MRD as a Compass: A New Framework for Post-Transplant Blood Cancer Care

In blood cancers, remission is no longer a simple question of whether cancer is present or absent; it can exist at levels too small to see, but still biologically meaningful. Minimal Residual Disease (MRD) exposes microscopic levels of residual cancer cells after treatment, sometimes as few as one malignant cell among a million. This level of sensitivity provides clinicians with insights beyond imaging and routine blood tests, allowing much earlier assessment of relapse risk and treatment effectiveness.

Across leukemia, lymphoma, and myeloma, studies consistently show that achieving MRD-negative status is associated with a lower risk of relapse and longer overall survival. In some settings, MRD-negative patients demonstrate nearly double the long-term survival rates compared with those who remain MRD-positive[1].

In this article we explore how MRD is detected, how it guides treatment decisions across blood cancers, and why linking MRD detection to immune-based strategies may be critical to preventing relapse, particularly after stem cell transplantation, where relapse remains a devastating outcome.

MRD and how it’s Detected

MRD is measured using highly sensitive techniques capable of identifying microscopic numbers of cancer cells based on their proteins, genetic signatures, or circulating DNA fragments. These technologies include multiparameter flow cytometry, next-generation sequencing (NGS), PCR-based molecular assays, and liquid biopsy approaches, each with its distinct strengths depending on the disease context.

Flow cytometry (a laser-based analysis that examines individual cells from blood or bone marrow samples) is particularly effective in acute lymphoblastic leukemia (ALL), where surface antigen patterns are stable, well characterized, and reliably distinguish cancer cells from normal ones. NGS (a method for detecting specific DNA mutations in cancer cells) is particularly well-suited to acute myeloid leukemia (AML), allowing clinicians to track defined mutations such as FLT3-ITD or NPM1 over time. PCR-based testing (which amplifies microscopic amounts of disease-specific DNA so they can be measured) remains the backbone of chronic myeloid leukemia (CML) monitoring, because the disease is driven by a single, well-defined genetic abnormality; this allows clinicians to track changes in disease burden with extraordinary precision over time. Liquid biopsies (minimally invasive blood tests that detect tumor-derived DNA shed into the bloodstream) have emerged over the past decade as a powerful diagnostic approach across hematologic malignancies, offering a less invasive alternative to repeated bone marrow biopsies for longitudinal disease monitoring.

This degree of precision offers clinicians greater confidence in treatment decisions. MRD results can inform whether therapy should be intensified, maintained, modified, paused, or whether a patient should proceed to stem cell transplantation. Patients themselves often describe MRD results as more reassuring and meaningful than imaging, because MRD confirms remission at a molecular level, not just a visual one[1, 2].

MRD as a Treatment Compass

In ALL, MRD does more than predict outcomes: it actively directs therapy. This is because decades of clinical data have established that low levels of MRD are strongly predictive of relapse in ALL, making early intervention both necessary and clinically meaningful. Consistent with this strong prognostic value, the immunotherapy blinatumomab is approved for individuals with ALL who are in remission but remain MRD-positive, targeting residual disease before overt clinical relapse occurs. In clinical studies, blinatumomab has been shown to eliminate detectable MRD in approximately 78% of treated individuals and has been associated with significant improvements in relapse-free survival[1]. Despite these advances, however, relapse remains a significant challenge, particularly in settings of compromised immune function, and because eliminating detectable MRD does not necessarily address the underlying biology that drives disease recurrence.

AML illustrates a different, but equally important, application of MRD. Rather than directing a specific therapy, post-treatment MRD status plays a central role in risk stratification, helping determine whether stem cell transplantation (SCT) is warranted and how intensive conditioning should be. Persistent MRD after treatment signals a higher risk of relapse and may justify more aggressive transplant-based approaches, whereas MRD negativity can support less invasive strategies. Following SCT, continued MRD monitoring enables earlier detection of molecular relapse and creates opportunity for pre-emptive intervention[2]. However, even with MRD-guided risk stratification, relapse remains common in AML, reflecting the limitations of transplant-based strategies and the persistence of the disease beyond current detection thresholds.

In multiple myeloma, MRD assessment is beginning to change long-term treatment strategies, with MRD negativity opening up the possibility of treatment de-escalation in selected settings. Several trials showing prolonged remission even when the therapy is reduced or paused[1]. By contrast, chronic lymphocytic leukemia (CLL) illustrates the limits of MRD as a universal marker: some therapies improve survival without achieving MRD negativity, underscoring that while MRD is a powerful tool, it must be interpreted in the context of disease biology and the treatment mechanism rather than applied uniformly across all blood cancers[1].

Why MRD Matters More in Blood Cancers

Blood cancers are unique because the malignant cells originate from the immune system itself. This creates a distinct biological challenge: immune cells must recognize and eliminate a mutated version of themselves. Many hematologic malignancies exploit immune checkpoints to survive, suppressing T-cell activity through inhibitory receptors such as PD-1 and Tim-3. In AML, exhausted T cells are particularly enriched in the bone marrow, where their function is further dampened by a local microenvironment that limits nutrient availability, increases inhibitory signaling, and favors leukemia survival over immune activation[3].

The tumor microenvironment (referring, in blood cancers, to the bone marrow and surrounding cellular niches) adds another layer of protection. For instance, in multiple myeloma, cancer-associated fibroblasts form a physical and biochemical barrier around malignant cells, releasing factors that impair immune function, including the activity of CAR-T cells (T cells collected from the patient and genetically engineered in the lab to target cancer cells). These mechanisms help explain why relapse can occur, even in patients who initially respond well to treatment[4].

Stem Cell Transplantation and the Power of Donor Immunity

SCT adds a critical immune dimension to MRD-guided cancer care. Beyond the initial reduction of cancer cell burden through chemotherapy or conditioning regimens, allogeneic SCT introduces donor immune cells capable of recognizing and eliminating residual malignant cells through a process known as graft-versus-leukemia (GvL) effect, the most effective path to long-term remission. MRD monitoring is central in this context, enabling detection of residual or re-emerging disease at the molecular level, often well before clinical relapse becomes apparent, and creating a window for proactive intervention.

Donor lymphocyte infusion (DLI) exemplifies MRD-guided immune modulation. In chronic myeloid leukemia (CML), DLI has successfully restored remission and MRD negativity following post-transplant relapse by reactivating donor immune responses. Similar strategies are being explored across other hematologic malignancies, although their effectiveness varies depending on disease biology and immune context.

At the same time, significant effort is focused on preserving the beneficial GvL effect while minimizing graft-versus host disease (GvHD), a complication in which donor immune cells attack the recipient’s own tissues. Approaches under investigation include regulatory T-cell (Treg) infusion, targeted modulation of immune signaling pathways, and refinement of conditioning regimens. In each case, MRD serves as a critical tool to guide timing, intensity, and patient selection for these interventions[5, 6].

The Challenge of Post-Transplant Relapse

Despite these advances in transplantation and MRD-guided monitoring, relapse after SCT remains one of the most difficult challenges in blood cancer care. Outcomes following post-transplant relapse of hematological malignancies are often dismal, and therapeutic options are limited.

This reflects the complex biology of relapse in the post-transplant setting. Residual malignant cells may persist below detection thresholds, evade immune surveillance, or exploit immunosuppressive microenvironments. At the same time, donor immune function can be compromised by exhaustion, prior therapies, or ongoing immunosuppression. Together, these factors limit the durability of existing strategies and underscore the need for approaches that go beyond chemotherapy and conventional targeted inhibitors.

LEUMUNA: Connecting MRD Detection to Immune Activation

LEUMUNA™ (ulodesine hemiglutarate, LR 09) is an oral metabolic immune checkpoint modulator in development by Helix BioPharma for patients who relapse with acute leukemia after allogeneic SCT. By inhibiting purine nucleoside phosphorylase (PNP), an enzyme responsible for the breakdown of guanosine (a naturally occurring building block of RNA), LEUMUNA increases the availability of guanosine within the immune environment and functionally enhances signaling through Toll-like receptor 7 (TLR7), an innate immune sensor that initiates inflammatory signaling and helps mobilize donor immune cells against residual leukemia. This activation promotes cytokine production and triggers a GvL response in the post-transplant setting.

Importantly, this mechanism is designed to address relapse, not simply minimal residual disease. Rather than acting through direct cytotoxicity or by targeting a specific leukemia antigen or mutation, LEUMUNA leverages innate immune sensing to engage donor-derived immune responses in settings where immune surveillance has failed and relapse has occurred. The mechanism builds on clinical experience with earlier PNP inhibitors like forodesine, which produced complete remissions in some post-transplant relapsed T-ALL patients[7]. In mouse models of B-cell ALL relapse conducted at the Fred Hutchinson Cancer Center, LEUMUNA has been shown to significantly reduce the risk of leukemia-related death by enhancing GvL activity, with no statistically significant increase in GvHD-associated death.

Looking Forward

MRD is redefining how remission is measured, how relapse risk is understood, and how therapy is timed across blood cancers. When combined with advances in immunology and transplantation science, MRD is helping shift care away from one-size-fits-all approaches toward treatment strategies that are increasingly informed by disease biology, immune context, and risk dynamics. The future of blood cancer care will integrate MRD testing, targeted therapies, immunotherapy, transplantation, and metabolic modulation into more coherent treatment pathways guided by disease biology and immune dynamics.

At Helix, our mission is to help accelerate this evolution. By advancing such as LEUMUNA and contributing to the scientific understanding of MRD-guided immune activation, we aim to expand options for patients facing post-transplant relapse, a setting where unmet need remains profound. Through continued research, collaboration, and engagement with the scientific community, we remain committed to supporting longer survival, deeper disease control, and more durable immune recovery.

References:

1. Chandhok NS, Sekeres MA. Measurable residual disease in hematologic malignancies: a biomarker in search of a standard. EClinicalMedicine. 2025 Jul 10;86:103348. doi: 10.1016/j.eclinm.2025.103348. PMID: 40666170; PMCID: PMC12257026.

2. Ngai LL, Kelder A, Janssen JJWM, Ossenkoppele GJ, Cloos J. MRD Tailored Therapy in AML: What We Have Learned So Far. Front Oncol. 2021;10:603636. Published 2021 Jan 15. doi:10.3389/fonc.2020.603636

3. Tan J, Yu Z, Huang J, Chen Y, Huang S, Yao D, Xu L, Lu Y, Chen S, Li Y. Increased PD-1+Tim-3+ exhausted T cells in bone marrow may influence the clinical outcome of patients with AML. Biomark Res. 2020 Feb 13;8:6. doi: 10.1186/s40364-020-0185-8. PMID: 32082573; PMCID: PMC7020501.

4. Duan J, Wang Y, Jiao S. Checkpoint blockade-based immunotherapy in the context of tumor microenvironment: Opportunities and challenges. Cancer Med. 2018 Sep;7(9):4517-4529. doi: 10.1002/cam4.1722. Epub 2018 Aug 7. PMID: 30088347; PMCID: PMC6144152.

5. Biernacki MA, Sheth VS, Bleakley M. T cell optimization for graft-versus-leukemia responses. JCI Insight. 2020 May 7;5(9):e134939. doi: 10.1172/jci.insight.134939. PMID: 32376800; PMCID: PMC7253012.

6. Pacini CP, Soares MVD, Lacerda JF. The impact of regulatory T cells on the graft-versus-leukemia effect. Front Immunol. 2024;15:1339318. Published 2024 Apr 22. doi:10.3389/fimmu.2024.1339318

7. Balakrishnan K, Verma D, O’Brien S, Kilpatrick JM, Chen Y, Tyler BF, Bickel S, Bantia S, Keating MJ, Kantarjian H, Gandhi V, Ravandi F. Phase 2 and pharmacodynamic study of oral forodesine in patients with advanced, fludarabine-treated chronic lymphocytic leukemia. Blood. 2010 Aug 12;116(6):886-92. doi: 10.1182/blood-2010-02-272039. Epub 2010 Apr 28. PMID: 20427701; PMCID: PMC2924226.

Jacek Antas

Chief Executive Officer


Jacek Antas is a shareholder of the Company, has spent more than 25 years in the financial services industry holding various positions in sales and consulting.

Mr. Antas obtained a master’s degree from the Warsaw School of Economics and has served as a board member of various
companies throughout his career.

This will close in 0 seconds

James B. Murphy

Chief Financial Officer


Mr. Murphy is a certified public accountant with over thirty years of experience in finance and operations management. He is currently a consultant with Danforth Advisors LLC (“Danforth”), a leading provider of outsourced strategic and operational specialists across functions in the life sciences industry. While at Danforth, Mr. Murphy has served over fifteen private and publicly held life sciences companies as CFO and CFO Advisor, helping them secure over USD 0.5 billion in financing and successfully execute pivotal asset transactions. Mr. Murphy functions as a consultant to Helix pursuant to a consulting agreement between the Company and Danforth.

This will close in 0 seconds

Thomas Mehrling

Medical Adviser


Thomas Mehrling (PhD in Pharmacology and MD) has over 20 years’ experience in multinational Pharma companies developing novel oncology compounds from preclinical research through to registration. Prior to entering the industry, he spent 13 years as an MD at the University Hospital in Frankfurt, working on preclinical and translational projects. He served as Director of European Oncology at Mundipharma International (2003–2013), building the company’s first European oncology business from the ground up out of Cambridge, UK, and completing the clinical development, registration and launch of two major products in Europe, DepoCyte® and Levact® (Ribomustin® and Treanda®). In 2013, he led the establishment of the Mundipharma Group’s start-up, Mundipharma EDO, developing anti-cancer therapeutics for solid tumours out of Basel, Switzerland.

This will close in 0 seconds

Kim Gaspar

Director Quality Assurance


Kim is the Director of Quality Assurance at Helix BioPharma Corp. An experienced quality assurance professional with expertise in Canadian, US, and EU regulations, she has been involved in all aspects of Phase I/II biopharmaceutical product development over the years, including regulatory submissions, QC laboratory compliance, tech transfer and third-party oversight of CMC activities, clinical QA, and bioanalytical data analysis. Kim joined Helix in 2000, transitioning into QA in 2003. She holds a B.Sc in Biochemistry and a Ph.D in Veterinary Physiological Sciences, both from the University of Saskatchewan.

This will close in 0 seconds

Brenda Lee

Director Clinical Operations


Brenda is the Clinical Operations Director at Helix Biopharma Corp. A clinical research operations professional with 25 years of experience managing clinical trials, ranging from early Phase I to late Phase IIIb/IV studies, she brings experience in clinical study protocol writing and development, trial start-up and vendor management, and a proven track record in planning and managing clinical trials to quality standards, timelines and budget. Brenda joined Helix Biopharma Corp. in 2018, working to advance the clinical program of L-DOS47. She holds B.Sc and M.Sc. degrees from the University of Toronto, specializing in Nutritional Sciences and Human Biology.

This will close in 0 seconds

Jerzy Leszczynski

Director


Jerzy Leszczynski is a shareholder of the Company, has spent more than 35 years developing businesses and has served in the capacity of board member of various real estate development companies. Mr. Leszczynski obtained his Master of Science in Chemistry from the Warsaw Institute of Technology.

This will close in 0 seconds

Janusz Grabski

Director, Chair of Audit Committee


Janusz (John) Grabski is a lawyer specialized in corporate and real estate law with over twenty years of experience.

This will close in 0 seconds

Malgorzata Laube

Director


Malgorzata Laube has over 19 years of experience in nuclear medicine. In her last role with Alberta Health Services, she was the Department Supervisor, Nuclear Medicine at Royal Alexandra Hospital. Ms. Laube obtained a MSc degree in Environmental Engineering from the Warsaw University of Technology and is based in Edmonton, Alberta, Canada.

This will close in 0 seconds

Jacek Antas

Chairman of the Board


Jacek Antas is a shareholder of the Company, has spent more than 25 years in the financial services industry holding various positions in sales and consulting.

Mr. Antas obtained a master’s degree from the Warsaw School of Economics and has served as a board member of various
companies throughout his career.

This will close in 0 seconds

Jonathan Davis

Advisor, ADC Discovery


Jonathan Davis received his Ph.D. from University of California, San Francisco, where he studied protein structure and function using NMR. After a post-doc at Harvard Medical School exploring RNA selection and structure in the labs of Jack Szostak and Gerhard Wagner, he went to work at EMD Serono, where his work involved improving antibody-based therapeutics, inventing a platform technology for generating heterodimeric Fcs as a basis for multifunctional molecules, and developing a novel scaffold based on an artificially-designed protein from David Baker’s lab. In 2008 he took a job at Bristol-Myers Squibb in Waltham/Cambridge MA, working on antibody discovery and platform development in a wide range of therapeutic areas, with a particular focus on multispecific therapeutics. He moved to Madison, WI in 2019 to take on the role of VP of Innovation and Strategy at Invenra, a biotech focused on bispecific antibodies, and where he is currently head of the Scientific Advisory Board. In early 2024 he left the corporate world to found Creative Antibodies, a consulting firm that helps guide companies to successful antibody discovery and development projects, from mAbs to multispecifics, ADCs, and other formats. Outside of science, Jonathan is a conservatory trained cellist, plays numerous other instruments, and founded the UCSF Orchestra (now Symphony Parnassus) in San Francisco, where he was Music Director for six years.

This will close in 0 seconds

Davide Guggi

Advisor, CMC


Davide graduated as a pharmacist and received his PhD in Pharmaceutical Technology and Biotechnology from the University of Vienna. He has over 20 years of experience in the pharmaceutical industry, principally in the field of oncology. At the beginning of his career, Davide led oncology business units and commercial departments at Mundipharma and Gilead across Austria and Eastern Europe. Since over 10 years he has been working as a CMC expert, covering operational and regulatory CMC functions on behalf of over 20 different small- and medium-sized biotech companies across the world. He has served as CMC Director and CSO/CTO for several years, developing both small molecules and biologics (mABs, Fab, ADCs and Radio-immuno-conjugates) from early discovery to NDA/BLA in the US, EU and Canada, with a focus on First-in-Human and Phase I/II studies in oncology indications.

This will close in 0 seconds

This will close in 0 seconds

Tumor Defense Breaker™, L-DOS47


L‑DOS47 is a first‑in‑class, clinical-stage antibody‑enzyme conjugate designed to deliver a game-changing assist to anti-cancer immunity and today’s leading cancer immunotherapies for the treatment of prevalent, hard-to-treat solid tumors. The compound precisely targets CEACAM6, a cell-surface protein overexpressed in non‑small cell lung cancer (NSCLC) and other aggressive tumors, where it delivers an enzymatic payload that raises the extracellular pH of the acidic tumor microenvironment (TME). By neutralizing tumor acidity, L-DOS47 restores immune cell infiltration and activity, helps turn immunologically “cold” tumors “hot”, and enhances the therapeutic reach of immune checkpoint inhibitors. With patented composition-of-matter coverage through 2036 and demonstrated synergy with PD-1 inhibitor, pembrolizumab, L-DOS47 is poised to significantly increase the efficacy of immune checkpoint blockade and unlock broader and more durable responses in NSCLC and other aggressive solid tumors.

This will close in 0 seconds

LEUMUNA™


LEUMUNA™ is an oral immune checkpoint modulator designed to activate the donor immune system to recognize and fight relapsing leukemia in patients who have undergone allogeneic stem cell transplantation (allo-SCT). Although a life-saving procedure, up to 30% of patients who undergo allo-SCT see their cancer return, facing a median survival of just four months. LEUMUNA aims to offer these patients a new lease on life, by activating an immune cascade and inciting graft-versus-leukemia (GvL) effect, potentially offering long-term remission. Backed by strong preclinical data and a promising safety record from trials with its precursor compound, ulodesine, LEUMUNA offers a patient‑friendly, oral approach to a difficult-to-treat condition, with patent protection through 2041 and an Orphan Drug Designation granted by the US FDA.

This will close in 0 seconds

GEMCEDA™


GEMCEDA is a first-in-class oral prodrug of gemcitabine that opens up the possibility for convenient at-home administration, metronomic dosing and seamless integration into combination regimens with immune checkpoint inhibitors. To date, gemcitabine is only administered intravenously because oral forms have shown poor bioavailability of about 10%. GEMCEDA was developed as a prodrug to enable new uses of gemcitabine by combining it with cedazuridine, an enzyme inhibitor that helps boost its bioavailability to 90%. This remarkable innovation allows for greater flexibility in dosing schedules, fewer clinic visits, and a better quality of life, while achieving bioavailability on par with intravenous gemcitabine. Supported by a well‑established safety profile, scalable manufacturing, and patent coverage to 2043, GEMCEDA reimagines how chemotherapy can fit into patients’ lives.

This will close in 0 seconds