About clonoSEQ

Detect subtle changes and manage patients decisively along the treatment continuum

clonoSEQ can powerfully detect measurable residual disease (MRD) throughout the treatment continuum—enabling you to confidently evaluate changes in disease burden and leap into action.1

clonoSEQ can powerfully detect measurable residual disease (MRD) throughout the treatment continuum—enabling you to confidently evaluate changes in disease burden and leap into action.1

Monitoring MRD status over time identifies changes in disease dynamics and provides valuable information for care decisions

Test at multiple timepoints across your patient’s journey to measure the effectiveness of treatment and inform changes as needed.

Identifying dominant clonal DNA sequences with the Clonality (ID) Test enables subsequent MRD monitoring to help inform personalized treatment decisions

Tracking (MRD) Reports show clonoSEQ MRD levels, which are highly prognostic of outcomes, throughout patient care

Determine whether to stay the course or adjust treatment based on your patient’s response to treatment

Be confident that the earliest signs of MRD resurgence are detected

MRD status may be helpful to determine whether a patient can stop or change therapy or if they should stay the course

See subtle changes in disease burden in patients who are on or off therapy

clonoSEQ can complement other measurement tools with a simple blood-based test that offers a clear view of disease burden13,24-26

How clonoSEQ works

The test leverages the basic biology of B- and T-cells to identify DNA sequences unique to the malignant cell population.27,28

The test leverages the basic biology of B- and T-cells to identify DNA sequences unique to the malignant cell population.27,28

These clones can then be tracked by their unique DNA signature or “barcode.”1

These clones can then be tracked by their unique DNA signature or “barcode.”1

clonoSEQ quantifies the malignant cell population comprising MRD so clonoSEQ results are a direct measure of cancer sequences, and not a surrogate marker of disease27,28

biopharma companies
routinely use clonoSEQ2
9

clinical trials
conducted to date
29

unique patients
tested29

Learn how clonoSEQ can help your patients across several disease states

FDA-cleared in bone marrow, CLIA-validated in peripheral blood

CLIA-validated in peripheral blood and bone marrow

FDA-cleared and CLIA-validated in bone marrow and peripheral blood

FDA-cleared in bone marrow, CLIA-validated in peripheral blood

FDA-cleared in bone marrow, CLIA-validated in peripheral blood

CLIA-validated in peripheral blood and bone marrow

MCL, mantle cell lymphoma.

*Testing for validated sample types other than bone marrow (in ALL, multiple myeloma, CLL) and peripheral blood (in CLL) is available via Adaptive’s CLIA-validated LDT service. Other sample types have not been cleared or approved by the FDA. Peripheral blood is FDA-cleared for CLL and plasma from blood is CLIA-validated/preferred for DLBCL.

NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.


This page is intended for a US-based audience.

clonoSEQ® is available as an FDA-cleared in vitro diagnostic (IVD) test service provided by Adaptive Biotechnologies to detect measurable residual disease (MRD) in bone marrow from patients with multiple myeloma or B-cell acute lymphoblastic leukemia (B-ALL) and blood or bone marrow from patients with chronic lymphocytic leukemia (CLL). Additionally, clonoSEQ is available for use in other lymphoid cancers and specimen types as a CLIA-validated laboratory developed test (LDT). To review the FDA-cleared uses of clonoSEQ, visit clonoSEQ.com/technical-summary.

References

  1. clonoSEQ®. [technical summary]. Seattle, WA: Adaptive Biotechnologies; 2020.
  2. Molica S, et al. Clin Lymphoma Myeloma Leuk. 2019;19(7):423-430.
  3. Kumar S, et al. Lancet Oncol. 2016;17(8):e328-e346.
  4. Akabane H, et al. Clin Adv Hematol Oncol. 2020;18(7):413-422.
  5. Martinez-Lopez J, et al. Blood. 2014;123(20):3073-3079.
  6. Perrot A, et al. Blood. 2018;132(23):2456-2464.
  7. Kovacs G, et al. J Clin Oncol. 2016;34(31):3758-3765.
  8. Al-Sawaf O, et al. Paper presented at: the 62nd ASH Annual Meeting and Exposition; December 5-8, 2020; virtual. Abstract 127. 
  9. Short N, et al. Blood Adv. 2022;6(13):4006-4014.
  10. Frank M, et al. J Clin Oncol. 2021;39(27):3034-3043.
  11. Costa L, et al. Lancet Haematol. 2023;10(11):e890-e901.
  12. Costa L, et al. Poster presented at: the 64th ASH Annual Meeting and Exposition; December 10-13, 2022; New Orleans, LA. Poster 3227. 
  13. Thompson P, et al. Blood. 2019;134(22):1951-1959. 
  14. Pulsipher M, et al. Blood Cancer Discov. 2022;3(1):66-81.
  15. Cavo M, et al. Blood. 2022;139(6):835-844. 
  16. Soumerai J, et al. Lancet Haematol. 2021;8(12):e879-e890.
  1. Logan A, et al. Biol Blood Marrow Transplant. 2014;20(9):1307-1313.
  2.  Roschewski M, et al. Lancet Oncol. 2015;16(5):541-549. 
  3. Munshi N, et al. Paper presented at: the 64th ASH Annual Meeting and Exposition; December 10-13, 2022; New Orleans, LA. Abstract 2030. 
  4. Merryman R, et al. Blood Adv. 2023;7(17):4748-4759. 
  5. Ananth S, et al. Paper presented at: the 65th ASH Annual Meeting and Exposition; December 9-12, 2023; San Diego, CA. Abstract 1673. 
  6. Kumar A, et al. Paper presented at: the 65th ASH Annual Meeting and Exposition; December 9-12, 2023; San Diego, CA. Abstract 738.
  7. Rezazadeh A, et al. Clin Lymphoma Myeloma Leuk. 2023. 10:S2152-2650(23)02195-X [online ahead of print]. doi: 10.1016/j.clml.2023.12.006
  8. Vij R, et al. Clin Lymphoma Myeloma Leuk. 2014;14(2):131-139.e1.
  9. Muffly L, et al. Blood Adv. 2021;5(16):3147-3151. 
  10. Muffly L, et al. Paper presented at: the 62nd ASH Annual Meeting and Exposition; December 5-8, 2020; virtual. Abstract 975.
  11. Carlson C, et al. Nat Commun. 2013;4:2680. 
  12. Faham M, et al. Blood. 2012;120(26):5173-5180. 
  13. Data on file. Adaptive Biotechnologies. 2023.