Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 1.
Published in final edited form as: Leuk Lymphoma. 2020 Jun 16;61(8):1777–1779. doi: 10.1080/10428194.2020.1779261

Exhausted and outnumbered: CD4+ T cells in the myeloma battlefield

Elisabet E Manasanch 1, Morie A Gertz 2
PMCID: PMC7665850  NIHMSID: NIHMS1643651  PMID: 32543918

The role of the immune system in the pathogenesis of hematologic malignancies has long been the subject of research studies (1). More recently, knowledge on the mechanisms by which immune cell subsets contribute to immune tolerance and help blood cancers gain control of the microenvironment has made considerable progress (2). Indeed, the discovery and role of checkpoint inhibitors as well as the ability to successfully manipulate effector immune cells has led to a revolution in cancer treatment (3). To our knowledge, seven immune checkpoint inhibitors are currently approved for treatment of at least fifteen different tumor types, including lymphoma (4). Tisagenlecleucel and axicabtagene, both CD19 targeted chimeric antigen receptor T cell product (CAR T) with different costimulatory domains were the first of such therapies to be approved for treatment of leukemia (tisagenlecleucel) and later non-Hodgkin’s lymphoma (both tisagenlecleucel and axicabtagene) by regulatory agencies (57). In the field of multiple myeloma, it is increasingly being recognized that dysregulation of the quantity and composition of immune cell subsets in the tumor microenvironment leads to progression from monoclonal gammopathy of unknown significance (MGUS) to overt myeloma (8). Immune dysregulation has also been linked to a higher risk of “graft versus host”-like syndrome after autologous stem cell transplantation (ASCT) and poor clinical outcomes in newly diagnosed myeloma (911).

In this issue of Leukemia and Lymphoma, Gu and colleagues evaluate the role of T lymphocyte subsets in the peripheral blood of one hundred and ten newly diagnosed myeloma patients (12). The study period (2012–2016) belongs to the era of novel agents with patients treated with bortezomib and immunomodulatory agents, however only a small subset of patients underwent ASCT (14%). The authors measured levels of CD3+CD4+ and CD3+CD8+ T cell subsets in peripheral blood at baseline for these patients. They found that the median CD3+CD4+ T cell count was low at 0.41 × 109/L. Patients who had higher CD3+CD4+ T cells (>0.5 × 109/L) had longer progression free survival (PFS) and overall survival (OS) compared to those who didn’t (PFS 66 vs 34 months and OS 31.5 vs 18.5 months). A higher CD4+/CD8+ T cell ratio (>0.7 × 109/L) was associated with longer PFS (24 vs 10 months) and OS (24 vs 10 months) in this cohort of patients. No relationship was seen between CD3+CD8+ T cells counts in peripheral blood and clinical outcomes. Overall, lower CD3+CD4+ T cell counts were associated with lower hemoglobin (hazard ratio (HR) 2.3) and higher LDH (HR 2.4), Beta-2-microglobulin (HR 2.7) and high-risk cytogenetics (HR 2.7).

Although similar findings had been reported elsewhere, the results of this study are important as they confirm that the composition of the immune system at baseline remains an important predictor of clinical outcomes in the era of novel agents in myeloma patients (1). The development of CD38 monoclonal antibodies daratumumab and isatuximab is one of the major recent advances in the treatment of myeloma. Studies investigating their use added to the standard of care in frontline myeloma are ongoing (IMROZ- NCT03319667 transplant ineligible patients and PERSEUS – NCT03710603 transplant eligible patients) or have been recently reported (GRIFFIN – transplant eligible patients) (13). It remains to be seen whether immune cell composition remains an independent predictor of clinical outcome in patients treated with quadruplet therapies. Immune dysregulation occurs early in myeloma pathogenesis and considerable research effort is in place to develop immunotherapy treatment strategies for early myeloma (14). For example, PD-L1 on myeloma cells has been associated with an increased risk of progression from smoldering to multiple myeloma (15). A pilot study of single agent pembrolizumab in 13 patients with smoldering myeloma led to an extraordinary responder who remains in complete remission minimal residual disease negative after only three doses of pembrolizumab 3 years post therapy. Extensive correlative analysis showed that this patient had a pre-existing immune response which was augmented by pembrolizumab, whereas the non-responders had features of T-cell exhaustion and may require combination immune therapy to restore checkpoint function (16). Strategies that enhance antitumor T-cell responses need to be further explored in early myeloma.

A biologics license application for idecabtagene vicleucel (bb2121), a BCMA targeted CAR T cell product, has been filed with the US Food and Drug Administration for approval in the treatment of relapsed myeloma (17). Given high responses in this patient population, trials are ongoing to investigate the role of cell therapy in high-risk newly diagnosed myeloma for whom standard options may not result in tumor remission (KarMMa-4; NCT04196491). In fact, the possibility exists that cell therapy in earlier lines will result in deeper and longer remissions compared to multiply relapsed/refractory myeloma. As in the manuscript by Gu and colleagues, higher CD4+/CD8+ ratio at the time of leukapheresis has also been associated with response to cell therapy and thus, improved clinical outcomes. Furthermore, a significantly higher median CD4+/CD8+ ratio in newly diagnosed myeloma patients after initial therapy compared to relapsed/refractory myeloma has been reported. This suggests that leukapheresis for T cell products should be done soon after initial therapy and not when the patient is multiply relapsed (18, 19). In myeloma, CD4+ T cells are often times exhausted and outnumbered. Strategies that can rebuild T cell function should be investigated in clinical trials.

References

  • 1.San Miguel JF, Gonzalez M, Gascon A, Moro MJ, Hernandez JM, Ortega F, et al. Lymphoid subsets and prognostic factors in multiple myeloma. Cooperative Group for the Study of Monoclonal Gammopathies. Br J Haematol. 1992;80(3):305–9. [DOI] [PubMed] [Google Scholar]
  • 2.Curran EK, Godfrey J, Kline J. Mechanisms of Immune Tolerance in Leukemia and Lymphoma. Trends Immunol. 2017;38(7):513–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Freyer CW, Porter DL. Advances in CAR T Therapy for Hematologic Malignancies. Pharmacotherapy. 2020. [DOI] [PubMed] [Google Scholar]
  • 4.Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers (Basel). 2020;12(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2019;380(1):45–56. [DOI] [PubMed] [Google Scholar]
  • 6.Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):439–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N Engl J Med. 2017;377(26):2531–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zavidij O, Haradhvala NJ, Mouhieddine TH, Sklavenitis-Pistofidis R, Cai S, Reidy M, et al. Single-cell RNA sequencing reveals compromised immune microenvironment in precursor stages of multiple myeloma. Nature Cancer. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Anagnostou T, Sakemura R, Forsman CL, Patnaik MS, Paludo J, Gastineau DA, et al. Baseline immune dysregulation in autologous stem cell transplant recipients is associated with a ‘graft versus host’-like syndrome and poor outcomes. Bone Marrow Transplant. 2020. [DOI] [PubMed] [Google Scholar]
  • 10.Zahran AM, Sayed MM, Shafik EA, Khallaf SM, Mohamed WM, Hetta HF. The Frequency and clinical Implications of Lymphocyte Subsets and Circulating Plasma Cells in Newly Diagnosed Multiple Myeloma Patients. Egypt J Immunol. 2019;26(2):117–31. [PubMed] [Google Scholar]
  • 11.Vela-Ojeda J, Esparza MAG, Majluf-Cruz A, Garcia-Chavez J, Montiel-Cervantes LA, Reyes-Maldonado E, et al. Post-treatment improvement of NK cell numbers predicts better survival in myeloma patients treated with thalidomide-based regimens. Int J Hematol. 2019;110(3):306–12. [DOI] [PubMed] [Google Scholar]
  • 12.Gu Y, Jin Y, Ding J, Yujie W, Shi Q, Qu X, et al. Low absolute CD4+ T cell counts in peripheral blood predict poor prognosis in patients with newly diagnosed multiple myeloma. Leukemia & Lymphoma. 2020:1–8. [DOI] [PubMed] [Google Scholar]
  • 13.Voorhees PM, Kaufman JL, Laubach JP, Sborov DW, Reeves B, Rodriguez C, et al. Daratumumab, Lenalidomide, Bortezomib, & Dexamethasone for Transplant-eligible Newly Diagnosed Multiple Myeloma: GRIFFIN. Blood. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kunacheewa C, Manasanch EE. High-risk smoldering myeloma versus early detection of multiple myeloma: Current models, goals of therapy, and clinical implications. Best Pract Res Clin Haematol. 2020;33(1):101152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dhodapkar MV, Sexton R, Das R, Dhodapkar KM, Zhang L, Sundaram R, et al. Prospective analysis of antigen-specific immunity, stem-cell antigens, and immune checkpoints in monoclonal gammopathy. Blood. 2015;126(22):2475–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Manasanch EE, Han G, Mathur R, Qing Y, Zhang Z, Lee H, et al. A pilot study of pembrolizumab in smoldering myeloma: report of the clinical, immune, and genomic analysis. Blood Adv. 2019;3(15):2400–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N Engl J Med. 2019;380(18):1726–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Garfall AL, Dancy EK, Cohen AD, Hwang WT, Fraietta JA, Davis MM, et al. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma. Blood Adv. 2019;3(19):2812–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cohen AD, Garfall AL, Stadtmauer EA, Melenhorst JJ, Lacey SF, Lancaster E, et al. B cell maturation antigen–specific CAR T cells are clinically active in multiple myeloma. The Journal of Clinical Investigation. 2019;129(6):2210–21. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES