We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Clinical Trial ProtocolOpen Accesscc iconby iconnc iconnd icon

BOREAS: a global, phase III study of the MDM2 inhibitor navtemadlin (KRT-232) in relapsed/refractory myelofibrosis

    Srdan Verstovsek

    *Author for correspondence: Tel.: +1 713 745 3429;

    E-mail Address: sverstov@mdanderson.org

    The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA

    ,
    Haifa Kathrin Al-Ali

    Krukenberg Cancer Center, University Hospital, Halle, 33790, Germany

    , ,
    Andrew Perkins

    Australian Centre for Blood Diseases, Monash University, Victoria, 3294, Australia

    , ,
    Sanjay R Mohan

    Vanderbilt University Medical Center, Nashville, TN 37232, USA

    ,
    Bart L Scott

    Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA

    ,
    Dariusz Woszczyk

    Hematology Department, University of Opole, Opole, 45-040, Poland

    ,
    Steffen Koschmieder

    Department of Hematology, Oncology, Haemostaseology & Stem Cell Transplantation (Department of Medicine IV), Faculty of Medicine, RWTH University Aachen, Aachen, 52062, Germany

    ,
    Regina García-Delgado

    Virgen de la Victoria University Hospital, Málaga, 29010, Spain

    ,
    Rejtő László

    Josa András Teaching Hospital, Nyíregyháza, 4400, Hungary

    ,
    Jesse S McGreivy

    Kartos Therapeutics, Inc., Redwood City, CA 94065, USA

    ,
    Wayne P Rothbaum

    Kartos Therapeutics, Inc., Redwood City, CA 94065, USA

    &
    Jean-Jacques Kiladjian

    Saint-Louis Hospital & Université Paris Cité, Paris, 75010, France

    Published Online:https://doi.org/10.2217/fon-2022-0901

    Abstract

    Patients with myelofibrosis (MF) who discontinue ruxolitinib due to progression/resistance have poor prognoses. JAK inhibitors control symptoms and reduce spleen volumes with limited impact on underlying disease pathophysiology. Murine double minute 2 (MDM2), a negative regulator of p53, is overexpressed in circulating malignant CD34+ MF cells. The oral MDM2 inhibitor navtemadlin (KRT-232) restores p53 activity to drive apoptosis of wild-type TP53 tumor cells by inducing expression of pro-apoptotic Bcl-2 family proteins. Navtemadlin demonstrated promising clinical and disease-modifying activity and acceptable safety in a phase II study in patients with relapsed/refractory MF. The randomized phase III BOREAS study compares the efficacy and safety of navtemadlin to best available therapy in patients with MF that is relapsed/refractory to JAK inhibitor treatment.

    Clinical Trial Registration:NCT03662126 ( ClinicalTrials.gov).

    Plain language summary

    Myelofibrosis (MF) is a rare blood cancer that disrupts normal blood cell production and causes fibrosis (tissue thickening/scarring) in bone marrow, reduced red blood cells in the circulation, and an enlarged spleen. Although currently approved treatments can help relieve some effects, they have limited impact on the underlying cause of the disease. Navtemadlin is a new therapy that inhibits a protein frequently overexpressed in cancer cells found in MF patients called murine double minute 2 (MDM2), which regulates a common tumor suppressor protein called p53. By inhibiting MDM2, navtemadlin restores normal p53 function and its ability to kill MF cancer cells. BOREAS is a large clinical study of navtemadlin for MF patients whose disease is not responding to current therapy.

    Tweetable abstract

    Navtemadlin (KRT-232), a potent, selective and orally available MDM2 inhibitor is being explored in the global phase III BOREAS trial in #myelofibrosis patients who are relapsed or refractory to JAK-inhibitor treatment #MPNSM.

    Graphical abstract

    Trial

    BOREAS is a randomized, open-label, global, phase III study that evaluates the efficacy and safety of navtemadlin versus best available therapy (BAT) for the treatment of primary or secondary myelofibrosis (MF) in patients who have relapsed or are refractory (R/R) to Janus kinase (JAK) inhibitor treatment (NCT03662126) [1]. This trial has been named BOREAS (BlOcking MDM2 to REActivate p53 in MyelofibrosiS) after the Greek god of the powerful North Wind. Just as winds can signal changes in weather patterns, the name BOREAS is metaphorically signaling that a potential change is coming for the treatment of MF.

    Background & rationale

    Unmet medical need in MF

    MF is a chronic myeloproliferative neoplasm, with an annual incidence of approximately 1/100,000 and an estimated prevalence of 4–6/100,000 in USA [2]. The disease is characterized by clonal proliferation of myeloid cells resulting in extramedullary hematopoiesis, hepatosplenomegaly, constitutional symptoms and cytopenias, along with bone marrow fibrosis, and an increased risk for transformation into acute myeloid leukemia (AML) [3–5]. Overproduction and dysregulation of inflammatory cytokines, particularly from monocytes or precursors of macrophages and dendritic cells, play an important role in the etiology and distinct clinical features of MF [6]. Circulating inflammatory cytokine levels, such as TNFα, IL-6, IL-8 and IL-10, are elevated in MF and may serve as prognostic markers for disease progression or poor outcomes [6,7]. TNFα is known to promote clonal dominance of malignant cells, whereas high IL-6 and IL-8 are associated with severity of constitutional symptoms [7] and elevated IL-8 is also with increased leukocytosis and higher-grade fibrosis [8]. IL-2, IL-6, IL-8, and beta-2 microglobulin, an important component for antigen presentation, have also been associated with blast transformation [7].

    Although the exact cause of MF is unknown, the pathophysiology in most patients is characterized by the acquisition of driver mutations that deregulate the JAK/signal transducers and activators of transcription (STAT) signaling pathway [9,10]. Aberrant pro-survival signaling through growth factor and cytokine receptors increases the proliferation rate of malignant myeloid cells [11]. Somatic mutations in three genes in this pathway, JAK2, MPL (thrombopoietin receptor), and/or CALR, have been shown to drive the development of MF [9,12]. The gain-of-function JAK2 mutation (V617F) has been identified in 40–75% of patients with primary MF (PMF) and can lead to the constitutive activation of downstream signaling through STAT, resulting in uncontrolled myeloproliferation [9,12]. Similarly, frame-shift mutations in the CALR gene and gain-of-function mutations in the MPL gene, which are found in 20–30% and less than 10% of patients with PMF, respectively [9,12], can lead to JAK/STAT activation. Approximately 10% of patients have no reported driver mutations; this triple-negative mutation status is associated with poor prognosis in patients with MF [9,12]. In addition, mutations in several high molecular risk (HMR) genes involved in epigenetic regulation (ASXL1, EZH2, IDH1/2) and RNA splicing (SRSF2) were identified as poor prognostic factors for overall survival and leukemia-free survival in patients with PMF [13].

    Although transplantation provides a potentially curative treatment option for high- to very high-risk patients with MF, the high morbidity and mortality associated with this approach limits its adoption to younger patients with good performance status [5,14]. As a result, JAK inhibitors are currently the standard-of-care for MF based on their ability to provide symptom control and reduce spleen volumes [15]. The four main JAK inhibitors currently being studied include ruxolitinib, fedratinib, pacritinib and momelotinib, the first three of which are approved by the US FDA for the treatment of myelofibrosis [15,16]. Although these agents mediate spleen volume reduction (SVR) and improvements in total symptom score (TSS), currently published data reports only a modest impact on bone marrow fibrosis and their disease-modifying effects are limited [15]. Even with recent approvals of novel JAK-inhibitors, the prognosis for patients who are R/R to JAK inhibitors is particularly poor [3,17]. Median overall survival in patients who discontinue ruxolitinib is 13–16 months, highlighting the need for novel therapies that target alternative pathways [17,18].

    Overview of MDM2 & navtemadlin mechanism of action

    p53 is a tumor suppressor protein that regulates cellular proliferation and apoptosis [19,20]. Murine double minute 2 (MDM2), the primary negative regulator of p53 plays a key role in maintaining low p53 levels in unstressed mammalian cells. In response to cellular stressors like DNA damage, hypoxia or oncogene activity, p53 initiates the transcription of various genes that culminate in either cell cycle arrest or apoptosis [20–24]. Reparable damage to cells induces low levels of p53 that mediate cell cycle arrest, whereas stressors that irreparably damage cells induce high levels of p53, inducing the intrinsic (mitochondrial) apoptosis pathway (Figure 1) [20–26].

    Figure 1. Navtemadlin (KRT-232) mechanism of action.

    p53 induces apoptosis by shifting the balance between pro-survival and pro-apoptotic Bcl-2 family members. MDM2 is the primary negative regulator of p53 cellular functions. MDM2 inhibition via navtemadlin (KRT-232) restores p53 activity to drive apoptosis of myelofibrosis cells harboring wild-type TP53.

    © 2022 Kartos Therapeutics, Inc. All rights reserved. Reproduced with permission from Kartos Therapeutics, Inc.

    More specifically, activated p53 upregulates selected pro-apoptotic Bcl-2 proteins (including Puma, Noxa and Bim) to overcome the effects of pro-survival Bcl-2 proteins (including Bcl-2, Bcl-XL and Mcl-1) to initiate cell death through the pore forming effectors (including Bax, Bak and Bok), which mediate the release of cytochrome c and induction of caspase-9 [22,24].

    As the key negative regulator of p53, MDM2 binds to and modulates the function of p53 by inhibiting its downstream transcriptional activity, promoting its transport out of the nucleus, and ubiquitinating the protein for proteasomal degradation [19]. In non-malignant cells, activated MDM2 plays an important role in maintaining p53 at low levels. Inactivating mutations in TP53 or overexpression of MDM2 can overcome p53-driven cell killing, promoting oncogenic transformation and conferring a strong survival advantage [20]. Although inactivating TP53 mutations are infrequent in MPNs (<5%), p53 activity is often suppressed by constitutive JAK2-mediated signaling in CD34+ cells, leading to the upregulation of the La antigen and MDM2 overexpression [27–29].

    Navtemadlin (KRT-232) is a potent, selective, orally available MDM2 inhibitor that restores p53 activity to drive apoptosis in TP53 wild-type (TP53WT) malignancies, including reductions in malignant CD34+ cells in MF (Figure 1) [30–32]. In TP53WT tumor cell lines, navtemadlin treatment increased protein levels of p53, p21, MDM2 and Puma in a dose-dependent manner; similar effects were observed on mRNA levels of these proteins in vivo [31]. Further studies in murine TP53WT xenograft models demonstrated that oral navtemadlin treatment resulted in significant inhibition of tumor growth and was associated with dose- and time-dependent induction of p21 mRNA in tumor samples, as well as increased apoptosis [31].

    Navtemadlin clinical & disease-modifying activity in MF

    Preclinical studies provided the basis for further evaluation of navtemadlin in phase I/II studies of patients with MF and other TP53WT hematologic malignancies and solid tumors [33–36]. Phase I studies demonstrated navtemadlin's dose proportional pharmacokinetic and pharmacodynamic profile, helped to characterize the optimal dose and schedule for future study, and provided early signals of antitumor activity among patients with TP53WT malignancies [33,35]. Consistent with the established mechanism of action, navtemadlin induced gene expression of several p53 pathway members in the bone marrow, including TP53, Bax, Puma, p21, and MDM2.

    Following these early studies, a proof-of-concept, two-part phase II study (KRT-232-101) was designed to identify the recommended phase II dose and schedule of navtemadlin for patients with MF who were R/R to JAK inhibitor treatment [34,37]. Patients were randomly assigned to 1 of 3 arms: 120 mg or 240 mg once daily (q.d.) on days 1–7 of 21-day cycles or 240 mg q.d. on days 1–7 of 28-day cycles. JAK inhibitor treatment washout was not required prior to initiation of navtemadlin. Phase II results showed promising activity of navtemadlin at a dose of 240 mg q.d. given days 1–7/28 with a best SVR ≥35% of 16% in 25 evaluable patients, best TSS response ≥50% in 30% of patients, and median reduction in peripheral blood CD34+ cell counts at week 24 of 87% (Figure 2) [34,37]. Navtemadlin also demonstrated a tolerable safety profile; the most frequently reported adverse events of any grade and regardless of causality were gastrointestinal (e.g., diarrhea, nausea, vomiting) and hematologic (e.g., thrombocytopenia, anemia, neutropenia). Gastrointestinal adverse events were managed in part with the use of prophylaxis for nausea and vomiting.

    Figure 2. Clinical proof-of-concept for navtemadlin (240 mg q.d., D1–7/28) in relapsed/refractory myelofibrosis.

    *SVR evaluable: patients must have baseline and at least one pre-planned post-baseline spleen MRI/CT (week 12, 24, 36).

    §TSS evaluable: requires patients to have a baseline TSS and >20-days within a 28-day period reported for post-baseline assessments.

    Best Modified TSS: Best change from baseline to trailing 28-day average at end of week 4, 8, 12, 16, 20, 24, etc.

    Modified MPN-SAF TSS includes early satiety, abdominal discomfort, night sweats, itching, bone pain and rib pain.

    CT: Computed tomography; MPN-SAF: Myeloproliferative Neoplasm Symptom Assessment Form; ORR: Overall response rate; Rux: Ruxolitinib; SVR: Spleen volume response; TP53MUT: Mutated tumor protein p53; TSS: Total symptom score.

    Spleen responses were superior in patients who were off ruxolitinib prior to their baseline MRI/CT scan versus those who remained on ruxolitinib therapy at the time of their baseline scan (best SVR ≥35%: 29% vs 0%) [34,37]. This effect may be attributed to ruxolitinib discontinuation syndrome, which occurs within 21 days of cessation of treatment and is characterized by an acute relapse of disease symptoms, including accelerated splenomegaly and worsening cytopenias [38]. Although JAK inhibitor washout was not required prior to initiating navtemadlin treatment [34]; the spleen flare that may have resulted from ruxolitinib discontinuation is likely to have confounded the reported overall SVRs in this study.

    A subsequent correlative analysis of data from the same study showed that navtemadlin treatment resulted in clinically relevant changes in measures reflective of disease modification [32]. An analysis of reductions in variant allele frequency (VAF) of driver- or HMR-gene following navtemadlin treatment demonstrated that among patients with evaluable samples, a best driver gene reduction ≥20% was observed in 34% patients, and 29% showed a complete reduction in high-molecular risk or driver genes. Reduction in driver VAF at any time on study significantly correlated with SVR responses, which were higher in patients with ≥20% versus <20% decreases in driver VAFs (32% vs 5%; p = 0.0072). Bone marrow biopsies collected after 24 weeks of treatment showed that 27% of navtemadlin-treated patients had an improved fibrosis score of ≥1 grade and 51% had stable fibrosis scores as assessed by central pathology review. Navtemadlin-treated patients also demonstrated a significant decrease in serum TNFα levels (median best decrease of 41% from baseline), which significantly correlated with improvements in TSS and SVR. Demonstrating the correlation between clinical and biologic responses to navtemadlin treatment, spleen volume responses correlated with decreased peripheral CD34+ cell counts, improved fibrosis scores, and reduction of myeloproliferative neoplasm (MPN)-driver mutation burden. These results suggest that changes in biomarkers of disease burden were associated with clinical benefit and support further exploration of navtemadlin's disease-modifying effects in MF.

    The BOREAS phase III study design

    Based on the promising activity reported in the phase II study, the phase III BOREAS study has been initiated to evaluate the efficacy and safety of navtemadlin versus BAT for patients with primary or secondary MF who are R/R to JAK inhibitor treatment (NCT03662126).

    Eligibility criteria

    Adults with confirmed TP53WT primary or secondary MF (including PMF, post-polycythemia vera [PV] MF, or post-essential thrombocythemia [ET] MF according to WHO criteria [39]) are being enrolled in the study. Patients must have intermediate-1, intermediate-2, or high-risk disease (defined by Dynamic International Prognostic Scoring System [DIPSS] [40]); Eastern Cooperative Oncology Group performance status (ECOG-PS) ≤2; and have adequate hematologic function (Table 1). Patients must be R/R to prior JAK inhibitor treatment. Key exclusion criteria include prior MDM2 inhibitor or p53-directed therapy, JAK inhibitor treatment within 28 days prior to screening MRI/CT, JAK inhibitor intolerance, and allogeneic stem-cell transplant.

    Table 1. Key eligibility criteria.
    Inclusion criteria
    • Age ≥18 years
    • Confirmed diagnosis of PMF, post-PV MF, or post-ET MF (WHO criteria [39])
    TP53WT status by central laboratory testing
    • High-risk, intermediate-2 risk or intermediate-1 risk (defined by DIPSS [40])
    • Relapsed/refractory to prior treatment with JAK inhibitor defined as:
      ○ Relapsed patients are those with progressive disease after JAK inhibitor treatment, defined by one of the following:
        ▪ Increase in spleen volume by ≥25% by radiographic imaging from nadir
        ▪ ≥100% increase in palpable distance below LLCM for baseline splenomegaly of 5 to 10 cm
        ▪ ≥50% increase in palpable distance below LLCM for baseline splenomegaly of >10 cm
        ▪ Regrowth after achieving complete response
      ○ Refractory patients are those with a lack of spleen response after ≥12 weeks of JAK inhibitor treatment, defined by either:
        ▪ <10% spleen volume reduction by radiographic imaging
        ▪ <30% decrease from baseline in spleen size by palpation
    • ECOG PS ≤2
    Exclusion criteria
    • Prior splenectomy
    • Splenic irradiation within 3 months prior to randomization
    • History of major hemorrhage or intracranial hemorrhage within 6 months prior to randomization
    • History of stroke, reversible ischemic neurological defect, or transient ischemic attack within 6 months prior to randomization
    • Prior MDM2 inhibitor therapy or p53-directed therapy
    • Prior allogeneic stem cell transplant or plans for allogeneic stem cell transplant
    • History of major organ transplant
    • Grade 2 or higher QTc prolongation (>480 milliseconds per NCI-CTCAE v5.0)

    ANC: Absolute neutrophil count; CT: Computed tomography; DIPSS: Dynamic International Prognostic Scoring System; ECOG PS: Eastern Cooperative Oncology Group performance status; JAK: Janus kinase; LLCM: Left lower costal margin; MDM2: Murine double minute 2; MF: Myelofibrosis; MFSAF v4.0: Myelofibrosis Symptom Assessment Form version 4.0; NCI-CTCAE v5.0: National Cancer Institute Common Terminology Criteria for Adverse Events, version 5.0; PMF: Primary MF; Post-ET MF: Post-essential thrombocythemia MF; Post-PV MF: Post-polycythemia vera MF; QTc: Corrected QT interval; TP53WT: Wild-type tumor protein p53 gene; TSS: Total symptom score; WHO: World Health Organization.

    Study design

    Approximately 282 eligible patients are randomized in a 2:1 ratio to navtemadlin (Arm 1: 188 patients) or BAT (Arm 2: 94 patients). Patients are stratified by primary versus secondary MF (PMF vs post-PV-MF or post-ET-MF) and baseline TSS (≤10 vs >10). Navtemadlin treatment is given at a dose of 240 mg orally once daily on days 1–7 in 28-day treatment cycles (Figure 3) based on the dose and schedule established in the phase II study [34]. Patients randomized to BAT will receive one of the following treatment options at the discretion of the investigator in 28-day cycles: anti-cancer therapy, including hydroxyurea, thalidomide, lenalidomide, pomalidomide, cladribine, busulfan, methotrexate, interferon or supportive care, including the erythropoiesis-stimulating agent, danazol, red blood cell (RBC) transfusion, corticosteroids, anagrelide, low-dose aspirin or granulocyte-colony stimulating factor. JAK inhibitors are not permitted. BAT may be given sequentially or in combination as per local treatment guidelines and regional standard of care. Patients who receive BAT with documented disease progression at any time or who complete week 24 assessments may crossover to the navtemadlin arm. Patients will be treated until disease progression, unacceptable toxicity, death, or withdrawal of consent. Patients who discontinue study treatment for any reason will be monitored in long-term follow-up for survival and subsequent anticancer therapy.

    Figure 3. BOREAS phase III study design.

    *Treatment selection is at the discretion of the investigator. Patients with documented disease progression at any time or those who complete week-24 assessments may crossover to the navtemadlin arm.

    D: Day; MF: Myelofibrosis; R/R: Relapsed/refractory; TSS: Total symptom score.

    All patients receive anti-emetic prophylaxis for nausea/vomiting 30 min prior to and 8 h after each navtemadlin dose on days 1–7 of the treatment cycle when navtemadlin is administered. Anti-diarrheal prophylaxis is given on each day that navtemadlin is taken for at least the first two cycles of therapy.

    Study end points & outcome measures

    The primary end point is the proportion of patients achieving an SVR ≥35% at week 24 by MRI/CT (central review; Table 2). Key secondary end points include the proportion of patients with ≥50% reduction in TSS (per Myelofibrosis Symptom Assessment Form version 4.0 [MFSAF v4.0] [41]) from baseline to week 24, overall survival (OS), progression-free survival (PFS), overall SVR ≥35% at any time by MRI/CT, spleen response duration (time from initial SVR ≥35% by MRI/CT to first occurrence of disease progression), rate of conversion from RBC transfusion dependence to independence at week 24, and safety.

    Table 2. Study end points.
    Primary end point• Spleen volume reduction (SVR) ≥35% at week 24 by MRI/CT (central review)
    Key secondary end points• ≥50% reduction in TSS rate at week 24 (per MFSAF v4.0)
    • Overall survival
    • Progression-free survival
    • Overall SVR ≥35% at any time by MRI/CT (central review)
    • Spleen response duration
    • Rate of conversion from red blood cell transfusion dependence to independence at week 24
    • Safety

    CT: Computed tomography; MFSAF v4.0: Myelofibrosis Symptom Assessment Form version 4.0; SVR: Spleen volume reduction; TSS: Total symptom score.

    BOREAS will be considered complete 2 years after the last patient is enrolled, at which time patients who remain on study treatment may be eligible to enroll in a rollover study. Presently, the estimated study completion date is 31 December 2025. A list of trial locations across the globe is available on the BOREAS trial website: www.boreas-trial.com.

    Conclusion

    The BOREAS study will evaluate the safety and efficacy of navtemadlin versus BAT for the treatment of patients with MF whose disease is R/R to standard of care JAK inhibitor treatment. Patients with TP53WT may also be recruited for the open-label, multicenter phase Ib/II study of navtemadlin combined with ruxolitinib in patients who have shown a suboptimal response following ≥18 week of ruxolitinib treatment (NCT04485260) [42]. The BOREAS study has the potential to establish navtemadlin as a novel therapeutic approach for MF patients who are R/R to JAK inhibitor treatment and would otherwise have limited treatment options.

    Executive summary

    Myelofibrosis

    • Myelofibrosis (MF) is a progressive, myeloproliferative neoplasm (MPN) characterized by ineffective clonal hematopoiesis, splenomegaly, progressive bone marrow fibrosis, profound cytopenia and a propensity for transformation into acute myeloid leukemia.

    • Although the exact cause of MF is unknown, the pathophysiology of MF in most patients is characterized by driver mutations that dysregulate the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway.

    • JAK inhibitors are the standard of care for MF. Although therapies targeting the JAK/STAT pathway provide symptom control and reduce spleen volumes, they have limited impact on the underlying pathophysiology of the disease.

    • There remains an unmet medical need for novel therapeutic approaches with disease-modifying effects in MF, especially for patients who have failed JAK inhibitor treatment.

    Navtemadlin (KRT-232)

    • Navtemadlin is a potent, selective, orally available murine double minute 2 (MDM2) inhibitor that restores p53 activity to drive apoptosis in TP53 wild-type MF.

    • Elevated circulating CD34+ cells that overexpress MDM2 are a characteristic feature of MF.

    • By inhibiting MDM2, navtemadlin restores p53 activity, which regulates Bcl-2 family proteins to induce apoptosis.

    • Navtemadlin has demonstrated a manageable safety profile and promising clinical activity, including reductions in spleen volume and improvements in symptom burden in a phase II study, and has the potential for disease-modifying effects in MF patients who are relapsed/refractory (R/R) to JAK inhibitor treatment.

      Spleen volume responses correlated with decreased peripheral CD34+ cell counts and TNFα levels, improved fibrosis scores, and reduction of MPN driver mutation burden.

    BOREAS phase III study

    • BOREAS is an open-label, phase III randomized, controlled study of navtemadlin versus best available therapy (BAT) in patients with PMF, post-PV MF, or post-ET MF who are R/R to JAK inhibitor treatment.

    • Eligible patients will be randomized 2:1 to receive oral navtemadlin 240 mg once daily on days 1–7 of 28-day treatment cycles or BAT.

    • The primary end point is the proportion of patients with a spleen volume reduction ≥35% at week 24.

    • Secondary end points will include the proportion of patients with ≥50% reduction in total symptom score, overall and progression-free survival, spleen response duration, red blood cell transfusion dependency conversion and safety.

    Conclusion

    • The BOREAS phase III study has the potential to establish navtemadlin as a novel treatment for patients with MF whose disease is R/R to JAK inhibitor treatment.

    Supplementary data

    An infographic accompanies this paper. To view or download this infographic in your browser please click here: https://www.futuremedicine.com/doi/suppl/10.2217/fon-2022-0901

    Author contributions

    All the authors were involved in the design and/or conduct of the study. All authors have contributed to the preparation and writing of the manuscript and approved the final manuscript.

    Acknowledgments

    The authors thank the coinvestigators on the clinical study, as well as patients, families, and caregivers who are participating in the study. This study is funded by Kartos Therapeutics, (CA, USA).

    Financial & competing interests disclosure

    S Verstovsek has received consultancy fees from Celgene, Constellation Pharmaceuticals, Incyte, Novartis, Pragmatist, and Sierra Oncology; and research funding from Blueprint Medicines, Celgene, CTI BioPharma Corp, Genentech, Gilead Sciences, Incyte, Novartis, NS Pharma, Promedior, and Roche. HK Al-Ali has received honoraria from Novartis, Bristol Myers Squibb, and AbbVie; has received consultancy fees from Novartis, Bristol Myers Squibb, and AbbVie; and has received research funding from Incyte and Bristol Myers Squibb. J Mascarenhas has consultancy fees from Incyte, Kartos Therapuetics, Celgene, Bristol Myers Squibb, Constellation, CTI BioPharma Corp, Geron, AbbVie, Imago, Galecto, Karyopharm, GSK, Roche, Novartis, PharrmaEssentia; and research funding from Novartis, Roche, Merck, CTI BioPharma Corp, Incyte, Kartos Therapeutics, AbbVie, PharmaEssentia, Celgene, Bristol Myers Squibb, and Geron. A Perkins has received consultancy fees from Novartis, Sierra Oncology, Abbvie, and CTI BioPharma Corp and has been paid to participate in a speaker's bureau for Novartis. AM Vannucchi has received consultancy fees from; and has been paid to participate in a speaker's bureau for Celgene, Incyte, Novartis, AbbVie, Blueprint Medicines, CTI BioPharma Corp, and Roche. S Mohan has received research funding Incyte, Karyopharm, Kartos Therapeutics, and Taiho. B Scott has received consultancy fees from CTI BioPharma Corp and Incyte. D Woszczyk has received research funding from AbbVie, Acerta, Astellas, Celgene, Janssen, Kartos Therapeutics, Roche, Sierra Oncology, and Telios Pharma. S Koschmieder has received research funding from Geron, Janssen, AOP Pharma, and Novartis, consulting fees from Pfizer, Incyte, Ariad, Novartis, AOP Pharma, Bristol Myers Squibb, Celgene, Geron, Janssen, CTI BioPharma Corp, Roche, Bayer, and PharmaEssentia; honoraria from Novartis, Bristol Myers Squibb/Celgene, Pfizer; travel and accommodation support from Alexion, Novartis, Bristol Myers Squibb, Incyte, AOP Pharma, CTI BioPharma, Pfizer, Celgene, Janssen, Geron, Roche, AbbVie, Sierra Oncology, and Kartos Therapeutics; consultancy fees for Pfizer, Incyte, Ariad, Novartis, AOP Pharma, BMS, Celgene, Geron, Janssen, CTI BioPharma Corp, Roche, Bayer, Sierra Oncology, and PharmaEssentia; and had a patent issued for a BET inhibitor at RWTH Aachen University. R Garcia Delgado has been paid to participate in a speaker's bureau for; has provided expert testimony on behalf of; and has received travel, accommodations, and expenses from Novartis and Janssen. R László has no financial relationships to disclose. JS McGreivy is currently employed by; is in a leadership position with; has stock and other ownership interests; and has received travel, accommodations, and expenses from Kartos Therapeutics. WP Rothbaum is currently employed by Quogue Capital; is a current equity holder in Iovance Biotherapeutics and Acerta Pharma/AstraZeneca; has individual stock in Kartos Therapeutics and Telios Pharma, has patents and royalties in Iovance Biotherapeutics and Acerta, and Quogue IP Holdings, has served on the Board of Directors or advisory committees for and received travel, accommodations, and expenses from Iovance Biotherapeutics, Kartos Therapeutics, and Telios Pharma. J-J Kiladjian has had a consulting or advisory role for Novartis, AbbVie, BMS, Incyte, AOP Orphan and has received research funding from Kartos Therapeutics. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Editorial support was provided by S Thomas, PhD with Team 9 Science and funded by Kartos Therapeutics. The authors directed development of the publication and are fully responsible for all content and editorial decisions.

    Ethical conduct of research

    Signed informed consent is required by patients prior to study initiation. The BOREAS study protocol is approved by the institutional ethics review board of each participating study site. The study is being performed in accordance with the ethical principles of the Declaration of Helsinki and conducted in line with the International Conference for Harmonisation Guidelines for GCP and region-specific laws and regulations.

    Data sharing statement

    Queries about the ongoing trial can be addressed to boreas-trial@kartosthera.com.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. KRT-232 versus best available therapy for the treatment of subjects with myelofibrosis who are relapsed or refractory to JAK inhibitor treatment (BOREAS). (2021). (Accessed 12 Aug) https://clinicaltrials.gov/ct2/show/NCT03662126
    • 2. Mehta J, Wang H, Iqbal SU, Mesa R. Epidemiology of myeloproliferative neoplasms in the United States. Leuk. Lymphoma 55(3), 595–600 (2014).
    • 3. Cervantes F, Dupriez B, Pereira A et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 113(13), 2895–2901 (2009).
    • 4. Masarova L, Verstovsek S. The evolving understanding of prognosis in post-essential thrombocythemia myelofibrosis and post-polycythemia vera myelofibrosis vs primary myelofibrosis. Clin. Adv. Hematol. Oncol. 17(5), 299–307 (2019).
    • 5. Tefferi A. Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management. Am. J. Hematol. 96(1), 145–162 (2021).
    • 6. Fisher DAC, Miner CA, Engle EK et al. Cytokine production in myelofibrosis exhibits differential responsiveness to JAK-STAT, MAP kinase, and NFkappaB signaling. Leukemia. 33(8), 1978–1995 (2019).
    • 7. Fisher DAC, Fowles JS, Zhou A, Oh ST. Inflammatory pathophysiology as a contributor to myeloproliferative neoplasms. Front Immunol. 12, 683401 (2021).
    • 8. Dunbar A, Lu M, Farina M et al. Increased interleukin-8 (IL-8)-CXCR2 signaling promotes progression of bone marrow fibrosis in myeloproliferative neoplasms. Blood 136, 6–7 (2020).
    • 9. Shammo JM, Stein BL. Mutations in MPNs: prognostic implications, window to biology, and impact on treatment decisions. Hematology Am. Soc. Hematol. Educ. Program. 2016(1), 552–560 (2016).
    • 10. Kubesova B, Pavlova S, Malcikova J et al. Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status. Leukemia. 32(2), 450–461 (2018).
    • 11. Lussana F, Rambaldi A. Inflammation and myeloproliferative neoplasms. J. Autoimmun. 85, 58–63 (2017).
    • 12. Ferreira Cristina S, Polo B, Lacerda JF. Somatic mutations in Philadelphia chromosome-negative myeloproliferative neoplasms. Semin. Hematol. 55(4), 215–222 (2018).
    • 13. Vannucchi AM, Lasho TL, Guglielmelli P et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 27(9), 1861–1869 (2013).
    • 14. Harrison CN, Schaap N, Mesa RA. Management of myelofibrosis after ruxolitinib failure. Ann. Hematol. 99(6), 1177–1191 (2020).
    • 15. Bose P, Verstovsek S. JAK inhibition for the treatment of myelofibrosis: limitations and future perspectives. Hemasphere. 4(4), e424 (2020).
    • 16. Sureau L, Orvain C, Ianotto JC et al. Efficacy and tolerability of Janus kinase inhibitors in myelofibrosis: a systematic review and network meta-analysis. Blood Cancer J. 11(7), 135 (2021).
    • 17. Newberry KJ, Patel K, Masarova L et al. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood 130(9), 1125–1131 (2017).
    • 18. Kuykendall AT, Shah S, Talati C et al. Between a rux and a hard place: evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation. Ann. Hematol. 97(3), 435–441 (2018).
    • 19. Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature 408(6810), 307–310 (2000).
    • 20. Kastenhuber ER, Lowe SW. Putting p53 in context. Cell 170(6), 1062–1078 (2017).
    • 21. Bensaad K, Vousden KH. Savior and slayer: the two faces of p53. Nat. Med. 11(12), 1278–1279 (2005).
    • 22. Vaseva AV, Moll UM. The mitochondrial p53 pathway. Biochim. Biophys. Acta 1787(5), 414–420 (2009).
    • 23. Kracikova M, Akiri G, George A, Sachidanandam R, Aaronson SA. A threshold mechanism mediates p53 cell fate decision between growth arrest and apoptosis. Cell Death Differ. 20(4), 576–588 (2013).
    • 24. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20(3), 175–193 (2019).
    • 25. Chen L, Willis SN, Wei A et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol. Cell. 17(3), 393–403 (2005).
    • 26. Ghiotto F, Fais F, Bruno S. BH3-only proteins: the death-puppeteer's wires. Cytometry A. 77(1), 11–21 (2010).
    • 27. Nakatake M, Monte-Mor B, Debili N et al. JAK2(V617F) negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene 31(10), 1323–1333 (2012).
    • 28. Raza S, Viswanatha D, Frederick L et al. TP53 mutations and polymorphisms in primary myelofibrosis. Am. J. Hematol. 87(2), 204–206 (2012).
    • 29. Marcellino BK, Hoffman R, Tripodi J et al. Advanced forms of MPNs are accompanied by chromosomal abnormalities that lead to dysregulation of TP53. Blood Adv. 2(24), 3581–3589 (2018).
    • 30. Sun D, Li Z, Rew Y et al. Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development. J. Med. Chem. 57(4), 1454–1472 (2014). • Original publication for the discovery of navtemadlin (AMG-232, KRT-232), a highly potent inhibitor of MDM2.
    • 31. Canon J, Osgood T, Olson SH et al. The MDM2 inhibitor AMG 232 demonstrates robust antitumor efficacy and potentiates the activity of p53-inducing cytotoxic agents. Mol. Cancer Ther. 14(3), 649–658 (2015). •• Identification of the mechanisms of action of navtemadlin (AMG-232, KRT-232) in preclinical in vitro and in vivo tumor models.
    • 32. Vachhani P, Lange A, Delgado RG et al. Potential disease-modifying activity of navtemadlin (KRT-232), a first-in-class MDM2 inhibitor, correlates with clinical benefits in relapsed/refractory myelofibrosis (MF). Blood 138(Suppl. 1), 3581 (2021).
    • 33. Erba HP, Becker PS, Shami PJ et al. Phase 1b study of the MDM2 inhibitor AMG 232 with or without trametinib in relapsed/refractory acute myeloid leukemia. Blood Adv. 3(13), 1939–1949 (2019).
    • 34. Al-Ali HK, Delgado RG, Lange A et al. KRT-232, a first-in-class, murine double minute 2 inhibitor (MDM2i), for myelofibrosis (MF) relapsed or refractory (R/R) to Janus-associated kinase inhibitor (JAKi) treatment (TX). HemaSphere. 4, 65. Abstract S215 (2020). •• Initial report of phase II dosing, safety, and efficacy results from the BOREAS trial, an open-label, single-arm, 2-part, global study of navtemadlin (KRT-232) in patients with MF that were relapsed/refractory to JAK inhibitor treatment.
    • 35. Gluck WL, Gounder MM, Frank R et al. Phase 1 study of the MDM2 inhibitor AMG 232 in patients with advanced p53 wild-type solid tumors or multiple myeloma. Invest. New Drugs 38(3), 831–843 (2020). •• First-in-human phase I study of navtemadlin (AMG-232, KRT-232) to demonstrate dose-proportional plasma concentrations, tolerable safety, and preliminary antitumor activity in TP53WT patients with solid tumors or hematologic malignancies.
    • 36. Wong MKK, Kelly CM, Burgess MA et al. KRT-232, a first-in-class, murine double minute 2 inhibitor (MDM2i), for TP53 wild-type (p53WT) Merkel cell carcinoma (MCC) after anti-PD-1/L1 immunotherapy. J. Clin. Oncol. 38(Suppl. 15), 10072 (2020).
    • 37. Verstovsek S, Al-Ali HK, Mascarenhas J et al. BOREAS: A global phase III study of KRT-232, a first-in-class murine double minute 2 (MDM2) inhibitor in TP53WT relapsed/refractory (R/R) myelofibrosis (MF). J. Clin. Oncol. 39(Suppl. 15), TPS7057 (2021).
    • 38. Palandri F, Palumbo GA, Elli EM et al. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis. Blood Cancer J. 11(1), 4 (2021).
    • 39. Arber DA, Orazi A, Hasserjian R et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127(20), 2391–2405 (2016).
    • 40. Gangat N, Caramazza D, Vaidya R et al. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J. Clin. Oncol. 29(4), 392–397 (2011).
    • 41. Mesa RA, Schwager S, Radia D et al. The Myelofibrosis Symptom Assessment Form (MFSAF): an evidence-based brief inventory to measure quality of life and symptomatic response to treatment in myelofibrosis. Leuk. Res. 33(9), 1199–1203 (2009). • An evidence-based inventory to measure quality of life and symptomatic treatment response in patients with myelofibrosis.
    • 42. An open-label, multicenter, phase Ib/II study of the safety and efficacy of KRT-232 combined with ruxolitinib in patients with primary myelofibrosis (PMF), post-polycythemia vera MF (post-PV-MF), or post-essential thrombocythemia MF (post-ET-MF) who have a suboptimal response to ruxolitinib. Accessed 24 Oct, 2022. https://clinicaltrials.gov/ct2/show/NCT04485260.