not impair the anti-tumor activities of ICIs [183, 184].

Advertisement

5. Conclusions

ICIs therapies have demonstrated remarkable efficacy in several subtypes of HL and NHL, and some ICIs (e.g., pembrolizumab) have been approved to use in HL and PMBCL. Especially, anti-PD-1/PD-L1 antibodies in a combination with other therapies have acquired promising results, and AEs are common in these treatments. Thus, we need to do more clinical trials and real-world studies to further explore the effectiveness and safety of ICIs treatment in lymphoma.

Advertisement

Funding

This work was financially supported by National Natural Science Foundation of China (NSFC 81500173).

References

  1. 1. Lenschow DJ, Bluestone JA. T cell co-stimulation and in vivo tolerance. Current Opinion in Immunology. 1993;5(5):747-752
  2. 2. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nature Reviews. Immunology. 2015;15(8):486-499
  3. 3. Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunological Reviews. 2020;294(1):164-176
  4. 4. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews. Cancer. 2012;12(4):252-264
  5. 5. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537-1544
  6. 6. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: Current researches in cancer. American Journal of Cancer Research. 2020;10(3):727-742
  7. 7. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nature Immunology. 2001;2(3):261-268
  8. 8. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nature Medicine. 2002;8(8):793-800
  9. 9. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nature Medicine. 2003;9(5):562-567
  10. 10. Dermani FK, Samadi P, Rahmani G, Kohlan AK, Najafi R. PD-1/PD-L1 immune checkpoint: Potential target for cancer therapy. Journal of Cellular Physiology. 2019;234(2):1313-1325
  11. 11. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nature Reviews. Immunology. 2018;18(3):153-167
  12. 12. Patsoukis N, Wang Q , Strauss L, Boussiotis VA. Revisiting the PD-1 pathway. Science Advances. 2020;6(38):eabd2712
  13. 13. Quigley M, Pereyra F, Nilsson B, Porichis F, Fonseca C, Eichbaum Q , et al. Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nature Medicine. 2010;16(10):1147-1151
  14. 14. Clark CA, Gupta HB, Sareddy G, Pandeswara S, Lao S, Yuan B, et al. Tumor-intrinsic PD-L1 signals regulate cell growth, pathogenesis, and autophagy in ovarian Cancer and melanoma. Cancer Research. 2016;76(23):6964-6974
  15. 15. Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, Peach R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity. 1994;1(9):793-801
  16. 16. Linsley PS, Bradshaw J, Greene J, Peach R, Bennett KL, Mittler RS. Intracellular trafficking of CTLA-4 and focal localization towards sites of TCR engagement. Immunity. 1996;4(6):535-543
  17. 17. Schneider H, Smith X, Liu H, Bismuth G, Rudd CE. CTLA-4 disrupts ZAP70 microcluster formation with reduced T cell/APC dwell times and calcium mobilization. European Journal of Immunology. 2008;38(1):40-47
  18. 18. Guntermann C, Alexander DR. CTLA-4 suppresses proximal TCR signaling in resting human CD4(+) T cells by inhibiting ZAP-70 Tyr(319) phosphorylation: A potential role for tyrosine phosphatases. Journal of Immunology. 2002;168(9):4420-4429
  19. 19. Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, et al. Trans-endocytosis of CD80 and CD86: A molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332(6029):600-603
  20. 20. Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Molecular and Cellular Biology. 2005;25(21):9543-9553
  21. 21. Kubsch S, Graulich E, Knop J, Steinbrink K. Suppressor activity of anergic T cells induced by IL-10-treated human dendritic cells: Association with IL-2- and CTLA-4-dependent G1 arrest of the cell cycle regulated by p27Kip1. European Journal of Immunology. 2003;33(7):1988-1997
  22. 22. Brunner MC, Chambers CA, Chan FK, Hanke J, Winoto A, Allison JP. CTLA-4-mediated inhibition of early events of T cell proliferation. Journal of Immunology. 1999;162(10):5813-5820
  23. 23. Finn OJ. Immuno-oncology: Understanding the function and dysfunction of the immune system in cancer. Annals of Oncology. 2012;23(Suppl. 8):viii6-viii9
  24. 24. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of Cancer: Clinical impact and mechanisms of response and resistance. Annual Review of Pathology. 2021;16:223-249
  25. 25. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. The Journal of Experimental Medicine. 1999;190(3):355-366
  26. 26. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734-1736
  27. 27. Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Current Opinion in Immunology. 2006;18(2):206-213
  28. 28. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England Journal of Medicine. 2010;363(8):711-723
  29. 29. Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O’Donnell E, et al. 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. 2010;116(17):3268-3277
  30. 30. U.S. Food and Drug Administration (FDA). Nivolumab (Opdivo) for Hodgkin Lymphoma. 2016. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/nivolumab-opdivo-hodgkin-lymphoma [Accessed: Jun 17, 2022]
  31. 31. U.S. Food and Drug Administration (FDA). Pembrolizumab (KEYTRUDA) for classical Hodgkin lymphoma. 2017. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/pembrolizumab-keytruda-classical-hodgkin-lymphoma [Accessed: Jun 17, 2022]
  32. 32. Liput J, Guler E, Smith DA, Tirumani SH, Hoimes C, Caimi PF, et al. Clinical, imaging findings, responses, and outcomes of patients with classical Hodgkin lymphoma and non-Hodgkin lymphoma undergoing immune checkpoint inhibitor therapy: A single-institution experience. Journal of Computer Assisted Tomography. 2020;44:619-626. DOI: 10.1097/RCT.0000000000001043
  33. 33. Davis KL, Fox E, Merchant MS, Reid JM, Kudgus RA, Liu X, et al. Nivolumab in children and young adults with relapsed or refractory solid tumours or lymphoma (ADVL1412): A multicentre, open-label, single-arm, phase 1-2 trial. The Lancet Oncology. 2020;21:541-550. DOI: 10.1016/S1470-2045(20)30023-1
  34. 34. Kasamon YL, de Claro RA, Wang Y, Shen YL, Farrell AT, Pazdur R. FDA approval summary: Nivolumab for the treatment of relapsed or progressive classical Hodgkin lymphoma. The Oncologist. 2017;22:585-591. DOI: 10.1634/theoncologist.2017-0004
  35. 35. Bair SM, Strelec LE, Feldman TA, Ahmed G, Armand P, Shah NN, et al. 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. 2019;24:955-962. DOI: 10.1634/theoncologist.2018-0538
  36. 36. Armand P, Engert A, Younes A, Fanale M, Santoro A, Zinzani PL, et al. Nivolumab for relapsed/refractory classic Hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: Extended follow-up of the multicohort single-arm phase II CheckMate 205 trial. Journal of Clinical Oncology. 2018;36(14):1428-1439. DOI: 10.1200/JCO.2017.76.0793
  37. 37. Falchi L, Sawas A, Deng C, Amengual JE, Colbourn DS, Lichtenstein EA, et al. High rate of complete responses to immune checkpoint inhibitors in patients with relapsed or refractory Hodgkin lymphoma previously exposed to epigenetic therapy. Journal of Hematology & Oncology. 2016;9(1):132. DOI: 10.1186/s13045-016-0363-1
  38. 38. Armand P, Shipp MA, Ribrag V, Michot JM, Zinzani PL, Kuruvilla J, et al. Programmed Death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab Vedotin failure. Journal of Clinical Oncology. 2016;34(31):3733-3739. DOI: 10.1200/JCO.2016.67.3467
  39. 39. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. The New England Journal of Medicine. 2015;372:311-319. DOI: 10.1056/NEJMoa1411087
  40. 40. Chen R, Zinzani PL, Lee HJ, Armand P, Johnson NA, Brice P, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood. 2019;134(14):1144-1153. DOI: 10.1182/blood.2019000324
  41. 41. Younes A, Santoro A, Shipp M, Zinzani PL, Timmerman JM, Ansell S, 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. The Lancet Oncology. 2016;17:1283-1294. DOI: 10.1016/S1470-2045(16)30167-X
  42. 42. Bekoz H, Ozbalak M, Karadurmus N, Paydas S, Turker A, Toptas T, et al. Nivolumab for relapsed or refractory Hodgkin lymphoma: Real-life experience. Annals of Hematology. 2020;99:2565-2576. DOI: 10.1007/s00277-020-04077-4
  43. 43. Bekoz H, Karadurmus N, Paydas S, Turker A, Toptas T, Firatli Tuglular T, et al. Nivolumab for relapsed or refractory Hodgkin lymphoma: Real-life experience. Annals of Oncology. 2017;28:2496-2502. DOI: 10.1093/annonc/mdx341
  44. 44. Geoerger B, Kang HJ, Yalon-Oren M, Marshall LV, Vezina C, Pappo A, et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): Interim analysis of an open-label, single-arm, phase 1-2 trial. The Lancet Oncology. 2020;21:121-133. DOI: 10.1016/S1470-2045(19)30671-0
  45. 45. Maruyama D, Terui Y, Yamamoto K, Fukuhara N, Choi I, Kuroda J, et al. Final results of a phase II study of nivolumab in Japanese patients with relapsed or refractory classical Hodgkin lymphoma. Japanese Journal of Clinical Oncology. 2020;50:1265-1273. DOI: 10.1093/jjco/hyaa117
  46. 46. Ramchandren R, Domingo-Domenech E, Rueda A, Trneny M, Feldman TA, Lee HJ, et al. Nivolumab for newly diagnosed advanced-stage classic Hodgkin lymphoma: Safety and efficacy in the phase II CheckMate 205 study. Journal of Clinical Oncology. 2019;37:1997-2007. DOI: 10.1200/JCO.19.00315
  47. 47. Chan TSY, Hwang YY, Khong PL, Leung AYH, Chim CS, Tse EWC, et al. Low-dose pembrolizumab and nivolumab were efficacious and safe in relapsed and refractory classical Hodgkin lymphoma: Experience in a resource-constrained setting. Hematological Oncology. 2020;38:726-736. DOI: 10.1002/hon.2787
  48. 48. Dada R, Zabani Y. Nivolumab induces impressive responses in relapsed/refractory classic Hodgkin lymphoma: Single institutional experience. Journal of Oncology Pharmacy Practice. 2019;25:1586-1589. DOI: 10.1177/1078155218800150
  49. 49. Kuruvilla J, Ramchandren R, Santoro A, Paszkiewicz-Kozik E, Gasiorowski R, Johnson NA, et al. Pembrolizumab versus brentuximab vedotin in relapsed or refractory classical Hodgkin lymphoma (KEYNOTE-204): An interim analysis of a multicentre, randomised, open-label, phase 3 study. The Lancet Oncology. 2021;22:512-524. DOI: 10.1016/S1470-2045(21)00005-X
  50. 50. Momotow J, Buhnen I, Trautmann-Grill K, Kobbe G, Hahn D, Schroers R, et al. Outcomes of anti-programmed death 1 treatment for relapsed/refractory Hodgkin lymphoma: A German Hodgkin study group multicentre real-world analysis. British Journal of Haematology. 2022;198(2):401-404. DOI: 10.1111/bjh.18231
  51. 51. Hur JY, Yoon SE, Kim SJ, Kim WS. Immune checkpoint inhibitors in patients with pretreated HodgkinEs lymphoma: A Korean single-center, retrospective study. Blood Res. 2020;55:85-90. DOI: 10.5045/br.2020.2020014
  52. 52. Lepik KV, Mikhailova NB, Moiseev IS, Kondakova EV, Tsvetkova LA, Zalyalov YR, et al. Nivolumab for the treatment of relapsed and refractory classical Hodgkin lymphoma after ASCT and in ASCT-naive patients. Leukemia & Lymphoma. 2019;60:2316-2319. DOI: 10.1080/10428194.2019.1573368
  53. 53. Armand P, Kuruvilla J, Michot JM, Ribrag V, Zinzani PL, Zhu Y, et al. KEYNOTE-013 4-year follow-up of pembrolizumab in classical Hodgkin lymphoma after brentuximab vedotin failure. Blood Advances. 2020;4:2617-2622. DOI: 10.1182/bloodadvances.2019001367
  54. 54. Manson G, Mear JB, Herbaux C, Schiano JM, Casasnovas O, Stamatoullas A, et al. Long-term efficacy of anti-PD1 therapy in Hodgkin lymphoma with and without allogenic stem cell transplantation. European Journal of Cancer. 2019;115:47-56
  55. 55. Merryman RW, Castagna L, Giordano L, Ho VT, Corradini P, Guidetti A, et al. Allogeneic transplantation after PD-1 blockade for classic Hodgkin lymphoma. Leukemia. 2021;35(9):2672-2683
  56. 56. Martinez C, Carpio C, Heras I, Rios-Herranz E, Buch J, Gutierrez A, et al. Potential survival benefit for patients receiving allogeneic hematopoietic stem cell transplantation after nivolumab therapy for relapse/refractory Hodgkin lymphoma: Real-life experience in Spain. Biology of Blood and Marrow Transplantation. 2020;26(8):1534-1542
  57. 57. Armand P, Chen YB, Redd RA, Joyce RM, Bsat J, Jeter E, et al. PD-1 blockade with pembrolizumab for classical Hodgkin lymphoma after autologous stem cell transplantation. Blood. 2019;134(1):22-29
  58. 58. Casadei B, Argnani L, Morigi A, Lolli G, Broccoli A, Pellegrini C, et al. Potential survival benefit for patients receiving autologous hematopoietic stem cell transplantation after checkpoint inhibitors for relapsed/refractory Hodgkin lymphoma: A real-life experience. Hematological Oncology. 2020;38(5):737-741
  59. 59. Ito A, Kim SW, Matsuoka KI, Kawakita T, Tanaka T, Inamoto Y, et al. Safety and efficacy of anti-programmed cell death-1 monoclonal antibodies before and after allogeneic hematopoietic cell transplantation for relapsed or refractory Hodgkin lymphoma: A multicenter retrospective study. International Journal of Hematology. 2020;112(5):674-689
  60. 60. Singh AK, Porrata LF, Aljitawi O, Lin T, Shune L, Ganguly S, et al. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin’s lymphoma. Bone Marrow Transplantation. 2016;51(9):1268-1270
  61. 61. Charles J, Giovannini D, Terzi N, Schwebel C, Sturm N, Masson D, et al. Multi-organ failure induced by nivolumab in the context of Allo-stem cell transplantation. Experimental Hematology & Oncology. 2019;8:8
  62. 62. Steinerova K, Jindra P, Lysak D, Karas M. Development of resistant GvHD in a patient treated with nivolumab for Hodgkins lymphoma relapse after allogeneic unrelated transplantation. Klinická Onkologie;32(1):66-69
  63. 63. Minson A, Douglas G, Bilmon I, Grigg A. Low dose PD-1 inhibition in relapsed refractory Hodgkin lymphoma after allogeneic stem cell transplant with concomitant active GVHD. British Journal of Haematology. 2019;184(5):840-844
  64. 64. Amerikanou R, Neill L, Shafat M, Roddy H, Hyare H, Hughes S, et al. Multi-organ graft-versus-host disease after nivolumab for relapsed Hodgkin lymphoma: The role of plasma exchange. Lancet Haematol. 2021;8(11):e862
  65. 65. Sasse S, Brockelmann PJ, Goergen H, Plutschow A, Muller H, Kreissl S, et al. Long-term follow-up of contemporary treatment in early-stage Hodgkin lymphoma: Updated analyses of the German Hodgkin study group HD7, HD8, HD10, and HD11 trials. Journal of Clinical Oncology. 2017;35(18):1999-2007
  66. 66. Brockelmann PJ, Goergen H, Keller U, Meissner J, Ordemann R, Halbsguth TV, et al. Efficacy of nivolumab and AVD in early-stage Unfavorable classic Hodgkin lymphoma: The randomized phase 2 German Hodgkin study group NIVAHL trial. JAMA Oncology. 2020;6(6):872-880
  67. 67. Allen PB, Savas H, Evens AM, Advani RH, Palmer B, Pro B, et al. Pembrolizumab followed by AVD in untreated early unfavorable and advanced-stage classical Hodgkin lymphoma. Blood. 2021;137(10):1318-1326
  68. 68. Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin's lymphoma. Journal of Clinical Oncology. 2012;30(18):2183-2189
  69. 69. Advani RH, Moskowitz AJ, Bartlett NL, Vose JM, Ramchandren R, Feldman TA, et al. Brentuximab vedotin in combination with nivolumab in relapsed or refractory Hodgkin lymphoma: 3-year study results. Blood. 2021;138(6):427-438
  70. 70. Cheson BD, Bartlett NL, LaPlant B, Lee HJ, Advani RJ, Christian B, et al. Brentuximab vedotin plus nivolumab as first-line therapy in older or chemotherapy-ineligible patients with Hodgkin lymphoma (ACCRU): A multicentre, single-arm, phase 2 trial. Lancet Haematology. 2020;7(11):e808-e815
  71. 71. Fedorova LV, Lepik KV, Volkov NP, Kotselyabina PV, Borzenkova ES, Popova MO, et al. Efficacy and safety of nivolumab combined with brentuximab vedotin after nivolumab monotherapy failure in patients with relapsed and refractory classic Hodgkin lymphoma. International Journal of Clinical Oncology. 2022;27(3):626-632
  72. 72. Lucchini E, Rusconi C, Levis M, Ricci F, Santoro A, Ricardi U, et al. Immune checkpoint inhibitors in combination with radiotherapy as salvage treatment for relapsed/refractory classical Hodgkin lymphoma: A retrospective analysis in 12 patients. Hematology Reports. 2021;13(2):9080
  73. 73. de Forceville L, Deau-Fischer B, Franchi P, Arsene-Henry A, Cassou Mounat T, Bouscary D, et al. Radiotherapy in combination with nivolumab for relapsed/refractory classical Hodgkin lymphoma: About two cases. Cancer Radiothérapie. 2019;23(3):232-239
  74. 74. Bai B, Huang HQ , Cai QQ , Wang XX, Cai QC, Lin ZX, et al. Promising long-term outcome of gemcitabine, vinorelbine, liposomal doxorubicin (GVD) in 14-day schedule as salvage regimen for patients with previously heavily treated Hodgkin's lymphoma and aggressive non-Hodgkin's lymphoma. Medical Oncology. 2013;30(1):350
  75. 75. Moskowitz AJ, Shah G, Schoder H, Ganesan N, Drill E, Hancock H, et al. Phase II trial of pembrolizumab plus gemcitabine, vinorelbine, and liposomal doxorubicin as second-line therapy for relapsed or refractory classical Hodgkin lymphoma. Journal of Clinical Oncology. 2021;39(28):3109-3117
  76. 76. Zhang YC, Wang JN, Ma SY, Cai J, Su N, Huang HQ , et al. Combination of PD-1 inhibitor with GVD (gemcitabine, vinorelbine, liposomal doxorubicin) versus GVD regimen as second-line therapy for relapsed/refractory classical Hodgkin lymphoma. British Journal of Haematology. 2022;196(1):127-135
  77. 77. Xu J, Zhang Y, Jia R, Yue C, Chang L, Liu R, et al. Anti-PD-1 antibody SHR-1210 combined with Apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: An open-label, dose escalation and expansion study. Clinical Cancer Research. 2019;25(2):515-523
  78. 78. Huang J, Mo H, Zhang W, Chen X, Qu D, Wang X, et al. Promising efficacy of SHR-1210, a novel anti-programmed cell death 1 antibody, in patients with advanced gastric and gastroesophageal junction cancer in China. Cancer. 2019;125(5):742-749
  79. 79. Huang J, Xu B, Mo H, Zhang W, Chen X, Wu D, et al. Safety, activity, and biomarkers of SHR-1210, an anti-PD-1 antibody, for patients with advanced Esophageal carcinoma. Clinical Cancer Research. 2018;24(6):1296-1304
  80. 80. Mo H, Huang J, Xu J, Chen X, Wu D, Qu D, et al. Safety, anti-tumour activity, and pharmacokinetics of fixed-dose SHR-1210, an anti-PD-1 antibody in advanced solid tumours: A dose-escalation, phase 1 study. British Journal of Cancer. 2018;119(5):538-545
  81. 81. Fang W, Yang Y, Ma Y, Hong S, Lin L, He X, et al. Camrelizumab (SHR-1210) alone or in combination with gemcitabine plus cisplatin for nasopharyngeal carcinoma: Results from two single-arm, phase 1 trials. The Lancet Oncology. 2018;19(10):1338-1350
  82. 82. Song Y, Wu J, Chen X, Lin T, Cao J, Liu Y, et al. A single-arm, multicenter, phase II study of Camrelizumab in relapsed or refractory classical Hodgkin lymphoma. Clinical Cancer Research. 2019;25(24):7363-7369
  83. 83. Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354(6316):1160-1165
  84. 84. Ghoneim HE, Fan Y, Moustaki A, Abdelsamed HA, Dash P, Dogra P, et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell. 2017;170(1):142-157 e119
  85. 85. Gore SD, Jones C, Kirkpatrick P. Decitabine. Nature Reviews. Drug Discovery. 2006;5(11):891-892
  86. 86. Nie J, Wang C, Liu Y, Yang Q , Mei Q , Dong L, et al. Addition of low-dose decitabine to anti-PD-1 antibody Camrelizumab in relapsed/refractory classical Hodgkin lymphoma. Journal of Clinical Oncology. 2019;37(17):1479-1489
  87. 87. Liu Y, Wang C, Li X, Dong L, Yang Q , Chen M, et al. Improved clinical outcome in a randomized phase II study of anti-PD-1 camrelizumab plus decitabine in relapsed/refractory Hodgkin lymphoma. Journal for Immunotherapy of Cancer. 2021;9(4):e002347
  88. 88. Yan Z, Ma J, Yao S, Yao Z, Wang H, Chu J, et al. Anti-angiogenic agent combined with anti-PD-1 immunotherapy showed activity in patients with classical Hodgkin lymphoma who have failed immunotherapy: A retrospective case report study. Frontiers in Immunology. 2021;12:727464
  89. 89. Shi Y, Su H, Song Y, Jiang W, Sun X, Qian W, et al. Safety and activity of sintilimab in patients with relapsed or refractory classical Hodgkin lymphoma (ORIENT-1): A multicentre, single-arm, phase 2 trial. Lancet Haematology. 2019;6(1):e12-e19
  90. 90. Dahan R, Sega E, Engelhardt J, Selby M, Korman AJ, Ravetch JV. FcgammaRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell. 2015;28(3):285-295
  91. 91. Song Y, Gao Q , Zhang H, Fan L, Zhou J, Zou D, et al. Treatment of relapsed or refractory classical Hodgkin lymphoma with the anti-PD-1, tislelizumab: Results of a phase 2, single-arm, multicenter study. Leukemia. 2020;34(2):533-542
  92. 92. Ding XS, Mi L, Song YQ , Liu WP, Yu H, Lin NJ, et al. Relapsed/refractory classical Hodgkin lymphoma effectively treated with low-dose decitabine plus tislelizumab: A case report. World Journal of Clinical Cases. 2021;9(21):6041-6048
  93. 93. Lin N, Zhang M, Bai H, Liu H, Cui J, Ke X, et al. Efficacy and safety of GLS-010 (zimberelimab) in patients with relapsed or refractory classical Hodgkin lymphoma: A multicenter, single-arm, phase II study. European Journal of Cancer. 2022;164:117-126
  94. 94. Shen L, Gong J, Xu Y, Zhang X, Peng Z, Qi C, et al. A novel recombinant human anti-PD-1 monoclonal antibody GLS-010 in patients with advanced cancer: Result of a phase Ia clinical trial. Annals of Oncology. 2018;29:x22-x23
  95. 95. Lin S, Gong J, Xu Y, Zhang X, Peng Z, Qi C, et al. Gls-010, a novel anti-PD-1 mAb in Chinese advanced gastrointestinal tumor: Result of a phase Ib clinical trial. Journal of Clinical Oncology. 2019;37(4_suppl):125-125
  96. 96. Song Y, Zhu J, Lin N, Zhang C, Zhang M, Bai H, et al. GLS-010, a novel anti-PD-1 mAb in Chinese patients with relapsed or refractory classical Hodgkin’s lymphoma: Preliminary result of a phase II clinical trial. Journal of Clinical Oncology. 2019;37(15_suppl):e14072-e14072
  97. 97. Song Y, Zhou K, Jin C, Qian Z, Hou M, Fan L, et al. A phase II study of penpulimab, an anti-PD-1 antibody, in patients with relapsed or refractoryclassic Hodgkin lymphoma (cHL). Journal of Clinical Oncology. 2021;39(15_suppl):7529-7529
  98. 98. Dhillon S. Penpulimab: First approval. Drugs. 2021;81(18):2159-2166
  99. 99. Chen X, Kong H, Luo L, Han S, Lei T, Yu H, et al. High efficacy of PD-1 inhibitor after initial failure of PD-L1 inhibitor in relapsed/refractory classical Hodgkin lymphoma. BMC Cancer. 2022;22(1):9
  100. 100. Fujii R, Friedman ER, Richards J, Tsang KY, Heery CR, Schlom J, et al. Enhanced killing of chordoma cells by antibody-dependent cell-mediated cytotoxicity employing the novel anti-PD-L1 antibody avelumab. Oncotarget. 2016;7(23):33498-33511
  101. 101. Boyerinas B, Jochems C, Fantini M, Heery CR, Gulley JL, Tsang KY, et al. Antibody-dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunology Research. 2015;3(10):1148-1157
  102. 102. Herrera AF, Burton C, Radford J, Miall F, Townsend W, Santoro A, et al. Avelumab in relapsed/refractory classical Hodgkin lymphoma: Phase 1b results from the JAVELIN Hodgkins trial. Blood Advances. 2021;5(17):3387-3396
  103. 103. Gong J, Cao J, Zhang Q , Xu N, Zhao Y, Xing B, et al. Safety, antitumor activity and biomarkers of sugemalimab in Chinese patients with advanced solid tumors or lymphomas: Results from the first-in-human phase 1 trial. Cancer Immunology, Immunotherapy. 2022;71(8):1897-1908
  104. 104. Dhillon S, Duggan S. Sugemalimab: First Approval. Drugs. 2022;82(5):593-599
  105. 105. Ogasawara K, Newhall K, Maxwell SE, Dell'Aringa J, Komashko V, Kilavuz N, et al. Population pharmacokinetics of an anti-PD-L1 antibody, durvalumab in patients with hematologic malignancies. Clinical Pharmacokinetics. 2020;59(2):217-227
  106. 106. Geoerger B, Zwaan CM, Marshall LV, Michon J, Bourdeaut F, Casanova M, et al. Atezolizumab for children and young adults with previously treated solid tumours, non-Hodgkin lymphoma, and Hodgkin lymphoma (iMATRIX): A multicentre phase 1-2 study. The Lancet Oncology. 2020;21(1):134-144
  107. 107. Ansell SM, Hurvitz SA, Koenig PA, LaPlant BR, Kabat BF, Fernando D, et al. 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. 2009;15(20):6446-6453
  108. 108. Wolchok JD, Neyns B, Linette G, Negrier S, Lutzky J, Thomas L, et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: A randomised, double-blind, multicentre, phase 2, dose-ranging study. The Lancet Oncology. 2010;11(2):155-164
  109. 109. Weber JS, O'Day S, Urba W, Powderly J, Nichol G, Yellin M, et al. Phase I/II study of ipilimumab for patients with metastatic melanoma. Journal of Clinical Oncology. 2008;26(36):5950-5956
  110. 110. Phan GQ , Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(14):8372-8377
  111. 111. Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113(7):1581-1588
  112. 112. Diefenbach CS, Hong F, Ambinder RF, Cohen JB, Robertson MJ, David KA, et al. Ipilimumab, nivolumab, and brentuximab vedotin combination therapies in patients with relapsed or refractory Hodgkin lymphoma: Phase 1 results of an open-label, multicentre, phase 1/2 trial. Lancet Haematology. 2020;7(9):e660-e670
  113. 113. Zeldis JB, Knight R, Hussein M, Chopra R, Muller G. A review of the history, properties, and use of the immunomodulatory compound lenalidomide. Annals of the New York Academy of Sciences. 2011;1222:76-82
  114. 114. Sakamuri D, Glitza IC, Betancourt Cuellar SL, Subbiah V, Fu S, Tsimberidou AM, et al. Phase I dose-escalation study of anti-CTLA-4 antibody ipilimumab and lenalidomide in patients with advanced cancers. Molecular Cancer Therapeutics. 2018;17(3):671-676
  115. 115. El Halabi L, Adam J, Gravelle P, Marty V, Danu A, Lazarovici J, et al. Expression of the immune checkpoint regulators LAG-3 and TIM-3 in classical Hodgkin lymphoma. Clinical Lymphoma, Myeloma and Leukemia. 2021;21(4):257-266 e253
  116. 116. Annibali O, Bianchi A, Grifoni A, Tomarchio V, Tafuri M, Verri M, et al. A novel scoring system for TIGIT expression in classic Hodgkin lymphoma. Scientific Reports. 2021;11(1):7059
  117. 117. Armand P, Lesokhin A, Borrello I, Timmerman J, Gutierrez M, Zhu L, et al. A phase 1b study of dual PD-1 and CTLA-4 or KIR blockade in patients with relapsed/refractory lymphoid malignancies. Leukemia. 2021;35(3):777-786
  118. 118. Cao Y, Wang X, Jin T, Tian Y, Dai C, Widarma C, et al. Immune checkpoint molecules in natural killer cells as potential targets for cancer immunotherapy. Signal Transduction and Targeted Therapy. 2020;5(1):250
  119. 119. Tun AM, Ansell SM. Immunotherapy in Hodgkin and non-Hodgkin lymphoma: Innate, adaptive and targeted immunological strategies. Cancer Treatment Reviews. 2020;88:102042
  120. 120. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503-511
  121. 121. Modi D, Potugari B, Uberti J. Immunotherapy for diffuse large B-cell lymphoma: Current landscape and future directions. Cancers (Basel). 2021;13(22):5827
  122. 122. Gu Q , Li J, Chen Z, Zhang J, Shen H, Miao X, et al. Expression and prognostic significance of PD-L2 in diffuse large B-cell lymphoma. Frontiers in Oncology. 2021;11:664032
  123. 123. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nature Medicine. 1999;5(12):1365-1369
  124. 124. Li S, Young KH, Medeiros LJ. Diffuse large B-cell lymphoma. Pathology. 2018;50(1):74-87
  125. 125. Grimm KE, O’Malley DP. Aggressive B cell lymphomas in the 2017 revised WHO classification of tumors of hematopoietic and lymphoid tissues. Annals of Diagnostic Pathology. 2019;38:6-10
  126. 126. Godfrey J, Tumuluru S, Bao R, Leukam M, Venkataraman G, Phillip J, et al. PD-L1 gene alterations identify a subset of diffuse large B-cell lymphoma harboring a T-cell-inflamed phenotype. Blood. 2019;133(21):2279-2290
  127. 127. Kiyasu J, Miyoshi H, Hirata A, Arakawa F, Ichikawa A, Niino D, et al. Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood. 2015;126(19):2193-2201
  128. 128. Wang Y, Farooq U, Link BK, Larson MC, King RL, Maurer MJ, et al. Late relapses in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Journal of Clinical Oncology. 2019;37(21):1819-1827
  129. 129. Ansell SM, Minnema MC, Johnson P, Timmerman JM, Armand P, Shipp MA, et al. Nivolumab for relapsed/refractory diffuse large B-cell lymphoma in patients ineligible for or having failed autologous transplantation: A single-arm, phase II study. Journal of Clinical Oncology. 2019;37(6):481-489
  130. 130. Zheng Y, Si J, Yuan T, Ding S, Tian C. Immune targeted therapy for diffuse large B cell lymphoma. Blood Science. 2021;3(4):136-148
  131. 131. Smith SD, Till BG, Shadman MS, Lynch RC, Cowan AJ, Wu QV, et al. Pembrolizumab with R-CHOP in previously untreated diffuse large B-cell lymphoma: Potential for biomarker driven therapy. British Journal of Haematology. 2020;189(6):1119-1126
  132. 132. Younes A, Burke JM, Cheson BD, Diefenbach C, Ferrari S, Hahn UH, et al. Safety and efficacy of atezolizumab in combination with rituximab plus CHOP in previously untreated patients with diffuse large B-cell lymphoma (DLBCL): Updated analysis of a phase I/II study. Blood. 2019;134(Supplement_1):2874-2874
  133. 133. Herrera AF, Goy A, Mehta A, Ramchandren R, Pagel JM, Svoboda J, et al. Safety and activity of ibrutinib in combination with durvalumab in patients with relapsed or refractory follicular lymphoma or diffuse large B-cell lymphoma. American Journal of Hematology. 2020;95(1):18-27
  134. 134. Younes A, Burke JM, Cheson B, Diefenbach C, Ferrari S, Hahn U, et al. Safety and efficacy of atezolizumab in combination with rituximab plus CHOP in previously untreated patients with diffuse large B-cell lymphoma (DLBCL): Primary analysis of a phase I/II study. Blood. 2018;132(Supplement 1):2969-2969
  135. 135. Schuster SJ, Bartlett NL, Assouline S, Yoon S-S, Bosch F, Sehn LH, et al. Mosunetuzumab induces complete remissions in poor prognosis non-Hodgkin lymphoma patients, including those who are resistant to or relapsing after chimeric antigen receptor T-cell (CAR-T) therapies, and is active in treatment through multiple lines. Blood. 2019;134(Supplement_1):6-6
  136. 136. Hutchings M, Gritti G, Sureda A, Terol MJ, Dyer MJ, Iacoboni G, et al. CD20-TCB, a novel T-cell-engaging bispecific antibody, can be safely combined with the anti-PD-L1 antibody atezolizumab in relapsed or refractory B-cell non-Hodgkin lymphoma. Blood. 2019;134(Supplement_1):2871-2871
  137. 137. Hirayama AV, Gauthier J, Hay KA, Sheih A, Cherian S, Chen X, et al. Efficacy and toxicity of JCAR014 in combination with durvalumab for the treatment of patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Blood. 2018;132(Supplement 1):1680-1680
  138. 138. Argnani L, Casadei B, Pelusi C, Lo Preiato V, Pagotto U, Bertoni F, et al. Immune-related adverse events in the treatment of non-Hodgkin lymphoma with immune checkpoint inhibitors. Scientific Reports. 2022;12(1):1753
  139. 139. Ansell S, Gutierrez ME, Shipp MA, Gladstone D, Moskowitz A, Borello I, et al. A phase 1 study of nivolumab in combination with ipilimumab for relapsed or refractory hematologic malignancies (CheckMate 039). Blood. 2016;128(22):183-183
  140. 140. Savage KJ. Primary mediastinal large B-cell lymphoma. Blood. 2021;140(9):955-970
  141. 141. Chen H, Pan T, He Y, Zeng R, Li Y, Yi L, et al. Primary mediastinal B-cell lymphoma: Novel precision therapies and future directions. Frontiers in Oncology. 2021;11:654854
  142. 142. Al Hadidi SA, Lee HJ. Pembrolizumab for the treatment of Hodgkin lymphoma. Expert Opinion on Biological Therapy. 2020;20(11):1275-1282
  143. 143. Armand P, Rodig S, Melnichenko V, Thieblemont C, Bouabdallah K, Tumyan G, et al. Pembrolizumab in relapsed or refractory primary mediastinal large B-cell lymphoma. Journal of Clinical Oncology. 2019;37(34):3291-3299
  144. 144. Zinzani PL, Santoro A, Gritti G, Brice P, Barr PM, Kuruvilla J, et al. Nivolumab combined with brentuximab Vedotin for relapsed/refractory primary mediastinal large B-cell lymphoma: Efficacy and safety from the phase II CheckMate 436 study. Journal of Clinical Oncology. 2019;37(33):3081-3089
  145. 145. Fakhri B, Ai W. Current and emerging treatment options in primary mediastinal B-cell lymphoma. Therapeutic Advances in Hematology. 2021;12:20406207211048959
  146. 146. Beltran BE, Castro D, Paredes S, Miranda RN, Castillo JJ. EBV-positive diffuse large B-cell lymphoma, not otherwise specified: 2020 update on diagnosis, risk-stratification and management. American Journal of Hematology. 2020;95(4):435-445
  147. 147. Chen BJ, Chapuy B, Ouyang J, Sun HH, Roemer MG, Xu ML, et al. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clinical Cancer Research. 2013;19(13):3462-3473
  148. 148. Satou A, Nakamura S. EBV-positive B-cell lymphomas and lymphoproliferative disorders: Review from the perspective of immune escape and immunodeficiency. Cancer Medicine. 2021;10(19):6777-6785
  149. 149. Quan L, Chen X, Liu A, Zhang Y, Guo X, Yan S, et al. PD-1 blockade can restore functions of T-cells in Epstein-Barr virus-positive diffuse large B-cell lymphoma In vitro. PLoS One. 2015;10(9):e0136476
  150. 150. Soussain C, Malaise D, Cassoux N. Primary vitreoretinal lymphoma: A diagnostic and management challenge. Blood. 2021;138(17):1519-1534
  151. 151. Ferreri AJM, Cwynarski K, Pulczynski E, Fox CP, Schorb E, La Rosee P, et al. Whole-brain radiotherapy or autologous stem-cell transplantation as consolidation strategies after high-dose methotrexate-based chemoimmunotherapy in patients with primary CNS lymphoma: Results of the second randomisation of the international Extranodal lymphoma study Group-32 phase 2 trial. Lancet Haematology. 2017;4(11):e510-e523
  152. 152. Yamanaka R, Morii K, Shinbo Y, Sano M, Homma J, Tsuchiya N, et al. Late relapse of primary central nervous system lymphoma. Leukemia & Lymphoma. 2017;58(2):475-477
  153. 153. Schaff LR, Grommes C. Update on novel therapeutics for primary CNS lymphoma. Cancers (Basel). 2021;13(21):5372
  154. 154. Nayak L, Iwamoto FM, LaCasce A, Mukundan S, Roemer MGM, Chapuy B, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood. 2017;129(23):3071-3073
  155. 155. Zain JM, Hanona P. Aggressive T-cell lymphomas: 2021 updates on diagnosis, risk stratification and management. American Journal of Hematology. 2021;96(8):1027-1046
  156. 156. Hathuc V, Kreisel F. Genetic landscape of peripheral T-cell lymphoma. Life (Basel). 2022;12(3):410
  157. 157. Kim J, Cho J, Byeon S, Kim WS, Kim SJ. Comparison of first-line treatments of peripheral T-cell lymphoma according to regimen: A systematic review and meta-analysis. Hematological Oncology. 2021;39(5):664-673
  158. 158. Liu X, Yang M, Wu M, Zheng W, Xie Y, Zhu J, et al. A retrospective study of the CHOP, CHOPE, and CHOPE/G regimens as the first-line treatment of peripheral T-cell lymphomas. Cancer Chemotherapy and Pharmacology. 2019;83(3):443-449
  159. 159. Boku N, Ryu MH, Kato K, Chung HC, Minashi K, Lee KW, et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: Interim results of a randomized, phase II trial (ATTRACTION-4). Annals of Oncology. 2019;30(2):250-258
  160. 160. Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: Preliminary results of a phase Ib study. Journal of Clinical Oncology. 2016;34(23):2698-2704
  161. 161. Yang R, Sun L, Li CF, Wang YH, Yao J, Li H, et al. Galectin-9 interacts with PD-1 and TIM-3 to regulate T cell death and is a target for cancer immunotherapy. Nature Communications. 2021;12(1):832
  162. 162. Wang H, Kaur G, Sankin AI, Chen F, Guan F, Zang X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. Journal of Hematology & Oncology. 2019;12(1):59
  163. 163. Peiro I, Palmero R, Iglesias P, Diez JJ, Simo-Servat A, Marin JA, et al. Thyroid dysfunction induced by nivolumab: Searching for disease patterns and outcomes. Endocrine. 2019;64(3):605-613
  164. 164. Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Annals of Oncology. 2015;26(12):2375-2391
  165. 165. Abdel-Rahman O, Fouad M. Risk of pneumonitis in cancer patients treated with immune checkpoint inhibitors: A meta-analysis. Therapeutic Advances in Respiratory Disease. 2016;10(3):183-193
  166. 166. Asada M, Mikami T, Niimura T, Zamami Y, Uesawa Y, Chuma M, et al. The risk factors associated with immune checkpoint inhibitor-related pneumonitis. Oncology. 2021;99(4):256-259
  167. 167. Vardhana S, Cicero K, Velez MJ, Moskowitz CH. Strategies for recognizing and managing immune-mediated adverse events in the treatment of Hodgkin lymphoma with checkpoint inhibitors. The Oncologist. 2019;24(1):86-95
  168. 168. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al. 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. 2018;36(17):1714-1768
  169. 169. Miyahara K, Noda T, Ito Y, Hidaka H, Fujimoto S, Takedomi H, et al. An investigation of nine patients with gastrointestinal immune-related adverse events caused by immune checkpoint inhibitors. Digestion. 2020;101(1):60-65
  170. 170. Jain P, Gutierrez Bugarin J, Guha A, Jain C, Patil N, Shen T, et al. Cardiovascular adverse events are associated with usage of immune checkpoint inhibitors in real-world clinical data across the United States. ESMO Open. 2021;6(5):100252
  171. 171. Lal JC, Brown SA, Collier P, Cheng F. A retrospective analysis of cardiovascular adverse events associated with immune checkpoint inhibitors. Cardiooncology. 2021;7(1):19
  172. 172. Hu J, Tian R, Ma Y, Zhen H, Ma X, Su Q , et al. Risk of cardiac adverse events in patients treated with immune checkpoint inhibitor regimens: A systematic review and Meta-analysis. Frontiers in Oncology. 2021;11:645245
  173. 173. Wen L, Zou X, Chen Y, Bai X, Liang T. Sintilimab-induced autoimmune diabetes in a patient with the anti-tumor effect of partial regression. Frontiers in Immunology. 2020;11:2076
  174. 174. Samoa RA, Lee HS, Kil SH, Roep BO. Anti-PD-1 therapy-associated type 1 diabetes in a Pediatric patient with relapsed classical Hodgkin lymphoma. Diabetes Care. 2020;43(9):2293-2295
  175. 175. Munakata W, Ohashi K, Yamauchi N, Tobinai K. Fulminant type I diabetes mellitus associated with nivolumab in a patient with relapsed classical Hodgkin lymphoma. International Journal of Hematology. 2017;105(3):383-386
  176. 176. Hughes MS, Pietropaolo M, Vasudevan MM, Marcelli M, Nguyen H. Checking the checkpoint inhibitors: A case of autoimmune diabetes after PD-1 inhibition in a patient with HIV. Journal of the Endocrine Society. 2020;4(12):bvaa150
  177. 177. Nalbantoglu M, Altunrende B, Tuncer OG, Akman G. Autoimmune encephalitis after treatment of Hodgkin's lymphoma with the immune checkpoint inhibitor nivolumab. Noro Psikiyatri Arsivi. 2021;58(2):163-165
  178. 178. Erol-Yildiz R, Kizilay T, Tuzun E, Misirli H, Turkoglu R. Nivolumab-induced autoantibody negative limbic encephalitis in a patient with Hodgkin lymphoma. Leukemia & Lymphoma. 2020;61(6):1519-1521
  179. 179. De la Hoz A, Foolad F, Gallegos C, Kornblau S, Kontoyiannis DP. Nivolumab-induced encephalitis post allogeneic stem cell transplant in a patient with Hodgkin's disease. Bone Marrow Transplantation. 2019;54(5):749-751
  180. 180. Tardy MP, Gastaud L, Boscagli A, Peyrade F, Gallamini A, Thyss A. Autoimmune hemolytic anemia after nivolumab treatment in Hodgkin lymphoma responsive to immunosuppressive treatment. A case report. Hematological Oncology. 2017;35(4):875-877
  181. 181. Palla AR, Kennedy D, Mosharraf H, Doll D. Autoimmune Hemolytic Anemia as a complication of nivolumab therapy. Case Reports in Oncology. 2016;9(3):691-697
  182. 182. Rogado J, Sanchez-Torres JM, Romero-Laorden N, Ballesteros AI, Pacheco-Barcia V, Ramos-Levi A, et al. Immune-related adverse events predict the therapeutic efficacy of anti-PD-1 antibodies in cancer patients. European Journal of Cancer. 2019;109:21-27
  183. 183. Weber JS, Hodi FS, Wolchok JD, Topalian SL, Schadendorf D, Larkin J, et al. Safety profile of nivolumab monotherapy: A pooled analysis of patients with advanced melanoma. Journal of Clinical Oncology. 2017;35(7):785-792
  184. 184. Eggermont AMM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, et al. Association between immune-related adverse events and recurrence-free survival among patients with stage III melanoma randomized to receive pembrolizumab or placebo: A secondary analysis of a randomized clinical trial. JAMA Oncology. 2020;6(4):519-527

Written By

Jiawen Huang and Juan Huang

Submitted: 25 July 2022 Reviewed: 29 August 2022 Published: 31 October 2022