Educational content on VJHemOnc is intended for healthcare professionals only. By visiting this website and accessing this information you confirm that you are a healthcare professional.

The Community Focus Channel on VJHemOnc is an independent medical education platform, supported with funding from Johnson & Johnson (Gold). Supporters have no influence on the production of content. The levels of sponsorship listed are reflective of the amount of funding given.

The Lymphoma Channel on VJHemOnc is an independent medical education platform, supported with funding from AstraZeneca (Diamond), BMS (Gold), Johnson & Johnson (Gold), Takeda (Silver) and Galapagos (Bronze). Supporters have no influence on the production of content. The levels of sponsorship listed are reflective of the amount of funding given.

The Myeloproliferative Neoplasms Channel on VJHemOnc is an independent medical education platform, supported with funding from Takeda (Gold) and Kartos Therapeutics, Inc. (Bronze). Supporters have no influence on the production of content. The levels of sponsorship listed are reflective of the amount of funding given.

The Non-Malignant Channel on VJHemOnc is an independent medical education platform, supported with funding from Agios (Gold). Supporters have no influence on the production of content. The levels of sponsorship listed are reflective of the amount of funding given.

The Sickle Cell Disease Channel on VJHemOnc is an independent medical education platform, supported with funding from Agios (Gold). Supporters have no influence on the production of content. The levels of sponsorship listed are reflective of the amount of funding given.

Tafasitamab plus lenalidomide and rituximab in R/R FL: results from the Phase III inMIND Study

Follicular lymphoma (FL) is an indolent yet incurable form of non-Hodgkin lymphoma (NHL), with many patients eventually relapsing or becoming refractory to treatment.1 Chemoimmunotherapy is commonly used in the frontline setting, with immunotherapy-based approaches being preferred in the relapsed/refractory (R/R) setting. Despite advances in immunotherapy, achieving durable responses remains a challenge in patients with R/R FL.2 Currently, the combination of lenalidomide and rituximab is approved for patients who have received at least one prior line of therapy. Tafasitamab, a humanized anti-CD19 monoclonal antibody, has demonstrated efficacy in other lymphomas and is now under investigation for R/R FL.2

At the 66th American Society of Hematology (ASH) Annual Meeting, Laurie Sehn, MD, MPH, BC Cancer Agency, Vancouver, Canada, shared results from the Phase III randomized, double-blind, placebo-controlled inMIND study (NCT04680052), which evaluated the safety and efficacy of adding tafasitamab to lenalidomide and rituximab in patients with R/R FL or marginal zone lymphoma. The primary endpoint was progression-free survival (PFS) in patients with FL, with secondary endpoints including overall response rate (ORR), PET-complete response (CR), duration of response (DOR), and overall survival (OS). A total of 548 patients with FL were randomized to receive either tafasitamab or placebo, along with lenalidomide and rituximab.

At a median follow-up of 14.1 months, the addition of tafasitamab to lenalidomide and rituximab resulted in a significant improvement in PFS compared with placebo (22.4 months versus 13.9 months; hazard ratio [HR] [95% CI], 0.43 [0.32, 0.58]; p<0.0001). In addition, tafasitamab demonstrated a higher ORR (83.5% versus 72.4%, p=0.0014) and PET-CR rate (49.4% versus 39.8%, p=0.029). Overall, these findings suggest that the addition of tafasitamab to lenalidomide and rituximab results in a clinically significant and durable improvement in PFS in patients with R/R FL.2

Lack of benefit of autoSCT in patients with MCL with undetectable measurable residual disease

Mantle cell lymphoma (MCL) remains a challenging hematologic malignancy to treat due to its heterogeneity and aggressive nature.3 Autologous stem cell transplantation (autoSCT) has been a cornerstone of treatment for MCL, particularly for younger patients.4 However, the role of autoSCT in patients with undetectable measurable residual disease (uMRD) remains unclear. The Phase III ECOG-ACRIN EA4151 study (NCT03267433) evaluated whether autoSCT offers an advantage for patients with MCL who achieve deep first remission, particularly those with uMRD as measured by a highly sensitive MRD assay.5

Timothy Fenske, MD, Medical College of Wisconsin, Milwaukee, WI, shared results from an initial report of this study, which was conducted through the U.S. National Clinical Trials Network (NCTN) and the Blood and Marrow Transplant Clinical Trials Network (BMT-CTN). Patients aged 18-70 with MCL and CR after induction therapy were eligible for inclusion. The primary and secondary endpoints were OS and PFS, respectively. Patients with uMRD at a sensitivity of 1 in 10-6 were randomized to receive either autoSCT plus three years of maintenance rituximab (Arm A) or maintenance rituximab alone (Arm B). Patients with MRD-positive CR or MRD-indeterminate CR received autoSCT plus maintenance rituximab (Arms C and D, respectively).5

From August 2017 to July 2024, 650 patients were assigned to a treatment arm, with 257, 259, 49, and 85 patients enrolled on Arms A, B, C, and D, respectively. Results demonstrated that the three-year OS was 82.1% in Arm A and 82.7% in Arm B for all randomized patients and 86.2% and 84.8% in patients treated as assigned. The three-year PFS for Arms A and B were 76.6% and 77.4% in all randomized patients and 81.5% and 80.4% in patients treated as assigned. Results from this interim analysis suggest that in the current era of highly effective induction and maintenance regimens, patients with MCL in first CR and uMRD do not benefit from consolidative autoSCT, although further studies are warranted.5

Andean enriched NFKB1 haplotype and response to ropeginterferon alfa-2b in PV and ET

Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPNs) characterized by chronic inflammation and increased thrombotic risk.6 Recent studies have investigated the genetic factors that modulate the inflammatory pathways in these diseases, with a particular focus on the NF-κB pathway, which regulates pro-inflammatory cytokines.7

Josef Prchal, MD, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, shared findings from a study investigating the genetic basis for elevated hemoglobin levels in Andean natives (Aymara) using granulocyte transcriptome data. Three novel, alternatively spliced NFKB1 transcripts (AS-NFKB1) were identified and found to be associated with inflammatory and HIF pathways. The study revealed that the Aymara-enriched NFKB1 rs230511 T allele is associated with reduced inflammation, lower expression of prothrombotic and HIF-targeted genes, and a better response to ropeginterferon alfa-2b.6 The novel insights from this study offer valuable implications for PV and ET treatment.

Outcome of cerebral vasculopathy and cognitive performances 10 years post-enrollment in the DREPAGREFFE trials

Children with sickle cell anemia (SCA) often require long-term chronic transfusion therapy to prevent stroke risk.8  However, chronic transfusion exposes patients to several potential complications, including alloimmunization and iron overload, and is not curative.9

The DREPAGREFFE-1 trial (NCT01340404) was the first prospective study comparing allogeneic stem cell transplantation (alloSCT) with standard of care (SoC) for children with SCA and high cerebral arterial velocities, followed by the DREPAGREFFE-2 trial (NCT05053932), which assessed long-term outcomes at 10 years.

In this study, 67 children, including seven with a history of stroke, were enrolled. 32 children with matched sibling donors (MSD) were transplanted, while those without (n=35) were maintained on chronic transfusion for at least one year and then switched to hydroxyurea if cerebral arterial velocities normalized and no stenosis was present. Françoise Bernaudin, MD, Paris-East Créteil University (UPEC), Créteil, France, presented results from these trials.

At years one and three, the alloSCT group reported better physical and school functioning, though no significant differences in ischemic lesions or cognitive performance were observed. Furthermore, at 10 years, sickle cell hemoglobin (HbS%), reticulocytes, bilirubin, and white blood cell (WBC) counts were lower in the alloSCT group (p<0.001). These results demonstrate that alloSCT provides sustained benefits over SoC in children with SCA, supporting alloSCT as the preferred treatment option for this patient population.10

References

  1. Singh D, Singh A, Mukkamalla SKR. Relapsed and Refractory Follicular Lymphoma. Statpearls. 2023 Jan.
  2. Sehn L, Luminari S, Scholz C, et al. Tafasitamab Plus Lenalidomide and Rituximab for Relapsed or Refractory Follicular Lymphoma: Results from a Phase 3 Study (inMIND). Blood. 2024 Nov; 144 (Supplement 2): LBA-1.
  3. Yang P, Cai QQ, Zhang W, et al. Real-world treatment and outcome patterns of patients with mantle cell lymphoma in China: A large, multicenter retrospective analysis. Cancer Med. 2023 Jun;12(12):13204-13216.
  4. Kumar A. What is the role of up-front autologous stem cell transplantation in mantle cell lymphoma? Hematology Am Soc Hematol Educ Program. 2022 Dec; (1): 155–162.
  5. Fenske T, Wang XV, Till BG, et al. Lack of Benefit of Autologous Hematopoietic Cell Transplantation (auto-HCT) in Mantle Cell Lymphoma (MCL) Patients (pts) in First Complete Remission (CR) with Undetectable Minimal Residual Disease (uMRD): Initial Report from the ECOG-ACRIN EA4151 Phase 3 Randomized Trial. Blood. 2024 Nov; 144 (Supplement 2): LBA-6.
  6. Song J, Kim SJ, Prchal JT. Andean Enriched NFKB1 Haplotype Reduces Inflammation and Improves Response to Ropeginterferon Alfa-2b in Polycythemia Vera (PV) and Essential Thrombocythemia (ET)Blood. 2024 Nov; 144 (Supplement 2): LBA-4.
  7. Laranjeira ABA, Kong T, Snyder SC, et al. In vivo ablation of NF-κB cascade effectors alleviates disease burden in myeloproliferative neoplasms. Blood. 2024 Jun; 143 (23): 2414–2424.
  8. Howard J. Sickle cell disease: when and how to transfuse. Hematology Am Soc Hematol Educ Program. 2016 Dec; (1):625-631.
  9. Guilliams KP, Fields ME, Ragan DK, et al. Red cell exchange transfusions lower cerebral blood flow and oxygen extraction fraction in pediatric sickle cell anemia. Blood. 2018 March; 131 (9): 1012–1021.
  10. Bernaudin F, Verlhac S, Ducros-Miralles E, et al. Outcome of Cerebral Vasculopathy and Cognitive Performances 10 Years Post-Enrollment in the Drepagreffe Trial Comparing Allogeneic Stem Cell Transplantation to Standard-Care in Children with Sickle Cell Anemia and History of Abnormal Cerebral Velocities. Blood. 2024 Nov; 144 (Supplement 2): LBA-5.
Written by Anya Dragojlovic Kerkache
Edited by Simon Ng & Sol Yohannes
Publishing date: 19/12/2024