Breaking Research MRD Positive Status Predicts Leukemia Outcomes

Introduction
Minimal residual disease (MRD) status has emerged as one of the most powerful prognostic markers in leukemia management, fundamentally transforming how clinicians assess treatment response and long-term outcomes. MRD-positive status profoundly influences survival outcomes, as demonstrated by compelling evidence from recent clinical trials and meta-analyses. Patients who remain MRD-positive after therapy exhibit notably poorer prognoses compared to those who achieve MRD negativity. Data from large-scale analyses reveal that the 5-year disease-free survival rate among MRD-negative patients is approximately 64%, whereas those with MRD positivity experience a markedly reduced rate of about 25%. Similarly, the 5-year overall survival rate stands at roughly 68% for MRD-negative patients, compared to only 34% for individuals who remain MRD-positive.
The concept of MRD reflects the persistence of leukemic cells that survive initial therapy but exist below the detection limit of traditional morphological assessment. These residual malignant cells, though few in number, represent a critical determinant of relapse risk and treatment outcome. The prognostic impact of MRD has been consistently validated across multiple leukemia subtypes, including acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). It remains a reliable marker regardless of the detection methodology used—whether through multiparametric flow cytometry, quantitative polymerase chain reaction (qPCR), or next-generation sequencing (NGS). The sensitivity of these methods allows clinicians to detect one leukemic cell among 10,000 to 100,000 normal cells, providing an early and precise indicator of therapeutic efficacy.
For clinicians and researchers, the interpretation of MRD-positive results carries substantial clinical implications. MRD positivity has been shown to supersede several conventional prognostic indicators, including baseline white blood cell count, cytogenetic profile, and immunophenotypic characteristics. As such, MRD assessment is increasingly being integrated into clinical guidelines to guide treatment intensification, transplantation decisions, and post-remission monitoring strategies. Patients who fail to achieve MRD negativity after induction or consolidation therapy are considered at high risk for relapse and may benefit from alternative therapeutic approaches, such as allogeneic hematopoietic stem cell transplantation (HSCT) or targeted immunotherapy.
Ongoing research continues to refine the role of MRD as both a prognostic and predictive biomarker. Advances in translational models are helping to elucidate mechanisms of MRD persistence, including clonal evolution and microenvironmental protection within the bone marrow niche. Innovative treatment modalities are also being explored to eradicate residual disease, with promising results emerging from immune-based strategies such as chimeric antigen receptor (CAR) T-cell therapy, bispecific T-cell engagers, and antibody-drug conjugates designed to specifically target leukemic cells that evade standard chemotherapy.
In summary, MRD-positive status represents a critical inflection point in leukemia management, reflecting minimal yet clinically significant disease burden. Its detection provides clinicians with actionable insights that can guide individualized treatment decisions, improve prognostic accuracy, and ultimately enhance patient survival outcomes. The integration of MRD monitoring into standard practice underscores a paradigm shift in oncology—from reliance on morphological remission to the pursuit of molecular and immunological eradication of disease.
Keywords: minimal residual disease, leukemia, acute myeloid leukemia, prognosis, survival outcomes, MRD monitoring, CAR T-cell therapy, translational oncology
Understanding MRD Positive Status in Leukemia
Measurable residual disease (MRD) represents a critical metric that shapes treatment decisions and prognostication in modern leukemia management. The presence of residual leukemic cells below morphological detection thresholds yet identifiable through specialized techniques forms the foundation of this concept that has transformed leukemia care.
What does MRD positive mean in clinical terms?
MRD positive status indicates the presence of sub-microscopic leukemic cells that persist despite treatment appearing successful by conventional assessment methods. In clinical practice, MRD positivity typically refers to the detection of leukemic cells at levels below the traditional 5% blast threshold used for morphological remission assessment [1]. Although a patient may present with approximately 10^12 leukemic blasts at diagnosis, standard microscopy can only detect around 1% of blasts [2]. Consequently, MRD represents anything from 10^10 cells down to the detection limit of the assay employed [2].
The threshold for defining MRD positivity varies across studies but has generally standardized around 0.01% (10^-4) leukemic cells. This cutoff has proven remarkably consistent across research, with patients showing even minimal levels between 0.01% and 0.1% demonstrating substantially worse outcomes than those achieving complete MRD clearance [3]. Additionally, higher MRD levels correlate with progressively worse prognosis—patients with 0.1% to 1% MRD showing 49% five-year event-free survival (EFS), while those with more than 1% MRD experiencing only 30% EFS [3].
MRD positive leukemia vs MRD negative: outcome differences
The contrast between outcomes for MRD positive versus MRD negative patients is stark across all leukemia subtypes. In adult ALL patients, those achieving MRD negativity within the first three cycles of chemotherapy demonstrated 64% ten-year event-free survival, whereas MRD positive patients faced a remarkably reduced rate of only 21% [3]. For AML, the median five-year overall survival reaches 68% in MRD negative patients but drops dramatically to 34% in MRD positive cases [4].
Timing of MRD clearance also plays a key role in outcome prediction. In a study of high-risk Philadelphia-negative ALL patients, those achieving early MRD negativity (within 1.5 months of therapy) had a remarkable 100% two-year relapse-free survival compared to only 38% for those remaining MRD positive [1]. Notably, even patients who eventually cleared MRD but did so late (after 1.5 months) still experienced poor outcomes, with two-year relapse-free survival of just 34% [1].
The impact of MRD status extends through all treatment phases. For transplant candidates, pre-transplant MRD positivity predicts substantially higher relapse rates—patients entering transplant with MRD positivity experiencing 33.7% relapse at 12 months versus only 7.3% for those transplanted in MRD negative status [5].
Prevalence of MRD positivity in AML and ALL
MRD positivity occurs at variable rates depending on leukemia subtype, treatment protocol, and assessment timing. Initially, during therapy, MRD positivity rates are quite high. For instance, approximately 68.9% of pediatric ALL patients have detectable MRD in peripheral blood at day 8 of treatment [3]. Following induction therapy, this prevalence decreases but remains clinically significant—42.2% of patients still exhibit detectable MRD on day 33 of therapy [6].
For adult patients with ALL, studies from the German Multicenter Study Group demonstrated distinct patterns of MRD clearance. Among standard-risk patients receiving asparaginase-based chemotherapy, only 10% achieved early complete MRD negativity (both day 11 and day 24 negative), while a concerning proportion remained persistently positive [3]. Among those persistently MRD positive at both day 24 and week 16, 94% experienced relapse within three years [3].
In AML, the prevalence patterns differ somewhat from ALL. MRD persistence in AML patients often correlates with specific genetic markers. Particularly informative are NPM1 mutations and fusion genes like RUNX1-RUNX1T1, CBFB-MYH11, and PML-RARA, where persistent detection following therapy strongly predicts relapse [7]. Patients achieving at least 4-log reduction in NPM1 mutational burden demonstrated superior outcomes, with 58% achieving bone marrow MRD negativity as their best response [5].
Comparative Overview of MRD Detection Techniques
The accurate detection of minimal residual disease requires sophisticated laboratory techniques that have evolved substantially over the past decade. Current MRD assessment methods differ in sensitivity, applicability, and turnaround time—factors that directly influence their clinical utility for monitoring MRD positive leukemia.
Flow cytometry sensitivity: 10^-4 to 10^-5
Multiparameter flow cytometry (MFC) remains a cornerstone technique for MRD detection, offering rapid results within hours of sample collection. Conventional MFC typically achieves a sensitivity of 10^-4 (0.01%), making it sufficient for many clinical decision points [8]. However, this sensitivity level depends heavily on the quality of the specimen and the expertise of the operator performing the analysis [7].
The EuroFlow Consortium has pioneered standardized 8-color antibody panels that considerably improve MFC sensitivity. When analyzing samples containing more than 4 million cells, this approach can reach sensitivities of 10^-5, making it comparable to PCR-based methods [9]. Moreover, next-generation flow cytometry (NGF) further enhances sensitivity to approximately 2 × 10^-6 through optimized combinations of fluorochromes and antibody reagents [1].
In AML diagnostics, flow cytometry identifies leukemia-associated immunophenotypes (LAIPs)—marker expression combinations present on >10% of blasts but absent in healthy bone marrow. These LAIPs can be identified in approximately 90% of patients, enabling subsequent MRD tracking [8]. Nevertheless, MFC faces challenges including the requirement for fresh samples, subjective interpretation, and potential phenotypic shifts during treatment [7].
PCR-based detection: Ig/TCR rearrangements and fusion genes
PCR techniques offer enhanced sensitivity compared to standard flow cytometry, typically detecting one leukemic cell among 100,000 normal cells (10^-5) [2]. Two principal PCR approaches exist for MRD assessment: detection of immunoglobulin (IG) and T-cell receptor (TCR) gene rearrangements, and quantification of fusion transcripts.
Real-time quantitative PCR (RQ-PCR) targeting IG/TCR rearrangements remains the gold standard for MRD detection in ALL [7]. This method identifies unique sequences arising from V(D)J recombination in diagnostic specimens, then tracks these sequences in follow-up samples. Remarkably, IG rearrangements can be found in both B-cell malignancies and approximately 20% of T-ALL cases, whereas TCR rearrangements appear in up to 90% of B-ALL cases—a phenomenon called cross-lineage rearrangement [7].
For patients with fusion gene-positive leukemia, reverse transcription PCR (RT-PCR) provides an effective alternative. This approach is especially valuable in Philadelphia chromosome-positive ALL, where BCR-ABL1 transcript detection through RT-PCR demonstrates superior sensitivity compared to flow cytometry [2]. Additional fusion targets include ETV6-RUNX1 in pediatric B-ALL and TCF3-PBX1 in both pediatric and adult populations, each showing variable concordance with IG/TCR-based methods [10].
NGS-based MRD detection: 10^-6 sensitivity and clonotype tracking
Next-generation sequencing represents the newest frontier in MRD assessment, offering unprecedented sensitivity of 10^-6 (one leukemic cell per million normal cells) [1]. The clonoSEQ assay, which targets IG/TCR rearrangements through NGS, has received FDA clearance for MRD detection in B-cell ALL, chronic lymphocytic leukemia, and multiple myeloma [2].
NGS-MRD delivers several distinct advantages beyond sensitivity alone. First, it allows comprehensive characterization of the IG/TCR repertoire, enabling detection of clones present at low frequency at diagnosis that may expand later [7]. Second, NGS facilitates clonal tracking, providing insight into clonal evolution patterns during treatment and relapse [1]. Third, studies demonstrate strong correlation between blood and bone marrow NGS-MRD results, potentially enabling less invasive monitoring [1].
The clinical value of this enhanced sensitivity has been validated in multiple studies. Patients with ALL who test MRD-negative at 10^-4 via flow cytometry but MRD-positive via NGS show increased relapse risk compared to those negative by both methods [1]. Furthermore, in the transplant setting, detectable pre-transplant NGS-MRD at levels below 10^-4 significantly predicts post-transplant relapse and inferior survival [1].
Digital PCR represents yet another advancement, offering improved precision without requiring reference curves. This technique separates samples into thousands of water-oil emulsion droplets before PCR amplification, further enhancing detection limits [7].
Clinical Impact of MRD Positive Status in AML
Among risk factors for disease recurrence, MRD positive status stands out as a powerful determinant of patient outcomes in acute myeloid leukemia (AML). The prognostic value of MRD has emerged as one of the strongest indicators of adverse outcomes across different treatment settings and disease stages, often superseding baseline genetic classifications.
MRD positive AML and relapse risk
MRD positivity in AML patients correlates directly with substantially higher relapse rates. Patients who test MRD positive on day +100 after transplantation face an extremely poor prognosis, with a cumulative incidence of relapse reaching 93.3% [11]. This stark contrast underscores the clinical relevance of MRD status post-treatment.
The impact of MRD positivity persists regardless of when it’s detected during the treatment course. Patients showing MRD positivity after the first consolidation chemotherapy (MRDcon1) demonstrate markedly higher relapse rates than those achieving MRD negativity [12]. Similarly, MRD positivity after the second consolidation cycle (MRDcon2) independently predicts adverse outcomes after adjustment for other variables [12].
The hazard ratios for MRD positivity quantify this elevated risk precisely:
- Pre-HSCT MRD ≥ 0.1%: HR 2.29 for decreased leukemia-free survival [11]
- Post-induction MRD: HR 0.50 (0.23-0.82) for overall survival [4]
- Post-induction MRD: HR 0.44 (0.23-0.82) for leukemia-free survival [4]
Post-induction MRD and survival outcomes
MRD status after induction therapy shows clear correlation with survival metrics. Patients achieving MRD negativity experience a median overall survival of 23.3 months versus only 15.2 months for MRD positive patients (p = 0.006) [4]. This survival advantage extends to leukemia-free survival, with MRD negative patients maintaining disease control for 20.3 months compared to just 9.2 months for those with detectable disease (p = 0.01) [4].
Timing of MRD clearance plays a vital role in outcome prediction. Patients who achieve early MRD negativity (immediately after induction) show superior outcomes compared to those who clear MRD later during consolidation. The median relapse-free survival reaches 73 months, 22 months, and 5 months for patients with MRD negative/negative, positive/negative, and positive/positive status after induction and consolidation, respectively [5]. Hence, undetectable flow MRD after induction associates with better relapse-free survival than undetectable MRD achieved later during consolidation [5].
In multivariable analysis, MRD status retains its independent prognostic value for both overall survival (HR: 0.50, 95% CI: 0.23–0.82; p = 0.04) and leukemia-free survival (HR: 0.44, 95% CI: 0.23–0.82; p = 0.01) after adjustment for other clinical variables [4].
MRD-guided transplant decisions in AML
MRD status profoundly influences transplantation decisions and outcomes. For MRD positive patients, allogeneic hematopoietic stem cell transplantation (allo-HSCT) in first complete remission provides substantial survival benefit compared to no transplantation (HR, 0.39; 95% CI, 0.24–0.64) [5]. Conversely, this transplant benefit diminishes considerably for MRD negative patients (HR, 0.82; 95% CI, 0.50–1.33) [5].
Pre-transplant MRD status stratifies patients into distinct risk categories. The 2-year and 5-year overall survival rates reach 76% and 66.7% for patients with MRD <0.1%, compared to 50.5% and 58.4% for those with MRD ≥0.1% (p<0.001) [11]. Likewise, leukemia-free survival at 2 years shows similar stratification: 67.6% for MRD negative patients, 49.7% for low-level MRD (<0.1%), and only 36.6% for high-level MRD (≥0.1%) patients [11].
Conditioning intensity represents another critical consideration for MRD positive patients. Myeloablative conditioning demonstrates improved survival compared to reduced intensity conditioning in young patients with detectable pre-transplant MRD [13]. Moreover, among patients with persistent FLT3-ITD or NPM1 mutations receiving reduced intensity conditioning, melphalan-based regimens yield superior survival compared to other reduced-intensity approaches [13].
As transplant strategies evolve, MRD status increasingly guides decisions about conditioning regimens, graft manipulation, maintenance therapy, and immunosuppression management—with the ultimate goal of reducing relapse risk in this vulnerable population [13].
MRD Positive Status in ALL: Prognostic Implications
The measurement of minimal residual disease has revolutionized risk stratification in acute lymphoblastic leukemia, providing unprecedented precision in outcome prediction. Studies consistently demonstrate that MRD status after therapy outweighs traditional prognostic factors in determining patient outcomes.
MRD positivity after induction therapy in ALL
Post-induction MRD assessment yields critical prognostic information in ALL treatment protocols. A comprehensive meta-analysis revealed that adults achieving MRD negativity experienced 64% ten-year disease-free survival in contrast to merely 21% for those with detectable disease after initial therapy [3]. In pediatric populations, approximately 42.2% of patients maintain detectable MRD following induction therapy [6]. Children with negative MRD status post-induction demonstrated 94.6% five-year event-free survival compared to 76.1% for those with persistent disease [6].
The German Multicenter Study Group on Adult ALL (GMALL) established that molecular response to standard induction and consolidation emerged as the sole factor with independent prognostic value for remission duration throughout both standard-risk and high-risk patient categories [14]. Correspondingly, the French, Northern Italian, and Spanish study groups confirmed this strong prognostic impact [14].
Risk classification increasingly incorporates MRD status alongside conventional factors. Patients stratified by MRD-based risk demonstrate markedly different outcomes:
- Standard risk (42%): 92.3% five-year EFS
- Intermediate risk (52%): 77.6% five-year EFS
- High risk (6%): 50.1% five-year EFS [6]
Philadelphia chromosome-positive ALL and MRD tracking
In Philadelphia chromosome-positive ALL, MRD status has emerged as an exceptionally valuable predictive measure [15]. Short et al. found that Ph+ ALL patients achieving MRD negativity by three months experienced substantially improved median overall survival (127 months versus 38 months) and relapse-free survival (126 versus 18 months) compared to those with inferior responses [15].
Besides survival metrics, treatment pathways now incorporate MRD status in Ph+ ALL decision-making. Current therapeutic strategies including tyrosine kinase inhibitors, targeted antibody therapies, CAR T-cell therapy, and hematopoietic cell transplantation increasingly rely on MRD status for optimization [15].
Recent data suggests that certain Ph+ patients who achieve complete molecular response may safely avoid transplantation. In a study of patients treated with hyper-CVAD plus ponatinib who did not undergo transplant in first remission, those achieving complete molecular response demonstrated an impressive 87% six-year overall survival in landmark analysis [9].
MRD levels and timing: early vs late clearance
The timing of MRD clearance proves vital in ALL prognostication. Patients can be categorized by their MRD clearance pattern into distinct prognostic groups. In one study, five-year overall survival rates were 64.75% for those negative after first induction, 52.15% for those clearing after second induction, 43.85% after third induction, and merely 24.32% for persistently positive patients [16].
Early clearance consistently predicts superior outcomes. Among patients with Ph-negative ALL, those achieving MRD negativity immediately after induction had three-year overall survival of 76%, whereas patients clearing MRD during later consolidation showed reduced survival of 58% [3].
Pre-transplant MRD status likewise influences post-transplant outcomes. The cumulative incidence of relapse ranges from approximately 10% in patients with undetectable pre-HSCT MRD to 30% with MRD between 1 × 10^-6 and 1 × 10^-3, and exceeds 50% with MRD above 1 × 10^-3 [3].
Ultimately, the growing adoption of MRD-guided treatment has shifted focus toward dynamic risk assessment throughout therapy. Rather than relying solely on baseline characteristics, treatment intensification decisions increasingly depend on how rapidly patients achieve MRD negativity – recognizing that early MRD response represents one of the strongest determinants of long-term cure.
MRD as a Predictor of Post-Treatment Relapse
Detecting minimal residual disease enables physicians to predict impending relapse months before clinical manifestations appear. This temporal advantage offers critical windows for intervention that may improve long-term outcomes for leukemia patients.
Correlation between MRD levels and relapse timing
The relationship between MRD positivity and subsequent relapse follows predictable patterns in most leukemic conditions. In a prospective analysis of standard-risk ALL patients by the German Multicenter Study Group, 28 of 105 patients (27%) converted to MRD positivity after achieving initial remission, with 17 of these 28 patients (61%) subsequently experiencing relapse [17]. The median interval between MRD detection and clinical relapse was 9.5 months, yet this timeframe shortened considerably to just 4.1 months for patients with MRD within the quantitative range of PCR [17].
MRD level directly influences relapse kinetics. Patients with higher MRD loads typically demonstrate more rapid progression to clinical relapse. In AML, relapse kinetics after transplantation vary based on the “functional class” of mutations and their stability [18]. Mutations in epigenetic modifier and splicosome genes exhibit greater stability before relapse and slower progression to clinical disease, whereas signaling pathway mutations (like FLT3-ITD) accelerate relapse timelines [18].
In essence, MRD positivity almost universally predicts poorer outcomes across hematologic malignancies, with MRD positive patients showing markedly reduced 5-year overall survival—34% versus 68% for MRD negative patients [19].
MRD monitoring post-transplant and post-CAR T therapy
Post-transplant MRD assessment provides vital information about relapse risk. Upon examining 935 AML patients who underwent transplantation, researchers found that 136 (15%) had MRD before transplant, yet merely 11 (1%) remained MRD positive at day +70 to +100 [7]. Nevertheless, pre-transplant MRD positivity maintained prognostic value even after conversion to MRD negativity—these patients experienced 3-year relapse rates of 40% versus 15% in consistently MRD negative patients [7].
Subsequently, MRD detection after CAR T-cell therapy demonstrates similar predictive power. In a study of 27 mantle cell lymphoma patients post-Brexucabtagene autoleucel (Brexu-cel), early MRD assessment effectively predicted outcome [10]. Patients with detectable MRD on day 28 had lower median progression-free survival (10.74 months) compared to those with undetectable MRD (17.69 months) [10]. Primarily, MRD predicted impending relapses approximately 294 days (range 15-332 days) before radiological evidence appeared [10].
This predictive value extends to ALL patients receiving tisagenlecleucel, where negative bone marrow MRD status at 3 and 6 months post-infusion correlated with reduced relapse risk [2].
Case studies: relapse despite MRD negativity
Certainly, not all relapses follow MRD detection. Among 77 consistently MRD negative ALL patients in the GMALL study, 5 patients (6%) still experienced relapse [17]. Upon examining 935 patients post-transplant who were alive without relapse at day +100, multiple factors influenced relapse despite MRD negativity, including specific genetic mutations [7].
The possibility of false negatives remains a challenge, with one study finding many MRD negative cases (36%) that proceeded to relapse [18]. This phenomenon occurs for several reasons:
- Limited sensitivity of testing methodologies
- Spatial heterogeneity of residual disease
- Clonal evolution below detection thresholds
- Potential for extramedullary relapse
Ultimately, even with modern techniques, MRD status captures a moment in time rather than guaranteeing long-term outcomes. One investigation demonstrated that the highest risk period for conversion from MRD negative to positive status occurs between 2 and 5 years post-treatment [19]. This highlights the need for continued vigilance, as achieving MRD negativity—while reassuring—does not eliminate relapse risk entirely.
Therapeutic Strategies for MRD Positive Patients
Targeted immunotherapeutic approaches now offer viable options for patients with MRD positive leukemia, shifting treatment paradigms toward personalized strategies based on disease burden and molecular characteristics.
Blinatumomab for MRD clearance in B-ALL
Blinatumomab demonstrates remarkable efficacy in eliminating minimal residual disease in B-ALL. In the GIMEMA LAL2317 protocol, this bispecific T-cell engager increased MRD negativity rates from 72% to 93% after administration, with 73% of previously MRD positive patients becoming MRD negative [20]. Patients receiving blinatumomab achieved impressive survival outcomes—the 3-year survival reached 82% [20]. Remarkably, survival and relapse rates differed based on subsequent therapy: 91% survival with 15% relapse for patients assigned to standard chemotherapy versus 59% survival with 35% relapse for those proceeding to hematopoietic stem cell transplantation [20].
Response rates vary based on clinical context. In patients with Ph-negative disease, blinatumomab produces 31% NGS MRD negativity, whereas Ph-positive patients show 64% response rates [1]. Inasmuch as disease status matters, first remission patients demonstrate 52% response rates compared to 0% in later remissions [1]. Achieving NGS MRD negativity translates into substantially better outcomes: 71% versus 25% two-year relapse-free survival and 100% versus 46% overall survival for responders versus non-responders [1].
Inotuzumab ozogamicin in MRD positive settings
Inotuzumab ozogamicin represents another potent option for MRD eradication. In a phase 2 trial, 69% of B-ALL patients with persistent MRD achieved MRD negativity [21]. Most compelling evidence demonstrates durable responses, with two-year relapse-free survival of 54% and overall survival of 60% [22]. Presently, inotuzumab shows effectiveness even after blinatumomab failure, offering treatment flexibility [21].
Administration advantages exist—inotuzumab requires only a one-hour weekly infusion compared to continuous infusion with blinatumomab [21]. Side effects primarily involve manageable low-grade adverse events, with sinusoidal obstruction syndrome occurring in 8% of patients [22].
CAR T-cell therapy for persistent MRD
CAR T-cell therapy effectively eliminates MRD in leukemia patients with suboptimal responses. Given that persistent MRD predicts relapse, this approach tackles disease at its molecular roots. Clinical trials report 90.7% MRD negativity achievement post-CAR-T infusion [8]. Actually, patients receiving CAR-T therapy experienced higher three-year leukemia-free survival than non-recipients (77.8% versus 51.1%) [8].
Prior to widespread adoption, several considerations warrant attention. First, among patients receiving CAR-T without subsequent transplant, 75% maintained remission at median 23-month follow-up [8]. Second, disease burden before CAR-T therapy influences outcomes, with MRD-guided timing potentially optimizing results [2]. Lastly, MRD assessment following CAR-T therapy provides important prognostic information regarding remission durability [2].
Limitations and Challenges in MRD Interpretation
Despite advances in MRD detection technologies, several critical challenges impact result interpretation, potentially affecting treatment decisions for leukemia patients.
False positives due to clonal hematopoiesis
Clonal hematopoiesis (CH) emerges as a key concern in MRD assessment, occurring when somatic mutations accumulate in blood cells with age. These mutations can masquerade as leukemia-associated changes, creating false positive results. Approximately 10% of prostate cancer patients exhibit CH interference in DNA repair genes [23]. Even more troubling, in a study of 16,812 patients with advanced cancer across 49 tumor types, 42.3% had CH mutations in clinically reportable genes including BRCA2, BRCA1, KRAS, and BRAF [23].
In particular, mutations in DNMT3A, TET2, and ASXL1 (known as DTA mutations) frequently persist after therapy yet do not correlate with adverse outcomes [24]. These mutations appear in approximately 10% of healthy individuals older than 65 years and 18.4% of those older than 90 years [24]. First and foremost, to overcome this challenge, paired genotyping of plasma cfDNA and peripheral blood cell DNA could screen out hematopoietic-derived mutations [25].
Sample quality and assay sensitivity issues
Sample collection techniques substantially affect MRD detection accuracy. Peripheral blood hemodilution and patchy bone marrow sampling may both result in failure to identify the tumor clone [26]. For optimal results, early-pull bone marrow aspirate should be used, ideally followed by CD138+ cell enrichment [26]. To illustrate, for the clonoSEQ assay, at least 1.9 million nucleated cells are required to provide 95% confidence of MRD negativity at 10^-6 sensitivity [3].
Simultaneously, the interpretation of results with MRD below the limit of detection presents a diagnostic conundrum—potentially representing either clinically relevant low-level disease or inconsequential background sequences [3]. For MFC assays, sample processing timing matters; specimens should be analyzed within 24-48 hours of collection, with 2-5 mL from the first pull of bone marrow aspirate recommended [27].
Standardization gaps across labs
Lack of standardization remains a persistent obstacle in MRD assessment. A PETHEMA study examining MRD testing in AML found striking heterogeneity among hospitals in:
- Number of markers used
- Tube combinations
- Methodological approaches (LAIP vs DfN vs both) [28]
The consequence of this variability is reduced prognostic value compared to studies performed at single centers [28]. In AML particularly, the absence of a universal immunophenotype increases difficulties in establishing standardized MRD assays [24]. MFC analyzes require high-level expertise and remain operator-dependent to some degree, hampering inter-laboratory harmonization [24].
Ultimately, these limitations emphasize the need for caution when interpreting MRD results. As testing methodologies continue to evolve, ongoing efforts through collaborative groups like the ELN MRD Working Party and EuroMRD aim to develop consensus recommendations and quality control for molecular MRD targets [19].
Future Directions in MRD Research and Technology
Emerging technologies continue to push the boundaries of MRD detection sensitivity, addressing fundamental limitations of conventional approaches.
Digital PCR and ultra-sensitive detection
Digital PCR revolutionizes MRD assessment through absolute quantification of target molecules. Unlike standard qPCR, this technology partitions samples into thousands of oil-water emulsion droplets, each hosting individual reactions. This approach yields remarkable advantages: improved standardization across laboratories, independence from normalization target fluctuations, and more straightforward result interpretation by clinicians [29]. The sensitivity reaches impressive thresholds—up to 10^-5 according to validation studies, often surpassing conventional qPCR [30]. In clinical applications, ddPCR has demonstrated effectiveness with BCR::ABL1 variants, achieving sensitivity between 10^-4.5 and 10^-5 [31]. Fundamentally, its universal applicability enables adoption across various tumor types [30].
Single-cell MRD profiling
Single-cell MRD assays combine flow cytometric enrichment with integrated DNA sequencing and immunophenotyping at cellular resolution. This methodology achieves approximately 0.01% sensitivity while concurrently deconvoluting clonal architecture and providing clone-specific immunophenotypic data [5]. Throughout remission samples, these techniques have detected sequence variants in 75-80% of patients [32]. Importantly, decreasing clonal diversity at remission correlates with longer relapse-free survival [32]. Unlike traditional approaches, single-cell profiling clearly distinguishes between residual leukemic clones, preleukemic populations, and normal precursors [5].
AI-assisted MRD interpretation tools
Artificial intelligence dramatically reduces MRD analysis time to approximately one minute per case [33]. Recent AI pipelines utilize cluster-informed downsampling, reducing flowcytometry files by 87% while preserving small MRD populations [34]. These systems demonstrate exceptional concordance with conventional analysis—100% positive agreement for MRD ≥0.01% and excellent quantitative correlation (R^2=0.92) [34]. The DeepFlow algorithm processes nearly one million events within seconds compared to minutes or hours with other tools [35]. By minimizing subjective interpretation elements, these platforms potentially increase accessibility of MRD testing across clinical laboratories [33].
Conclusion 
Measurable residual disease assessment has transformed leukemia management, establishing itself as a cornerstone of modern treatment protocols and prognostic evaluation. The stark contrast between outcomes for MRD positive versus negative patients underscores the critical importance of achieving MRD clearance across leukemia subtypes. Patients with MRD negative status consistently demonstrate superior survival metrics—sometimes doubling or even tripling the survival rates of their MRD positive counterparts. Additionally, the timing of MRD clearance plays a key role, with early negativity conferring substantially better outcomes than delayed clearance.
Detection methodologies continue to evolve, each offering distinct advantages. Flow cytometry provides rapid results with reasonable sensitivity, though next-generation flow approaches now rival molecular techniques. PCR-based methods remain the gold standard for many applications, particularly in ALL, where IG/TCR rearrangements offer reliable targets. Next-generation sequencing has further expanded detection capabilities, pushing sensitivity boundaries to unprecedented levels—one leukemic cell among a million normal cells can now be identified. This enhanced sensitivity allows clinicians to detect impending relapse months before clinical manifestation, thus creating crucial windows for intervention.
The prognostic power of MRD status transcends traditional risk classifications. MRD assessment after induction therapy often outweighs baseline genetic factors in predicting outcomes. Transplant decisions likewise increasingly depend on MRD status, with MRD positive patients deriving greater benefit from allogeneic transplantation compared to their MRD negative counterparts.
Therapeutic options for MRD positive patients have expanded dramatically. Blinatumomab demonstrates remarkable efficacy in eliminating minimal residual disease in B-ALL, converting many patients from MRD positive to negative status. Inotuzumab ozogamicin represents another potent option, while CAR T-cell therapy effectively targets persistent disease with promising durability.
Despite these advances, several challenges persist. Clonal hematopoiesis can lead to false positive results, sample quality issues may compromise detection accuracy, and standardization gaps across laboratories hinder result interpretation. Therefore, ongoing research aims to address these limitations through digital PCR, single-cell profiling, and AI-assisted interpretation tools.
MRD testing has undoubtedly revolutionized leukemia care, yet clinicians must recognize both its strengths and limitations. As technologies advance and become more accessible, MRD assessment will continue refining risk stratification, guiding treatment decisions, and ultimately improving outcomes for leukemia patients. The future of leukemia management rests on our ability to detect and eliminate these final traces of disease—perhaps the most crucial battle in achieving lasting remissions and potential cures.
Key Takeaways
MRD positive status dramatically impacts leukemia survival outcomes, with compelling research revealing critical insights for treatment decisions and patient prognosis.
- MRD positive patients face dramatically worse outcomes: 5-year survival drops from 68% (MRD negative) to just 34% (MRD positive), making MRD status a powerful predictor of relapse.
- Early MRD clearance is crucial for survival: Patients achieving MRD negativity within 1.5 months show 100% two-year survival versus only 38% for those remaining positive.
- Next-generation sequencing achieves unprecedented sensitivity: NGS detects one leukemic cell among one million normal cells (10^-6), enabling relapse prediction months before clinical symptoms appear.
- Targeted therapies effectively eliminate MRD: Blinatumomab converts 73% of MRD positive B-ALL patients to negative status, while CAR T-cell therapy achieves 90.7% MRD clearance rates.
- MRD status guides transplant decisions: MRD positive patients benefit significantly from allogeneic transplantation (HR 0.39), while MRD negative patients show minimal transplant advantage.
The integration of MRD assessment into routine clinical practice represents a paradigm shift toward precision medicine in leukemia care, enabling personalized treatment strategies based on molecular disease burden rather than traditional risk factors alone.
Frequently Asked Questions:
FAQs
Q1. What is considered MRD-positive in leukemia? MRD-positive typically refers to the detection of leukemic cells at levels of 0.01% (10^-4) or higher. This means finding at least 1 leukemic cell among 10,000 normal cells in a bone marrow or blood sample using sensitive detection methods.
Q2. How does MRD status impact survival rates in leukemia patients? MRD status affects survival outcomes. Patients who achieve MRD-negative status have much higher 5-year survival rates (around 68%) compared to MRD-positive patients (about 34%). Early MRD clearance is particularly crucial for improved survival prospects.
Q3. What are the latest technologies used for MRD detection? Next-generation sequencing (NGS) is at the forefront of MRD detection, offering unprecedented sensitivity of up to 10^-6 (one leukemic cell among a million normal cells). Other advanced methods include digital PCR and single-cell MRD profiling, which provide ultra-sensitive detection and detailed insights into residual disease.
Q4. How does MRD status influence transplant decisions in leukemia treatment? MRD status plays a crucial role in transplant decisions. MRD-positive patients generally benefit more from allogeneic stem cell transplantation compared to MRD-negative patients. For MRD-positive individuals, transplantation in first complete remission can offer substantial survival benefits.
Q5. What targeted therapies are effective for eliminating MRD in leukemia? Several targeted therapies have shown effectiveness in eliminating MRD. Blinatumomab has demonstrated high efficacy in converting MRD-positive B-ALL patients to MRD-negative status. CAR T-cell therapy is another powerful option, achieving MRD clearance in a high percentage of patients with persistent disease.
References:
[1] – https://ashpublications.org/blood/article/144/Supplement 1/1465/532757/Clearance-of-Very-Low-Levels-of-Measurable
[2] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10798764/
[3] – https://ashpublications.org/bloodadvances/article/9/6/1442/535272/Clinical-use-of-measurable-residual-disease-in
[4] – https://www.nature.com/articles/s41408-025-01298-6
[5] – https://pubmed.ncbi.nlm.nih.gov/37729414/
[6] – https://pmc.ncbi.nlm.nih.gov/articles/PMC6901875/
[7] – https://ashpublications.org/bloodadvances/article/9/3/558/518094/Measurable-residual-disease-as-predictor-of-post
[8] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8777073/
[9] – https://www.targetedonc.com/view/mrd-in-ph-all-how-do-you-measure-it-and-what-does-it-mean-
[10] – https://ashpublications.org/blood/article/142/Supplement 1/1673/502211/Post-CAR-T-Minimal-Residual-Disease-MRD-Monitoring
[11] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10000405/
[12] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10791633/
[13] – https://www.nature.com/articles/s41409-024-02493-y
[14] – https://ashpublications.org/bloodadvances/article/1/25/2456/15757/Minimal-residual-disease-in-adult-ALL-technical
[15] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11316691/
[16] – https://ashpublications.org/blood/article/140/Supplement 1/11850/492799/Effect-of-MRD-Level-after-Induction-Therapy-and
[17] – https://www.sciencedirect.com/science/article/pii/S0006497120519721
[18] – https://ashpublications.org/blood/article/144/3/245/516962/Transplant-MRD-and-predicting-relapse-in-AML
[19] – https://pmc.ncbi.nlm.nih.gov/articles/PMC12257026/
[20] – https://ashpublications.org/blood/article/145/21/2447/535764/Up-front-blinatumomab-improves-MRD-clearance-and
[21] – https://ashpublications.org/blood/article/143/5/417/498427/Phase-2-study-of-inotuzumab-ozogamicin-for
[22] – https://pubmed.ncbi.nlm.nih.gov/37879077/
[23] – https://www.carislifesciences.com/physicians/physician-tests/caris-assure/incidental-clonal-hematopoiesis/
[24] – https://pmc.ncbi.nlm.nih.gov/articles/PMC6893483/
[25] – https://aacrjournals.org/clincancerres/article/24/18/4437/80899/False-Positive-Plasma-Genotyping-Due-to-Clonal
[26] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7133460/
[27] – https://haematologica.org/article/view/10871
[28] – https://www.sciencedirect.com/science/article/abs/pii/S0740257023000333
[29] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9713561/
[30] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10225364/
[31] – https://www.nature.com/articles/s41598-024-57016-y
[32] – https://pubmed.ncbi.nlm.nih.gov/32150611/
[33] – https://ashpublications.org/bloodadvances/article/doi/10.1182/
bloodadvances.2025016126/547303/Artificial-Intelligence-Accelerates-the
[34] – https://pubmed.ncbi.nlm.nih.gov/40966417/
[35] – https://onlinelibrary.wiley.com/doi/full/10.1002/cyto.b.22116