HemaSphere (Apr 2024)

Circulating tumor plasma cells and peripheral blood measurable residual disease assessment in multiple myeloma patients not planned for upfront transplant

  • Prashant R. Tembhare,
  • Harshini Sriram,
  • Twinkle Khanka,
  • Sanghamitra Gawai,
  • Bhausaheb Bagal,
  • Sitaram G. Ghogale,
  • Nilesh Deshpande,
  • Karishma Girase,
  • Jagruti Patil,
  • Syed Khaizer Hasan,
  • Dhanalaxmi Shetty,
  • Kinjalka Ghosh,
  • Gaurav Chatterjee,
  • Sweta Rajpal,
  • Nikhil V. Patkar,
  • Hasmukh Jain,
  • Sachin Punatar,
  • Anant Gokarn,
  • Lingaraj Nayak,
  • Sumeet Mirgh,
  • Nishant Jindal,
  • Manju Sengar,
  • Navin Khattry,
  • Papagudi G. Subramanian,
  • Sumeet Gujral

DOI
https://doi.org/10.1002/hem3.63
Journal volume & issue
Vol. 8, no. 4
pp. n/a – n/a

Abstract

Read online

Abstract Circulating tumor plasma cells (CTPCs) provide a noninvasive alternative for measuring tumor burden in newly diagnosed multiple myeloma (NDMM). Moreover, measurable residual disease (MRD) assessment in peripheral blood (PBMRD) can provide an ideal alternative to bone marrow MRD, which is limited by its painful nature and technical challenges. However, the clinical significance of PBMRD in NDMM still remains uncertain. Additionally, data on CTPC in NDMM patients not treated with transplant are scarce. We prospectively studied CTPC and PBMRD in 141 NDMM patients using highly sensitive multicolor flow cytometry (HS‐MFC). PBMRD was monitored at the end of three cycles (PBMRD1) and six cycles (PBMRD2) of chemotherapy in patients with detectable baseline CTPC. Patients received bortezomib‐based triplet therapy and were not planned for an upfront transplant. Among baseline risk factors, CTPC ≥ 0.01% was independently associated with poor progression‐free survival (PFS) (hazard ratio [HR] = 2.77; p = 0.0047) and overall survival (OS) (HR = 2.9; p = 0.023) on multivariate analysis. In patients with detectable baseline CTPC, undetectable PBMRD at both subsequent time points was associated with longer PFS (HR = 0.46; p = 0.0037), whereas detectable PBMRD at any time point was associated with short OS (HR = 3.25; p = 0.004). Undetectable combined PBMRD (PBMRD1 and PBMRD2) outperformed the serum‐immunofixation‐based response. On multivariate analysis, detectable PBMRD at any time point was independently associated with poor PFS (HR = 2.0; p = 0.025) and OS (HR = 3.97; p = 0.013). Thus, our findings showed that CTPC and PBMRD assessment using HS‐MFC provides a robust, noninvasive biomarker for NDMM patients not planned for an upfront transplant. Sequential PBMRD monitoring has great potential to improve the impact of the existing risk stratification and response assessment models.