Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4237 2023-06-29 12:39:47 |
2 format change + 3 word(s) 4240 2023-06-29 12:48:50 | |
3 No changes were made Meta information modification 4240 2023-07-21 20:53:49 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Perdikis-Prati, S.; Sheikh, S.; Bouroumeau, A.; Lang, N. Immune Checkpoint Blockade in Lymphoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/46217 (accessed on 27 July 2024).
Perdikis-Prati S, Sheikh S, Bouroumeau A, Lang N. Immune Checkpoint Blockade in Lymphoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/46217. Accessed July 27, 2024.
Perdikis-Prati, Sarah, Semira Sheikh, Antonin Bouroumeau, Noémie Lang. "Immune Checkpoint Blockade in Lymphoma" Encyclopedia, https://encyclopedia.pub/entry/46217 (accessed July 27, 2024).
Perdikis-Prati, S., Sheikh, S., Bouroumeau, A., & Lang, N. (2023, June 29). Immune Checkpoint Blockade in Lymphoma. In Encyclopedia. https://encyclopedia.pub/entry/46217
Perdikis-Prati, Sarah, et al. "Immune Checkpoint Blockade in Lymphoma." Encyclopedia. Web. 29 June, 2023.
Immune Checkpoint Blockade in Lymphoma
Edit

Immune checkpoint blockade (ICB) has revolutionized the prognosis of several advanced-stage solid tumors. However, its success has been far more limited in hematological malignancies and is mostly restricted to classical Hodgkin lymphoma (cHL) and primary mediastinal B cell lymphoma (PMBCL). In patients with non-Hodgkin lymphoma (NHL), response to PD-1/PD-L1 ICB monotherapy has been relatively limited, although some subtypes are more sensitive than others. Numerous predictive biomarkers have been investigated in solid malignancies, such as PD-L1 expression, tumor mutational burden (TMB) and microsatellite instability (MSI), among others. 

Hodgkin lymphoma non-Hodgkin lymphoma PD-1/PD-L1 checkpoint inhibitors tumor mutational burden predictive biomarkers

1. Introduction

The tumor microenvironment (TME), consisting of T cells, tumor-associated macrophages (TAMs), dendritic cells (DCs), neutrophils and natural killer (NK) and stromal cells, is thought to play a significant role in the development and progression of many cancers, as well as in tumor escape from the immune system. Accumulation of somatic mutations during oncogenesis has been shown to result in the presentation of neoantigens at the tumor cell surface, which can elicit tumor-specific CD4+ and CD8+ T cells with antitumor potential [1]. Many studies have demonstrated that tumor evasion is, at least in part, mediated by the inhibition of antitumor T-cell responses, mostly via upregulation of immune checkpoint molecules [2][3][4]. The understanding of these resistance mechanisms led to the delineation of the concept of immune checkpoint blockade (ICB).
More precisely, interaction between the immune checkpoint (IC) molecule programmed cell death protein 1 (PD-1) expressed by activated tumor-infiltrating T cells and its ligands (PD-L1 and PD-L2) expressed by the surrounding tumor and TME cells commonly leads to downregulation of neoantigen-specific T-cell responses. Blocking these interactions is a frequently used therapeutic approach to restore the antitumor effect of the host immune system [5][6].
In lymphoproliferative disorders, PD-1 is frequently expressed on tumor cells themselves as in tumor-infiltrating lymphocytes (TILs), while its ligands may be upregulated by tumor cells (some B-cell or T-cell lymphomas) but also TME cells such as TAMs, mast cells and mesenchymal cells [7][8][9]. PD-1 blockade using nivolumab or pembrolizumab has dramatically improved the prognosis of relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) and is now a well-recognized therapeutic option in this setting [10][11][12]. On the other hand, the efficacy of PD-1 or PD-L1 blockades in non-Hodgkin lymphoma (NHL) has shown disappointingly low response rates, except for some specific subsets of NHL, such as primary mediastinal B cell lymphoma (PMBCL) [13], primary testicular lymphoma (PTL) or primary central nervous system lymphoma (PCNSL) [14][15]. Interestingly, these NHL subsets have been shown to be highly infiltrated by T cells [16][17]. Of note, the different anti-PD-1/PD-L1 antibodies used in the clinic vary in their IgG isotypes and affinity to the various Fc gamma receptors (FcγRs) expressed on immune cells; however, whether these changes translate into different clinical benefits is not well established [18]. A persistent challenge remains the identification of predictive biomarkers of response to PD-1/PD-L1 ICB in this setting.

2. Clinical Efficacy of ICB in Lymphomas

2.1. Classical Hodgkin Lymphoma (cHL)

2.1.1. PD-1/PD-L1 ICB as Monotherapy

The safety and activity of PD-1 ICB was first tested by Ansell et al. (2015) in a phase 1 study on 23 heavily pretreated cHL patients who received biweekly nivolumab, a PD-1 inhibitor, demonstrating an impressive 87% overall response rate (ORR) and a 17% complete metabolic response rate (CMR) [11][19]. Larger studies subsequently confirmed the efficacy of PD-1 ICB monotherapy in R/R cHL using either nivolumab or pembrolizumab [20][21][22][23]. All trials showed similar results in R/R cHL patients, including those relapsing after autologous stem cell transplantation (ASCT) and those ineligible for ASCT. At a 5-year follow-up, both CheckMate 205 and KEYNOTE-087 trials demonstrated sustained responses, with median PFSs of 15 months and 13.7 months, respectively. Additionally, a subgroup analysis of the KEYNOTE-087 trial demonstrated the efficacy of pembrolizumab after treatment with the anti-CD30 antibody drug conjugate brentuximab vedotin (BV) [24]. Based on these results, the FDA approved nivolumab (2016) and pembrolizumab (2017) for R/R cHL patients relapsing after ASCT and BV [25]. More recently, the phase 3 KEYNOTE-204 study (2021) compared pembrolizumab and BV in patients with R/R cHL either following ASCT or in patients ineligible for ASCT and showed an increased median progression-free survival (PFS) for pembrolizumab over BV [26].
In addition to nivolumab and pembrolizumab, several other PD-1 inhibitors have been investigated in R/R cHL early-phase trials. PD-1/PD-L1 antibodies vary in their IgG isotypes. PD-1 IgG4 antibodies include nivolumab, pembrolizumab, sintilimab, camrelizumab, tislelizumab and zimberelimab, with ORRs ranging from 42% to 91% [27][28][29][30][31][32][33][34][35], while penpulimab, an IgG1 antibody, demonstrated an ORR of 89% with a 47% CR in a multicenter phase 1/2 trial [36]. To the researchers' knowledge, avelumab, an IgG1 anti-PD-L1 antibody, is the only anti-PD-L1 antibody that has been tested in this setting, achieving a 42% ORR and a 19% CR [37].

2.1.2. PD-1/PD-L1 ICB in Combination with Chemotherapy

Frontline Setting

Assuming that PD-1/PD-L1 ICB may prime the TME for the induction of antitumor T-cell responses before patients receive cytotoxic agents, both sequential and concomitant combinations of ICB with conventional chemotherapy have been evaluated in the frontline setting. Ramchandren et al. (2019) evaluated a sequential approach of four cycles of single-agent nivolumab followed by twelve cycles of nivolumab given concomitantly with doxorubicin, vinblastine and dacarbazine (N-AVD) in patients with newly diagnosed advanced-stage cHL (cohort D of CheckMate 205) [38]. N-AVD has also been investigated in early-stage unfavorable cHL in a phase 2 trial conducted by the German Hodgkin Study Group (2020), achieving similar response rates [39]. N-AVD regimen is currently being evaluated in a frontline randomized phase 3 study against BV-AVD in patients with advanced-stage cHL, with an estimated completion date in 2024 (NCT03907488). Similarly, a sequential strategy of pembrolizumab monotherapy followed by combination with AVD has been evaluated in unfavorable or advanced-stage cHL patients [40]. Another ongoing nonrandomized PET-adapted phase 2 study (NCT03617666) is investigating the safety and efficacy of sequential avelumab followed by ABVD in first-line high-risk cHL [41]. As approximatively twenty percent of newly diagnosed cHL patients are ineligible for intensive chemotherapy regimens and consequently at risk of experiencing worse outcomes [42][43], Cheson and colleagues (2020) investigated the combination of 8 cycles of BV with nivolumab for older (>60 years) or chemo-ineligible newly diagnosed cHL patients. The trial was prematurely closed after an interim analysis failed to show that the combination met the predefined ORR criteria (ORR > 68%); however, 61% of all evaluable patients displayed an objective response and 48% achieved CMRs, demonstrating that this well-tolerated combination is active in this frail population [44].

Relapsed/Refractory Setting

Herrera and colleagues (2018) evaluated a nivolumab and BV combination for up to four cycles in the first-salvage-setting phase 1/2 study; this regimen was demonstrated to be well tolerated, achieving an ORR of 82%, including a 61% CMR [45]. An extended 3-year follow-up confirmed the durability of responses, with an estimated 77% 3-year PFS after a median follow-up of 34.3 months [46]. A phase 3 trial further investigating this regimen versus BV alone in R/R or ASCT-ineligible cHL patients was closed due to insufficient enrolment (NCT03138499). Three other trials demonstrated the efficacy of PD-1 ICB combined with standard salvage chemotherapy (ICE or gemcitabine, vinorelbine and liposomal doxorubicin (GVD) prior to ASCT) [47][48][49][50]. Atezolizumab, a PD-L1 inhibitor, scarcely tested as monotherapy in lymphoma ([51], NCT03120676), is currently being investigated in combination with a BeGEV regimen (bendamustine, gemcitabine and vinorelbine) (NCT05300282).
Patients with R/R HL who undergo ASCT have an expected 60% 18-month PFS [52][53][54]. Lepik et al. evaluated the utility of nivolumab and bendamustine administered for up to three cycles in heavily pretreated R/R cHL patients who had failed at least two lines of prior therapy, including nivolumab monotherapy. Among all enrolled patients (n = 30), 26 achieved a response (ORR: 87%) and 17 a CMR (57%) [55]. To the researchers' knowledge, only one trial phase 2 (Armand et al.) investigated the role of PD-1/PD-L1 ICB (pembrolizumab) as a consolidative therapy in the post-ASCT setting, achieving PFS and OS rates at 18 months of 82% and 100%, respectively [56].

2.1.3. PD-1/PD-L1 ICB Combined with Other Agents

The combination of nivolumab with other ICBs, such as ipilumumab, an anticytotoxic T-lymphocyte-associated protein 4 (CTLA4) inhibitor, or lirilumab, an antibody targeting the killer cell Ig-like receptors (KIR) expressed by NK cells, did not seem to significantly improve the response rate in 31 R/R cHL patients enrolled in phase 1 of the CheckMate 039 trial (2016, 2021) [57][58]. The safety and efficacy of a triple combination of nivolumab, ipilimumab and BV was tested in a phase 1/2 trial conducted by Diefenbach et al. (2020) on patients with R/R cHL in comparison with nivolumab-BV or ipilimumab-BV. The triple regimen was associated with increased toxicity without clinical benefit [59]. Potentially more promising are PD-1/PD-L1 ICB and histone deacetylase inhibitor (HDACi) combinations. The combination of pembrolizumab with entinostat was investigated in 22 R/R cHL patients by Sermer et al. (2020, 2021) in a phase 2 trial with an ORR of 86% [60][61]. Similarly, a phase 2 trial with camrelizumab, another PD-1 inhibitor, combined with decitabine, demonstrated improved response rates (ORR 95%, CMR 79%) compared with camrelizumab monotherapy (ORR 89%, CMR 32%) [33][34].
Various combinations of PD-1/PD-L1 ICB with novel immunotherapies are currently under investigation in early-phase trials. A first report of pembrolizumab combined with AFM-13, a CD30-CD16 bispecific antibody stimulating innate immune cells, such as NK and macrophages, achieved an ORR of 83% in R/R cHL patients who had received a median of three prior lines of therapy [62][63]. A recent phase 1 study evaluating the benefit of adoptive cellular therapy consisting of tumor-associated antigen (TAA)-specific T cells enrolled 10 patients with R/R HL (n = 8 active disease, n = 2 adjuvant after ASCT) to receive TAA-specific T cells (autologous or allogenic) with nivolumab given as a priming agent in 6 out of the 10 patients. Among the patients with active disease, one patient achieved CMR and seven had stable disease (SD) at 3 months [62]. In addition, there is growing evidence that chimeric antigen receptor (CAR) T-cell fitness may be improved by the adjunction of PD-1 ICB. With the limitation of a small sample size (n = 12), PD-1 ICB administration after CD30 CAR T-cell therapy in CD30+ lymphoma patients (n = 9 cHL, n = 1 angioimmunoblastic T-cell lymphoma, n = 2 gray zone lymphoma) suggested improved efficacy (ORR 86% versus 100%; CR 27% versus 80%) [64]. Similarly, the role of anti-PD-1 therapy after CD30 CAR T-cell treatment is currently being evaluated in R/R cHL patients (NCT04134325) [65][66]. Recently, Timmerman et al. (2022) demonstrated that the association of favezelimab (lymphocyte activating gene-3 (LAG-3) ICB) and pembrolizumab could be an effective therapeutic option for patients progressing under PD-1 ICB (ORR 31%, CR 7%) [67]. Other combination approaches in the R/R cHL setting are ongoing, such as nivolumab with ruxolitinib (NCT03681561) and nivolumab/pembrolizumab combined with radiation (NCT04419441).

2.2. Non-Hodgkin Lymphoma (NHL)

2.2.1. PD-1/PD-L1 ICB as Monotherapy

Aggressive NHL

First tested on a range of various hematological malignancies, nivolumab achieved an ORR of 36% and a CMR of 18% in the 11 R/R diffuse large B cell lymphoma (DLBCL) patients enrolled in a phase 1 study [68]. Single-agent activity was further evaluated in DLBCL patients who were relapsing after ASCT or ineligible for ASCT [69] and in patients with various hematologic malignancies relapsing after allogeneic stem cell transplantation (allo-SCT) [70], demonstrating modest benefits. In contrast to cHL, maintenance treatment with pembrolizumab administered after ASCT in DLBCL and PMBCL patients did not show any clinical benefit [71]. A randomized phase 3 trial investigating tislelizumab, a PD-1 inhibitor, as maintenance in DLBCL after ASCT is planned (NCT04799314). Even though the benefit of PD-1/PD-L1 inhibitors as monotherapy in R/R aggressive NHL has been disappointing, better activity has been observed in patient subsets, such as PMBCL, PCNSL and PTL patients [14][15][68]. Benefits from pembrolizumab were demonstrated in R/R PMBCL with 45–48% ORR and 13–33% CMR, leading to the accelerated approval of pembrolizumab by the FDA in this setting [23][72]. Nivolumab demonstrated activity in four patients with R/R PCNSL and one patient with PTL CNS recurrence; all five patients had clinical and radiographic responses to the monotherapy [15]. The efficacy in these specific aggressive NHL subgroups is likely due to their particular biology. This is described in further detail in the biomarker section. Pembrolizumab has also been tested on nine patients experiencing Richter transformation (RT) [73].
PD-1/PD-L1 ICB demonstrated modest results in natural killer NK/T-cell NHL. In a phase 1 trial, nivolumab monotherapy achieved no observed CR [68]. On the other hand, pembrolizumab showed slightly improved activity in cutaneous T-cell lymphoma (CTCL) [74] and in a series of seven R/R extranodal NK/T-cell lymphoma (ENKTL) patients, with two of them achieving CR [75]. This initial signal of activity in R/R ENKTL was recently confirmed by two phase 2 trials evaluating avelumab, a PD-L1 inhibitor, and sintilimab, a PD-1 inhibitor [76][77].

Indolent B NHL

PD-1/PD-L1 ICB has also been evaluated in indolent lymphomas. Lesokhin et al. (2016) administered single-agent nivolumab to 10 R/R FL patients (ORR 40%) [68], but this result was not confirmed in a larger phase 2 study conducted by Armand et al. (CheckMate-140) (ORR 4%) [78]. To date, other indolent lymphoid malignancies such as chronic lymphocytic leukemia (CLL), marginal cell lymphoma (MZL) and Waldenström macroglobulinemia (WM) have not shown significant response rates with PD-1/PD-L1 ICB [73][79][80].

2.2.2. PD-1/PD-L1 ICB in Combination with Other Agents

Aggressive NHL

  • Frontline setting
PD-1/PD-L1 ICB does not seem to add much benefit to frontline immunochemotherapy in newly diagnosed DLBCL. The combination of atezolizumab with frontline R-CHOP was tested on DLBCL patients [81][82] as a combination of pembrolizumab with R-CHOP in patients with either DLBCL or grade 3b FL [83]. Similarly, a durvalumab (another PD-L1 inhibitor) and R-CHOP combination did not seem to add any benefit [84]. An ongoing phase 3 trial is currently investigating nivolumab combined with DA-EPOCH-R versus DA-EPOCH-R in newly diagnosed patients with PMCBL (NCT04759586).
  • Relapse/refractory setting
Various therapeutic combinations, including PD-1/PD-L1 ICB, have been investigated in the R/R NHL setting, most of them showing disappointing results. Nivolumab with ipilimumab or lirilumab did not show any improved clinical activity [57][58], and nor did combinations of either nivolumab, pembrolizumab or durvalumab with BTKi (ibrutinib [85][86] or acalabrutinib [87]) or dinaciclib, a cycline kinase inhibitor (CDKi) [88]. Similarly, durvalumab in combination with ibrutininb, lenalidomide +/− rituximab or bendamustine +/− rituximab only adds a small benefit [89].
With the exceptions of specific NHL subtypes and potentially CAR T-cell association, PD-1/PD-L1 ICBs generally do not add much clinical benefit when combined with other active agents in the R/R NHL setting.

Indolent B NHL

  • Frontline setting
The combination of PD-1/PD-L1 ICB with anti-CD20 monoclonal antibodies +/− bendamustin has been tested on advanced-grade 1-3A FL [90][91]; the addition of chemotherapy resulted in an inacceptable toxicity profile, with fatal adverse events occurring in five patients (pneumonia, sudden death, cardiac arrest (due to severe immune-mediated myocarditis and bronchiolitis obliterans), gastrointestinal tract/biliary adenocarcinoma, progressive multifocal leukoencephalopathy) [92][93].
  • Relapse/refractory setting
The addition of PD-1/PD-L1 ICB to anti-CD 20 monoclonal antibodies has been extensively tested in the R/R setting of indolent lymphomas, with no or only modest results [94][95][96][97]. Atezolizumab with obinutuzumab and lenalidomide resulted in a 78% ORR and a 72% CR rate [98]. The LYSA group investigated atezolizumab, obinutuzumab and venetoclax in R/R FL and MZL with reported ORRs of 54% and 67%, respectively [79]. Durvalumab or nivolumab in combination with ibrutinib resulted in modest response rates in R/R FL patients in two trials [86][99][100].

3. Predictive Biomarkers of Response to ICB in Lymphoma

Predictive biomarkers of response to ICBs have been identified in different solid tumors, but our ability to accurately predict response in lymphoma remains suboptimal. Mechanisms of immune evasion may differ from one lymphoma subtype to another. Several biomarkers have been investigated in lymphoid malignancies, including the PD-L1 H-score, alterations/amplification of the 9p24.1 gene, microsatellite instability (MSI), tumor mutational burden (TMB), the density of intratumoral CD8+ T lymphocyte infiltrates, genetic alterations in MHC classes I and II, and miRNA-21, among others [101].

3.1. Tissue Expression and Plasma Levels of PD-Ls

PD-L1 and PD-L2 expression of TME can be influenced by two main signaling pathways. The extrinsic pathway relies on the release of inflammatory signals (i.e., IFN-γ) by TILs after tumor antigen recognition, consequently upregulating the expression of PD-L1/PD-L2 in tumor cells and TME cells [102]. On the other hand, the intrinsic pathway is mainly driven by genomic alteration of the 9p24.1 gene, EBV infection and the activation of the JAK/STAT transcription pathway [13][103].

3.2. Gene Alterations to 9p24.1

Copy number alterations (CNAs) to chromosome 9p24.1 (i.e., polysomy, copy gain, amplification, rarely translocation), leading to increased expression of PD-1 ligands in cHL, PMBCL and some extranodal large B-cell NHLs, are an important mechanism of tumor immune evasion. These alterations have been reported in the majority of cHLs (97%) [104], 63–75% of PMBCLs [105][106], over 40% of PTLs and roughly 20% of PCNSLs [107]. Unlike cHL, there is generally a low incidence (10–27%) of structural variations in PD-L1/PD-L2 in DLBCL [108][109]. The H-score could be used as a surrogate marker of the level of 9p24.1 gene alteration and predict the response to ICB [110]. cHL patients presenting 9p24.1 CNAs are more likely to present with advanced-stage disease and worse prognosis, with significantly shorter PFS [38]. On the other hand, they also tend to benefit from therapy with a PD-1 inhibitor [38][104][111]. However, Green et al. (2010) described that Hodgkin Reed–Sternberg cell lines harboring low levels of 9p24.1 CNAs still expressed PD-L1, suggesting that PD-L1 expression could be driven by other mechanisms [104]. Although the vast majority of NHLs have a considerably low sensitivity to PD-1/PD-L1 ICB, particular subtypes, such as PCNSL, PTL and PMBCL harbor 9p24.1 CNAs, conferring them an increased vulnerability to these agents [15][107]. In PMBCL, the magnitude of 9p24.1 CNAs is significantly associated with PD-L1 expression and survival outcome [14].

3.3. Epstein–Barr Virus (EBV) and JAK/STAT Signaling Pathway

In cHL and PMBCL cell lines, Green et al. (2010) observed that broader 9p24.1 amplifications also included the Janus kinase 2 (JAK2) locus located upstream from PD-1 ligand genes [104]. As a consequence, 9p24.1 CNAs directly enhanced the JAK/STAT intrinsic signaling pathway, promoting PD-1 ligand transcription [104]. Additionally, the JAK/STAT signaling pathway can also be activated by various cytokines secreted by cells within the TME (i.e., IFN-γ), leading to PD-L1 upregulation on tumor cells [112].
Even though the underlying mechanisms have not yet been fully elucidated, recent studies indicate that viruses also use the PD-1 signaling pathway to escape immune detection [113]. Thus, EBV-positive lymphomas tend to benefit from ICB therapy [114][115]. The expression of EBV latent membrane protein 1 (LMP1) or latent membrane protein 2a (LMP2a) was shown to be sufficient to activate the signaling cascade of the JAK/STAT pathway, leading to PD-L1 overexpression on tumor cells [113]. The expression of EBV has been implicated in approximately 40% of cHLs, 50–70% of post-transplant lymphoproliferative disorders (PTLDs), more than 95% of endemic Burkitt lymphomas (BLs), 20–30% of sporadic BLs, 25–40% of immunodeficiency-associated BLs, EBV-positive DLBCL NOS and most NK/T-cell lymphomas [116][117][118][119]. In a cohort of 1253 patients with DLBCL, PD-L1 protein expression was significantly associated with EBV positivity in the non-GCB subtype and showed a trend toward inferior OS in these patients [120].

3.4. Tissue Tumor Mutational Burden (TMB) and Plasma Tumor Mutational Burden (pTMB)

Based on the concept that higher mutation loads could yield to improved T-cell recognition of tumors via increased neoantigen production, tumor mutational burden (TMB), defined as the number of somatic mutations per megabase (mut/Mb) [121], was investigated as a predictive biomarker of response to PD-1/PD-L1 ICB in several solid malignancies [122][123][124]. Surprisingly, TMB has only been reported in a few lymphoma patient series, and its relationship to response to PD-1/PD-L1 ICB is not well established. Using a 406-DNA gene panel, Galanina and colleagues showed a median TMB of 1.7 mut/Mb across all hematologic malignancies [125]. Applying whole-exome sequencing (WES), Wienand et al. described a median TMB of 7.7 mut/Mb in cHL, although TMB may differ per EBV status, with EBV-negative cHL harboring a higher mutational level, almost similar to NSCLC (median 9.8 mut/Mb) and melanoma (13.5 mt/Mb) TMB [126][127][128]. The median TMB of PMBCL has been shown to be within the same range as cHL (7.0 mut/Mb) [129]. Using LymphomaScan, a 405-gene panel, Cho et al. (2021) investigated TMB in different NHLs (B-cell neoplasms n = 243, T- and NK-cell neoplasms n = 53, precursor lymphoid neoplasms n = 4), reporting single-nucleotide variant (SNV) and insertion–deletion mutations (Indels) for each subtype. Overall, they found that B-cell lymphomas had statistically more mutations (24 SNV/Indel) than T- and NK-cell lymphomas (17 SNV/Indel). PMBCL accounted for the highest TMB load (32 SNV/Indel), followed by PCNSL (30 SNV/Indel), DLBCL NOS (23 SNV/Indel), ALK-negative anaplastic large cell lymphoma (ALCL) (23 SNV/Indel), ALK-positive ALCL (14 SNV/Indel), follicular T-cell lymphoma (14 SNV/Indel) and nodal peripheral T-cell lymphoma with TFH phenotype (PTCL TFH) (14.5 SNV/Indel) [130].
Circulating tumor DNA (ctDNA), sometimes referred to as liquid biopsy, reflects tumor DNA spread within the circulation [131]. Plasma TMB (pTMB) has been investigated in solid tumors and DLBCL as a surrogate to quantify overall tumor burden and mutational load and more accurately capture molecular tumor heterogeneity and clonal evolution [132][133]. To the researchers' knowledge, only a limited number of clinical studies have been conducted on lymphoma. One study evaluated pTMB in cHL, showing that higher baseline ctDNA and a sharper ctDNA decrease (>40%) significantly correlated with better clinical responses to sintilimab [134]. Another study, reported as an abstract only, compared TMB and pTMB in several NHL subtypes and concluded that there is a higher TMB in DLBCL than in other lymphoma subtypes [135]. Based on these results, it seems that a higher TMB seems to correlate to PD-1 ICB response, as this is the case for other solid cancers [136].

3.5. MSI and d-MMR

Microsatellite instability (MSI), caused by deficiency of the DNA mismatch repair (MMR) system, results in a higher mutational load and tumor antigen expression, leading to increased antitumor T-cell activation. MSI and MMR status are well-recognized prognostic factors in several solid tumors. The National Cancer Institute recommends a panel of five microsatellite markers for MSI tumor detection (two mononucleotide repeats: BAT-25, BAT-26; three dinucleotide repeats: D2S123, D5S346, D17S250). MSI-high tumors are defined as having instability in two or more of these markers [137]. Like those harboring a high TMB, patients with MSI-high tumors achieve durable responses to ICB [138]. Pembrolizumab and dostarlimab, both anti-PD-1 antibodies, are now approved by the FDA for the treatment of unselected advanced-stage tumors with MSI/dMMR [139]. Even though MSI-associated hypermutation represents a potential biomarker for the efficacy of PD-1 blockade, it is likely infrequent in lymphoma; reported frequencies are low, occurring in only 0.46% of cHLs, 3.2% of DLBCLs and 8% of PMBCLs [128][129][140].

3.6. MHC Expression

Antigen presentation by major histocompatibility complex (MHC) molecules is an essential step in T-cell recognition and tumor cell eradication, as T-cell activation does not occur at the tumor site but in the lymph nodes. The MHC I complex, including the beta-2-microglobulin (B2M) subunit, presents tumor-generated peptides at the cell surface, which can be recognized by cytotoxic CD8+ T cells. On the other hand, MHC II complexes present tumor antigens to CD4+ T cells [141]. Acquired mutations in the antigen processing and presentation molecules are a potential mechanism of tumor escape [142]. B2M mutations resulting in decreased MHC I expression are a well-described acquired resistance mechanism to PD-1/PD-L1 ICB in melanoma [143]. By contrast, in a study on cHL, Roemer et al. (2016) discovered that over 75% of patients had decreased or absent expression of B2M/MHCI and MHCII on tumor cells [144]. Nonetheless, patients with cHL are known to have a high response rate to PD-1 ICB. The postulated alternate mechanism triggering a response to PD-1/PD-L1 ICB could be MHC class II expression. Indeed, melanoma patients with low or absent B2M/MHCI expression but increased MHCII expression demonstrated higher responses to ICB [145]. This suggests that MHCII expression could be a potential biomarker for PD-1/PD-L1 ICB response and requires further evaluation.

3.7. Intratumoral CD8+ T Lymphocyte Infiltrate Density

Several studies have demonstrated that response to PD-1/PD-L1 ICB seems tightly related to immune cell infiltration. High levels of cytotoxic CD8+ T lymphocytes at the TME have been independently linked to improved outcomes in several lymphomas (i.e., DLBCL [146], PTL [147], FL [148], MZL [149] and HL [150][151]). The association between tumor CD8+ T lymphocyte infiltration and response to PD-1/PD-L1 ICB has been confirmed for solid tumors through a large metanalysis conducted by Li et al. (2021) [152]. Such data have not yet been reported for lymphoma.

3.8. MicroRNAs

MicroRNAs (miRNAs) are small sequences of noncoding RNAs acting as gene expression regulators. miRNAs are involved in various physiologic and pathologic processes, including immune responses [153]. Numerous miRNAs are recognized as prognostic biomarkers and are investigated as potential therapeutic targets in solid and hematologic tumors [154]. Among all miRNAs, microRNA-21 (miR-21) is one of the most frequently overexpressed miRNAs in solid tumors and B-NHL [155]. High plasma miR-21 levels have been associated with poor prognosis in several B-NHL subtypes (e.g., primary gastrointestinal DLBCL [156], DLBCL [157], Burkitt lymphoma [158], PCNSL [159]). miR-21 depletion may enhance antitumor activity through the polarization of macrophages into an M1-like phenotype; on the other hand, miR-21 also upregulates PD-L1 expression [160]. Taking advantage of these properties, Xi et al. recently showed in a preclinical model a synergetic effect of miR-21-depleting therapy and PD-1 ICB [160].

3.9. Gut Microbiome

Recent evidence shows that the biodiversity of the gut microbiome could influence the antitumor activity of PD-1/PD-L1 ICB; this was notably investigated in melanoma [161][162]. Namely, Liu et al. (2021) demonstrated that a favorable gut microbiome characterized by its diversity and the presence of specific bacteria species could influence the innate and adaptive immune system by increasing antigen presentation and augmenting T-cell response. On the other hand, antibiotic use may disrupt the gut microbiome and impair cytotoxic T-cell responses against tumor cells [163]. Hwang et al. studied this association in a small retrospective cohort of 62 cHL patients treated with ICB and observed that prior and/or current antibiotherapy was linked to inferior outcomes [164]. Recently, Casadei et al. (2021) prospectively collected feces (at baseline, before each treatment, at response assessment and for grade >2 adverse events) from cHL (n = 12) and PMBCL (n = 5) patients undergoing PD-1PD-L1 ICB. They reported that the results of the first six patients (all cHL) showed clear differences in their microbiomes, with a depletion of health-promoting microbial components compared with healthy controls [165].

References

  1. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608.
  2. Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15.
  3. Armengol, M.; Santos, J.C.; Fernández-Serrano, M.; Profitós-Pelejà, N.; Ribeiro, M.L.; Roué, G. Immune-Checkpoint Inhibitors in B-Cell Lymphoma. Cancers 2021, 13, 214.
  4. Petitprez, F.; Meylan, M.; de Reyniès, A.; Sautès-Fridman, C.; Fridman, W.H. The Tumor Microenvironment in the Response to Immune Checkpoint Blockade Therapies. Front. Immunol. 2020, 11, 784.
  5. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264.
  6. Kyi, C.; Postow, M.A. Immune checkpoint inhibitor combinations in solid tumors: Opportunities and challenges. Immunotherapy 2016, 8, 821–837.
  7. Gravelle, P.; Burroni, B.; Péricart, S.; Rossi, C.; Bezombes, C.; Tosolini, M.; Laurent, C. Mechanisms of PD-1/PD-L1 expression and prognostic relevance in non-Hodgkin lymphoma: A summary of immunohistochemical studies. Oncotarget 2017, 8, 44960.
  8. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704.
  9. Xie, W.; Medeiros, L.J.; Li, S.; Yin, C.C.; Khoury, J.D.; Xu, J. PD-1/PD-L1 Pathway and Its Blockade in Patients with Classic Hodgkin Lymphoma and Non-Hodgkin Large-Cell Lymphomas. Curr. Hematol. Malig. Rep. 2020, 15, 372–381.
  10. Younes, A.; Santoro, A.; Shipp, M.; Zinzani, P.L.; Timmerman, J.M.; Ansell, S.; Armand, P.; Fanale, M.; Ratanatharathorn, V.; Kuruvilla, J.; et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: A multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016, 17, 1283–1294.
  11. Ansell, S.; Armand, P.; Timmerman, J.M.; Shipp, M.A.; Bradley Garelik, M.B.; Zhu, L.; Lesokhin, A.M. Nivolumab in Patients (Pts) with Relapsed or Refractory Classical Hodgkin Lymphoma (R/R cHL): Clinical Outcomes from Extended Follow-up of a Phase 1 Study (CA209-039). Blood 2015, 126, 583.
  12. Chen, R.; Zinzani, P.L.; Fanale, M.A.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 2125–2132.
  13. Ok, C.Y.; Young, K.H. Targeting the programmed death-1 pathway in lymphoid neoplasms. Cancer Treat. Rev. 2017, 54, 99–109.
  14. Armand, P.; Rodig, S.; Melnichenko, V.; Thieblemont, C.; Bouabdallah, K.; Tumyan, G.; Özcan, M.; Portino, S.; Fogliatto, L.; Caballero, M.D.; et al. Pembrolizumab in Relapsed or Refractory Primary Mediastinal Large B-Cell Lymphoma. J. Clin. Oncol. 2019, 37, 3291–3299.
  15. Nayak, L.; Iwamoto, F.M.; LaCasce, A.; Mukundan, S.; Roemer, M.G.M.; Chapuy, B.; Armand, P.; Rodig, S.J.; Shipp, M.A. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017, 129, 3071–3073.
  16. Zhou, H.; Xu-Monette, Z.Y.; Xiao, L.; Strati, P.; Hagemeister, F.B.; He, Y.; Chen, H.; Li, Y.; Manyam, G.C.; Li, Y.; et al. Prognostic factors, therapeutic approaches, and distinct immunobiologic features in patients with primary mediastinal large B-cell lymphoma on long-term follow-up. Blood Cancer J. 2020, 10, 49.
  17. Riemersma, S.A.; Oudejans, J.J.; Vonk, M.J.; Dreef, E.J.; Prins, F.A.; Jansen, P.M.; Vermeer, M.H.; Blok, P.; Kibbelaar, R.E.; Muris, J.J.; et al. High numbers of tumour-infiltrating activated cytotoxic T lymphocytes, and frequent loss of HLA class I and II expression, are features of aggressive B cell lymphomas of the brain and testis. J. Pathol. 2005, 206, 328–336.
  18. Chen, X.; Song, X.; Li, K.; Zhang, T. FcγR-Binding Is an Important Functional Attribute for Immune Checkpoint Antibodies in Cancer Immunotherapy. Front. Immunol. 2019, 10, 292.
  19. Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin’s Lymphoma. N. Engl. J. Med. 2015, 372, 311–319.
  20. Armand, P.; Shipp, M.A.; Ribrag, V.; Michot, J.-M.; Zinzani, P.L.; Kuruvilla, J.; Snyder, E.S.; Ricart, A.D.; Balakumaran, A.; Rose, S.; et al. Programmed Death-1 Blockade with Pembrolizumab in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure. J. Clin. Oncol. 2016, 34, 3733–3739.
  21. Maruyama, D.; Hatake, K.; Kinoshita, T.; Fukuhara, N.; Choi, I.; Taniwaki, M.; Ando, K.; Terui, Y.; Higuchi, Y.; Onishi, Y.; et al. Multicenter phase II study of nivolumab in Japanese patients with relapsed or refractory classical Hodgkin lymphoma. Cancer Sci. 2017, 108, 1007–1012.
  22. Maruyama, D.; Terui, Y.; Yamamoto, K.; Fukuhara, N.; Choi, I.; Kuroda, J.; Ando, K.; Hattori, A.; Tobinai, K. Final results of a phase II study of nivolumab in Japanese patients with relapsed or refractory classical Hodgkin lymphoma. Jpn. J. Clin. Oncol. 2020, 50, 1265–1273.
  23. Armand, P.; Kuruvilla, J.; Michot, J.-M.; Ribrag, V.; Zinzani, P.L.; Zhu, Y.; Marinello, P.; Nahar, A.; Moskowitz, C.H. KEYNOTE-013 4-year follow-up of pembrolizumab in classical Hodgkin lymphoma after brentuximab vedotin failure. Blood Adv. 2020, 4, 2617–2622.
  24. Zinzani, P.L.; Chen, R.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; Tomita, A.; et al. Pembrolizumab monotherapy in patients with primary refractory classical hodgkin lymphoma who relapsed after salvage autologous stem cell transplantation and/or brentuximab vedotin therapy: KEYNOTE-087 subgroup analysis. Leuk. Lymphoma 2020, 61, 950–954.
  25. Vaddepally, R.K.; Kharel, P.; Pandey, R.; Garje, R.; Chandra, A.B. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers 2020, 12, 738.
  26. Kuruvilla, J.; Ramchandren, R.; Santoro, A.; Paszkiewicz-Kozik, E.; Gasiorowski, R.; Johnson, N.A.; Fogliatto, L.M.; Goncalves, I.; de Oliveira, J.S.R.; Buccheri, V.; 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. Lancet Oncol. 2021, 22, 512–524.
  27. Song, Y.; Gao, Q.; Zhang, H.; Fan, L.; Zhou, J.; Zou, D.; Li, W.; Yang, H.; Liu, T.; Wang, Q.; 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, 533–542.
  28. Song, Y.; Wu, J.; Chen, X.; Lin, T.; Cao, J.; Liu, Y.; Zhao, Y.; Jin, J.; Huang, H.; Hu, J.; et al. A Single-Arm, Multicenter, Phase II Study of Camrelizumab in Relapsed or Refractory Classical Hodgkin Lymphoma. Clin. Cancer Res. 2019, 25, 7363–7369.
  29. Shi, Y.; Su, H.; Song, Y.; Jiang, W.; Sun, X.; Qian, W.; Zhang, W.; Gao, Y.; Jin, Z.; Zhou, J.; 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 Haematol. 2019, 6, e12–e19.
  30. Lin, N.; Zhang, M.; Bai, H.; Liu, H.; Cui, J.; Ke, X.; Zhang, H.; Liu, L.; Yan, D.; Jiang, Y.; 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. Eur. J. Cancer 2022, 164, 117–126.
  31. Song, Y.; Gao, Q.; Zhang, H.; Fan, L.; Zhou, J.; Zou, D.; Li, W.; Yang, H.; Liu, T.; Wang, Q.; et al. Tislelizumab for Relapsed/Refractory Classical Hodgkin Lymphoma: 3-Year Follow-up and Correlative Biomarker Analysis. Clin. Cancer Res. 2022, 28, 1147–1156.
  32. Wu, J.; Song, Y.; Chen, X.; Lin, T.; Cao, J.; Liu, Y.; Zhao, Y.; Jin, J.; Huang, H.; Hu, J.; et al. Camrelizumab for relapsed or refractory classical Hodgkin lymphoma: Extended follow-up of the multicenter, single-arm, Phase 2 study. Int. J. Cancer 2022, 150, 984–992.
  33. Nie, J.; Wang, C.; Liu, Y.; Yang, Q.; Mei, Q.; Dong, L.; Li, X.; Liu, J.; Ku, W.; Zhang, Y.; et al. Addition of Low-Dose Decitabine to Anti–PD-1 Antibody Camrelizumab in Relapsed/Refractory Classical Hodgkin Lymphoma. J. Clin. Oncol. 2019, 37, 1479–1489.
  34. Liu, Y.; Wang, C.; Li, X.; Dong, L.; Yang, Q.; Chen, M.; Shi, F.; Brock, M.; Liu, M.; Mei, Q.; et al. Improved clinical outcome in a randomized phase II study of anti-PD-1 camrelizumab plus decitabine in relapsed/refractory Hodgkin lymphoma. J. Immunother. Cancer 2021, 9, e002347.
  35. Su, H.; Song, Y.; Jiang, W.; Sun, X.; Qian, W.; Zhang, W.; Gao, Y.; Jin, Z.; Zhou, J.; Jin, C.; et al. Sintilimab for relapsed/refractory classical Hodgkin’s lymphoma: Long-term follow-up on the multicenter, single-arm phase II ORIENT-1 study. J. Clin. Oncol. 2020, 38, 8034.
  36. Song, Y.; Zhou, K.; Jin, C.; Qian, Z.; Hou, M.; Fan, L.; Li, F.; Ding, K.; Zhou, H.; Li, X.; et al. Penpulimab for Relapsed or Refractory Classical Hodgkin Lymphoma: A Multicenter, Single-Arm, Pivotal Phase I/II Trial (AK105-201). Front. Oncol. 2022, 12, 925236.
  37. Herrera, A.F.; Burton, C.; Radford, J.; Miall, F.; Townsend, W.; Santoro, A.; Zinzani, P.L.; Lewis, D.; Fowst, C.; Brar, S.; et al. Avelumab in relapsed/refractory classical Hodgkin lymphoma: Phase 1b results from the JAVELIN Hodgkins trial. Blood Adv. 2021, 5, 3387–3396.
  38. Ramchandren, R.; Domingo-Domènech, E.; Rueda, A.; Trněný, M.; Feldman, T.A.; Lee, H.J.; Provencio, M.; Sillaber, C.; Cohen, J.B.; Savage, K.J.; et al. Nivolumab for Newly Diagnosed Advanced-Stage Classic Hodgkin Lymphoma: Safety and Efficacy in the Phase II CheckMate 205 Study. J. Clin. Oncol. 2019, 37, 1997–2007.
  39. Bröckelmann, P.J.; Goergen, H.; Keller, U.; Meissner, J.; Ordemann, R.; Halbsguth, T.V.; Sasse, S.; Sökler, M.; Kerkhoff, A.; Mathas, S.; 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 Oncol. 2020, 6, 872.
  40. Allen, P.B.; Savas, H.; Evens, A.M.; Advani, R.H.; Palmer, B.; Pro, B.; Karmali, R.; Mou, E.; Bearden, J.; Dillehay, G.; et al. Pembrolizumab followed by AVD in untreated early unfavorable and advanced-stage classical Hodgkin lymphoma. Blood 2021, 137, 1318–1326.
  41. Johnson, P.; Federico, M.; Kirkwood, A.; Fosså, A.; Berkahn, L.; Carella, A.; d’Amore, F.; Enblad, G.; Franceschetto, A.; Fulham, M.; et al. Adapted Treatment Guided by Interim PET-CT Scan in Advanced Hodgkin’s Lymphoma. N. Engl. J. Med. 2016, 374, 2419–2429.
  42. Evens, A.M.; Carter, J.; Loh, K.P.; David, K.A. Management of older Hodgkin lymphoma patients. Hematology 2019, 2019, 233–242.
  43. Moccia, A.A.; Aeppli, S.; Güsewell, S.; Bargetzi, M.; Caspar, C.; Brülisauer, D.; Ebnöther, M.; Fehr, M.; Fischer, N.; Ghilardi, G.; et al. Clinical characteristics and outcome of patients over 60 years with Hodgkin lymphoma treated in Switzerland. Hematol. Oncol. 2021, 39, 196–204.
  44. Cheson, B.D.; Bartlett, N.L.; LaPlant, B.; Lee, H.J.; Advani, R.J.; Christian, B.; Diefenbach, C.S.; Feldman, T.A.; Ansell, S.M. 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 Haematol. 2020, 7, e808–e815.
  45. Herrera, A.F.; Moskowitz, A.J.; Bartlett, N.L.; Vose, J.M.; Ramchandren, R.; Feldman, T.A.; LaCasce, A.S.; Ansell, S.M.; Moskowitz, C.H.; Fenton, K.; et al. Interim results of brentuximab vedotin in combination with nivolumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2018, 131, 1183–1194.
  46. Advani, R.H.; Moskowitz, A.J.; Bartlett, N.L.; Vose, J.M.; Ramchandren, R.; Feldman, T.A.; LaCasce, A.S.; Christian, B.A.; Ansell, S.M.; Moskowitz, C.H.; et al. Brentuximab vedotin in combination with nivolumab in relapsed or refractory Hodgkin lymphoma: 3-year study results. Blood 2021, 138, 427–438.
  47. Herrera, A.F.; Chen, R.W.; Palmer, J.; Tsai, N.-C.; Mei, M.; Popplewell, L.L.; Nademanee, A.P.; Nikolaenko, L.; McBride, K.; Ortega, R.; et al. PET-Adapted Nivolumab or Nivolumab Plus ICE as First Salvage Therapy in Relapsed or Refractory Hodgkin Lymphoma. Blood 2019, 134, 239.
  48. Mei, M.G.; Lee, H.J.; Palmer, J.M.; Chen, R.; Tsai, N.-C.; Chen, L.; McBride, K.; Smith, D.L.; Melgar, I.; Song, J.Y.; et al. Response-adapted anti-PD-1–based salvage therapy for Hodgkin lymphoma with nivolumab alone or in combination with ICE. Blood 2022, 139, 3605–3616.
  49. Bryan, L.J.; Casulo, C.; Allen, P.; Smith, S.E.; Savas, H.; Karmali, R.; Winter, J.N. Pembrolizumab (PEM) Added to ICE Chemotherapy Results in High Complete Metabolic Response Rates in Relapsed/Refractory Classic Hodgkin Lymphoma (cHL): A Multi-Institutional Phase II Trial. Blood 2021, 138, 229.
  50. Moskowitz, A.J.; Shah, G.; Schöder, H.; Ganesan, N.; Drill, E.; Hancock, H.; Davey, T.; Perez, L.; Ryu, S.; Sohail, S.; et al. Phase II Trial of Pembrolizumab Plus Gemcitabine, Vinorelbine, and Liposomal Doxorubicin as Second-Line Therapy for Relapsed or Refractory Classical Hodgkin Lymphoma. J. Clin. Oncol. 2021, 39, 3109–3117.
  51. Geoerger, B.; Zwaan, C.M.; Marshall, L.V.; Michon, J.; Bourdeaut, F.; Casanova, M.; Corradini, N.; Rossato, G.; Farid-Kapadia, M.; Shemesh, C.S.; 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. Lancet Oncol. 2020, 21, 134–144.
  52. Crump, M. Management of Hodgkin Lymphoma in Relapse after Autologous Stem Cell Transplant. Hematology 2008, 2008, 326–333.
  53. Moskowitz, C.H.; Nademanee, A.; Masszi, T.; Agura, E.; Holowiecki, J.; Abidi, M.H.; Chen, A.I.; Stiff, P.; Gianni, A.M.; Carella, A.; et al. Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015, 385, 1853–1862.
  54. Schmitz, N.; Pfistner, B.; Sextro, M.; Sieber, M.; Carella, A.M.; Haenel, M.; Boissevain, F.; Zschaber, R.; Müller, P.; Kirchner, H.; et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: A randomised trial. Lancet 2002, 359, 2065–2071.
  55. Lepik, K.V.; Mikhailova, N.B.; Kondakova, E.V.; Zalyalov, Y.R.; Fedorova, L.V.; Tsvetkova, L.A.; Kotselyabina, P.V.; Borzenkova, E.S.; Babenko, E.V.; Popova, M.O.; et al. A Study of Safety and Efficacy of Nivolumab and Bendamustine (NB) in Patients With Relapsed/Refractory Hodgkin Lymphoma after Nivolumab Monotherapy Failure. HemaSphere 2020, 4, e401.
  56. Armand, P.; Chen, Y.-B.; Redd, R.A.; Joyce, R.M.; Bsat, J.; Jeter, E.; Merryman, R.W.; Coleman, K.C.; Dahi, P.B.; Nieto, Y.; et al. PD-1 blockade with pembrolizumab for classical Hodgkin lymphoma after autologous stem cell transplantation. Blood 2019, 134, 22–29.
  57. Ansell, S.; Gutierrez, M.E.; Shipp, M.A.; Gladstone, D.; Moskowitz, A.; Borello, I.; Popa-Mckiver, M.; Farsaci, B.; Zhu, L.; Lesokhin, A.M.; et al. A Phase 1 Study of Nivolumab in Combination with Ipilimumab for Relapsed or Refractory Hematologic Malignancies (CheckMate 039). Blood 2016, 128, 183.
  58. Armand, P.; Lesokhin, A.; Borrello, I.; Timmerman, J.; Gutierrez, M.; Zhu, L.; Popa McKiver, M.; Ansell, S.M. A phase 1b study of dual PD-1 and CTLA-4 or KIR blockade in patients with relapsed/refractory lymphoid malignancies. Leukemia 2021, 35, 777–786.
  59. Diefenbach, C.S.; Hong, F.; Ambinder, R.F.; Cohen, J.B.; Robertson, M.J.; David, K.A.; Advani, R.H.; Fenske, T.S.; Barta, S.K.; Palmisiano, N.D.; 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 Haematol. 2020, 7, e660–e670.
  60. Sermer, D.J.; Vardhana, S.A.; Ames, A.; Biggar, E.; Moskowitz, A.J.; Batlevi, C.L.; Caron, P.; Hamilton, A.M.; Moskowitz, C.H.; Matasar, M.J.; et al. Early data from a phase II trial investigating the combination of pembrolizumab (PEM) and entinostat (ENT) in relapsed and refractory (R/R) Hodgkin lymphoma (HL). J. Clin. Oncol. 2020, 38, e20018.
  61. Sermer, D.J.; Vardhana, S.; Biggar, E.; Moskowitz, A.J.; Joffe, E.; Khan, N.; Straus, D.J.; Kumar, A.; Zelenetz, A.D.; Horwitz, S.M.; et al. Interim Efficacy Analysis of a Phase II Study Demonstrates Promising Activity of the Combination of Pembrolizumab (PEM) and Entinostat (ENT) in Relapsed and Refractory (R/R) Hodgkin Lymphoma (HL). Blood 2021, 138, 2447.
  62. Ansell, S.M.; Bartlett, N.L.; Chen, R.W.; Herrera, A.; Domingo-Domenech, E.; Mehta, A.; Forero-Torres, A.; Garcia-Sanz, R.; Armand, P.; Devata, S.; et al. Investigating safety and preliminary efficacy of AFM13 plus pembrolizumab in patients with relapsed/refractory Hodgkin lymphoma after brentuximab vedotin failure. Hematol. Oncol. 2019, 37, 177–178.
  63. Bartlett, N.L.; Herrera, A.F.; Domingo-Domenech, E.; Mehta, A.; Forero-Torres, A.; Garcia-Sanz, R.; Armand, P.; Devata, S.; Izquierdo, A.R.; Lossos, I.S.; et al. A phase 1b study of AFM13 in combination with pembrolizumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2020, 136, 2401–2409.
  64. Sang, W.; Wang, X.; Geng, H.; Li, T.; Li, D.; Zhang, B.; Zhou, Y.; Song, X.; Sun, C.; Yan, D.; et al. Anti-PD-1 Therapy Enhances the Efficacy of CD30-Directed Chimeric Antigen Receptor T Cell Therapy in Patients with Relapsed/Refractory CD30+ Lymphoma. Front. Immunol. 2022, 13, 858021.
  65. Wang, H.; Kaur, G.; Sankin, A.I.; Chen, F.; Guan, F.; Zang, X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J. Hematol. Oncol. J. Hematol. Oncol. 2019, 12, 59.
  66. Li, A.M.; Hucks, G.E.; Dinofia, A.M.; Seif, A.E.; Teachey, D.T.; Baniewicz, D.; Callahan, C.; Fasano, C.; McBride, B.; Gonzalez, V.; et al. Checkpoint Inhibitors Augment CD19-Directed Chimeric Antigen Receptor (CAR) T Cell Therapy in Relapsed B-Cell Acute Lymphoblastic Leukemia. Blood 2018, 132, 556.
  67. Timmerman, J.; Lavie, D.; Johnson, N.A.; Avigdor, A.; Borchmann, P.; Andreadis, C.; Bazargan, A.; Gregory, G.; Keane, C.; Inna, T.; et al. Favezelimab (anti–LAG-3) plus pembrolizumab in patients with relapsed or refractory (R/R) classical Hodgkin lymphoma (cHL) after anti–PD-1 treatment: An open-label phase 1/2 study. J. Clin. Oncol. 2022, 40, 7545.
  68. Lesokhin, A.M.; Ansell, S.M.; Armand, P.; Scott, E.C.; Halwani, A.; Gutierrez, M.; Millenson, M.M.; Cohen, A.D.; Schuster, S.J.; Lebovic, D.; et al. Nivolumab in Patients with Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016, 34, 2698–2704.
  69. Ansell, S.M.; Minnema, M.C.; Johnson, P.; Timmerman, J.M.; Armand, P.; Shipp, M.A.; Rodig, S.J.; Ligon, A.H.; Roemer, M.G.M.; Reddy, N.; 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. J. Clin. Oncol. 2019, 37, 481–489.
  70. Davids, M.S.; Kim, H.T.; Costello, C.; Herrera, A.F.; Locke, F.L.; Maegawa, R.O.; Savell, A.; Mazzeo, M.; Anderson, A.; Boardman, A.P.; et al. A multicenter phase 1 study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation. Blood 2020, 135, 2182–2191.
  71. Frigault, M.J.; Armand, P.; Redd, R.A.; Jeter, E.; Merryman, R.W.; Coleman, K.C.; Herrera, A.F.; Dahi, P.; Nieto, Y.; LaCasce, A.S.; et al. PD-1 blockade for diffuse large B-cell lymphoma after autologous stem cell transplantation. Blood Adv. 2020, 4, 122–126.
  72. Zinzani, P.L.L.; Thieblemont, C.; Melnichenko, V.; Bouabdallah, K.; Waleswski, J.; Majlis, A.; Fogliatto, L.; Martin Garcia-Sancho, A.; Christian, B.; Gulbas, Z.; et al. Final Analysis of Keynote-170: Pembrolizumab in Relapsed or Refractory Primary Mediastinal Large B-Cell Lymphoma (PMBCL). Blood 2021, 138, 306.
  73. Ding, W.; LaPlant, B.R.; Call, T.G.; Parikh, S.A.; Leis, J.F.; He, R.; Shanafelt, T.D.; Sinha, S.; Le-Rademacher, J.; Feldman, A.L.; et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017, 129, 3419–3427.
  74. Khodadoust, M.S.; Rook, A.H.; Porcu, P.; Foss, F.; Moskowitz, A.J.; Shustov, A.; Shanbhag, S.; Sokol, L.; Fling, S.P.; Ramchurren, N.; et al. Pembrolizumab in Relapsed and Refractory Mycosis Fungoides and Sézary Syndrome: A Multicenter Phase II Study. J. Clin. Oncol. 2020, 38, 20–28.
  75. Kwong, Y.-L.; Chan, T.S.Y.; Tan, D.; Kim, S.J.; Poon, L.-M.; Mow, B.; Khong, P.-L.; Loong, F.; Au-Yeung, R.; Iqbal, J.; et al. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing l-asparaginase. Blood 2017, 129, 2437–2442.
  76. Kim, S.J.; Lim, J.Q.; Laurensia, Y.; Cho, J.; Yoon, S.E.; Lee, J.Y.; Ryu, K.J.; Ko, Y.H.; Koh, Y.; Cho, D.; et al. Avelumab for the treatment of relapsed or refractory extranodal NK/T-cell lymphoma: An open-label phase 2 study. Blood 2020, 136, 2754–2763.
  77. Tao, R.; Fan, L.; Song, Y.; Hu, Y.; Zhang, W.; Wang, Y.; Xu, W.; Li, J. Sintilimab for relapsed/refractory extranodal NK/T cell lymphoma: A multicenter, single-arm, phase 2 trial (ORIENT-4). Signal Transduct. Target. Ther. 2021, 6, 365.
  78. Armand, P.; Janssens, A.; Gritti, G.; Radford, J.; Timmerman, J.; Pinto, A.; Mercadal Vilchez, S.; Johnson, P.; Cunningham, D.; Leonard, J.P.; et al. Efficacy and safety results from CheckMate 140, a phase 2 study of nivolumab for relapsed/refractory follicular lymphoma. Blood 2021, 137, 637–645.
  79. Herbaux, C.; Ghesquieres, H.; Bouabdallah, R.; Guidez, S.; Gyan, E.; Gressin, R.; Morineau, N.; Ysebaert, L.; Le Gouill, S.; Laurent, C.; et al. Atezolizumab + obinutuzumab + venetoclax in patients with relapsed or refractory indolent non-Hodgkin’s lymphoma (R/R iNHL): Primary analysis of a phase 2 trial from LYSA. J. Clin. Oncol. 2021, 39, 7544.
  80. Panayiotidis, P.; Tumyan, G.; Thieblemont, C.; Ptushkin, V.V.; Marin-Niebla, A.; García-Sanz, R.; Le Gouill, S.; Stathis, A.; Bottos, A.; Hamidi, H.; et al. A phase-II study of atezolizumab in combination with obinutuzumab or rituximab for relapsed or refractory mantle cell or marginal zone lymphoma or Waldenström’s macroglobulinemia. Leuk. Lymphoma 2022, 63, 1058–1069.
  81. Younes, A.; Burke, J.M.; Cheson, B.; Diefenbach, C.; Ferrari, S.; Hahn, U.; Hawkes, E.; Khan, C.; Lossos, I.S.; Musuraka, G.; 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, 2969.
  82. Younes, A.; Burke, J.M.; Cheson, B.D.; Diefenbach, C.; Ferrari, S.; Hahn, U.H.; Hawkes, E.A.; Khan, C.; Lossos, I.S.; Musuraca, G.; 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, 2874.
  83. Smith, S.D.; Till, B.G.; Shadman, M.S.; Lynch, R.C.; Cowan, A.J.; Wu, Q.V.; Voutsinas, J.; Rasmussen, H.A.; Blue, K.; Ujjani, C.S.; et al. Pembrolizumab with R-CHOP in previously untreated diffuse large B-cell lymphoma: Potential for biomarker driven therapy. Br. J. Haematol. 2020, 189, 1119–1126.
  84. Nowakowski, G.S.; Willenbacher, W.; Greil, R.; Larsen, T.S.; Patel, K.; Jäger, U.; Manges, R.F.; Trümper, L.; Everaus, H.; Kalakonda, N.; et al. Safety and efficacy of durvalumab with R-CHOP or R2-CHOP in untreated, high-risk DLBCL: A phase 2, open-label trial. Int. J. Hematol. 2022, 115, 222–232.
  85. Younes, A.; Brody, J.; Carpio, C.; Lopez-Guillermo, A.; Ben-Yehuda, D.; Ferhanoglu, B.; Nagler, A.; Ozcan, M.; Avivi, I.; Bosch, F.; et al. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: A phase 1/2a study. Lancet Haematol. 2019, 6, e67–e78.
  86. Herrera, A.F.; Goy, A.; Mehta, A.; Ramchandren, R.; Pagel, J.M.; Svoboda, J.; Guan, S.; Hill, J.S.; Kwei, K.; Liu, E.A.; 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. Am. J. Hematol. 2020, 95, 18–27.
  87. Witzig, T.E.; Maddocks, K.J.; De Vos, S.; Lyons, R.M.; Edenfield, W.J.; Sharman, J.P.; Vose, J.; Yimer, H.A.; Wei, H.; Chan, E.M.; et al. Phase 1/2 trial of acalabrutinib plus pembrolizumab (Pem) in relapsed/refractory (r/r) diffuse large B-cell lymphoma (DLBCL). J. Clin. Oncol. 2019, 37, 7519.
  88. Gregory, G.P.; Kumar, S.; Wang, D.; Mahadevan, D.; Walker, P.; Wagner-Johnston, N.; Escobar, C.; Bannerji, R.; Bhutani, D.; Chang, J.; et al. Pembrolizumab plus dinaciclib in patients with hematologic malignancies: The phase 1b KEYNOTE-155 study. Blood Adv. 2022, 6, 1232–1242.
  89. Casulo, C.; Santoro, A.; Cartron, G.; Ando, K.; Munoz, J.; Le Gouill, S.; Izutsu, K.; Rule, S.; Lugtenburg, P.; Ruan, J.; et al. Durvalumab as monotherapy and in combination therapy in patients with lymphoma or chronic lymphocytic leukemia: The FUSION NHL 001 trial. Cancer Rep. 2023, 6, e1662.
  90. Barraclough, A.; Chong, G.; Gilbertson, M.; Grigg, A.; Churilov, L.; Fancourt, T.; Ritchie, D.; Koldej, R.; Agarwal, R.; Manos, K.; et al. Immune Priming with Single-Agent Nivolumab Followed by Combined Nivolumab & Rituximab Is Safe and Efficacious for First-Line Treatment of Follicular Lymphoma; Interim Analysis of the “1st FLOR” Study. Blood 2019, 134, 1523.
  91. Hawkes, E.A.; Lee, S.T.; Chong, G.; Gilbertson, M.; Grigg, A.; Churilov, L.; Fancourt, T.; Keane, C.; Ritchie, D.; Koldej, R.; et al. Immune priming with nivolumab followed by nivolumab and rituximab in first-line treatment of follicular lymphoma: The phase 21st FLOR study. J. Clin. Oncol. 2021, 39, 7560.
  92. Younes, A.; John, B.M.; Diefenbach, C.S.; Ferrari, S.; Kahn, C.; Sharman, J.P.; Tani, M.; Ujjani, C.S.; Vitolo, U.; Yuen, S.; et al. Safety and Efficacy of Atezolizumab in Combination with Obinutuzumab and Bendamustine in Patients with Previously Untreated Follicular Lymphoma: An Interim Analysis. Blood 2017, 130 (Suppl. S1), 481.
  93. Younes, A.; Burke, J.M.; Diefenbach, C.S.; Ferrari, S.; Khan, C.; Sharman, J.P.; Tani, M.; Ujjani, C.; Vitolo, U.; Yuen, S.L.S.; et al. Safety and efficacy of atezolizumab with obinutuzumab and bendamustine in previously untreated follicular lymphoma. Blood Adv. 2022, 6, 5659–5667.
  94. Westin, J.R.; Chu, F.; Zhang, M.; Fayad, L.E.; Kwak, L.W.; Fowler, N.; Romaguera, J.; Hagemeister, F.; Fanale, M.; Samaniego, F.; et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: A single group, open-label, phase 2 trial. Lancet Oncol. 2014, 15, 69–77.
  95. Nastoupil, L.J.; Westin, J.R.; Fowler, N.H.; Fanale, M.A.; Samaniego, F.; Oki, Y.; Obi, C.; Cao, J.; Cheng, X.; Ma, M.C.J.; et al. Response rates with pembrolizumab in combination with rituximab in patients with relapsed follicular lymphoma: Interim results of an on open-label, phase II study. J. Clin. Oncol. 2017, 35, 7519.
  96. Nastoupil, L.J.; Chin, C.K.; Westin, J.R.; Fowler, N.H.; Samaniego, F.; Cheng, X.; Ma, M.C.J.; Wang, Z.; Chu, F.; Dsouza, L.; et al. Safety and activity of pembrolizumab in combination with rituximab in relapsed or refractory follicular lymphoma. Blood Adv. 2022, 6, 1143–1151.
  97. Palomba, M.L.; Till, B.G.; Park, S.I.; Morschhauser, F.; Cartron, G.; Marks, R.; Shivhare, M.; Hong, W.-J.; Raval, A.; Chang, A.C.; et al. Combination of Atezolizumab and Obinutuzumab in Patients with Relapsed/Refractory Follicular Lymphoma and Diffuse Large B-Cell Lymphoma: Results from a Phase 1b Study. Clin. Lymphoma Myeloma Leuk. 2022, 22, e443–e451.
  98. Morschhauser, F.; Ghosh, N.; Lossos, I.S.; Palomba, M.L.; Mehta, A.; Casasnovas, O.; Stevens, D.; Katakam, S.; Knapp, A.; Nielsen, T.; et al. Obinutuzumab-atezolizumab-lenalidomide for the treatment of patients with relapsed/refractory follicular lymphoma: Final analysis of a Phase Ib/II trial. Blood Cancer J. 2021, 11, 147.
  99. Jain, N.; Basu, S.; Thompson, P.A.; Ohanian, M.; Ferrajoli, A.; Pemmaraju, N.; Cortes, J.E.; Estrov, Z.; Burger, J.A.; Neelapu, S.S.; et al. Nivolumab Combined with Ibrutinib for CLL and Richter Transformation: A Phase II Trial. Blood 2016, 128, 59.
  100. Jain, N.; Ferrajoli, A.; Basu, S.; Thompson, P.A.; Burger, J.A.; Kadia, T.M.; Estrov, Z.E.; Pemmaraju, N.; Lopez, W.; Thakral, B.; et al. A Phase II Trial of Nivolumab Combined with Ibrutinib for Patients with Richter Transformation. Blood 2018, 132, 296.
  101. Jeong, A.-R.; Ball, E.D.; Goodman, A.M. Predicting Responses to Checkpoint Inhibitors in Lymphoma: Are We Up to the Standards of Solid Tumors? Clin. Med. Insights Oncol. 2020, 14, 1–13.
  102. Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201.
  103. Cha, J.-H.; Chan, L.-C.; Li, C.-W.; Hsu, J.L.; Hung, M.-C. Mechanisms Controlling PD-L1 Expression in Cancer. Mol. Cell 2019, 76, 359–370.
  104. Green, M.R.; Monti, S.; Rodig, S.J.; Juszczynski, P.; Currie, T.; O’Donnell, E.; Chapuy, B.; Takeyama, K.; Neuberg, D.; Golub, T.R.; 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, 3268–3277.
  105. Mottok, A.; Hung, S.S.; Chavez, E.A.; Woolcock, B.; Telenius, A.; Chong, L.C.; Meissner, B.; Nakamura, H.; Rushton, C.; Viganò, E.; et al. Integrative genomic analysis identifies key pathogenic mechanisms in primary mediastinal large B-cell lymphoma. Blood 2019, 134, 802–813.
  106. Shi, M.; Roemer, M.G.M.; Chapuy, B.; Liao, X.; Sun, H.; Pinkus, G.S.; Shipp, M.A.; Freeman, G.J.; Rodig, S.J. Expression of Programmed Cell Death 1 Ligand 2 (PD-L2) Is a Distinguishing Feature of Primary Mediastinal (Thymic) Large B-cell Lymphoma and Associated with PDCD1LG2 Copy Gain. Am. J. Surg. Pathol. 2014, 38, 1715–1723.
  107. Chapuy, B.; Roemer, M.G.M.; Stewart, C.; Tan, Y.; Abo, R.P.; Zhang, L.; Dunford, A.J.; Meredith, D.M.; Thorner, A.R.; Jordanova, E.S.; et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood 2016, 127, 869–881.
  108. Godfrey, J.; Tumuluru, S.; Bao, R.; Leukam, M.; Venkataraman, G.; Phillip, J.; Fitzpatrick, C.; McElherne, J.; MacNabb, B.W.; Orlowski, R.; et al. PD-L1 gene alterations identify a subset of diffuse large B-cell lymphoma harboring a T-cell–inflamed phenotype. Blood 2019, 133, 2279–2290.
  109. Wang, Y.; Wenzl, K.; Manske, M.K.; Asmann, Y.W.; Sarangi, V.; Greipp, P.T.; Krull, J.E.; Hartert, K.; He, R.; Feldman, A.L.; et al. Amplification of 9p24.1 in diffuse large B-cell lymphoma identifies a unique subset of cases that resemble primary mediastinal large B-cell lymphoma. Blood Cancer J. 2019, 9, 73.
  110. Igarashi, T.; Teramoto, K.; Ishida, M.; Hanaoka, J.; Daigo, Y. Scoring of PD-L1 expression intensity on pulmonary adenocarcinomas and the correlations with clinicopathological factors. ESMO Open 2016, 1, e000083.
  111. Roemer, M.G.M.; Advani, R.H.; Ligon, A.H.; Natkunam, Y.; Redd, R.A.; Homer, H.; Connelly, C.F.; Sun, H.H.; Daadi, S.E.; Freeman, G.J.; et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J. Clin. Oncol. 2016, 34, 2690–2697.
  112. Liu, J.; Hamrouni, A.; Wolowiec, D.; Coiteux, V.; Kuliczkowski, K.; Hetuin, D.; Saudemont, A.; Quesnel, B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-γ and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007, 110, 296–304.
  113. Luo, Y.; Liu, Y.; Wang, C.; Gan, R. Signaling pathways of EBV-induced oncogenesis. Cancer Cell Int. 2021, 21, 93.
  114. Kim, S.-J.; Hyeon, J.; Cho, I.; Ko, Y.H.; Kim, W.S. Comparison of Efficacy of Pembrolizumab between Epstein-Barr Virus-Positive and -Negative Relapsed or Refractory Non-Hodgkin Lymphomas. Cancer Res. Treat. 2019, 51, 611–622.
  115. Biggi, A.F.B.; Elgui de Oliveira, D. The Epstein-Barr Virus Hacks Immune Checkpoints: Evidence and Consequences for Lymphoproliferative Disorders and Cancers. Biomolecules 2022, 12, 397.
  116. Satou, A.; Nakamura, S. EBV-positive B-cell lymphomas and lymphoproliferative disorders: Review from the perspective of immune escape and immunodeficiency. Cancer Med. 2021, 10, 6777–6785.
  117. Ligeti, K.; Müller, L.P.; Müller-Tidow, C.; Weber, T. Risk factors, diagnosis, and management of posttransplant lymphoproliferative disorder: Improving patient outcomes with a multidisciplinary treatment approach. Transpl. Res. Risk Manag. 2017, 9, 1–14.
  118. Steiner, R.E.; Kridel, R.; Giostra, E.; McKee, T.A.; Achermann, R.; Mueller, N.J.; Dietrich, P.Y. Low 5-year cumulative incidence of post-transplant lymphoproliferative disorders after solid organ transplantation in Switzerland. Swiss Med. Wkly. 2018, 148, w14596.
  119. Tse, E.; Zhao, W.-L.; Xiong, J.; Kwong, Y.-L. How we treat NK/T-cell lymphomas. J. Hematol. Oncol. J. Hematol. Oncol. 2022, 15, 74.
  120. Kiyasu, J.; Miyoshi, H.; Hirata, A.; Arakawa, F.; Ichikawa, A.; Niino, D.; Sugita, Y.; Yufu, Y.; Choi, I.; Abe, Y.; 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, 2193–2201.
  121. Strickler, J.H.; Hanks, B.A.; Khasraw, M. Tumor Mutational Burden as a Predictor of Immunotherapy Response: Is More Always Better? Clin. Cancer Res. 2021, 27, 1236–1241.
  122. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2014, 371, 2189–2199.
  123. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015, 348, 124–128.
  124. Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R.; et al. Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. Oncologist 2020, 25, e147–e159.
  125. Galanina, N.; Bejar, R.; Choi, M.; Goodman, A.; Wieduwilt, M.; Mulroney, C.; Kim, L.; Yeerna, H.; Tamayo, P.; Vergilio, J.-A.; et al. Comprehensive Genomic Profiling Reveals Diverse but Actionable Molecular Portfolios across Hematologic Malignancies: Implications for Next Generation Clinical Trials. Cancers 2018, 11, 11.
  126. Ricciuti, B.; Wang, X.; Alessi, J.V.; Rizvi, H.; Mahadevan, N.R.; Li, Y.Y.; Polio, A.; Lindsay, J.; Umeton, R.; Sinha, R.; et al. Association of High Tumor Mutation Burden in Non–Small Cell Lung Cancers with Increased Immune Infiltration and Improved Clinical Outcomes of PD-L1 Blockade Across PD-L1 Expression Levels. JAMA Oncol. 2022, 8, 1160–1168.
  127. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34.
  128. Wienand, K.; Chapuy, B.; Stewart, C.; Dunford, A.J.; Wu, D.; Kim, J.; Kamburov, A.; Wood, T.R.; Cader, F.Z.; Ducar, M.D.; et al. Genomic analyses of flow-sorted Hodgkin Reed-Sternberg cells reveal complementary mechanisms of immune evasion. Blood Adv. 2019, 3, 4065–4080.
  129. Chapuy, B.; Stewart, C.; Dunford, A.J.; Kim, J.; Wienand, K.; Kamburov, A.; Griffin, G.K.; Chen, P.-H.; Lako, A.; Redd, R.A.; et al. Genomic analyses of PMBL reveal new drivers and mechanisms of sensitivity to PD-1 blockade. Blood 2019, 134, 2369–2382.
  130. Cho, J.; Yoon, S.E.; Kim, S.J.; Ko, Y.H.; Kim, W.S. Comparison of tumor mutation burden of 300 various non-Hodgkin lymphomas using panel based massively parallel sequencing. BMC Cancer 2021, 21, 972.
  131. Russano, M.; Napolitano, A.; Ribelli, G.; Iuliani, M.; Simonetti, S.; Citarella, F.; Pantano, F.; Dell’Aquila, E.; Anesi, C.; Silvestris, N.; et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: The potentiality of blood samples. J. Exp. Clin. Cancer Res. 2020, 39, 95.
  132. Zhu, G.; Guo, Y.A.; Ho, D.; Poon, P.; Poh, Z.W.; Wong, P.M.; Gan, A.; Chang, M.M.; Kleftogiannis, D.; Lau, Y.T.; et al. Tissue-specific cell-free DNA degradation quantifies circulating tumor DNA burden. Nat. Commun. 2021, 12, 2229.
  133. Roschewski, M.; Dunleavy, K.; Pittaluga, S.; Moorhead, M.; Pepin, F.; Kong, K.; Shovlin, M.; Jaffe, E.S.; Staudt, L.M.; Lai, C.; et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: A correlative biomarker study. Lancet Oncol. 2015, 16, 541–549.
  134. Shi, Y.; Su, H.; Song, Y.; Jiang, W.; Sun, X.; Qian, W.; Zhang, W.; Gao, Y.; Jin, Z.; Zhou, J.; et al. Circulating tumor DNA predicts response in Chinese patients with relapsed or refractory classical hodgkin lymphoma treated with sintilimab. EBioMedicine 2020, 54, 102731.
  135. Zhou, H.; Du, X.; Zhao, T.; Ouyang, Z.; Liu, W.; Deng, M.; He, Q.; Yi, Y.; Dai, L.; Yang, L.; et al. Distribution and influencing factors of tumor mutational burden in different lymphoma subtypes. J. Clin. Oncol. 2019, 37, e19053.
  136. Marcus, L.; Fashoyin-Aje, L.A.; Donoghue, M.; Yuan, M.; Rodriguez, L.; Gallagher, P.S.; Philip, R.; Ghosh, S.; Theoret, M.R.; Beaver, J.A.; et al. FDA Approval Summary: Pembrolizumab for the Treatment of Tumor Mutational Burden–High Solid Tumors. Clin. Cancer Res. 2021, 27, 4685–4689.
  137. Li, K.; Luo, H.; Huang, L.; Luo, H.; Zhu, X. Microsatellite instability: A review of what the oncologist should know. Cancer Cell Int. 2020, 20, 16.
  138. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413.
  139. Center for Devices and Radiological Health. List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools); FDA: Silver Spring, MD, USA, 2019. Available online: https://www.fda.gov/medical-devices/in-vitro-diagnostics/list-cleared-or-approved-companion-diagnostic-devices-in-vitro-and-imaging-tools (accessed on 6 June 2023).
  140. Tian, T.; Li, J.; Xue, T.; Yu, B.; Li, X.; Zhou, X. Microsatellite instability and its associations with the clinicopathologic characteristics of diffuse large B-cell lymphoma. Cancer Med. 2020, 9, 2330–2342.
  141. Wieczorek, M.; Abualrous, E.T.; Sticht, J.; Álvaro-Benito, M.; Stolzenberg, S.; Noé, F.; Freund, C. Major Histocompatibility Complex (MHC) Class I and MHC Class II Proteins: Conformational Plasticity in Antigen Presentation. Front. Immunol. 2017, 8, 292.
  142. Hazini, A.; Fisher, K.; Seymour, L. Deregulation of HLA-I in cancer and its central importance for immunotherapy. J. Immunother. Cancer 2021, 9, e002899.
  143. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 2018, 10, eaar3342.
  144. Roemer, M.G.M.; Advani, R.H.; Redd, R.A.; Pinkus, G.S.; Natkunam, Y.; Ligon, A.H.; Connelly, C.F.; Pak, C.J.; Carey, C.D.; Daadi, S.E.; et al. Classical Hodgkin Lymphoma with Reduced β2 M/MHC Class I Expression Is Associated with Inferior Outcome Independent of 9p24.1 Status. Cancer Immunol. Res. 2016, 4, 910–916.
  145. Weber, J.S.; Gibney, G.; Sullivan, R.J.; Sosman, J.A.; Slingluff, C.L.; Lawrence, D.P.; Logan, T.F.; Schuchter, L.M.; Nair, S.; Fecher, L.; et al. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): An open-label, randomised, phase 2 trial. Lancet Oncol. 2016, 17, 943–955.
  146. Autio, M.; Leivonen, S.-K.; Brück, O.; Mustjoki, S.; Jørgensen, J.M.; Karjalainen-Lindsberg, M.-L.; Beiske, K.; Holte, H.; Pellinen, T.; Leppä, S. Immune cell constitution in the tumor microenvironment predicts the outcome in diffuse large B-cell lymphoma. Haematologica 2020, 106, 718–729.
  147. Leivonen, S.-K.; Pollari, M.; Brück, O.; Pellinen, T.; Autio, M.; Karjalainen-Lindsberg, M.-L.; Mannisto, S.; Kellokumpu-Lehtinen, P.-L.; Kallioniemi, O.; Mustjoki, S.; et al. T-cell inflamed tumor microenvironment predicts favorable prognosis in primary testicular lymphoma. Haematologica 2019, 104, 338–346.
  148. Wu, H.; Tang, X.; Kim, H.J.; Jalali, S.; Pritchett, J.C.; Villasboas, J.C.; Novak, A.J.; Yang, Z.-Z.; Ansell, S.M. Expression of KLRG1 and CD127 defines distinct CD8+ subsets that differentially impact patient outcome in follicular lymphoma. J. Immunother. Cancer 2021, 9, e002662.
  149. Nygren, L.; Wasik, A.M.; Baumgartner-Wennerholm, S.; Jeppsson-Ahlberg, Å.; Klimkowska, M.; Andersson, P.; Buhrkuhl, D.; Christensson, B.; Kimby, E.; Wahlin, B.E.; et al. T-Cell Levels Are Prognostic in Mantle Cell Lymphoma. Clin. Cancer Res. 2014, 20, 6096–6104.
  150. Alonso-Álvarez, S.; Vidriales, M.B.; Caballero, M.D.; Blanco, O.; Puig, N.; Martin, A.; Peñarrubia, M.J.; Zato, E.; Galende, J.; Bárez, A.; et al. The number of tumor infiltrating T-cell subsets in lymph nodes from patients with Hodgkin lymphoma is associated with the outcome after first line ABVD therapy. Leuk. Lymphoma 2017, 58, 1144–1152.
  151. Maimela, N.R.; Liu, S.; Zhang, Y. Fates of CD8+ T cells in Tumor Microenvironment. Comput. Struct. Biotechnol. J. 2019, 17, 1–13.
  152. Li, F.; Li, C.; Cai, X.; Xie, Z.; Zhou, L.; Cheng, B.; Zhong, R.; Xiong, S.; Li, J.; Chen, Z.; et al. The association between CD8+ tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: A systematic review and meta-analysis. eClinicalMedicine 2021, 41, 101134.
  153. Hammond, S.M. An overview of microRNAs. Adv. Drug Deliv. Rev. 2015, 87, 3–14.
  154. Fuertes, T.; Ramiro, A.R.; de Yebenes, V.G. miRNA-Based Therapies in B Cell Non-Hodgkin Lymphoma. Trends Immunol. 2020, 41, 932–947.
  155. Volinia, S.; Calin, G.A.; Liu, C.-G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261.
  156. Ji, Q.; Jiang, T.; Su, J.; Zhang, S.; Li, C.; Yang, X.; Wu, X.; Yao, J.; Yuan, D.; Wang, J. Serum miR-21 predicts the prognosis of patients with primary gastrointestinal diffuse large B-cell lymphoma. Acta Biochim. Pol. 2022, 69, 379–385.
  157. Li, J.; Fu, R.; Yang, L.; Tu, W. miR-21 expression predicts prognosis in diffuse large B-cell lymphoma. Int. J. Clin. Exp. Pathol. 2015, 8, 15019–15024.
  158. Li, J.; Zhai, X.-W.; Wang, H.-S.; Qian, X.-W.; Miao, H.; Zhu, X.-H. Circulating MicroRNA-21, MicroRNA-23a, and MicroRNA-125b as Biomarkers for Diagnosis and Prognosis of Burkitt Lymphoma in Children. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2016, 22, 4992–5002.
  159. Mao, X.; Sun, Y.; Tang, J. Serum miR-21 is a diagnostic and prognostic marker of primary central nervous system lymphoma. Neurol. Sci. 2014, 35, 233–238.
  160. Xi, J.; Huang, Q.; Wang, L.; Ma, X.; Deng, Q.; Kumar, M.; Zhou, Z.; Li, L.; Zeng, Z.; Young, K.H.; et al. miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy. Oncogene 2018, 37, 3151–3165.
  161. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103.
  162. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108.
  163. Liu, X.; Chen, Y.; Zhang, S.; Dong, L. Gut microbiota-mediated immunomodulation in tumor. J. Exp. Clin. Cancer Res. 2021, 40, 221.
  164. Hwang, S.R.; Higgins, A.; Castillo Almeida, N.E.; LaPlant, B.; Maurer, M.J.; Ansell, S.M.; Witzig, T.E.; Thanarajasingam, G.; Bennani, N.N. Effect of antibiotic use on outcomes in patients with Hodgkin lymphoma treated with immune checkpoint inhibitors. Leuk. Lymphoma 2021, 62, 247–251.
  165. Casadei, B.; Guadagnuolo, S.; Barone, M.; Turroni, S.; Argnani, L.; Brigidi, P.; Zinzani, P.L. Gut Microbiota Role in Response to Checkpoint Inhibitor Treatment in Patients with Relapsed/Refractory B-Cell Hodgkin Lymphoma: The MICRO-Linf Study. Blood 2021, 138, 2957.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 297
Revisions: 3 times (View History)
Update Date: 21 Jul 2023
1000/1000
Video Production Service