All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit the Lymphoma Coalition.

The Lymphoma Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your Lymphoma Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The Lymphoma Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the Lymphoma Hub cannot guarantee the accuracy of translated content. The Lymphoma Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2021-11-01T14:00:10.000Z

Hodgkin Lymphoma and adoptive immunotherapy: An overview of current research

Nov 1, 2021
Share:

Bookmark this article

A recent article by Ho, et al. in Blood Advances1 discusses adoptive immunotherapy for the treatment of classic Hodgkin lymphoma (cHL). Despite treatment options such as brentuximab vedotin and programmed cell death-1 (PD-1) checkpoint inhibitors, significant challenges remain in treating patients who are ineligible for transplant, or those who progress after transplant. Ho and colleagues’ discussion of relevant clinical trials, current challenges, and possible future strategies to improve treatment options for cHL are summarized in this article.

Chimeric antigen receptor (CAR) T-cell therapy, a type of adoptive immunotherapy, involves the genetic engineering of T cells to express CARs, and these receptors redirecting T cells toward tumor antigens. Autologous T cells are harvested from patients, engineered, and expanded ex vivo, then re-infused back into the patient to target and kill cancer cells. CAR T-cell therapy has been used to successfully treat B-cell malignancies, specifically non-Hodgkin lymphomas and relapsed or refractory (R/R) acute lymphoblastic leukemia. The role of CAR T-cell therapy in patients with R/R cHL however remains uncertain.

The immunosuppressive tumor microenvironment in cHL 

cHL is a lymphoid neoplasm characterized by the presence of Hodgkin Reed-Sternberg (HRS) cells admixed with a much larger population of non-malignant inflammatory cells.2 To consider novel adoptive T-cell treatment strategies, an understanding of the immune evasion in cHL and its immunosuppressive tumor microenvironment (TME) is important. HRS cells are characterized by cell surface markers CD15 and CD30 and loss of typical B-cell surface markers such as CD19. Although they account for only 1–2% of tumor cell mass, they thrive in a lymphocyte rich TME. Mechanisms of HRS immune evasion in cHL include:

  • Downregulation and alteration of HLA class I and II molecules causing interruption of tumor-antigen presentation.3-5
  • Secretion of various chemokines (CCL5, CCL17, and CCL22) and cytokines (IL-7) to recruit regulatory T cells, which in turn secrete IL-10 and TGF-β which inhibit cytotoxic T-lymphocyte (CTL) activity.6,7
  • Expression of Fas ligand which can induce apoptosis of CTLs.6,7
  • Recruitment of tumor-associated macrophages which support the immunosuppressive TME.
  • Frequent expression of checkpoint ligands—through various mechanisms including Epstein Barr virus (EBV) infection—such as programmed death receptor ligand-1 (PD-L1) and PD-L2 which inhibit T cell activation and proliferation.5,8,9

EBV-targeted T cells

Infection with EBV is often involved in the pathogenesis of cHL: approximately 40% of patients with cHL have EBV-positive disease.10 EBV-specific T cells have been used with significant clinical success to treat EBV-associated post-transplant lymphoproliferative disease. However, the expression of EBV antigens on HRS cells in EBV positive cHL follows the type II latency pattern, in which only weakly immunogenic EBV antigens are expressed. These include latent membrane protein (LMP)1, LMP2, EBV nuclear antigen 1 and BamH1-A right frame 1. EBV-positive HRS cells also secrete IL-10, which further inhibits cytotoxic T-cell response to viral antigens. This is in contrast to post-transplant lymphoproliferative disease in which the entire repertoire of EBV antigens is expressed (type III latency pattern).

After an early study of autologous CTLs directed against a nonspecific array of EBV antigens demonstrated limited clinical responses in cHL,11 LMP-specific CTLs were developed to improve specificity for antigens seen in the type II latency pattern of cHL. Study findings are summarized in Table 1. Patients did not undergo lymphodepletion prior to infusions.

Table 1. Results of EBV-targeted T cells in clinical trials*

Therapy

Reference

Phase

Patients (n)

Disease status

Response

LMP-1/2 specific T cells

Bollard,
 et al.12

1

50

EBV+ lymphoma in remission but considered high risk of relapse, or active R/R disease

25 with cHL (8 active, 17 in remission)

Active disease cohort:
ORR 62%;
CR 52%,
PR 10%

Remission cohort: 2-year EFS 82%

Response rates similar cHL vs NHL

LMP-1/2 specific T cells with DNRII

Bollard,
 et al.13

1

8

Extensively pre-treated R/R EBV-positive cHL

Active disease cohort: ORR 43%; CR 29%; PR 14%; SD 57%

Patient in remission (n = 1) remained in remission for 2+ years

cHL, classic Hodgkin lymphoma; CR, complete response; DNRII, dominant-negative TGF-β type 2 receptor; EBV, Epstein Barr virus; EFS, event free survival; LMP, latent membrane protein; NHL, non-Hodgkin lymphoma; ORR, overall response rate; PR, partial response; R/R, relapsed or refractory; SD, stable disease.
*Adapted from Ho,
et al.1

In the study by Bollard, et al.,12 50 patients with EBV+ lymphoma received infusions of LMP-specific T cells. Of these, 25 had cHL, eight with active disease. The overall response rate (ORR) for patients with active cHL was 50%. Interestingly, although cHL is associated with the type II latency pattern of EBV antigen expression, a small number of patients were found to have type III latency pattern secondary to comorbid immunosuppression. In the eight patients with active disease, three had type III latency and five had type II. All three patients with type III latency achieved complete response (CR) whereas the ORR for type II latency pattern was just 10%. This suggests differences in tumor immune evasion may exist, that are more easily overcome in patients that are immunosuppressed.

To further the efficacy of EBV-directed therapy, Bollard, et al.13 targeted the immunosuppressive TME by transducing EBV-specific CTLs with a dominant-negative TGF-β type 2 receptor (DNRII). Through forcing DNRII expression, T cells were resistant to TGF-β in vitro, retaining their tumor antigen-specific activity. Although only small a phase I trial of patients with extensively pre-treated R/R EBV-positive cHL, four of seven patients with active disease achieved a response to treatment (two achieved CR, one achieved PR and four had stable disease [SD]). Two patients who had previously received LMP-specific CTLs without DNRII14 showed a greater response to infusion with DNRII CTLs. In both studies, infusions were found to be safe.

Infusion of donor-derived allogenic EBV-specific CTLs has been shown to be safe and produce clinical responses in some small clinical trials.15,16 The use of partially matched banked EBV-specific CTLs (targeting LMP, BARF-1 and EBNA-1) is currently being explored (NCT02287311) and may be beneficial for those with rapidly progressive disease requiring urgent treatment.

CD30 CAR T cells

CD30, a member of the tumor necrosis factor receptor family, has anti-apoptotic and pro-survival effects on cells. It is expressed highly on HRS cells and only minimally on non-malignant activated B and T cells, thus limiting the possibility of on-target off-tumor toxicity. The success of brentuximab vedotin, a CD30 monoclonal antibody-drug conjugate, suggests it is an ideal therapeutic target.

Three published trials have considered CD30-targeting CAR-T cells and are summarized in Table 2. Of note, all three studies found that treatment was well tolerated. Furthermore, despite previous concern that CD30 CAR-T cells could impair cell-mediated immunity given that a small fraction of activated B and T cells express CD30, no significant increase in infections was seen in any of the trials.

Table 2. Results of CD30-targeted T cells in clinical trials*

Therapy

Reference

Phase

Patients (n)

Disease status

Lymphodepletion

Response

CD30 CAR-T cells

Wang, et al.17

1

18

CD30+ lymphoma:

17 with cHL
1 with ALCL

All had progressive disease

Fludarabine and cyclophosphamide; gemcitabine, mustargen, and cyclophosphamide; or ab-paclitaxel and cyclophosphamide

ORR 39%; CR 0%; PR 39%; SD 33%

PFR: 6 months (range, 3–14 months)

CD30 CAR-T cells

Ramos, et al.18

1

9

CD30+ lymphoma:
6 with cHL,
1 with DLBCL evolved to cHL,
2 with ALCL

All heavily pre-treated

None

ORR 33%; CR 33%; SD 33%; PD 33%. Durable CR up to 2.5+ years

CD30 CAR-T cells

Ramos, et al.19

1/2

41

R/R cHL

Median of 7 prior lines of therapy

90% previously received BV

81% previously received checkpoint inhibitor

Bendamustine; bendamustine and fludarabine; or cyclophosphamide and fludarabine

No responses with bendamustine-alone LD cohort

Responses with fludarabine-based regime (n = 32): ORR 72%; CR 59%; PR 13%; SD 9%; PD 19%

ALCL, anaplastic large cell lymphoma; CAR, chimeric antigen receptor; cHL, classic Hodgkin lymphoma; CR, complete response; EFS, event-free survival; NHL, non-Hodgkin lymphoma; ORR, overall response rate; PFS, progression-free survival; PR, partial response; R/R, relapsed or refractory; SD, stable disease.
*Adapted from Ho,
et al.1

Multiple clinical trials of CD30-targeted CAR-T cells are ongoing. One such study (NCT03602157) is investigating CD30 CAR-T cells that have been engineered to co-express CCR4. CCR4 is the ligand for chemokines CCL17 and CCL22, which are secreted by HRS cells and aid immune evasion and inhibition of immune response. These CD30 CAR-T cells with CCR4 have been shown to improve tumor-trafficking and anti-tumor activity in a mouse model of cHL.20 Furthermore, anti-CCR4 monoclonal antibodies were effective in depleting CCR4+ T cells and preventing migration of regulatory T cells in vitro and thus may present a further novel combination treatment strategy. Finally, several studies (NCT04288726, NCT01192464) are investigating EBV-specific CTLs that have been engineered to express anti-CD30 CAR.

Targeting the tumor microenvironment

Although CD19 is not expressed on HRS cells, CD19 targeted CAR-T cells have been considered in R/R cHL as a means of targeting non-malignant B cells in the TME, as well as CD19+ B cells in circulation that may represent HRS precursors. In a pilot study of four patients with R/R cHL and no curative therapy options, therapy was well tolerated; however, observed responses were transient: one had CR, one had PR, one had SD and one had PD at 1 month follow up.

CD123 is present on approximately 60% of cHL cell populations (as alpha chain of IL-3 receptor) as well as other cells in the TME such as M2-type tumor-associated macrophages (which suppress immune response and promote tumor infiltration). A pre-clinical study of anti-CD123 CAR-T cells demonstrated their ability to eradicate cHL cells, develop immunological memory and resist suppression by M2-macrophages.21

The potential to combine infusion of CD30 CAR-T cells with CD19 or CD123 CAR-T cells may offer further treatment strategies and is currently being explored (NCT03125577).

Conclusion

The development of adoptive immunity for the treatment of cHL has not accelerated as quickly as with other B cell malignancies; however, EBV-specific CTLs and CD30 CAR-T cells have shown promising responses in early clinical trials in patients with R/R cHL. Understanding the unique immunosuppressant tumor microenvironment in cHL and its mechanisms of immune evasion may offer opportunity to further improve treatment strategies, such as the addition of CD19 or CD123 CAR-T cells. Building on the success of adoptive immunotherapy in EBV+ patients, trials examining specific tumor-associated antigens—regardless of EBV status—are also now underway.

Furthermore, treatment strategies that combine CAR-T cells with checkpoint inhibitors, CAR-T cells that have been engineered to secrete anti-PD-1 checkpoint inhibitors, or T cells that have been genetically altered to disrupt PD-1 expression (via CRISPR-Cas9 gene editing) may enhance anti-tumor action and improve persistence of transferred T cells.

  1. Ho C, Ruella M,Levine B, et al. Adoptive T-cell Therapy for Hodgkin Lymphoma. Blood Adv. 2021. Online ahead of print. DOI: 10.1182/bloodadvances.2021005304
  2. Küppers R, Engert A, Hansmann ML. Hodgkin lymphoma. J Clin Invest. 2012;122(10):3439-3447. DOI: 1172/JCI61245
  3. Nijland M, Veenstra RN, Visser L, et al. HLA dependent immune escape mechanisms in B-cell lymphomas: Implications for immune checkpoint inhibitor therapy? Oncoimmunology. 2017;6(4):e1295202. DOI: 1080/2162402X.2017.1295202
  4. Diepstra A, Imhoff GW, Karim-Kos HE, et al. HLA class II expression by Hodgkin Reed-Sternberg cells is an independent prognostic factor in classical Hodgkin’s lymphoma. J Clin Oncol. 2007;25(21):3101-3108. DOI: 1200/JCO.2006.10.0917
  5. Roemer MGM, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol. 2016;34(23):2690-2697. DOI: 1200/JCO.2016.66.4482
  6. Marshall NA, Christie LE, Munro LR, et al. Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma. Blood. 2004;103(5):1755-1762. DOI: 1182/blood-2003-07-2594
  7. Dotti G, Savoldo B, Pule M, et al. Human cytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis. Blood. 2005;105(12):4677-4684. DOI: 1182/blood-2004-08-3337
  8. Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD- 1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116(17):3268-3277. DOI: 1182/blood-2010-05-282780
  9. Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and post-transplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012;18(16):1611-1618. DOI: 1158/1078-0432.CCR-11-1942
  10. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375-382. DOI: 1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t
  11. Bollard CM, Aguilar L, Straathof KC, et al. Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin’s disease. J Exp Med. 2004;200(12):1623-1633. DOI: 1084/jem.20040890
  12. Bollard CM, Straathof KC, Huls MH, et al. The generation and characterization of LMP2-specific CTLs for use as adoptive transfer from patients with relapsed EBV-positive Hodgkin disease. J Immunother. 2004;27(4):317-327. DOI: 1097/00002371-200407000-00008
  13. Bollard CM, Tripic T, Cruz CR, et al. Tumor-specific T-cells engineered to overcome tumor immune evasion induce clinical responses in patients with relapsed Hodgkin lymphoma. J Clin Oncol. 2018;36(11):1128-1139. DOI: 1200/JCO.2017.74.3179
  14. Bollard CM, Gottschalk S, Torrano V, et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol. 2014;32(8):798-808. DOI: 1200/JCO.2013.51.5304
  15. McLaughlin LP, Rouce R, Gottschalk S, et al. EBV/LMP-specific T cells maintain remissions of T- and B-cell EBV lymphomas after allogeneic bone marrow transplantation. Blood. 2018;132(22):2351-2361. DOI: 1182/blood-2018-07-863654
  16. Lucas KG, Salzman D, Garcia A, et al. Adoptive immunotherapy with allogeneic Epstein-Barr virus (EBV)-specific cytotoxic T-lymphocytes for recurrent, EBV-positive Hodgkin disease. Cancer. 2004;100(9):1892-1901. DOI: 1002/cncr.20188
  17. Wang CM, Wu ZQ, Wang Y, et al. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res. 2017;23(5):1156-1166. DOI: 1158/1078-0432.CCR-16-1365
  18. Ramos CA, Ballard B, Zhang H, et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor–redirected lymphocytes. J Clin Invest. 2017;127(9):3462-3471. DOI: 1172/JCI94306
  19. Ramos CA, Grover NS, Beaven AW, et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma. J Clin Oncol. 2020;38(32):3794-3804. DOI: 1200/JCO.20.01342
  20. Di Stasi A, De Angelis B, Rooney CM, et al. T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model. Blood. 2009;113(25):6392-6402. DOI: 1182/blood-2009-03-209650
  21. Ruella M, Klichinsky M, Kenderian SS, et al. Overcoming the immunosuppressive tumor microenvironment of Hodgkin lymphoma using chimeric antigen receptor T cells. Cancer Discov. 2017;7(10):1154-1167. DOI: 1158/2159-8290.CD-16-0850

Understanding your specialty helps us to deliver the most relevant and engaging content.

Please spare a moment to share yours.

Please select or type your specialty

  Thank you

Newsletter

Subscribe to get the best content related to lymphoma & CLL delivered to your inbox