Next Article in Journal
Deep Sequencing of Early T Stage Colorectal Cancers Reveals Disruption of Homologous Recombination Repair in Microsatellite Stable Tumours with High Mutational Burdens
Next Article in Special Issue
Novel Salvage Therapy Options for Initial Treatment of Relapsed/Refractory Classical Hodgkin’s Lymphoma: So Many Options, How to Choose?
Previous Article in Journal
A Cohort Study of Free Light Chain Ratio in Combination with Serum Protein Electrophoresis as a First-Line Test in General Practice
Previous Article in Special Issue
An Update on the Pathology and Molecular Features of Hodgkin Lymphoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Updates in the Role of Checkpoint Inhibitor Immunotherapy in Classical Hodgkin’s Lymphoma

1
Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
2
Department of Internal Medicine, Temple University Hospital, Philadelphia, PA 19140, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(12), 2936; https://doi.org/10.3390/cancers14122936
Submission received: 1 May 2022 / Revised: 23 May 2022 / Accepted: 30 May 2022 / Published: 14 June 2022
(This article belongs to the Special Issue Therapy of Hodgkin Lymphoma)

Abstract

:

Simple Summary

The introduction of immunotherapy into the treatment options for Hodgkin’s lymphoma has improved survival in patients with recurrence of their cancer. These agents help the body’s immune system respond and clear cancer cells. Currently, these agents are only approved in patients who have had their cancer return multiple times or have progressed through multiple therapies. However, the evaluation of these agents as part of therapy in the upfront setting or second line setting has been conducted. This study will review these clinical studies and provide insights into the future directions of immunotherapy use by physicians in treatment of patients with Hodgkin’s lymphoma.

Abstract

Classical Hodgkin’s lymphoma is a highly curable disease, but 10–25% of patients with higher-risk disease relapse. The introduction of checkpoint inhibitors (CPIs) targeting PD-1 have changed the landscape of treatment for patients with relapsed/refractory disease to multiple lines of therapy. The depth of response to CPI as a monotherapy is highest in the first relapse as salvage therapy based on outcomes reported in several phase II studies. With earlier use of CPI and brentuximab vedotin, the optimal sequencing of therapy is evolving. In this review, we will summarize clinical investigation of anti-PD-1 mAb in earlier line settings to provide insights on utilizing these agents as chemotherapy- and radiation-sparing approaches, increasing depth of response, and as part of combination regimens.

1. Introduction

Classical Hodgkin’s lymphoma (cHL) accounts for roughly 11% of all lymphomas in the United States [1] and is defined by its distinct pathologic features consistent with Reed Sternberg cells in a background of inflammatory cells, which enable the evasion of immune-mediated antitumor mechanisms [2,3,4]. Although the majority of patients with cHL can be cured with standard chemoradiation, patients who are unresponsive to initial treatment or who are not candidates for curative regimens have poor outcomes [5,6,7]. Immune checkpoint inhibitors (CPIs) enable the reconstitution of the immune response to target malignant cells, offering an alternative mechanism of action from traditional cytotoxic chemotherapy and have thus transformed the treatment landscape for relapsed/refractory (R/R) cHL.
Approximately 10–25% of patients with advanced or early-stage unfavorable risk disease have disease relapse [8,9]. The standard of care on relapse consists of salvage chemotherapy followed by autologous stem cell transplant (ASCT). This approach cures only about 50% of patients and is associated with significant risk for treatment-related toxicity and secondary malignancy [10,11]. Older patients and those with medical comorbidities who are not candidates for intensive therapy remain at a particularly high risk for relapse and inferior survival [12]. With modern frontline treatment regimens, patients over the age of 60 years had a ~15% worse 3-year progression-free survival (PFS) compared with younger patients [13].
Although the addition of the anti-CD30 monoclonal antibody drug conjugate brentuximab vedotin (BV) to the cHL treatment armamentarium has improved outcomes, the rate of relapse in patients treated with BV-containing frontline therapy is still 18% at 5 year follow-up [14]. In the high-risk R/R population who received BV as maintenance therapy after ASCT, 41% of patients have progression of their disease [15]. In the multiple-relapse setting, these responses are not durable, with only 38% of patients treated with BV maintaining their complete response (CR) at 5 year follow-up [16,17]. Immunotherapy with CPI treatment has demonstrated remarkable efficacy in patients with relapse after multiple lines of therapy and primary refractory disease and in patients who are not candidates for intensive salvage treatment (Table 1). This has led to the investigation of these agents in earlier lines of treatment to improve survival rates while also reducing treatment-related toxicity. Herein, we will review the clinical trial data on anti-PD1 mAb in the upfront and R/R settings over the past 5 years. We will also discuss considerations for sequencing, combination regimens, and the roles of chemotherapy- and/or radiation-sparing approaches.

2. Immune Checkpoints and the Tumor Microenvironment (TME) in cHL

The interaction of the programmed death receptor ligands 1 and 2 (PDL1/CD274 and PDL2/CD273) on tumor cells with PD-1 receptors on T cells leads to reduced T-cell activation and proliferation, thus enabling tumors to evade immune response (see Figure 1) [42]. The amplification of PDL1 and PDL2 on tumor cells, coded on chromosome 9p24.1, is mediated by PIM serine/threonine kinases on tumor cells via the constitutive activation of the NFkB and JAK-STAT signaling pathways [43].
Anti-PD-1 monoclonal antibody (mAb) therapies reconstitute the immune surveillance of malignant cells, enabling a rapid reduction in Reed-Sternberg cells and PDL1+ inflammatory cells as well as a durable PD-1 blockade that persists even after treatment cessation based on patient samples during and after treatment [44]. A rise in CD4+ T cell receptors, dendritic cells, and mature, highly differentiated NK cells is seen in peripheral blood after anti-PD1 mAb treatment, correlating with earlier and more robust clinical response [44,45,46,47].
PDL-1 expression is a well-defined marker of response to anti-PD-1 mAb across multiple cancer subtypes. In addition to this, 9p24.1 copy number amplification and the expression of major histocompatibility complex class II are associated with improved response to CPI in cHL [48]. In the frontline setting, these biomarkers did not demonstrate an association with early response rates with anti-PD-1 mAb-based therapy [49]. Emerging data on the role of metabolic disorders and patient demographic factors such as sex, age, and gender on effector T cell activity and PD-1 expression suggest that these factors may also contribute to CPI efficacy [50]. The further investigation of these tumoral biomarkers and the effects of extra-tumoral risk factors on TME may help guide treatment decision-making in the future.
Lastly, other immune checkpoints have been identified as potential targets in the TME of cHL, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), lymphocyte-activation gene-3 (LAG-3), and T-cell immunoglobulin and mucin-domain containing 3 (TIM-3) [51,52]. The CD68 ligand that binds CTLA-4 is commonly expressed on HRS cells, and this expression persists after treatment with PD-1 blockade. This has therefore served as the rationale for targeting this protein with the monoclonal antibody ipilimumab to overcome anti-PD-1 mAb resistance [51]. Although LAG-3 and TIM-3 are not as frequently expressed on HRS cells, these proteins are commonly seen within the TME surrounding HRS cells [52]. However, ongoing investigation is needed to identify the clinical utility of targeting these alternative immune checkpoints.

3. Checkpoint Inhibitors in Multiple Relapsed/Refractory Disease

For patients with R/R disease after multiple lines of therapy including ASCT +/− BV, anti-PD-1 mAb monotherapy has an overall response rate (ORR) of ~70% with a median PFS of 14–15 months. Although only 20–30% of responders have CR in this setting, those who do achieve CR have a prolonged remission with a median duration of response (mDOR) of 37 months with nivolumab; additionally, mDOR was not reached with pembrolizumab at 5 year follow-up [23,53]. In a study of anti-PD-1 mAb tislelizumab in patients with relapse after ASCT or 2 systemic regimens in a Chinese population, 67% of patients achieved CR, and ~50% of patients had ongoing response at 30 month follow-up, confirming the correlation of depth of response to duration of response with this class of agents [27]. Novel combination regimens of immunotherapy combined with other targeted therapies have deepened response rates in the multiple-relapse setting. The combinations of BV with the checkpoint inhibitors nivolumab and ipilimumab achieved CRs of 61% and 57%, respectively. Although the combination of all three agents achieved the highest CR of 73%, the inclusion of ipilimumab resulted in higher toxicity than BV + Nivo in this phase I/II study. The rates of grade 3–4 adverse events were reported at 43% and 50%, respectively, in the BV-Ipi and triplet combination arms but only 16% in the BV-Nivo group. [31]. Therefore, anti-PD-1 mAb based therapies are preferred.
In early clinical investigation, the addition of hypomethylating agent to anti-PD-1 mAb improved CR from 32 to 71% in patients with R/R cHL to at least 2 prior lines of therapy, with 100% of patients still responding to combination therapy at 6 month follow-up. Investigational regimens evaluating nivolumab in combination with JAK2 inhibitor, and BTK inhibitor are also underway (Table 2). Additionally, retreatment with anti PD-1 mAb has been described in a small number of patients with multiply relapsed disease, demonstrating ORRs of 70–100% and CRs of 33–73% [54,55,56,57]. Phase II investigation of combination BV + Nivo in patients with prior treatment of either agent is underway and may provide more insight into the roles of sequencing and retreatment as these agents are introduced to patients in earlier lines of therapy (Table 2).

4. Immunotherapy as First Salvage and Bridge to ASCT

Treatment with standard chemotherapy options in first relapse followed by ASCT leads to long-term cure in only half of patients, with reported 6 year event-free survival and overall survival (OS) of 45% and 55%, respectively [58]. Standard cytotoxic chemotherapy salvage achieved an ORR of ~70–90% and a CR of ~20–30% [59,60,61,62,63]. In patients who fail to respond to initial salvage chemotherapy, the outcomes after ASCT are particularly dismal. At 10 year follow-up, the PFS and OS are only 7–23% and 11–17%, respectively [60,61] Achieving complete metabolic response (CMR) to salvage therapy prior to ASCT is associated with superior outcomes and has served as a rationale for incorporating targeted therapies into first salvage lines of therapy. While the addition of BV into first-line salvage regimens has improved CR to 70–80% [62,64,65,66], single-agent BV induces CRs of only 27–43% when assessed as monotherapy or as part of PET-adapted bridging therapy ASCT [67,68].
On the other hand, the investigation of anti-PD-1 mAb in the first relapse in several phase II studies has demonstrated increased depth of response both as a single agent and as combination therapy (Table 1) [33,34,35]. Combination BV and Nivo demonstrated ORR of 82–85% and CR of 59–67% [32,69,70] At 3 year follow-up after combination achieved a PFS of 77%, and in patients who responded to induction and proceeded to ASCT, PFS improved to 91% [33]. In a small phase II PET-adapted study evaluating nivolumab monotherapy followed by escalation to nivolumab with ifosfamide, carboplatin, and etoposide (ICE) in patients with positive interim PET scans, 71% of patients achieved CR to the single agent nivolumab and were eligible to proceed directly to ASCT, sparing 26 of the total 42 evaluable patients from requiring cytotoxic treatments prior to ASCT. A total of 9 of the 42 patients required escalation of their treatment to N-ICE, and of these, 89% achieved CR [35]. This approach demonstrated overall 2 year PFS and OS of 72% and 95%, respectively (Table 1) [35]. Larger studies are underway to evaluate nivolumab monotherapy as salvage prior to ASCT (Table 2).
Other small studies evaluating combination of immunotherapy with standard regimens as bridge to ASCT, including pembrolizumab combined with ICE or gemcitabine, vinorelbine, and pegylated liposomal doxorubicin (GVD), showed similarly high CR rates of 86–95% [36]. The active investigation of other anti-PD-1 mAb combination regimens via a PET-adapted approach using anti-PD1 mAb with GVD are underway to assess efficacy with less toxic or chemotherapy-sparing therapies (Table 2).

5. Immunotherapy in Primary Refractory Disease

Historical data in patients with R/R disease within 1 year of frontline therapy who are treated with standard salvage chemotherapy followed by ASCT demonstrated a 10 year freedom from second failure (FF2F) and OS of 34 and 43%, respectively [9]. The incorporation of CPI has significantly improved outcomes in this population. In a subgroup analysis of R/R patients treated with pembrolizumab, patients with primary refractory disease achieved a CR of 35% with mDOR of 17 months at a median of 28 month follow-up, achieving 2 year PFS and OS of 32% and 94%, respectively. In a phase II study of pembrolizumab with chemotherapy (GVD) as initial salvage therapy as a bridge to ASCT, 79% of patients had either primary refractory disease or relapse within 1 year. All patients responded to this combination treatment with a remarkable CR of 95%, and 36 of the 39 patients enrolled proceeded to ASCT. All remained in remission at 13.5 month follow-up [34].

6. Immunotherapy as Maintenance after ASCT

Lastly, CPI has also been explored as consolidation therapy after ASCT similar to BV, as investigated in the phase III ATHERA study [15,37]. In the ATHERA study, high-risk patients, defined as primary refractory or relapse/progression of disease within 12 months of frontline therapy or extra-nodal involvement at relapse, were included. Extended follow-up of BV maintenance demonstrated a 5-year PFS of 59% vs 41% with placebo [37]. Phase II evaluation of pembrolizumab after ASCT demonstrated an estimated 18 month PFS of 82% overall, and among patients in the subgroup who met the criteria for high-risk disease per the ATHERA protocol, PFS was 85% [15,37].
In the absence of long-term follow-up and head-to-head comparison, the PFS with post-ASCT consolidative pembrolizumab compares favorably with that of BV. However, as BV has moved to the upfront setting as the standard of care for advanced-stage cHL and nivolumab is being used in earlier lines of therapy, the role of maintenance BV or anti-PD-1 mAb is less clear. Ongoing phase II investigation of the combination of BV and nivolumab in patients with relapse who previously received either agent may provide insight into the roles of sequencing and retreatment with these agents as consolidation post ASCT (Table 2).

7. Immunotherapy in Frontline Treatment

Frontline chemotherapy with doxorubicin, vinblastine, and dacarbazine with bleomycin (ABVD) or BV (A-AVD) with and without radiation (RT) achieve high cure rates. However, these therapies are associated with significant toxicity such as bleomycin- and/or radiation-associated lung toxicity, anthracycline-associated cardiotoxicity, infections, and secondary malignancy [7,11,71,72] Whereas the reduction or elimination of bleomycin as part of a PET-adapted ABVD regimen or the A-AVD regimen has reduced rates of pulmonary toxicity and the use of G-CSF has decreased the risk of infectious complications [13,73,74] older and medically frail patients remain at high risk for complications [72,75,76]. An analysis of patients treated with ABVD from the German Hodgkin Study Group demonstrated that 68% of older patients suffered from major toxicities, with a treatment-related mortality of 5% [77].
Although anti-PD-1 mAb is associated with unique immune-related adverse events (irAEs), the majority are mild and can be well managed with observation or immunosuppression based on severity. Grade 3–4 irAE with anti-PD-1 mAb is reported at rate of 12–28% in patients with cHL which is similar to rates described in patients with solid tumors [22,40,78] In a meta-analysis of patients with cancer treated with anti-PD-1 mAb, the rate of treatment-related mortality was 0.36% [79]. This has therefore led to investigation of anti-PD-1 mAb in the frontline setting.
Unlike in the second-line salvage setting, single-agent anti-PD-1 mAb and mAb in combination with BV failed to improve response rates in the frontline setting compared with standard therapy [39,41]. However, 37–51% of patients did achieve CR to single-agent PDL-1 mAb when given nivolumab or pembrolizumab initially as part of sequential therapy followed by AVD, identifying a population that may be spared from cytotoxic treatment [39,41]. In the NIVAHL study, patients received N-AVD as concomitant therapy or sequential treatment with nivolumab followed by AVD, both as part of combined modality treatment for early-stage unfavorable disease. Combination N-AVD induced a CR of 87% compared with 51% after the initial course of nivolumab. Although this study failed to meet its efficacy benchmark in the concomitant cohort, the 1 year PFS rates were 100% and 98% in the concomitant and sequential therapy arms, respectively [39].
In a study evaluating pembrolizumab with AVD as a radiation-sparing (RT) approach in early unfavorable or advanced-stage disease, all patients received sequential therapy. Specifically, 37% had CR to initial pembrolizumab monotherapy, and all patients achieved CR after 2 additional cycles of AVD. At the median follow-up of 22 months, all patients had sustained response, and none were treated with RT. Of the 30 total patients, 12 had bulky disease [41].
In a study of upfront BV and nivolumab in older patients and those unsuitable for standard chemotherapy, only 64% of patients responded, and this study was closed early due to failure to meet the primary endpoint [38]. At the median follow-up of 21 months, the mPFS was 22 months, and mDOR was not reached. Although this study did not improve standard of care therapy, it demonstrates high activity as a chemotherapy-sparing approach in those who are not candidates for more intensive therapy. Based on the remarkable efficacy of anti-PD-1 mAb therapy in chemotherapy-refractory disease, treatment with immunotherapy in the frontline as part of a PET-adapted approach in patients with suboptimal response to anthracycline-based treatment is under investigation (Table 1). In early-stage non-bulky disease, PET-adapted treatment with nivolumab-based therapy as an RT-sparing option or in combination with RT is also underway (Table 2).

8. Future Directions

The optimal sequencing of CPI, brentuximab vedotin, cytotoxic therapy, and ASCT in the R/R setting remains unclear as targeted agents are now being used in the upfront and 2nd-line setting. Therefore, the future treatment paradigm of cHL will continue to evolve based on the multiple active clinical trials summarized in Table 2, particularly the comparison of upfront N-AVD vs. A-AVD in advanced-stage disease (NCT03907488). Determining which patients are eligible for retreatment with anti-PD-1 mAb is crucial and will likely be dependent on the duration of the initial response and the reason for discontinuation. Novel anti-PD-1-based combination regimens with a hypomethylating agent, a Bruton’s tyrosine kinase inhibitor, and a JAK2 inhibitor may be effective in heavily pretreated patients and/or those with prior progression on CPI regimens.
In future trial designs for the treatment of cHL in the upfront setting, efforts to reduce short- and long-term toxicity rates must be prioritized in addition to malignancy-specific survival metrics. Although small phase II studies have demonstrated favorable efficacy of anti-PD-1 mAb with chemotherapy as a RT-sparing approach in early-stage bulky disease, ongoing studies using a PET-adapted approach are underway. By incorporating anti-PD-1 mAb as treatment escalation in patients with positive interim PET findings after upfront chemotherapy, patients have the potential of avoiding additional toxic chemotherapy and radiation (NCT04866654, NCT03233347, NCT03712202).

9. Conclusions

Classic HL is characterized by the dysregulation of TME-enabling pro-tumoral signaling and the evasion of immune surveillance by cancer cells, for which immune checkpoint inhibitor therapy is an ideal target. The inclusion of anti-PD-L1 mAb into the treatment armamentarium of cHL has provided patients with R/R disease who are insensitive to cytotoxic chemotherapy with the potential for long-term remission, particularly in those achieving deep response to treatment.
Single-agent anti-PD-1 mAb offers a CR of ~20–30% in the multiply relapsed/refractory setting, ~60–70% in the first relapse setting, and ~40–50% in the frontline setting based on various phase II studies (Table 1). It can be concluded that although standard chemoradiation can cure most patients in the upfront setting, those who do relapse have disease biology driven primarily by immune dysregulation and insensitivity to cytotoxic chemotherapy. This therefore serves as the ideal rationale for PET-adapted approaches in the frontline setting where patients who fail to have CMR on PET imaging with standard cytotoxic therapy proceed to anti-PD-1 mAb-based treatment (Table 2). It also serves as rationale for the utilization of immunotherapy in the 2nd-line salvage setting prior to ASCT as anti-PD-1 mAb utilizes an intact immune system that may be compromised with repeated lines of cytotoxic chemotherapy.
The specific populations that have the greatest potential for benefit from anti-PD-1 mAb-based therapy are patients deemed poor candidates for standard chemotherapy due to age or medical comorbidities and those with refractory or early relapsing disease. Based on the current data, a decision to incorporate immunotherapy must be tailored to individual patients based on their ASCT eligibility, their underlying comorbidities including those that put them at increased risk for immune-mediated toxicities related to anti-PD-1 mAb, and their personalized goals of care.

Author Contributions

Investigation, S.N., F.R.; writing—original draft preparation, S.N., F.R.; writing—review and editing, R.N.; visualization, A.V.; supervision, R.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. National Cancer Institute. Cancer Stat Facts: Hodgkin Lymphoma. In Cancer Stat.; 2016. Available online: https://seer.cancer.gov/statfacts/html/hodg.html (accessed on 30 April 2022).
  2. Vardiman, J.W.; Harris, N.L.; Brunning, R.D. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002, 100, 2292–2302. [Google Scholar] [CrossRef] [PubMed]
  3. Weniger, M.A.; Küppers, R. Molecular biology of Hodgkin lymphoma. Leukemia 2021, 35, 968–981. [Google Scholar] [CrossRef] [PubMed]
  4. Aldinucci, D.; Gloghini, A.; Pinto, A.; De Filippi, R.; Carbone, A. The classical Hodgkin’s lymphoma microenvironment and its role in promoting tumour growth and immune escape. J. Pathol. 2010, 221, 248–263. [Google Scholar] [CrossRef] [PubMed]
  5. Gordon, L.I.; Hong, F.; Fisher, R.I.; Bartlett, N.; Connors, J.M.; Gascoyne, R.D.; Wagner, H.; Stiff, P.J.; Cheson, B.D.; Gospodarowicz, M.; et al. Randomized Phase III Trial of ABVD Versus Stanford V With or Without Radiation Therapy in Locally Extensive and Advanced-Stage Hodgkin Lymphoma: An Intergroup Study Coordinated by the Eastern Cooperative Oncology Group (E2496). J. Clin. Oncol. 2013, 31, 684–691. [Google Scholar] [CrossRef] [Green Version]
  6. Carde, P.; Karrasch, M.; Fortpied, C.; Brice, P.; Khaled, H.; Casasnovas, O.; Caillot, D.; Gaillard, I.; Bologna, S.; Ferme, C.; et al. Eight Cycles of ABVD Versus Four Cycles of BEACOPP escalated Plus Four Cycles of BEACOPP baseline in Stage III to IV, International Prognostic Score ≥ 3, High-Risk Hodgkin Lymphoma: First Results of the Phase III EORTC 20012 Intergroup Trial. J. Clin. Oncol. 2016, 34, 2028–2036. [Google Scholar] [CrossRef] [Green Version]
  7. Sasse, S.; Bröckelmann, P.J.; Goergen, H.; Plütschow, A.; Müller, H.; Kreissl, S.; Buerkle, C.; Borchmann, S.; Fuchs, M.; Borchmann, P.; 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. J. Clin. Oncol. 2017, 35, 1999–2007. [Google Scholar] [CrossRef]
  8. Raemaekers, J.M.; André, M.P.; Federico, M.; Girinsky, T.; Oumedaly, R.; Brusamolino, E.; Brice, P.; Fermé, C.; Van Der Maazen, R.; Gotti, M.; et al. Omitting Radiotherapy in Early Positron Emission Tomography–Negative Stage I/II Hodgkin Lymphoma Is Associated With an Increased Risk of Early Relapse: Clinical Results of the Preplanned Interim Analysis of the Randomized EORTC/LYSA/FIL H10 Trial. J. Clin. Oncol. 2014, 32, 1188–1194. [Google Scholar] [CrossRef] [Green Version]
  9. Sibon, D.; Morschhauser, F.; Resche-Rigon, M.; Ghez, D.; Dupuis, J.; Marcais, A.; Deau-Fischer, B.; Bouabdallah, R.; Sebban, C.; Salles, G.; et al. Single or tandem autologous stem-cell transplantation for first-relapsed or refractory Hodgkin lymphoma: 10-year follow-up of the prospective H96 trial by the LYSA/SFGM-TC study group. Haematologica 2015, 101, 474–481. [Google Scholar] [CrossRef] [Green Version]
  10. Khera, N.; Storer, B.; Flowers, M.E.; Carpenter, P.A.; Inamoto, Y.; Sandmaier, B.M.; Martin, P.J.; Lee, S.J. Nonmalignant Late Effects and Compromised Functional Status in Survivors of Hematopoietic Cell Transplantation. J. Clin. Oncol. 2012, 30, 71–77. [Google Scholar] [CrossRef]
  11. Schaapveld, M.; Aleman, B.M.P.; Van Eggermond, A.M.; Janus, C.P.M.; Krol, S.; Van Der Maazen, R.W.M.; Roesink, J.M.; Raemaekers, J.M.M.; De Boer, J.P.; Zijlstra, J.M.; et al. Second Cancer Risk Up to 40 Years after Treatment for Hodgkin’s Lymphoma. N. Engl. J. Med. 2015, 373, 2499–2511. [Google Scholar] [CrossRef]
  12. Evens, A.M.; Hong, F.; Gordon, L.I.; Fisher, R.I.; Bartlett, N.; Connors, J.M.; Wagner, H.; Gospodarowicz, M.K.; Cheson, B.D.; Advani, R.; et al. Efficacy and tolerability of ABVD and Stanford V for elderly advanced-stage Hodgkin lymphoma (HL): Analysis from the phase III randomized U.S. Intergroup Trial E2496. J. Clin. Oncol. 2011, 29, 8035. [Google Scholar] [CrossRef]
  13. Straus, D.J.; Długosz-Danecka, M.; Alekseev, S.; Illés, Á.; Picardi, M.; Lech-Maranda, E.; Feldman, T.; Smolewski, P.; Savage, K.J.; Bartlett, N.; et al. Brentuximab vedotin with chemotherapy for stage III/IV classical Hodgkin lymphoma: 3-year update of the ECHELON-1 study. Blood 2020, 135, 735–742. [Google Scholar] [CrossRef] [PubMed]
  14. Straus, D.J.; Długosz-Danecka, M.; Connors, J.M.; Alekseev, S.; Illés, Á.; Picardi, M.; Lech-Maranda, E.; Feldman, T.; Smolewski, P.; Savage, K.J.; et al. Brentuximab vedotin with chemotherapy for stage III or IV classical Hodgkin lymphoma (ECHELON-1): 5-year update of an international, open-label, randomised, phase 3 trial. Lancet Haematol. 2021, 8, e410–e421. [Google Scholar] [CrossRef]
  15. Moskowitz, C.H.; Walewski, J.; Nademanee, A.; Masszi, T.; Agura, E.; Holowiecki, J.; Abidi, M.H.; Chen, A.I.; Stiff, P.; Viviani, S.; et al. Five-year PFS from the AETHERA trial of brentuximab vedotin for Hodgkin lymphoma at high risk of progression or relapse. Blood 2018, 132, 2639–2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Younes, A.; Gopal, A.K.; Smith, S.E.; Ansell, S.M.; Rosenblatt, J.D.; Savage, K.J.; Ramchandren, R.; Bartlett, N.L.; Cheson, B.D.; De Vos, S.; et al. Results of a Pivotal Phase II Study of Brentuximab Vedotin for Patients With Relapsed or Refractory Hodgkin’s Lymphoma. J. Clin. Oncol. 2012, 30, 2183–2189. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, R.; Gopal, A.K.; Smith, S.E.; Ansell, S.M.; Rosenblatt, J.D.; Savage, K.J.; Connors, J.M.; Engert, A.; Larsen, E.K.; Huebner, D.; et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood 2016, 128, 1562–1566. [Google Scholar] [CrossRef] [Green Version]
  18. 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. [Google Scholar] [CrossRef] [Green Version]
  19. Armand, P.; Engert, A.; Younes, A.; Fanale, M.; Santoro, A.; Zinzani, P.L.; Timmerman, J.M.; Collins, G.P.; Ramchandren, R.; Cohen, J.B.; 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. J. Clin. Oncol. 2018, 36, 1428–1439. [Google Scholar] [CrossRef]
  20. Ansell, S.; Bröckelmann, P.; von Keudell, G.; Lee, H.J.; Santoro, A.; Zinzani, P.L.; Collins, G.; Cohen, J.; De Boer, J.P.; Kuruvilla, J.; et al. HL-398: Five-Year Overall Survival from the CheckMate 205 Study of Nivolumab for Relapsed or Refractory (R/R) Classical Hodgkin Lymphoma (cHL). Clin. Lymphoma Myeloma Leuk. 2021, 21, S373–S374. [Google Scholar] [CrossRef]
  21. 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. [Google Scholar] [CrossRef]
  22. CChen, R.; Zinzani, P.L.; Lee, H.J.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood 2019, 134, 1144–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Armand, P.; Zinzani, P.L.L.; Lee, H.J.; Johnson, N.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; Tomita, A.; et al. Five-Year Follow-up of Keynote-087: Pembrolizumab Monotherapy in Relapsed/Refractory Classical Hodgkin Lymphoma (R/R cHL). Blood 2021, 138, 1366. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  25. Kuruvilla, J.; Ramchandren, R.; Santoro, A.; Paszkiewicz-Kozik, E.; Gasiorowski, R.; Johnson, A.N.; 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. [Google Scholar] [CrossRef]
  26. 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 2019, 34, 533–542. [Google Scholar] [CrossRef] [Green Version]
  27. 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. [Google Scholar] [CrossRef]
  28. 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. [Google Scholar] [CrossRef]
  29. 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. [Google Scholar] [CrossRef]
  30. 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. [Google Scholar] [CrossRef] [Green Version]
  31. Diefenbach, C.S.; Hong, F.; Ambinder, R.F.; Cohen, J.B.; Robertson, M.J.; David, A.K.; 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. [Google Scholar] [CrossRef]
  32. 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. [Google Scholar] [CrossRef] [PubMed]
  33. 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. [Google Scholar] [CrossRef] [PubMed]
  34. 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. [Google Scholar] [CrossRef] [PubMed]
  35. Mei, M.G.; Lee, H.J.; Palmer, J.; Chen, R.W.; Tsai, N.-C.; Chen, L.; McBride, K.; Smith, D.L.; Melgar, I.; Song, J.Y.; et al. Response-adapted anti-PD1 based salvage therapy for Hodgkin lymphoma with nivolumab +/− ICE (NICE). Blood 2022. [Google Scholar] [CrossRef]
  36. Bryan, L.J.; Casulo, C.; Allen, P.; Smith, S.E.; Savas, H.; Karmali, R.; Pro, B.; O’Shea, K.; Chmiel, J.; Palmer, B.A.; et al. 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. [Google Scholar] [CrossRef]
  37. Armand, P.; Chen, Y.-B.; Redd, R.A.; Joyce, R.M.; Bsat, J.; Jeter, E.; Merryman, R.; 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. [Google Scholar] [CrossRef]
  38. Cheson, B.D.; Bartlett, N.L.; LaPlant, B.; Lee, H.J.; Advani, R.J.; Christian, B.; Diefenbach, C.S.; Feldman, A.T.; 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. [Google Scholar] [CrossRef]
  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, 872–880. [Google Scholar] [CrossRef]
  40. 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. [Google Scholar] [CrossRef]
  41. 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. [Google Scholar] [CrossRef]
  42. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Szydłowski, M.; Prochorec-Sobieszek, M.; Szumera-Ciećkiewicz, A.; Derezińska, E.; Hoser, G.; Wasilewska, D.; Szymańska-Giemza, O.; Jabłońska, E.; Białopiotrowicz, E.; Sewastianik, T.; et al. Expression of PIM kinases in Reed-Sternberg cells fosters immune privilege and tumor cell survival in Hodgkin lymphoma. Blood 2017, 130, 1418–1429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Merryman, R.W.; Redd, R.; Jeter, E.; Wong, J.L.; McHugh, K.; Reynolds, C.; Nazzaro, M.; Varden, A.; Brown, J.R.; Crombie, J.L.; et al. Immune Reconstitution following High-Dose Chemotherapy and Autologous Stem Cell Transplantation with or without Pembrolizumab Maintenance Therapy in Patients with Lymphoma. Transplant. Cell. Ther. 2021, 28, 32.e1–32.e10. [Google Scholar] [CrossRef] [PubMed]
  45. Guolo, F.; Minetto, P.; Pesce, S.; Ballerini, F.; Clavio, M.; Cea, M.; Frello, M.; Garibotto, M.; Greppi, M.; Bozzo, M.; et al. Post-Transplant Nivolumab Plus Unselected Autologous Lymphocytes in Refractory Hodgkin Lymphoma: A Feasible and Promising Salvage Therapy Associated With Expansion and Maturation of NK Cells. Front. Immunol. 2021, 12, 4535. [Google Scholar] [CrossRef] [PubMed]
  46. Cader, F.Z.; Hu, X.; Goh, W.L.; Wienand, K.; Ouyang, J.; Mandato, E.; Redd, R.; Lawton, L.N.; Chen, P.-H.; Weirather, J.L.; et al. A peripheral immune signature of responsiveness to PD-1 blockade in patients with classical Hodgkin lymphoma. Nat. Med. 2020, 26, 1468–1479. [Google Scholar] [CrossRef]
  47. Garcia-Marquez, M.A.; Thelen, M.; Reinke, S.; Keller, D.; Wennhold, K.; Lehmann, J.; Veldman, J.; Borchmann, S.; Rosenwald, A.; Sasse, S.; et al. Reverted exhaustion phenotype of circulating lymphocytes as immune correlate of anti-PD1 first-line treatment in Hodgkin lymphoma. Leukemia 2021, 36, 760–771. [Google Scholar] [CrossRef]
  48. Roemer, M.G.; Redd, R.A.; Cader, F.Z.; Pak, C.J.; Abdelrahman, S.; Ouyang, J.; Sasse, S.; Younes, A.; Fanale, M.; Santoro, A.; et al. Major Histocompatibility Complex Class II and Programmed Death Ligand 1 Expression Predict Outcome After Programmed Death 1 Blockade in Classic Hodgkin Lymphoma. J. Clin. Oncol. 2018, 36, 942–950. [Google Scholar] [CrossRef]
  49. Gerhard-Hartmann, E.; Goergen, H.; Bröckelmann, P.J.; Mottok, A.; Steinmüller, T.; Grund, J.; Zamò, A.; Ben-Neriah, S.; Sasse, S.; Borchmann, S.; et al. 9p24.1 alterations and programmed cell death 1 ligand 1 expression in early stage unfavourable classical Hodgkin lymphoma: An analysis from the German Hodgkin Study Group NIVAHL trial. Br. J. Haematol. 2021, 196, 116–126. [Google Scholar] [CrossRef]
  50. Deshpande, R.P.; Sharma, S.; Watabe, K. The Confounders of Cancer Immunotherapy: Roles of Lifestyle, Metabolic Disorders and Sociological Factors. Cancers 2020, 12, 2983. [Google Scholar] [CrossRef]
  51. Patel, S.S.; Weirather, J.L.; Lipschitz, M.; Lako, A.; Chen, P.-H.; Griffin, G.K.; Armand, P.; Shipp, M.A.; Rodig, S.J. The microenvironmental niche in classic Hodgkin lymphoma is enriched for CTLA-4-positive T cells that are PD-1-negative. Blood 2019, 134, 2059–2069. [Google Scholar] [CrossRef]
  52. El Halabi, L.; Adam, J.; Gravelle, P.; Marty, V.; Danu, A.; Lazarovici, J.; Ribrag, V.; Bosq, J.; Camara-Clayette, V.; Laurent, C.; et al. Expression of the Immune Checkpoint Regulators LAG-3 and TIM-3 in Classical Hodgkin Lymphoma. Clin. Lymphoma Myeloma Leuk. 2020, 21, 257–266. [Google Scholar] [CrossRef] [PubMed]
  53. Ansell, S.M.; Bröckelmann, P.J.; von Keudell, G.; Lee, H.J.; Santoro, A.; Zinzani, P.L.; Collins, G.P.; Cohen, J.B.; De Boer, J.P.; Kuruvilla, J.; et al. Nivolumab for Relapsed or Refractory (R/R) Classical Hodgkin Lymphoma (CHL) after Autologous Transplantation: 5-year Overall Survival from the Phase 2 Checkmate 205 Study. Hematol. Oncol. 2021, 39. [Google Scholar] [CrossRef]
  54. Fedorova, L.V.; Lepik, K.V.; Mikhailova, N.B.; Kondakova, E.V.; Zalyalov, Y.R.; Baykov, V.V.; Babenko, E.V.; Kozlov, A.V.; Moiseev, I.S.; Afanasyev, B.V. Nivolumab discontinuation and retreatment in patients with relapsed or refractory Hodgkin lymphoma. Ann. Hematol. 2021, 100, 691–698. [Google Scholar] [CrossRef] [PubMed]
  55. Manson, G.; Brice, P.; Herbaux, C.; Bouabdallah, K.; Antier, C.; Poizeau, F.; Dercle, L.; Houot, R. Efficacy of anti-PD1 re-treatment in patients with Hodgkin lymphoma who relapsed after anti-PD1 discontinuation. Haematologica 2020, 105, 2664–2666. [Google Scholar] [CrossRef] [Green Version]
  56. Lepik, K.V.; Fedorova, L.V.; Kondakova, E.V.; Zalyalov, Y.R.; Babenko, E.V.; Lepik, E.E.; Kotselyabina, P.V.; Beynarovich, A.V.; Popova, M.O.; Volkov, N.P.; et al. A Phase 2 Study of Nivolumab Using a Fixed Dose of 40 mg (Nivo40) in Patients With Relapsed/Refractory Hodgkin Lymphoma. HemaSphere 2020, 4, e480. [Google Scholar] [CrossRef]
  57. Chan, T.S.Y.; Hwang, Y.; Khong, P.; Leung, A.Y.H.; Chim, C.; Tse, E.W.C.; Kwong, Y. Low-dose pembrolizumab and nivolumab were efficacious and safe in relapsed and refractory classical Hodgkin lymphoma: Experience in a resource-constrained setting. Hematol. Oncol. 2020, 38, 726–736. [Google Scholar] [CrossRef]
  58. Smith, S.D.; Moskowitz, C.H.; Dean, R.; Pohlman, B.; Sobecks, R.; Copelan, E.; Andresen, S.; Bolwell, B.; Maragulia, J.C.; Vanak, J.M.; et al. Autologous stem cell transplant for early relapsed/refractory Hodgkin lymphoma: Results from two transplant centres. Br. J. Haematol. 2011, 153, 358–363. [Google Scholar] [CrossRef]
  59. Baetz, T.; Belch, A.; Couban, S.; Imrie, K.; Yau, J.; Myers, R.; Ding, K.; Paul, N.; Shepherd, L.; Iglesias, J.; et al. Gemcitabine, dexamethasone and cisplatin is an active andnon-toxic chemotherapy regimen in relapsed or refractory Hodgkin’s disease: A phase II study by the National Cancer Institute of Canada Clinical Trials Group. Ann. Oncol. 2003, 14, 1762–1767. [Google Scholar] [CrossRef]
  60. Sirohi, B.; Cunningham, D.; Powles, R.; Murphy, F.; Arkenau, T.; Norman, A.; Oates, J.; Wotherspoon, A.; Horwich, A. Long-term outcome of autologous stem-cell transplantation in relapsed or refractory Hodgkin’s lymphoma. Ann. Oncol. 2008, 19, 1312–1319. [Google Scholar] [CrossRef]
  61. Moskowitz, C.H.; KewalRamani, T.; Nimer, S.D.; Gonzalez, M.; Zelenetz, A.; Yahalom, J. Effectiveness of high dose chemoradiotherapy and autologous stem cell transplantation for patients with biopsy-proven primary refractory Hodgkin’s disease. Br. J. Haematol. 2004, 124, 645–652. [Google Scholar] [CrossRef]
  62. Stamatoullas, A.; Ghesquieres, H.; Filliatre, L.C.; Quittet, P.; Morschhauser, F.; Ribrag, V.; Edeline, V.; Brice, P. Brentuximab Vedotin in First Refractory/Relapsed Classical Hodgkin Lymphoma Patients Treated By Chemotherapy (ICE) before Autologous Transplantation. Final Analysis of Phase II Study. Blood 2019, 134, 132. [Google Scholar] [CrossRef]
  63. Bartlett, N.; Niedzwiecki, D.; Johnson, J.; Friedberg, J.; Johnson, K.; van Besien, K.; Zelenetz, A.; Cheson, B.; Canellos, G. Gemcitabine, vinorelbine, and pegylated liposomal doxorubicin (GVD), a salvage regimen in relapsed Hodgkin’s lymphoma: CALGB. Ann. Oncol. 2007, 18, 1071–1079. [Google Scholar] [CrossRef] [PubMed]
  64. LaCasce, A.S.; Bociek, R.G.; Sawas, A.; Caimi, P.; Agura, E.; Matous, J.; Ansell, S.M.; Crosswell, H.E.; Islas-Ohlmayer, M.; Behler, C.; et al. Brentuximab vedotin plus bendamustine: A highly active first salvage regimen for relapsed or refractory Hodgkin lymphoma. Blood 2018, 132, 40–48. [Google Scholar] [CrossRef] [PubMed]
  65. Hagenbeek, A.; Zijlstra, J.M.; Plattel, W.J.; Morschhauser, F.; Lugtenburg, P.J.; Brice, P.; Hutchings, M.; Gastinne, T.; Liu, R.D.; Burggraaf, C.N.; et al. Combining Brentuximab Vedotin with DHAP as Salvage Treatment in Relapsed/Refractory Hodgkin Lymphoma: The Phase II HOVON/LLPC Transplant BRaVE study. Blood 2018, 132, 2923. [Google Scholar] [CrossRef]
  66. Cassaday, R.D.; Fromm, J.; Cowan, A.J.; Libby, E.N.; Philip, M.; Behnia, S.; Nartea, M.; Press, O.; Gopal, A.K. Safety and Activity of Brentuximab Vedotin (BV) Plus Ifosfamide, Carboplatin, and Etoposide (ICE) for Relapsed/Refractory (Rel/Ref) Classical Hodgkin Lymphoma (cHL): Initial Results of a Phase I/II Trial. Blood 2016, 128, 1834. [Google Scholar] [CrossRef]
  67. Herrera, A.; Palmer, J.; Martin, P.; Armenian, S.; Tsai, N.-C.; Kennedy, N.; Sahebi, F.; Cao, T.; Budde, L.; Mei, M.; et al. Autologous stem-cell transplantation after second-line brentuximab vedotin in relapsed or refractory Hodgkin lymphoma. Ann. Oncol. 2017, 29, 724–730. [Google Scholar] [CrossRef]
  68. Moskowitz, A.J.; Schöder, H.; Yahalom, J.; McCall, S.J.; Fox, S.Y.; Gerecitano, J.; Grewal, R.; Hamlin, P.; Horwitz, S.; Kobos, R.; et al. PET-adapted sequential salvage therapy with brentuximab vedotin followed by augmented ifosamide, carboplatin, and etoposide for patients with relapsed and refractory Hodgkin’s lymphoma: A non-randomised, open-label, single-centre, phase 2 study. Lancet Oncol. 2015, 16, 284–292. [Google Scholar] [CrossRef]
  69. 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. [Google Scholar] [CrossRef]
  70. Cole, P.D.; Mauz-Körholz, C.; Mascarin, M.; Michel, G.; Cooper, S.; Beishuizen, A.; Leger, K.J.; Amoroso, L.; Buffardi, S.; Rigaud, C.; et al. Nivolumab and brentuximab vedotin (BV)-based, response-adapted treatment in children, adolescents, and young adults (CAYA) with standard-risk relapsed/refractory classical Hodgkin lymphoma (R/R cHL): Primary analysis. J. Clin. Oncol. 2020, 38, 8013. [Google Scholar] [CrossRef]
  71. Sleijfer, S. Bleomycin-Induced Pneumonitis. Chest 2001, 120, 617–624. [Google Scholar] [CrossRef] [Green Version]
  72. Swerdlow, A.J.; Higgins, C.D.; Smith, P.; Cunningham, D.; Hancock, B.W.; Horwich, A.; Hoskin, P.; Lister, A.; Radford, J.; Rohatiner, A.Z.S.; et al. Myocardial Infarction Mortality Risk After Treatment for Hodgkin Disease: A Collaborative British Cohort Study. JNCI J. Natl. Cancer Inst. 2007, 99, 206–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Connors, J.M.; Jurczak, W.; Straus, D.J.; Ansell, S.M.; Kim, W.S.; Gallamini, A.; Younes, A.; Alekseev, S.; Illés, Á.; Picardi, M.; et al. Brentuximab Vedotin with Chemotherapy for Stage III or IV Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 378, 331–344. [Google Scholar] [CrossRef] [PubMed]
  74. 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. [Google Scholar] [CrossRef] [PubMed]
  75. Aleman, B.M.P.; van den Belt-Dusebout, A.W.; Bruin, M.L.d.; van’t Veer, M.B.; Baaijens, M.H.A.; de Boer, J.P.; Hart, A.A.M.; Klokman, W.J.; Kuenen, M.A.; Ouwens, G.M.; et al. Late cardiotoxicity after treatment for Hodgkin lymphoma. Blood 2007, 109, 1878–1886. [Google Scholar] [CrossRef] [Green Version]
  76. Proctor, S.J.; Wilkinson, J.; Jones, G.; Watson, G.C.; Lucraft, H.H.; Mainou-Fowler, T.; Culligan, D.; Galloway, M.J.; Wood, K.M.; McNally, R.J.Q.; et al. Evaluation of treatment outcome in 175 patients with Hodgkin lymphoma aged 60 years or over: The SHIELD study. Blood 2012, 119, 6005–6015. [Google Scholar] [CrossRef]
  77. Böll, B.; Görgen, H.; Fuchs, M.; Pluetschow, A.; Eich, H.T.; Bargetzi, M.J.; Weidmann, E.; Junghanß, C.; Greil, R.; Scherpe, A.; et al. ABVD in Older Patients With Early-Stage Hodgkin Lymphoma Treated Within the German Hodgkin Study Group HD10 and HD11 Trials. J. Clin. Oncol. 2013, 31, 1522–1529. [Google Scholar] [CrossRef] [Green Version]
  78. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  79. Wang, D.Y.; Salem, J.-E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Checkpoint inhibitor immunotherapy in Hodgkin’s lymphoma. Inactive T cells are activated through their TCR by encountering antigenic peptides presented by the MHC complex on the surfaces of APCs or tumor cells (Reed-Sternberg cells). In addition to TCR-MHC engagement, a co-stimulatory signal via B7 protein is required for target-cell lysis and effector cell responses. B7 protein on activated APCs can pair with either a CD28 on the surface of a T-cell to produce a costimulatory signal to enhance the activity of TCR-MHC signal and T-cell activation, or it can pair with CTLA-4 to produce an inhibitory signal to keep the T cell in the inactive state. Blocking the binding of B7 to CTLA-4 with an anti-CTLA-4 antibody allows T cells to be activated and to kill tumor cells. Other immune checkpoint proteins such as PD-1 on the surfaces of T cells and PD-L1 on tumor cells can also prevent T cells from killing tumor cells. Immune checkpoint blockade via monoclonal antibodies (anti-PD-L1 or anti-PD-1) can lead T cells to kill tumor cells. Abbreviations: MHC: major histocompatibility complex; APC: antigen-presenting cells; TCR: T-cell receptor; CTLA-4: cytotoxic T-lymphocyte associated antigen 4; PD-1: programmed death 1; PD-L1: programmed death-ligand 1.
Figure 1. Checkpoint inhibitor immunotherapy in Hodgkin’s lymphoma. Inactive T cells are activated through their TCR by encountering antigenic peptides presented by the MHC complex on the surfaces of APCs or tumor cells (Reed-Sternberg cells). In addition to TCR-MHC engagement, a co-stimulatory signal via B7 protein is required for target-cell lysis and effector cell responses. B7 protein on activated APCs can pair with either a CD28 on the surface of a T-cell to produce a costimulatory signal to enhance the activity of TCR-MHC signal and T-cell activation, or it can pair with CTLA-4 to produce an inhibitory signal to keep the T cell in the inactive state. Blocking the binding of B7 to CTLA-4 with an anti-CTLA-4 antibody allows T cells to be activated and to kill tumor cells. Other immune checkpoint proteins such as PD-1 on the surfaces of T cells and PD-L1 on tumor cells can also prevent T cells from killing tumor cells. Immune checkpoint blockade via monoclonal antibodies (anti-PD-L1 or anti-PD-1) can lead T cells to kill tumor cells. Abbreviations: MHC: major histocompatibility complex; APC: antigen-presenting cells; TCR: T-cell receptor; CTLA-4: cytotoxic T-lymphocyte associated antigen 4; PD-1: programmed death 1; PD-L1: programmed death-ligand 1.
Cancers 14 02936 g001
Table 1. Phase II and III Studies Evaluating Anti-PD1 mAb Therapies in Hodgkin’s Lymphoma.
Table 1. Phase II and III Studies Evaluating Anti-PD1 mAb Therapies in Hodgkin’s Lymphoma.
StudyPhaseKey InclusionAgentnORR (CR)PFS (Median Follow Up)OSmDOR (Median Follow Up)
Multiple R/R Disease: anti-PD-1 mAb monotherapy
Younes 2016 [18]
Armand 2018 [19]
Ansell 2021 [20]
IIPost ASCT +/− BVNivo24371% (21%)37% (24 m)
18% (60 m)
87%
(24 m)
71%
(60 m)
18 m (58 m)
Chen 2017 [21]
Chen 2019 [22]
Armand 2021 [23]
IIPost ASCT +/− BV or R/R to first line salvage
therapy
Pembro21071% (28%)44% (60 m)71%
(60 m)
7 m (60 m)
Zinzani 2020 [24] IIPrimary
Refractory
Disease
subgroup
Pembro7182% (35%)32% (24 m)94%
(24 m)
17 m (28 m)
Kuruvilla 2021 [25]IIIPost or ineligible for ASCTPembro vs. BV304N/A54% vs. 36 (12 m)N/AN/A
Song 2020 [26]
Song 2022 [27]
IIPost or ineligible for ASCTTislelizumab7087% (67%)41% (36 m)85%
(36 m)
32 m (10 m)
Nie 2019 [28]
Liu 2021 [29]
IIPost 2+ prior LOTCamrelizumab 1990% (32%)67% (24 m)63%
(24 m)
NR
Song 2019 [30]IIPost ASCTCamrelizumab7576.0% (28%)81% (6 m)
67% (12 m)
NRNR
Multiple R/R Disease: anti-PD-1 mAb combination therapies
Diefenbach 2020 [31]I/IIR/R after 1+ prior LOTIpi/BV2176% (57%)61% (12 m)NR N/A
Nivo/BV1889% (61%)70% (12 m)NRNR
Ipi/Nivo/BV2282% (73%)80% (12 m)NRNR
Lepik 2020 [32]IIR/R to Nivo monotherapyNivo + Benda3087% (57%)23% (25 m)97%
(24 m)
7 m
(25 m)
Nie 2019 [28]
Liu 2021 [29]
IIR/R in anti-PD-1 mAb naïve ptsCamrelizumab + Decitabine 4295% (71%)79% (6 m)
89% (12 m)
63%
(24 m)
NR
R/R in anti-PD-1 resistant ptsCamrelizumab + Decitabine 2552% (28%)79% (6 m)
59% (12 m)
N/A16 m (35 m)
First Salvage prior to Transplant
Advani 2021 [33]I/IIR/R in first
salvage therapy
Nivo+ BV9185% (67%)77% (36 m)93%
(36 m)
N/A
Moskowitz 2021 [34]IIR/R prior to ASCT Pembro + GVD38100% (95%)N/AN/AN/A
Mei 2022 [35]IIR/R bridge to ASCTNivo +/− ICE9100% (89%)72% (24 m)94%
(24 m)
N/A
Bryan 2021 [36]IIR/R prior to ASCTPembro + ICE42N/A88% (24 m)95%
(24 m)
N/A
Maintenance after ASCT
Armand 2019 [37]IIR/R after ASCTPembro30N/A82% (18 m)100%
(18 m)
N/A
Frontline
Cheson 2020 [38]II60+ years old and ineligible for chemotherapy Nivo + BV4661% (48%)N/AN/AN/A
Brockelmann 2020 [39]IIEarly stage
unfavorable
Nivo + AVD
(C and S)
10996% (87%)S: 95%
(24 m)
C: 100%
(24 m)
S: 100%
(24 m)
C: 100%
(24 m)
N/A
Ramchandren 2019 [40]IIAdvanced stage Nivo + AVD (S)5184% (67%)92% (9 m)98% (9 m)N/A
Allen 2021 [41]IIEarly stage
unfavorable and advanced stage
Pembro + AVD (S) 30100% (No CR)NRNRN/A
AVD: Adriamycin, vinblastine, dacabazine; C: combination; CR: complete response rate assessed by positron emission tomography (PET); GVD: gemcitabine, vinorelbine, doxorubicin (liposomal); ICE: ifosfamide, carboplatin, etoposide; Ipi: ipilimumab; LOT: line of therapy; mDOR: median duration of response; N/A: data not available; NR: not reached; Nivo: nivolumab; ORR: overall response rate assessed by PET; PFS: progression-free survival; Pembro: pembrolizumab; S: sequential; m: month.
Table 2. Phase II/III Pipeline Studies Evaluating Anti-PD1 mAb Therapies in Hodgkin’s Lymphoma.
Table 2. Phase II/III Pipeline Studies Evaluating Anti-PD1 mAb Therapies in Hodgkin’s Lymphoma.
ClinicalTrial.Gov
Identifier
PhaseRegimen
Frontline Setting Regimens
NCT04866654 IIABVD +/− PET adapted RT + Nivo in early stage nonbulky disease
NCT03233347IIA-AVD with PET adapted BV + Nivo; followed by Nivo maintenance in early-stage nonbulky disease
NCT03712202IIABVD with PET adapted BV/Nivo or ABVD in PET negative interim scan; A-AVD+ Nivo in PET positive interim scan in early-stage disease
NCT03907488IIIA-AVD vs. Nivo-AVD in stage III/IV disease
NCT03033914I/IIA(B)VD +/− PET adapted Nivo-AVD in stage III/IV disease
NCT03580408IINivo +/− Vinblastine in patients aged > 60 years and unfit for standard chemotherapy in any stage disease
Relapsed/Refractory setting: Combination with Chemotherapy Regimens
NCT03739619I/IINivo−Bendamustine + Gemcitabine
NCT04091490IINivo−DHAP
Relapsed/Refractory setting: Combination with Radiation Regimens
NCT03480334IINivo + RT in R/R cHL
NCT03495713IINivo + low dose RT in R/R cHL
Relapsed/Refractory setting: Chemotherapy Sparing Regimens
NCT04624984IIPD-1 inhibitor vs. PD-1 inhibitor + GVD as first salvage
NCT03337919IINivo monotherapy in disease refractory to 1st/2nd line salvage prior to ASCT
NCT04938232IIIpi +/− Nivo in R/R cHL
NCT04561206IIBV + Nivo as 2nd line tx in patients not candidates for ASCT
Relapsed/Refractory setting: Combination with Other Targeted Therapy Regimens
NCT01896999I/IINivo + BV +/− Ipi in R/R cHL
NCT05137886IITislelizumab + Decitabine
NCT03681561I/IINivo + Ruxolitinib in R/R cHL
NCT02940301IINivo + Ibrutinib in R/R cHL
NCT03057795IIBV + Nivo consolidation post ASCT
NCT05039073IIBV + Nivo combination in patients previously treated with BV or CPI
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nakhoda, S.; Rizwan, F.; Vistarop, A.; Nejati, R. Updates in the Role of Checkpoint Inhibitor Immunotherapy in Classical Hodgkin’s Lymphoma. Cancers 2022, 14, 2936. https://doi.org/10.3390/cancers14122936

AMA Style

Nakhoda S, Rizwan F, Vistarop A, Nejati R. Updates in the Role of Checkpoint Inhibitor Immunotherapy in Classical Hodgkin’s Lymphoma. Cancers. 2022; 14(12):2936. https://doi.org/10.3390/cancers14122936

Chicago/Turabian Style

Nakhoda, Shazia, Farsha Rizwan, Aldana Vistarop, and Reza Nejati. 2022. "Updates in the Role of Checkpoint Inhibitor Immunotherapy in Classical Hodgkin’s Lymphoma" Cancers 14, no. 12: 2936. https://doi.org/10.3390/cancers14122936

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop