Next Article in Journal
Utility of the Levonorgestrel-Releasing Intrauterine System in the Treatment of Abnormal Uterine Bleeding and Dysmenorrhea: A Narrative Review
Next Article in Special Issue
Targeting IRAK4 with Emavusertib in Lymphoma Models with Secondary Resistance to PI3K and BTK Inhibitors
Previous Article in Journal
The Role of Vascular Risk Factors in Post-Stroke Delirium: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Combining BeEAM with Brentuximab Vedotin for High-Dose Therapy in CD30 Positive Lymphomas before Autologous Transplantation—A Phase I Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Harnessing the Molecular Fingerprints of B Cell Lymphoma for Precision Therapy

by
Afua Adjeiwaa Mensah
1,* and
Patrizia Mondello
2,*
1
Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
2
Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(19), 5834; https://doi.org/10.3390/jcm11195834
Submission received: 22 September 2022 / Accepted: 27 September 2022 / Published: 1 October 2022
The last two decades have brought ground-breaking advances in genetics, culminating in deep profiling of the human genome and high resolution detection of genetic variants. These developments have rewired preclinical and clinical efforts in understanding cancer, paving the way for precision therapies [1]. Lymphomas are among the most frequent tumours worldwide, with B cell lymphomas accounting for approximately 85% of newly diagnosed cases [2]. They arise from the germinal centre (GC), a specialised structure within the B cell follicle that forms upon encounter of B cells with an external antigen. Similarly to other cancers, the ability of lymphoma cells to bypass physiological signalling pathways controlling proliferation, differentiation and cell death is primarily governed by genetic and epigenetic defects. In line with this, the most recent classifications of lymphoma subtypes largely rely on the presence or absence of specific mutated genes and concordant aberrant molecular signatures [3,4]. As we gain more insights into the genetic evolution of lymphomas, either as they progress towards a more aggressive disease or because of resistance mechanisms arising in response to treatment, it is becoming increasingly evident that therapies targeting lymphoma-specific proteins represent important components of the therapeutic arsenal [5,6]. Personalised treatments, also known as targeted or precision medicines, utilise the molecular fingerprint of a patient’s tumour to inform drug development and treatment choices [7]. Due to the increased antitumour specificity of targeted agents compared to their non-targeted counterparts, precision medicine is currently the focus of most efforts to develop anti-lymphoma drugs. The turning point in the development of effective targeted therapies for lymphoma came with our increased understanding of the molecular drivers of this disease. The massive amounts of data gleaned from high-throughput sequencing of samples from multiple patient cohorts have provided a molecular basis for the observed clinical differences in lymphoma progression, response to therapy, and survival in individual lymphoma patients, and have provided a rationale for the development of therapies targeting specific lymphoma drivers [3,4].
The first targeted agent approved for treating B cell lymphoma was the monoclonal antibody rituximab (R), which has significantly improved patient survival in combination with chemotherapy (R-CHOP), compared to chemotherapy alone [8]. Following on from rituximab, multiple monoclonal CD20 antibodies have been developed and approved for lymphoma treatment including ofatumumab and obinutuzumab, but none have shown superiority to R-CHOP [9,10,11,12]. More recently, bispecific T cell engager (BiTE) antibodies that bind a B-cell-specific antigen on tumour cells (usually CD19) and CD3 on T cells have shown promise. The first-in-class BiTE, blinatumomab, was first approved for B cell acute lymphoblastic leukaemia [13], but its use is now expanding to other B cell lymphomas in both relapsed and frontline settings [14,15]. By bringing tumour cells in proximity to T cells, BiTEs enhance tumour cell killing by direct cell-to-cell contact. BiTEs offer an off-the-shelf and more immediate alternative to cellular therapy with chimeric antigen receptor T-cells (CAR-T). The latter require weeks to engineer autologous T cells expressing a chimeric T cell receptor that targets a cell surface antigen such as CD19. Additionally, BiTEs show lower grade, albeit similar treatment-related toxicities, to CAR-T cells. The mechanism of action of a BiTE can also be its Achille’s heel—the reliance on the patient’s T cells to kill tumour cells makes BiTEs unsuitable when T cell activity is poor. While it might appear that BiTEs and CAR-T cells compete for the same clinical field, recent data suggest the two can improve patient outcome when used sequentially, with CD20 BiTEs showing efficacy after failure of CD19 CAR-T cell therapy [16]. Resistance mechanisms to both platforms are T cell exhaustion secondary to repeated and/or prolonged antigen exposure and increased expression of checkpoint inhibitor molecules such as PD-L1 [17,18].
Monoclonal antibodies conjugated to cytotoxic payloads via a chemical linker, so-called antibody-drug conjugates (ADCs), represent a promising advancement on monoclonal antibodies, since they combine the tumour-homing function of a monoclonal antibody with a highly cytotoxic payload [19]. As ADCs are internalized after antibody recognition of a tumour-specific antigen followed by payload release inside the cell, the toxins conjugated to the antibody moiety of ADCs typically target intracellular macromolecules including tubulin, DNA and RNA polymerase II. The CD79 ADC polatuzumab-vedotin combined with rituximab alone or with both rituximab and bendamustine has shown efficacy in relapsed/refractory (R/R) diffuse large B cell lymphoma (DLBCL). Notably, polatuzumab-vedotin confers a survival advantage when used as a replacement for vincristine within the frontline R-CHOP regimen [20,21,22].
The development of novel therapeutic agents aimed at disrupting well-defined oncogenic signalling pathways has also been extensively explored in the last decade. Small molecules targeting kinases have shown robust pre-clinical and clinical activity in lymphomas, leading to the approval of three Bruton’s tyrosine kinase (BTK) inhibitors and three phosphatidylinositol 3-kinase (PI3K) inhibitors [23,24,25,26,27,28]. The efficacy of these molecules in lymphoma treatment highlights the major role of aberrant signalling cascades in driving and/or sustaining lymphomagenesis. The dynamic nature of these pathways and their convergence on each other might explain the development of resistance mechanisms when one of these components is targeted [29]. This equally suggests the potential for novel combinations or sequential approaches that block putative feedback mechanisms and treatment escape routes. However, while these rational combinations enhance tumour cytotoxicity, they also increase treatment associated toxicities and adverse events. In line with this notion, a plethora of clinical trials have tried adding targeted therapies to frontline R-CHOP regimen in an attempt to enhance efficacy but none have succeeded in improving survival [7]. This was partly due to the additional toxicities associated with these novel agents that impaired the achievement of the target dose, thus limiting overall efficacy.
Another recently developed treatment strategy involves targeting adaptive immune escape mechanisms to unmask cancer cells and in turn reactivate immune surveillance. Monoclonal antibodies targeting PD-L1, PD-1 and CTLA-4 are the big players in this approach, but while they have been game changers in many solid tumours, lymphomas have failed to show robust responses. The factors governing the lack of response, as well as early progression in lymphoma patients treated with immune checkpoint therapy, remain obscure, but might be related to an unfavourable tumour microenvironment (TME) and/or genetic alterations that favour immune escape. Notorious exceptions to this are Hodgkin’s lymphoma and primary mediastinal B cell lymphoma [30,31], likely due to the robust expression of PD-L1 in these diseases. Among lymphomas unresponsive to immune checkpoint therapy, DLBCL and follicular lymphoma frequently present genetic alterations conducive to immune escape that target transcription factors and epigenetic modifiers with roles in shaping the lymphoma TME [32]. These include mutations in CREBBP and EZH2 that lead to loss of MHC-I and MHC-II, while selective inhibition of their counterbalance proteins, e.g., HDAC3, or of the mutated proteins themselves, e.g., EZH2, restores antigen presentation and, in turn, an anti-lymphoma immune response [33,34,35]. The correct functioning of epigenetic proteins can also be influenced by the availability of metabolic products such as S-adenosyl methionine and acetyl-CoA, which are essential donors for epigenetic methylation and acetylation reactions, respectively, thereby intricately linking epigenetic regulation and metabolism with the lymphoma TME. Since metabolic subtypes of DLBCL show differential responses to epigenetic agents [36], the combination of specific metabolic inhibitors with other targeted agents such as selective histone deacetylase inhibitors could be therapeutically beneficial.
The classes of targeted agents presented here, and their mechanisms of action, demonstrate that we are on the cusp of a new molecular era, whose success will require a deeper understanding of the processes underlying lymphomagenesis. Much more work needs to be done to identify predictive biomarkers and develop simpler platforms for the routine identification of molecular subgroups. Our improved understanding of these mechanisms will inform the design of more effective therapeutics with less toxicity, which target synthetic vulnerabilities while promoting antitumour immunity.
We have just begun to decipher the molecular fingerprints of B cell lymphomas and, as such, routine personalised medicine is still a long way off. However, the strides made thus far are encouraging and fuel an expectation for the future implementation of personalised treatments in standard clinical practice.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  2. Teras, L.R.; DeSantis, C.E.; Cerhan, J.R.; Morton, L.M.; Jemal, A.; Flowers, C.R. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J. Clin. 2016, 66, 443–459. [Google Scholar] [CrossRef]
  3. Chapuy, B.; Stewart, C.; Dunford, A.J.; Kim, J.; Kamburov, A.; Redd, R.A.; Lawrence, M.S.; Roemer, M.G.M.; Li, A.J.; Ziepert, M.; et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 2018, 24, 679–690. [Google Scholar] [CrossRef] [PubMed]
  4. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1396–1407. [Google Scholar] [CrossRef] [PubMed]
  5. Morin, R.D.; Mendez-Lago, M.; Mungall, A.J.; Goya, R.; Mungall, K.L.; Corbett, R.D.; Johnson, N.A.; Severson, T.M.; Chiu, R.; Field, M.; et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 2011, 476, 298–303. [Google Scholar] [CrossRef] [Green Version]
  6. Pasqualucci, L.; Trifonov, V.; Fabbri, G.; Ma, J.; Rossi, D.; Chiarenza, A.; Wells, V.A.; Grunn, A.; Messina, M.; Elliot, O.; et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat. Genet. 2011, 43, 830–837. [Google Scholar] [CrossRef] [Green Version]
  7. Mondello, P.; Ansell, S.M. PHOENIX rises: Genomic-based therapies for diffuse large B cell lymphoma. Cancer Cell 2021, 39, 1570–1572. [Google Scholar] [CrossRef]
  8. Coiffier, B.; Lepage, E.; Briere, J.; Herbrecht, R.; Tilly, H.; Bouabdallah, R.; Morel, P.; Van Den Neste, E.; Salles, G.; Gaulard, P.; et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002, 346, 235–242. [Google Scholar] [CrossRef]
  9. Czuczman, M.S.; Hess, G.; Gadeberg, O.V.; Pedersen, L.M.; Goldstein, N.; Gupta, I.; Jewell, R.C.; Lin, T.S.; Lisby, S.; Strange, C.; et al. Chemoimmunotherapy with ofatumumab in combination with CHOP in previously untreated follicular lymphoma. Br. J. Haematol. 2012, 157, 438–445. [Google Scholar] [CrossRef]
  10. Sehn, L.H.; Martelli, M.; Trneny, M.; Liu, W.; Bolen, C.R.; Knapp, A.; Sahin, D.; Sellam, G.; Vitolo, U. A randomized, open-label, Phase III study of obinutuzumab or rituximab plus CHOP in patients with previously untreated diffuse large B-Cell lymphoma: Final analysis of GOYA. J. Hematol. Oncol. 2020, 13, 71. [Google Scholar] [CrossRef]
  11. Vitolo, U.; Trneny, M.; Belada, D.; Burke, J.M.; Carella, A.M.; Chua, N.; Abrisqueta, P.; Demeter, J.; Flinn, I.; Hong, X.; et al. Obinutuzumab or Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone in Previously Untreated Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2017, 35, 3529–3537. [Google Scholar] [CrossRef] [PubMed]
  12. Marcus, R.; Davies, A.; Ando, K.; Klapper, W.; Opat, S.; Owen, C.; Phillips, E.; Sangha, R.; Schlag, R.; Seymour, J.F.; et al. Obinutuzumab for the First-Line Treatment of Follicular Lymphoma. N. Engl. J. Med. 2017, 377, 1331–1344. [Google Scholar] [CrossRef] [PubMed]
  13. Kantarjian, H.; Stein, A.; Gokbuget, N.; Fielding, A.K.; Schuh, A.C.; Ribera, J.M.; Wei, A.; Dombret, H.; Foa, R.; Bassan, R.; et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2017, 376, 836–847. [Google Scholar] [CrossRef] [PubMed]
  14. Katz, D.A.; Morris, J.D.; Chu, M.P.; David, K.A.; Thieblemont, C.; Morley, N.J.; Khan, S.S.; Viardot, A.; Martin Garcia-Sancho, A.; Rodriguez-Garcia, G.; et al. Open-label, phase 2 study of blinatumomab after frontline R-chemotherapy in adults with newly diagnosed, high-risk DLBCL. Leuk Lymphoma 2022, 1–11. [Google Scholar] [CrossRef] [PubMed]
  15. Dufner, V.; Sayehli, C.M.; Chatterjee, M.; Hummel, H.D.; Gelbrich, G.; Bargou, R.C.; Goebeler, M.E. Long-term outcome of patients with relapsed/refractory B-cell non-Hodgkin lymphoma treated with blinatumomab. Blood Adv. 2019, 3, 2491–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Subklewe, M. BiTEs better than CAR T cells. Blood Adv. 2021, 5, 607–612. [Google Scholar] [CrossRef]
  17. Zugmaier, G.; Gokbuget, N.; Klinger, M.; Viardot, A.; Stelljes, M.; Neumann, S.; Horst, H.A.; Marks, R.; Faul, C.; Diedrich, H.; et al. Long-term survival and T-cell kinetics in relapsed/refractory ALL patients who achieved MRD response after blinatumomab treatment. Blood 2015, 126, 2578–2584. [Google Scholar] [CrossRef]
  18. Chong, E.A.; Melenhorst, J.J.; Lacey, S.F.; Ambrose, D.E.; Gonzalez, V.; Levine, B.L.; June, C.H.; Schuster, S.J. PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: Refueling the CAR. Blood 2017, 129, 1039–1041. [Google Scholar] [CrossRef] [Green Version]
  19. Barreca, M.; Lang, N.; Tarantelli, C.; Spriano, F.; Barraja, P.; Bertoni, F. Antibody-drug conjugates for lymphoma patients: Preclinical and clinical evidences. Explor. Target. Anti-Tumor Ther. 2022. [Google Scholar]
  20. Sehn, L.H.; Herrera, A.F.; Flowers, C.R.; Kamdar, M.K.; McMillan, A.; Hertzberg, M.; Assouline, S.; Kim, T.M.; Kim, W.S.; Ozcan, M.; et al. Polatuzumab Vedotin in Relapsed or Refractory Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2020, 38, 155–165. [Google Scholar] [CrossRef]
  21. Tilly, H.; Morschhauser, F.; Bartlett, N.L.; Mehta, A.; Salles, G.; Haioun, C.; Munoz, J.; Chen, A.I.; Kolibaba, K.; Lu, D.; et al. Polatuzumab vedotin in combination with immunochemotherapy in patients with previously untreated diffuse large B-cell lymphoma: An open-label, non-randomised, phase 1b-2 study. Lancet Oncol. 2019, 20, 998–1010. [Google Scholar] [CrossRef]
  22. Tilly, H.; Morschhauser, F.; Sehn, L.H.; Friedberg, J.W.; Trneny, M.; Sharman, J.P.; Herbaux, C.; Burke, J.M.; Matasar, M.; Rai, S.; et al. Polatuzumab Vedotin in Previously Untreated Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2022, 386, 351–363. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, M.L.; Rule, S.; Martin, P.; Goy, A.; Auer, R.; Kahl, B.S.; Jurczak, W.; Advani, R.H.; Romaguera, J.E.; Williams, M.E.; et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 2013, 369, 507–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sharman, J.P.; Egyed, M.; Jurczak, W.; Skarbnik, A.; Pagel, J.M.; Flinn, I.W.; Kamdar, M.; Munir, T.; Walewska, R.; Corbett, G.; et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): A randomised, controlled, phase 3 trial. Lancet 2020, 395, 1278–1291. [Google Scholar] [CrossRef]
  25. Hillmen, P.; Brown, J.R.; Eichhorst, B.F.; Lamanna, N.; O’Brien, S.M.; Qiu, L.; Salmi, T.; Hilger, J.; Wu, K.; Cohen, A.; et al. ALPINE: Zanubrutinib versus ibrutinib in relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma. Future Oncol. 2020, 16, 517–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gopal, A.K.; Kahl, B.S.; de Vos, S.; Wagner-Johnston, N.D.; Schuster, S.J.; Jurczak, W.J.; Flinn, I.W.; Flowers, C.R.; Martin, P.; Viardot, A.; et al. PI3Kdelta inhibition by idelalisib in patients with relapsed indolent lymphoma. N. Engl. J. Med. 2014, 370, 1008–1018. [Google Scholar] [CrossRef] [Green Version]
  27. Matasar, M.J.; Capra, M.; Ozcan, M.; Lv, F.; Li, W.; Yanez, E.; Sapunarova, K.; Lin, T.; Jin, J.; Jurczak, W.; et al. Copanlisib plus rituximab versus placebo plus rituximab in patients with relapsed indolent non-Hodgkin lymphoma (CHRONOS-3): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 678–689. [Google Scholar] [CrossRef]
  28. Flinn, I.W.; Miller, C.B.; Ardeshna, K.M.; Tetreault, S.; Assouline, S.E.; Mayer, J.; Merli, M.; Lunin, S.D.; Pettitt, A.R.; Nagy, Z.; et al. DYNAMO: A Phase II Study of Duvelisib (IPI-145) in Patients With Refractory Indolent Non-Hodgkin Lymphoma. J. Clin. Oncol. 2019, 37, 912–922. [Google Scholar] [CrossRef]
  29. Chandarlapaty, S. Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov. 2012, 2, 311–319. [Google Scholar] [CrossRef] [Green Version]
  30. 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. [Google Scholar] [CrossRef] [Green Version]
  31. Armand, P.; Rodig, S.; Melnichenko, V.; Thieblemont, C.; Bouabdallah, K.; Tumyan, G.; Ozcan, 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. [Google Scholar] [CrossRef] [PubMed]
  32. Mondello, P.; Ansell, S.M.; Nowakowski, G.S. Immune Epigenetic Crosstalk Between Malignant B Cells and the Tumor Microenvironment in B Cell Lymphoma. Front. Genet. 2022, 13, 826594. [Google Scholar] [CrossRef] [PubMed]
  33. Ennishi, D.; Takata, K.; Beguelin, W.; Duns, G.; Mottok, A.; Farinha, P.; Bashashati, A.; Saberi, S.; Boyle, M.; Meissner, B.; et al. Molecular and Genetic Characterization of MHC Deficiency Identifies EZH2 as Therapeutic Target for Enhancing Immune Recognition. Cancer Discov. 2019, 9, 546–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Mondello, P.; Tadros, S.; Teater, M.; Fontan, L.; Chang, A.Y.; Jain, N.; Yang, H.; Singh, S.; Ying, H.Y.; Chu, C.S.; et al. Selective Inhibition of HDAC3 Targets Synthetic Vulnerabilities and Activates Immune Surveillance in Lymphoma. Cancer Discov. 2020, 10, 440–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bourne, C.M.; Mun, S.S.; Dao, T.; Aretz, Z.E.H.; Molvi, Z.; Gejman, R.S.; Daman, A.; Takata, K.; Steidl, C.; Klatt, M.G.; et al. Unmasking the suppressed immunopeptidome of EZH2-mutated diffuse large B-cell lymphomas through combination drug treatment. Blood Adv. 2022, 6, 4107–4121. [Google Scholar] [CrossRef]
  36. Mensah, A.A.; Spriano, F.; Sartori, G.; Priebe, V.; Cascione, L.; Gaudio, E.; Tarantelli, C.; Civanelli, E.; Aresu, L.; Rinaldi, A.; et al. Study of the antilymphoma activity of pracinostat reveals different sensitivities of DLBCL cells to HDAC inhibitors. Blood Adv. 2021, 5, 2467–2480. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mensah, A.A.; Mondello, P. Harnessing the Molecular Fingerprints of B Cell Lymphoma for Precision Therapy. J. Clin. Med. 2022, 11, 5834. https://doi.org/10.3390/jcm11195834

AMA Style

Mensah AA, Mondello P. Harnessing the Molecular Fingerprints of B Cell Lymphoma for Precision Therapy. Journal of Clinical Medicine. 2022; 11(19):5834. https://doi.org/10.3390/jcm11195834

Chicago/Turabian Style

Mensah, Afua Adjeiwaa, and Patrizia Mondello. 2022. "Harnessing the Molecular Fingerprints of B Cell Lymphoma for Precision Therapy" Journal of Clinical Medicine 11, no. 19: 5834. https://doi.org/10.3390/jcm11195834

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