Skip to main content

Advertisement

Log in

A multi-cohort phase 1b trial of rituximab in combination with immunotherapy doublets in relapsed/refractory follicular lymphoma

  • Original Article
  • Published:
Annals of Hematology Aims and scope Submit manuscript

Abstract

Antibodies targeting PD-1 or 4-1BB achieve objective responses in follicular lymphoma (FL), but only in a minority of patients. We hypothesized that targeting multiple immune receptors could overcome immune resistance and increase response rates in patients with relapsed/refractory FL. We therefore conducted a phase 1b trial testing time-limited therapy with different immunotherapy doublets targeting 4-1BB (utomilumab), OX-40 (ivuxolimab), and PD-L1 (avelumab) in combination with rituximab among patients with relapsed/refractory grade 1–3A FL. Patients were enrolled onto 2 of 3 planned cohorts (cohort 1 — rituximab/utomilumab/avelumab; cohort 2 — rituximab/ivuxolimab/utomilumab). 3+3 dose escalation was followed by dose expansion at the recommended phase 2 dose (RP2D). Twenty-four patients were enrolled (16 in cohort 1 and 9 in cohort 2, with one treated in both cohorts). No patients discontinued treatment due to adverse events and the RP2D was the highest dose level tested in both cohorts. In cohort 1, the objective and complete response rates were 44% and 19%, respectively (50% and 30%, respectively, at RP2D). In cohort 2, no responses were observed. The median progression-free survivals in cohorts 1 and 2 were 6.9 and 3.2 months, respectively. In cohort 1, higher density of PD-1+ tumor-infiltrating T-cells on baseline biopsies and lower density of 4-1BB+ and TIGIT+ T-cells in on-treatment biopsies were associated with response. Abundance of Akkermansia in stool samples was also associated with response. Our results support a possible role for 4-1BB agonist therapy in FL and suggest that features of the tumor microenvironment and stool microbiome may be associated with clinical outcomes (NCT03636503).

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Sureda A, Zhang MJ, Dreger P et al (2018) Allogeneic hematopoietic stem cell transplantation for relapsed follicular lymphoma: a combined analysis on behalf of the Lymphoma Working Party of the EBMT and the Lymphoma Committee of the CIBMTR. Cancer 124. https://doi.org/10.1002/cncr.31264

  2. Evens AM, Vanderplas A, Lacasce AS et al (2013) Stem cell transplantation for follicular lymphoma relapsed/refractory after prior rituximab: a comprehensive analysis from the NCCN lymphoma outcomes project. Cancer 119. https://doi.org/10.1002/cncr.28243

  3. Jacobson CA, Chavez JC, Sehgal AR et al (2022) Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol 23. https://doi.org/10.1016/S1470-2045(21)00591-X

  4. Fowler NH, Dickinson M, Dreyling M et al (2022) Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med 28. https://doi.org/10.1038/s41591-021-01622-0

  5. Budde LE, Sehn LH, Matasar M et al (2022) Safety and efficacy of mosunetuzumab, a bispecific antibody, in patients with relapsed or refractory follicular lymphoma: a single-arm, multicentre, phase 2 study. Lancet Oncol 23:1055–1065. https://doi.org/10.1016/S1470-2045(22)00335-7

    Article  PubMed  CAS  Google Scholar 

  6. Pangault C, Amé-Thomas P, Rossille D et al (2020) Integrative analysis of cell crosstalk within follicular lymphoma cell niche: towards a definition of the FL supportive synapse. Cancers (Basel) 12. https://doi.org/10.3390/cancers12102865

  7. Amé-Thomas P, le Priol J, Yssel H et al (2012) Characterization of intratumoral follicular helper T cells in follicular lymphoma: role in the survival of malignant B cells. Leukemia 26. https://doi.org/10.1038/leu.2011.301

  8. Yang ZZ, Kim HJ, Wu H et al (2020) TIGIT expression is associated with T-cell suppression and exhaustion and predicts clinical outcome and anti–PD-1 response in follicular lymphoma. Clin Cancer Res 26. https://doi.org/10.1158/1078-0432.CCR-20-0558

  9. Armand P, Janssens A, Gritti G et al (2021) Efficacy and safety results from CheckMate 140, a phase 2 study of nivolumab for relapsed/refractory follicular lymphoma. Blood 137:637–645. https://doi.org/10.1182/blood.2019004753

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Kuruvilla J, Armand P, Hamadani M et al (2022) Pembrolizumab for patients with non-Hodgkin lymphoma: phase 1b KEYNOTE-013 study. Leuk Lymphoma. https://doi.org/10.1080/10428194.2022.2136956

  11. Gopal AK, Levy R, Houot R et al (2020) First-in-human study of utomilumab, a 4-1BB/CD137 agonist, in combination with rituximab in patients with follicular and other CD20+ non-Hodgkin lymphomas. Clin Cancer Res 26. https://doi.org/10.1158/1078-0432.CCR-19-2973

  12. Nastoupil LJ, Chin CK, Westin JR et al (2022) Safety and activity of pembrolizumab in combination with rituximab in relapsed or refractory follicular lymphoma. Blood Adv 6. https://doi.org/10.1182/bloodadvances.2021006240

  13. Alfaro C, Echeveste JI, Rodriguez-Ruiz ME et al (2015) Functional expression of CD137 (4-1BB) on T helper follicular cells. Oncoimmunology 4. https://doi.org/10.1080/2162402X.2015.1054597

  14. Voo KS, Foglietta M, Percivalle E et al (2014) Selective targeting of toll-like receptors and OX40 inhibit regulatory T-cell function in follicular lymphoma. Int J Cancer 135. https://doi.org/10.1002/ijc.28937

  15. Hirano F, Kaneko K, Tamura H et al (2005) Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res 65. https://doi.org/10.1158/0008-5472.1089.65.3

  16. Lee S-J, Myers L, Muralimohan G et al (2004) 4-1BB and OX40 dual costimulation synergistically stimulate primary specific CD8 T cells for robust effector function. J Immunol 173. https://doi.org/10.4049/jimmunol.173.5.3002

  17. Cheson BD, Fisher RI, Barrington SF et al (2014) Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 32:3059–3067. https://doi.org/10.1200/JCO.2013.54.8800

    Article  PubMed  PubMed Central  Google Scholar 

  18. Carey CD, Gusenleitner D, Lipschitz M et al (2017) Topological analysis reveals a PD-L1 associated microenvironmental niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood. https://doi.org/10.1182/blood-2017-03-770719

  19. Klindworth A, Pruesse E, Schweer T et al (2013) Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41:e1. https://doi.org/10.1093/nar/gks808

    Article  PubMed  CAS  Google Scholar 

  20. Aronesty E (2013) Comparison of sequencing utility programs. Open Bioinform J 7:1–8. https://openbioinformaticsjournal.com/VOLUME/7/PAGE/1/

  21. Martin M (2011) Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J 17:10–12

    Article  Google Scholar 

  22. Callahan BJ, McMurdie PJ, Rosen MJ et al (2016) DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods 13:581–583. https://doi.org/10.1038/nmeth.3869

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Sievers F, Wilm A, Dineen D et al (2011) Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol 7:539. https://doi.org/10.1038/msb.2011.75

    Article  PubMed  PubMed Central  Google Scholar 

  24. Price MN, Dehal PS, Arkin AP (2010) FastTree 2—approximately maximum-likelihood trees for large alignments. PLoS One 5:e9490. https://doi.org/10.1371/journal.pone.0009490

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Quast C, Pruesse E, Yilmaz P et al (2013) The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 41:D590–D596. https://doi.org/10.1093/nar/gks1219

    Article  PubMed  CAS  Google Scholar 

  26. Mallick H et al (2021) Multivariable association discovery in population-scale meta-omics studies. PLOS Computational Biology 17(11):e1009442. https://doi.org/10.1371/journal.pcbi.1009442

  27. Tolcher AW, Sznol M, Hu-Lieskovan S et al (2017) Phase Ib study of utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in combination with pembrolizumab (MK-3475) in patients with advanced solid tumors. Clin Cancer Res 23:5349–5357. https://doi.org/10.1158/1078-0432.CCR-17-1243

    Article  PubMed  CAS  Google Scholar 

  28. Sehn LH, Goy A, Offner FC et al (2015) Randomized phase II trial comparing obinutuzumab (GA101) with rituximab in patients with relapsed CD20+ indolent B-cell non-Hodgkin lymphoma: final analysis of the GAUSS study. J Clin Oncol 33:3467–3474. https://doi.org/10.1200/JCO.2014.59.2139

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Leonard JP, Trneny M, Izutsu K et al (2019) AUGMENT: a phase III study of lenalidomide plus rituximab versus placebo plus rituximab in relapsed or refractory indolent lymphoma. J Clin Oncol 37(14):1188–1199. https://doi.org/10.1200/JCO.19.00010

    Article  PubMed  PubMed Central  Google Scholar 

  30. Rummel M, Kaiser U, Balser C et al (2016) Bendamustine plus rituximab versus fludarabine plus rituximab for patients with relapsed indolent and mantle-cell lymphomas: a multicentre, randomised, open-label, non-inferiority phase 3 trial. Lancet Oncol 17. https://doi.org/10.1016/S1470-2045(15)00447-7

  31. Dreyling M, Santoro A, Mollica L et al (2017) Phosphatidylinositol 3-kinase inhibition by copanlisib in relapsed or refractory indolent lymphoma. J Clin Oncol 35:3898–3905. https://doi.org/10.1200/JCO.2017.75.4648

    Article  PubMed  CAS  Google Scholar 

  32. Tobinai K, Igarashi T, Itoh K et al (2011) Rituximab monotherapy with eight weekly infusions for relapsed or refractory patients with indolent B cell non-Hodgkin lymphoma mostly pretreated with rituximab: a multicenter phase II study. Cancer Sci:102. https://doi.org/10.1111/j.1349-7006.2011.02001.x

  33. Diefenbach CS, Peters BA, Li H et al (2021) Microbial dysbiosis is associated with aggressive histology and adverse clinical outcome in B-cell non-Hodgkin lymphoma. Blood Adv:5. https://doi.org/10.1182/bloodadvances.2020003129

  34. Smith M, Dai A, Ghilardi G et al (2022) Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med 28. https://doi.org/10.1038/s41591-022-01702-9

  35. Routy B, Le Chatelier E, Derosa L et al (2018) Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359. https://doi.org/10.1126/science.aan3706

  36. Matson V, Fessler J, Bao R et al (2018) The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359. https://doi.org/10.1126/science.aao3290

  37. Derosa L, Routy B, Thomas AM et al (2022) Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat Med 28. https://doi.org/10.1038/s41591-021-01655-5

  38. Jin Y, Dong H, Xia L et al (2019) The diversity of gut microbiome is associated with favorable responses to anti–programmed death 1 immunotherapy in Chinese patients with NSCLC. J Thorac Oncol 14. https://doi.org/10.1016/j.jtho.2019.04.007

  39. Gopalakrishnan V, Spencer CN, Nezi L et al (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359. https://doi.org/10.1126/science.aan4236

  40. Bae M, Cassilly CD, Liu X et al (2022) Akkermansia muciniphila phospholipid induces homeostatic immune responses. Nature 608:168–173. https://doi.org/10.1038/s41586-022-04985-7

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

PA and RWM would like to acknowledge support from the Harold and Virginia Lash Grant Program. PA is supported by a Scholar Award from the Leukemia and Lymphoma Society. RWM would like to acknowledge support from an American Society of Hematology Research Training Award for Fellows, a Lymphoma Research Foundation (LRF) Clinical Investigator Career Development Award, and the LRF Lymphoma Clinical Research Mentoring Program. Multiplex immunofluorescence was performed in the Tissue Biomarker Laboratory of the Center for Immuno-Oncology at DFCI. Pfizer and the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100009945) reviewed this manuscript for medical accuracy only before journal submission. The authors are fully responsible for the content of this manuscript, and the views and opinions described in the publication reflect solely those of the authors.

Funding

Pfizer and the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100009945) provided funding for this investigator-sponsored clinical trial. The authors declare no other sources of funding.

Author information

Authors and Affiliations

Authors

Contributions

RWM and CAJ designed the research, performed research, analyzed the data, and wrote the paper; ASF, IEA, JRB, JC, MSD, DCF, EDJ, AIK, ASL, SN, OOO, EMP, II, JK, JBC, NM-S, NLB, and MM performed research and reviewed the paper. RAR, TK, SJR, and PA designed the research, performed research, analyzed the data, and reviewed the paper.

Corresponding author

Correspondence to Reid W. Merryman.

Ethics declarations

Conflict of interest

RWM — Consulting: Genmab, Adaptive Biotechnologies, Bristol Myers Squibb, Abbvie, Intellia, Epizyme. Research funding: Bristol Myers Squibb, Merck, Genentech/Roche, Genmab.

RAR — none.

ASF — none.

IEA — Consulting: BeiGene.

JRB — consultant for Abbvie, Acerta/Astra-Zeneca, BeiGene, Eli Lilly, Genentech/Roche, Grifols Worldwide Operations, Hutchmed, iOnctura, Janssen, MEI Pharma, Numab Therapeutics, Pfizer, Pharmacyclics; received research funding from BeiGene, Gilead, iOnctura, Loxo/Lilly, MEI Pharma, SecuraBio, TG Therapeutics.

JLC — Consultancy: Karyopharm, Kite, Morphosys, Incyte, ADC Therapeutics, Research Funding: Genentech/Roche, Merck, Abbvie, Bayer.

MSD — consulting fees from AbbVie, Adaptive Biosciences, Aptitude Health, Ascentage Pharma, AstraZeneca, BeiGene, Bio Ascend, BMS, Celgene, Curio Science, Eli Lilly, Genentech, Janssen, Merck, Ono Pharmaceuticals, Research to Practice, Secura Bio, TG Therapeutics, Takeda, and research support from AbbVie, AstraZeneca, Ascentage Pharma, Genentech, MEI Pharma, Novartis, Surface Oncology, TG Therapeutics.

DCF — none.

EDJ — Consulting: Syros, Takeda. Research funding: Acerta, Janssen, Novartis, Pharmacyclics.

AIK — none.

ASL — Consulting: Research to Practice. Advisory ad boards: Kite and Seagen.

SN — none.

OOO — none.

EMP — none.

II — consultant for Celgene/Jazz, Kite, Epizyme, Beam Therapeutics, and ADC Therapeutics.

JK — Consulting: Merck, ADC Therapeutics, Gilead, Daiichi Sankyo, BeiGene, SecuraBio. Research support: Merck, SecuraBio. Speakers’ Bureau: Kite/Gilead.

JBC — Advisory Board: Abbvie, Janssen, Astra Zeneca, BeiGene, Kite, ADCT, Hutchmed, Loxo/Lilly. Research Funding: Genentech, Takeda, Lilly/Loxo, Celgene/BMS, Astra Zeneca, HutchMed.

NM-S — institutional clinical trial funding from AstraZeneca, Bristol Myers-Squibb, Celgene, C4 Therapeutics, Corvus Pharmaceuticals, Daiichi Sankyo, Genentech/Roche, Innate Pharmaceuticals, Secura Bio/Verastem, Yingli Pharmaceuticals and Dizal Pharmaceuticals. She has received compensation for service as a consultant for AstraZeneca, Secura Bio/Verastem, Daiichi Sankyo, C4 Therapeutics, Genentech, Karyopharm Therapeutics, Kyowa Hakko Kirin, and Ono pharmaceuticals.

NLB — Consultancy/Advisory Boards: SeaGen, Roche/Genentech, Kite/Gilead, Foresight Diagnostics. Research funding (Institution): ADC Therapeutics, BMS/Celgene, Kite/Gilead, Merck, Millennium, Pharmacyclics, Roche/Genentech, SeaGen.

MM — Advisory board: Novartis, Seattle Genetics, CTI, Janssen, EUSA; Speaker’s bureau: Seattle Genetics, Monjuvi.

TMK — none.

JOW — none.

SJR — Research fundings: Bristol Myers Squibb and KITE/Gilead. Member of the SAB for Immunitas Therapeutics.

PA — Consultancy: Merck, BMS, Pfizer, Affimed, Adaptive, Infinity, ADC Therapeutics, Celgene, Morphosys, Daiichi Sankyo, Miltenyi, Tessa, GenMab, C4, Enterome, Regeneron, Epizyme, Astra Zeneca, Genentech, Xencor. Research funding: Kite. Research funding (institutional): Merck, BMS, Affimed, Adaptive, Tensha, Otsuka, Sigma Tau, Genentech/Roche, IGM. Honoraria: Merck, BMS.

CAJ — Consulting: Kite/Gilead, Novartis, BMS/Celgene, bluebird bio, Epizyme, Ipsen, Instil Bio, ImmPACT Bio, Caribou Bio, Morphosys, Miltenyi, Abintus Bio. Research funding: Kite/Gilead and Pfizer.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

ESM 1

(DOCX 189 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Merryman, R.W., Redd, R.A., Freedman, A.S. et al. A multi-cohort phase 1b trial of rituximab in combination with immunotherapy doublets in relapsed/refractory follicular lymphoma. Ann Hematol 103, 185–198 (2024). https://doi.org/10.1007/s00277-023-05475-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00277-023-05475-0

Keywords

Navigation