If you have trouble reading this newsletter, view the web version here

   

IACH NEWS OF THE WEEK

Highlights of the 51st EBMT Annual Meeting, held in Florence, Italy, from March 30 to April 2, 2025.

April 17, 2025
Prepared by Dr Edwin Uriel Suárez

In sunny Florence, hematology had a confluence, this time to expose and review the most relevant topics of hematopoietic transplantation and cell therapy. Here is a summary of some of the talks.

Use of machine learning techniques to predict poor survival after hematopoietic cell transplantation for myelofibrosis [Chronic Malignancies Working Party Session].  

Highlights: 

●      This collaborative group presented data from an interesting study that reflects the utility of artificial intelligence in hematology.

●      A machine learning (ML) model, available as an interactive web application, was created to predict survival after transplant in myelofibrosis (MF).

●      This tool identifies 25% of patients with poor transplantation outcomes.


Using data from 5,183 MF patients who underwent first allogeneic hematopoietic cell transplantation (allo-HCT) between 2005 and 2020 at European Society for Blood and Marrow Transplant (EBMT) centers, the authors examined different ML models to predict overall survival (OS) after transplant. The cohort was divided into a training set (75%) and a test set (25%) for model validation. A Random Survival Forests (RSF) model was developed based on 10 variables: patient age, comorbidity index, performance status, blood blasts, hemoglobin, leukocytes, platelets, donor type, conditioning intensity, and graft-versus-host disease prophylaxis. Its performance was compared with a four-level Cox regression-based score and other ML-based models derived from the same dataset, as well as with the Center for International Blood and Marrow Transplant Research (CIBMTR) score. The RSF outperformed all comparators, achieving better concordance indices across both primary and post-essential thrombocythemia/polycythemia vera MF subgroups.

A web application based on the RSF model offers a practical and powerful tool to identify patients at high risk for poor transplantation outcomes. Its primary objective is to inform and enhance clinical decision-making by offering a clearer understanding of risk profiles.

Click for the full article

The Phase 2 STARGAZE Trial of the Glucagon-like Peptide 2 (GLP-2) Analog Apraglutide in Combination with Ruxolitinib for Steroid-Refractory Gastrointestinal (GI) Acute Graft-Versus-Host Disease (aGvHD): Comparisons with a MAGIC Control Cohort  [Presidential Symposium].

Highlights: 

●      In preclinical studies, glucagon-like peptide 2 (GLP-2) stimulated intestinal stem cells and Paneth cells in damaged tissue, reduced graft-versus-host disease (GvHD) severity, and improved survival.

●      Interim results of the phase 2 STARGAZE trial (NCT05415410) showed that adding the selective GLP-2 analog apraglutide (APRA) to ruxolitinib (RUX) was well tolerated and effective in patients with steroid-refractory (SR) lower- gastrointestinal (GI) acute (a) GvHD.

●      STARGAZE short- and long-term interim results compared favorably to a MAGIC control cohort.


The data showed that the all-organ overall response rates were higher in the STARGAZE cohort (n = 31; APRA vs.  RUX) vs. the MAGIC cohort (n = 31; RUX monotherapy) at days 28 (STARGAZE, 58.1%; MAGIC, 38.8%), 56 (45.1%; 32.2%), and 91 (45.1%; 25.8%). The complete response rates at these respective time points were also higher in STARGAZE vs. MAGIC. More patients with SR lower-GI aGvHD treated with RUX + APRA had a response at day 28, more patients maintained their response over time, and fewer patients experienced day 180 non-relapse mortality compared with RUX alone.

Click for the full article

EBMT consensus on allogeneic transplantation in T-cell lymphoma [Lymphoma Working Party Session].  

●      Angioimmunoblastic T-cell lymphoma (AITL), Peripheral T-cell lymphoma not otherwise specified (PTCL-NOS), Anaplastic large cell lymphoma (ALCL)-ALK* negative:

○      1st line of treatment (e.g., CHOP/CHOEP/BV-CHP).

■      If complete response (CR):  clinical trials and/or autologous hematopoietic cell transplantation (auto-HCT); *including high-risk ALK-positive ALCL.

■      If no-CR and fitness for allogeneic hematopoietic cell transplantation (allo-HCT): donor search and 2nd line of treatment (bridge to allo-HCT).

●      CR, partial response (PR), stable disease (SD): allo-HCT.

●      Progressive disease (PR): 3rd line of treatment (bridge to allo-HCT). 


●      Hepatosplenic T-cell lymphoma:

○      Fitness patient: donor search.

○      1st line of treatment (e.g., ICE, IVAC).

■      Chemo-sensitive (CR, PR): allo-HCT consolidation.

■      Chemo-refractory (SD, PD): clinical trials, 2nd line of treatment (e.g., GEMOX, DHAX).

●      CR, PR, SD: allo-HCT consolidation.

●      PD: to be considered for allo-HCT.


●      Extranodal natural killer (NK)/T-cell lymphoma:

○      Fitness patient: donor search.

○      1st line of treatment (e.g., P-GEMOX, DDGP, anti-PD1, PD-L1-based).

■      Chemo-sensitive:

●      CR: auto-HCT consolidation.

●      PR and SD: allo-HCT consolidation.

■      Chemo-refractory: PD.

●      Clinical trials, 2nd line of treatment.

○      CR, PR, SD: allo-HCT

○      PD: to be considered allo-HCT


●      Enteropathy-associated T-cell lymphoma:

○      Fitness patient: donor search.

○      1st line of treatment (e.g., CHOP, CHOEP, ICE/M, BR-CHP).

■      CR: auto-HCT consolidation.

■      PR, SD, PD: clinical trial, 2nd line of treatment.

●      CR, PR, SD: allo-HCT

●      PD: to be considered for allo-HCT.


●      Does histology impact post-allo-HCT outcome? The histological subtype should not be a determining decision-making factor.


●      Pre-transplant considerations:

○      The preferred donor hierarchy is matched (matched sibling donor > matched unrelated donor > haploidentical donor  and miss-matched unrelated donor).

○      Graft sources: peripheral blood and bone marrow acceptable.

○      Graft-versus-host disease prophylaxis should be guided by  the degree of human leukocyte antigen-matching, the type of conditioning, and the source of stem cells.


●      Conditioning regimens:

○      There is no clear superiority of myeloablative conditioning over reduced-intensity conditioning (RIC) and vice-versa.

■      RIC is the preferred choice: broaden the accessibility to allo-HCT.

■      Advantage of the potentially lower non-relapse mortality.

○      Centers should stay with the locally established RIC regimens.


●      Post-transplant maintenance:

○      No data to support routine post-transplant interventions.

○      Routine minimal residual disease monitoring is not recommended.

○      There is no data supporting prophylactic/pre-emptive donor lymphocyte infusion use in PTCL.

Non-ICANS Neurological Complications after CAR-T-cell therapies: EBMT Practice Harmonization and Guidelines Committee [Transplant Complications Working Party Session].

●      Movement and neurocognitive toxicity:   

○      Associated with B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapy. 

○      Delayed onset, typically after the resolution of cytokine release syndrome and immune effector cell–associated neurotoxicity syndrome (ICANS).

○      Progressive course.

○      Movement disorder symptoms: bradykinesia, micrographia, tremor, cogwheel rigidity, motor dysfunction, speech disturbances, and gait changes.

○      Cognitive symptoms: reduced attention or memory impairment.

○      Personality change symptoms: reduced facial expression, apathy, or flat affect.

○      Normal dopamine active transporter scan. 

○      Variable results in brain magnetic resonance (MRI) image and [¹⁸F]fluorodeoxyglucose- positron emission tomography.

○      Associated with the presence of CAR T-cells in the blood or cerebrospinal fluid.

●      Cranial nerve palsy:  

○      Commonly associated with BCMA CAR T-cell therapy, less frequently with CD19. CAR T-cells.

○      Early onset after infusion.

○      The most common presentation is facial nerve palsy (seventh cranial nerve).

○      Can be unilateral or bilateral.

○      Associated with higher CAR T-cell expansion and exposure amounts.

●      Tumour inflammation-associated neurotoxicity: 

○      Localized neurotoxicity following CAR T-cell therapy for central nervous system tumors.

○      Clinical manifestations depend on tumour location. 

■      Type 1: signs of raised intracranial pressure (e.g., headache and herniation syndromes).

■      Type 2: transient worsening of pre-existing neurological symptoms.  Brain or spinal MRI (or both) required.

●      Ischaemic stroke: 

○      Mostly described after CD19 CAR T-cell therapy.

○      It can be asymptomatic.

○      Computational tomography with angiography is needed to rule out cranial vessel occlusion.

●      Associated with current or previous severe ICANS: 

○      Myelopathy.

○      Mostly described after CD19 CAR T-cell therapy. 

○      Occurs with or shortly after the onset of ICANS.

○      Acute presentation but can be unrecognized in severely ill patients.

○      Brain and spinal MRI with and without gadolinium and lumbar puncture.

●      Peripheral neuropathy and Guillain–Barré syndrome: 

○      Described in CD19-directed and BCMA-directed CAR T-cell therapies.

○      Nerve root enhancement in spinal MRI can be seen.

●      Fludarabine-associated neurotoxicity:  

○      Delayed onset after high-dose fludarabine conditioning regimens. 

○      Rapidly progressive syndrome starts with visual disturbances, progressing to quadriparesis, dementia, peripheral neuropathy, blindness, coma, and death.

○      Brain MRI shows acute toxic leukoencephalopathy.


See Figure 2 and Figure 3 in the original publication for flow diagrams for the management.

Click for the full article

Role of allo-HCT in ‘non-classical’ MPNs and MDS/MPNs: recommendations from the PH&G Committee and the CMWP of the EBMT [Chronic Malignancies Working Party Session].  

‘Non-classical’ Myeloproliferative Neoplasms (MPNs) and Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPNs) represent a heterogeneous group of malignancies characterized by a wide range of clinical manifestations. Unlike classical MPNs, there is no standardized management approach for these conditions, particularly concerning the indications for and management of allogeneic hematopoietic cell transplantation (HCT).


●      Chronic neutrophilic leukemia (CNL): 

○      All CNL patients should be assessed early following diagnosis for potential transplant eligibility and donor search.

○      Treatment aimed at optimizing disease control (control of leukocytosis, reduction in splenomegaly where relevant) is recommended as a ‘bridge’ prior to transplant, balancing the risks and benefits.

○      Given limited data, no recommendations on optimal transplant conditioning regimens and post-transplant disease monitoring and maintenance can be made. However, consideration needs to be given to considerable relapse rates and approaches tailored accordingly.

●      Chronic eosinophilic leukemia (CEL):

○      Given the poor prognosis of CEL (World Health Organization [WHO]) / CEL non otherwise specified ([NOS] International Consensus Classification [ICC])  and the risk of organ dysfunction, all patients should be considered for HCT at diagnosis with a prompt donor search.

○      Careful cardiac and pulmonary function assessment is advised for transplant eligibility and then for tailoring the transplant platform.

○      Given the lack of disease-modifying agents, no recommendation can be made on pre-treatment strategies.

○      Transplantation should not be delayed once a suitable donor is found.

●      Myeloid/lymphoid neoplasms (MLN) with eosinophilia and tyrosine kinase (TK) gene fusions: 

○      In patients with PDGFRA/B-rearranged MLN, both in chronic and blast phases, HCT is only considered after failure of TK inhibitor (TKI) treatment. However, in young patients (<60 years) presenting with blast-phase disease, HCT could be considered upon achieving a response.

○      Careful assessment of cardiac and pulmonary function is advised for transplant eligibility and then for tailoring transplant platforms.

○      HCT, after bridging with an alternative TKI with or without chemotherapy, seems to be the preferred option for the rare cases of MLN-PDGFRA/B with secondary resistance to imatinib.

○      HCT with donor search should be considered early after diagnosis for most eligible patients with FGFR1, JAK2, ABL1, and FLT3-rearranged MLN, given the low predictability and uncertain durability of responses to TKIs. TKI treatment directed to the specific molecular abnormality is recommended to decrease disease burden pre-HCT.

○      A thorough evaluation of cardiac and pulmonary function is essential, given the possible organ impairment associated with prior/ongoing eosinophilic infiltration.

○      The HCT strategy should be tailored to the predominant clinical features of the disease.

○       Monitoring of the underlying molecular abnormality using sensitive techniques is advised to inform treatment strategies to prevent overt disease relapse.

○      The role of TKI maintenance after HCT warrants investigation.

●      MPN, NOS (WHO)/ MPN-unclassified (ICC):  

○      Given disease heterogeneity, therapeutic approaches to MPN, NOS should be discussed in centers with expertise in MPN management.

○      In transplant-eligible MPN, NOS patients, HCT can be considered for those patients at higher risk according to standard MF-derived prognostic systems.

○      It is recommended that standard guidelines for MF pertaining to transplant are applied.

●      MDS/MPN with neutrophilia (WHO)/ atypical chronic myeloid leukemia (ICC): 

○      It is recommended that eligible patients are considered for transplant early after diagnosis with a prompt donor search.

○      No recommendations can be made on optimal transplant conditioning and post-transplant disease monitoring and maintenance.

●      MDS/MPN with SF3B1 mutation and thrombocytosis (WHO) / MDS/MPN with thrombocytosis and SF3B1 Mutation (ICC) & MDS/MPN with ring sideroblasts and thrombocytosis, NOS (ICC only)

○      Transplant should be considered in high risk eligible patients (e.g., refractory anemia, adverse cytogenetics, and/or presence of ASXL1 or SETBP1 mutations) in both MDS/MPN with thrombocytosis with or without SF3B1 mutation, with a prompt donor search.

○      It is recommended that standard guidelines for MDS pertaining to transplant platform are applied.

●      MDS/MPN, NOS: 

○      It is recommended that eligible patients are considered for transplant early after diagnosis, with a prompt donor search.

○      It is recommended that standard guidelines for chronic myelomonocytic leukemia (CMML) pertaining to transplant platform are applied.

Click for the full article

Update on the recommendations of the 10th European Conference on Infections in Leukaemia (ECIL) [Infectious Diseases Working Party Session].

➔    Viral infections: 

◆     Cytomegalovirus (CMV). Allogeneic stem cell transplantation (allo-HCT). 

●      No recommendation can be given regarding which viral load cut-off should be used to switch to preemptive therapy (PET).

●      Letermovir (LMV) is recommended as the strategy of choice for preventing CMV for CMV primary prophylaxis for CMV seropositive adult allo-HCT recipients.

●      There is no controlled data to support primary LMV prophylaxis in patients with CMV-negative status. Regardless of the donor serostatus and it is not recommended.

●      Sequential monitoring of Interferon (IFN)-γ-producing CMV-specific T-cells may provide potentially useful information for managing CMV infection after allo-HCT, which may be used to personalize PET.

●      Extended prophylaxis should be considered in patients at high risk for CMV disease and can continue for at least 200 days after transplantation.

●      For some individuals, prophylaxis for longer than 200 days after transplantation can be considered if, in the treating physician’s judgment, the benefit is stronger than the risk.

●      So called LMV blips (single test low level DNA positivity in plasma or whole blood samples occurring especially early during LMV prophylaxis) are common, and it is not recommended to interrupt LMV  prophylaxis unless there are repeated positive samples showing increased viral load.

●      After discontinuation of prophylaxis, “secondary” prophylaxis with LMV can be considered in the following situations:

○      After successful treatment (negative quantitative nucleic acid testing [qNAT]) of a CMV reactivation in patients perceived to be at increased risk for CMV disease.

○      In patients who, for some reason, have not received primary prophylaxis and who have reactivated CMV that has been successfully treated.

●      LMV is not indicated for the treatment of CMV reactivations or disease due to the high risk for resistance development and possible underdosing since a treatment dose has not been determined.

●      Either intravenous (IV) ganciclovir or foscarnet can be used for first line preemptive therapy. Valganciclovir can be used instead of IV ganciclovir or foscarnet (except in patients with severe gastrointestinal graft-versus-host disease [GVHD]).

●      Maribavir can be considered in patients with neutropenia or renal function impairment not appropriate for therapy with valganciclovir or foscarnet.

●      The choice of drug depends on time after transplant, risk of toxicity, and previous antiviral drug exposure but is not influenced by whether a patient has received LMV prophylaxis.

●      Genotyping should be performed in any case of non-response to allow adjustment of further therapy.

●      Repeated genotyping is recommended if the viral load does not improve within two weeks of appropriate therapy.

●      Genotyping results should be combined with clinical interpretation to guide clinical decisions.

●      Maribavir is not indicated for CMV disease involving the central nervous system (CNS) and the eyes.

●      Foscarnet is an alternative therapy for refractory/resistant (R/R) CMV infections, in particular in the CNS and eyes, but is associated with significant toxicity. 

●      Cidofovir is an option for the treatment of CMV retinitis.

●      CMV-specific T-cells are an option for treating R/R CMV infection/disease, if available.

●      Combination therapy for R/R CMV infections could be considered. The combination of maribavir and val(ganciclovir) should not be used.


◆     CMV. Chimeric antigen receptor (CAR) T-cell therapies. 

●      CMV monitoring is only required in patients being CMV-seropositive before CAR T-cell therapy.

●      In CMV-seropositive patients, a viral load determination should be performed before the start of lymphodepletion.  If the tests show evidence of CMV replication, close monitoring is recommended.

●      Risk factors for CMV reactivation after CAR  T-cell therapy are cytokine release syndrome (CRS) grade 3-4, receiving corticosteroids >3 days, persistent lymphocytopenia < 200/µl, or receiving ≥2 immunosuppressants. Such patients should be regarded as high-risk patients for CMV reactivation.

●      Active monitoring for CMV-DNA testing should be considered between 2 and 6 weeks after cell infusion in high-risk CAR T-cell recipients.

●      Preemptive antiviral treatment could be considered in case of “high level”/rapidly increasing level of CMV-DNA-emia.

●      It is currently unclear whether CAR T-cells directed against different antigens have the same risk for CMV reactivation, so the same strategy should be employed.

●      LMV prophylaxis is not recommended.


◆     CMV. T-cell-engaging antibodies. 

●      Currently, there is a lack of good data allowing risk assessment regarding CMV reactivation and CMV disease in this patient population. The recommendations should, therefore, be seen as provisional.

●      CMV testing is only required in patients being CMV-seropositive before treatment with bispecific antibodies.

●      In CMV-seropositive patients, a viral load determination could be performed before therapy with bispecific antibodies.

●      If the tests show evidence of CMV replication, close monitoring is recommended.

●      Risk factors for CMV reactivation after T-cell engaging therapy are CRS grade ≥ 2, corticosteroids >3 days, or combination therapy with Anti-CD38 antibodies, immunomodulators (IMiDs), and proteasome inhibitors. Such patients should be regarded as high-risk patients for CMV reactivation.

●      Testing for CMV-DNA-emia could be considered in febrile patients who have received bispecific antibodies for > 4 weeks.

●      Antiviral treatment could be considered in cases of symptoms and a “high level”/rapidly increasing level of CMV-DNA-emia.


➔    Antifungal prophylaxis. 


◆     Recommendations in allo-HCT recipients: pre-engraftment 

●      No significant changes, only:

1. The introduction of isavuconazole (can be used as second-line mould active prophylaxis, in case of intolerance to posaconazole/voriconazole, or QTc prolongation). Based on observational studies.

2. Haplo-HCT with cyclophosphamide post-transplant should be considered “low risk.” “High risk” includes active leukemia, cord blood transplantation, and unrelated donors.

●      Emphasis on monitoring azole levels (posaconazole, itraconazole, voriconazole).


◆     Recommendations in allo-HCT recipients: post-engraftment 

●      The same change is the introduction of isavuconazole.


◆     CAR T-cell. Panel proposes to endorse these recommendations 


●      Mold-active prophylaxis in: 

○      Pre-infusion neutropenia, invasive mold infection within 6 months, recent allo-HCT, acute leukemia.

○      Post-infusion grade ≥3 CRS/immune effector cell–associated neurotoxicity syndrome, OR  ≥7 days of corticosteroids (≥0.1mg/kg dexamethasone or equivalent), OR ≥3 weeks of neutropenia, OR other immunosuppressive agent.

●      Yeast-active prophylaxis and consider pre-emptive monitoring for molds (pre-emptive therapy consists of diagnostics such as fungal biomarkers and surveillance radiographic imaging).

 

➔    Neutropenic patient with severe sepsis. 


◆     Revision of recommendations for empirical antibiotic therapy: Escalation approach. 

●      Uncomplicated presentation

●      No known colonization with resistant bacteria

●      No previous infection with resistant bacteria

●      In centers where infections due to resistant pathogens are rarely seen at the onset of febrile neutropenia

●      Options for initial antibiotic therapy:

○      Anti-pseudomonal cephalosporin (cefepime, ceftazidime; avoid if extended-spectrum beta-lactamases are prevalent).

○      Piperacillin-tazobactam

○      Other possible options include cefoperazone-sulbactam or piperacillin plus gentamicin.


◆     Revision of recommendations for empirical antibiotic therapy: De-escalation approach. 

●      Sepsis/septic shock

●      Known colonization with resistant bacteria

●      Previous infection with resistant bacteria

●      In centers where resistant pathogens are regularly seen at the onset of febrile neutropenia.

●      Options for initial antibiotic therapy:

○      Carbapenem monotherapy.

○      Combination of anti-pseudomonal beta-lactam + aminoglycoside (e.g., sepsis/septic shock, pneumonia, local epidemiology, previous use of carbapenems within 30 days).

○      Beta-lactam targeting the suspected colonizing pathogen based on susceptibility testing. Situations for which novel anti-Gram-negative beta-lactams are indicated as the empirical regimen:

a.     KPC-producers: ceftazidime-avibactam, meropenem-vaborbactam, imipenem-cilastatin-relebactam, cefiderocol.

b.     OXA-48-producers: ceftazidime-avibactam, cefiderocol.

c.     MBL-producers: ceftazidime-avibactam plus aztreonam, cefiderocol.

d.     Difficult-to-treat Pseudomonas aeruginosa: high dose ceftolozane-tazobactam, ceftazidime-avibactam, imipenem/cilastatin/relebactam, cefiderocol.

-        Screening for resistant bacteria should be performed in high-risk setting.

-         Coverage against invasive streptococcal infections should be considered if antibiotics with limited activity against Gram-positive organisms are used (e.g., ceftazidime with or without avibactam or cefiderocol).  


◆     Addition of anti-Gram-positive agents. 

●      Hemodynamic instability, or other evidence of sepsis, septic shock or pneumonia in patients (especially in those with known methicillin-resistant Staphylococcus aureus [MRSA] colonization).

●      Colonization with MRSA.

●      Suspicion of severe/serious catheter-related infection (e.g., chills or rigors with infusion through catheter and cellulitis around the catheter exit site).

●      Skin or soft-tissue infection at any site.


◆     Recommended strategies for de-escalation approach: Patient stable at presentation and stable at 72-96 h, fever of unknown origin (FUO). 

○       Stop any aminoglycoside, or anti-Gram positive agent if given in combination.

○       Switch to a narrower-spectrum agent, e.g., cefepime, ceftazidime, piperacillin/tazobactam, cefoperazone/sulbactam, if carbapenem or novel beta-lactam were used initially.

** Irrespective of fever status at re-evaluation.


◆     Revision of recommendation for discontinuation of antibiotic treatment in neutropenic patients with FUO:

●      Empiric antibiotic therapy (EAT) can be discontinued at ≥ 72 hours of treatment in hemodynamically stable patients since presentation and who are afebrile ≥48 hours, irrespective of neutrophil count or expected duration of neutropenia.

●      They recommend continuing EAT in stable, high- or intermediate-risk neutropenic patients with FUO and persistent fever. Diagnostic efforts should be continued to search for an infectious focus or alternative explanation of fever.

●      Discontinuation of EAT can be considered later when the bacterial infectious source was reasonably excluded by microbiological tests and imaging.

●      They do not recommend adding coverage against resistant Gram-positive, or Gram-negative bacteria in a stable patient with persistent fever.


◆     Antibiotic therapy can be discontinued before recovery from neutropenia in patients with clinically documented infections or microbiologically documented infections (CDI/MDI) after completion of intended course of treatment, who are hemodynamically stable and afebrile for ≥72 h, with resolution of all clinical signs and symptoms and microbiological eradication of infection (when re-sampling possible).

●       In patients with Gram-negative bloodstream infections, we recommend a duration of antibiotic treatment of at least 7 days with recovery of neutropenia or without recovery of neutropenia.

Click for the full article

IACH Activities

Webinar Series

Dedicated to specific topics in the field of clinical hematology delivered by the top experts in the field

IACH Nurses

Activities specifically designed to advance education for nurses in the field of hematology

Journal Club

Discussions of recent research paper published in a peer-reviewed journal

Junior Club

On-line case study events hosted by experts in the field together with junior physicians

IACH News

Breaking news podcasts on new developments in the field of clinical hematology

IACH Giants

A series of webinars celebrating the work of key leaders in the field of hematology

For questions please contact:
info@clinical-hematology.org


If you prefer not to receive future email communications from us, please unsubscribe here

This newsletter was sent using ITnewsletter