94 results on '"Gabriel N. Mannis"'
Search Results
2. Epstein–Barr virus-positive lymphoproliferative disorder manifesting as pulmonary disease in a patient with acute myeloid leukemia: a case report
- Author
-
Ritika Dutta, Susanna Y. Miao, Paul Phan, Sebastian Fernandez-Pol, Parveen Shiraz, Dora Ho, Gabriel N. Mannis, and Tian Y. Zhang
- Subjects
Acute myeloid leukemia ,Epstein–Barr virus ,Post-transplant lymphoproliferative disorder ,Hematopoietic stem cell transplant ,Case report ,Medicine - Abstract
Abstract Background Patients with lymphoproliferative disorders following hematopoietic stem cell transplant (HSCT) most commonly present with fever and lymphadenopathy within the first 5 months of transplant. Pulmonary post-transplant lymphoproliferative disorder (PTLD) is a particularly aggressive and rapidly progressive disease, with high morbidity and mortality. There are a very limited number of reported pulmonary PTLD cases following HSCT in patients with acute myeloid leukemia (AML). Early diagnosis and detection of pulmonary PTLD is critical given its high lethality. However, variable clinical presentations and nonspecific radiographic findings make pulmonary PTLD difficult to distinguish from other more common causes of pulmonary disease in AML patients. Case presentation Here, we describe a 68-year-old Caucasian man who presented for salvage induction therapy following relapse of his AML after a haploidentical allogeneic HSCT 10 months earlier. He developed recurrent fevers, dry cough, and hypoxemia, with chest computed tomography (CT) showing bibasilar consolidations and increased nodularity without increased lymphadenopathy. His symptoms initially improved with antibiotic and antifungal therapy, but his follow-up chest CT showed progression of disease despite symptomatic improvement. Epstein–Barr virus (EBV) was detected in his blood by polymerase chain reaction (PCR), and a lung biopsy revealed monomorphic PTLD with B cells positive for EBV. Unfortunately, the patient’s condition rapidly deteriorated, and he passed away prior to treatment initiation. Conclusions To our knowledge, this is the first reported case of an AML patient developing pulmonary PTLD relatively late in his post-transplant course in the setting of relapsed disease and salvage therapy. Pulmonary PTLD, a rare but highly lethal disorder, can imitate the symptoms and radiographic findings of pneumonia, a common diagnosis in immunocompromised AML patients. This case illustrates the importance of considering pulmonary PTLD in the differential diagnosis for pulmonary disease in AML patients with a history of HSCT, especially in the setting of progressive radiographic findings despite broad antibacterial and antifungal therapy. Further, our case demonstrates the importance of biopsy and uninterrupted EBV DNA monitoring in the definitive diagnosis of PTLD, given nonspecific symptomatology and radiographic findings.
- Published
- 2021
- Full Text
- View/download PDF
3. Venetoclax in Combination with Decitabine for Relapsed T-Cell Acute Lymphoblastic Leukemia after Allogeneic Hematopoietic Cell Transplant
- Author
-
Leena T. Rahmat, Anna Nguyen, Haifaa Abdulhaq, Sonam Prakash, Aaron C. Logan, and Gabriel N. Mannis
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Long-term disease-free survival in adults with T-cell acute lymphoblastic leukemia (T-ALL) remains poor, particularly after relapse, with few available salvage options. Preclinical data suggest that inhibition of the antiapoptotic protein BCL-2 (B-cell lymphoma 2) either alone or in combination with other agents, may be a unique therapeutic approach for the treatment of T-ALL. We present a case of a young male with T-ALL, relapsed after allogeneic hematopoietic stem cell transplant, who achieved a second complete remission following salvage therapy with combined venetoclax and decitabine. Assessment of measurable residual disease by next generation sequencing showed no evidence of residual disease of a sensitivity of 1 × 10−6. While the combination of venetoclax and hypomethylating agents has shown promise in the treatment of relapsed/refractory AML, and to our knowledge, this is the first report of this combination demonstrating clinical activity in relapsed/refractory T-ALL.
- Published
- 2018
- Full Text
- View/download PDF
4. The Goldilocks Dilemma in AML: Too Young and Fit, but Not Young and Fit Enough
- Author
-
Emma C. St. Martin, Tian Yi Zhang, and Gabriel N. Mannis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
- Full Text
- View/download PDF
5. Data from Inhibition of MET Signaling with Ficlatuzumab in Combination with Chemotherapy in Refractory AML: Clinical Outcomes and High-Dimensional Analysis
- Author
-
Charalambos Andreadis, Frank McCormick, Gabriela K. Fragiadakis, Pamela N. Munster, David P. Carbone, Chun J. Ye, Vivian Weinberg, Michael Flanders, Emma McMahon, Karin M. Gaensler, Peter H. Sayre, Thomas G. Martin, Lloyd E. Damon, Rebecca Olin, Gabriel N. Mannis, Matthew Settles, Aaron C. Logan, Tommy Jiang, Blythe Durbin-Johnson, Arjun A. Rao, Gregory K. Behbehani, Ravi K. Patel, Bradley W. Blaser, and Victoria E. Wang
- Abstract
Acute myeloid leukemia (AML) patients refractory to induction therapy or relapsed within 1 year have poor outcomes. Autocrine production of hepatocyte growth factor by myeloid blasts drives leukemogenesis in preclinical models. A phase Ib trial evaluated ficlatuzumab, a first-in-class anti-HGF antibody, in combination with cytarabine in this high-risk population. Dose-limiting toxicities were not observed, and 20 mg/kg was established as the recommended phase II dose. The most frequent treatment-related adverse event was febrile neutropenia. Among 17 evaluable patients, the overall response rate was 53%, all complete remissions. Phospho-proteomic mass cytometry showed potent on-target suppression of p-MET after ficlatuzumab treatment and that attenuation of p-S6 was associated with clinical response. Multiplexed single-cell RNA sequencing using prospectively acquired patient specimens identified IFN response genes as adverse predictive factors. The ficlatuzumab and cytarabine combination is well tolerated, with favorable efficacy. High-dimensional analyses at single-cell resolution represent promising approaches for identifying biomarkers of response and mechanisms of resistance in prospective clinical studies.Significance:This study demonstrates a favorable safety profile and promising clinical activity of ficlatuzumab and cytarabine in high-risk AML, thus supporting further investigation of this combination in a randomized trial. It also shows the utility of a novel application using multiplexed single-cell analyses to detect on-target activity and identify biomarkers of response.This article is highlighted in the In This Issue feature, p. 403
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data from Inhibition of MET Signaling with Ficlatuzumab in Combination with Chemotherapy in Refractory AML: Clinical Outcomes and High-Dimensional Analysis
- Author
-
Charalambos Andreadis, Frank McCormick, Gabriela K. Fragiadakis, Pamela N. Munster, David P. Carbone, Chun J. Ye, Vivian Weinberg, Michael Flanders, Emma McMahon, Karin M. Gaensler, Peter H. Sayre, Thomas G. Martin, Lloyd E. Damon, Rebecca Olin, Gabriel N. Mannis, Matthew Settles, Aaron C. Logan, Tommy Jiang, Blythe Durbin-Johnson, Arjun A. Rao, Gregory K. Behbehani, Ravi K. Patel, Bradley W. Blaser, and Victoria E. Wang
- Abstract
Supplementary Data from Inhibition of MET Signaling with Ficlatuzumab in Combination with Chemotherapy in Refractory AML: Clinical Outcomes and High-Dimensional Analysis
- Published
- 2023
- Full Text
- View/download PDF
7. Measurable Residual Disease Conversion Rate with Consolidation Chemotherapy in Acute Myeloid Leukemia
- Author
-
Daria Gaut, Caspian Oliai, Jonathan Boiarsky, Shiliang Zhang, Tali Azenkot, Vanessa E. Kennedy, Vishesh Khanna, Karla Olmedo Gutierrez, Navika D. Shukla, Benjamin Moskoff, Anand Ashwin Patel, Deepa Jeyakumar, Gabriel N. Mannis, Aaron C. Logan, Brian A. Jonas, and Gary J. Schiller
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
8. V-FAST Master Trial: Subgroup Analysis of Outcomes with CPX-351 Plus Midostaurin in Adults with Newly Diagnosed Acute Myeloid Leukemia By FLT3 Mutation Type
- Author
-
James McCloskey, Vinod A. Pullarkat, Gabriel N. Mannis, Tara L. Lin, Stephen A Strickland, Amir T. Fathi, Stefan Faderl, Divya Chakravarthy, Yana Lutska, Vijayalakshmi Chandrasekaran, Ronald S. Cheung, and Mark J. Levis
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
9. Value of embedded palliative care: outpatient palliative care and healthcare utilization for hematologic malignancies
- Author
-
Mazie Tsang, Kara Elizabeth Bischoff, Kelly L Schoenbeck, Kim Berry, David O'Riordan, Bita Fakhri, Sandy W Wong, Nina Shah, Rebecca L. Olin, Charalambos Andreadis, Jules Vieaux, Eve Cohen, Nancy Shepard Lopez, Gabriel N. Mannis, and Michael Rabow
- Subjects
Hematology - Published
- 2023
- Full Text
- View/download PDF
10. Outcomes with Molecularly Targeted Agents as Salvage Therapy Following Frontline Venetoclax + Hypomethylating Agent in Adults with Acute Myeloid Leukemia: A Multicenter Retrospective Analysis
- Author
-
Vishesh Khanna, Tali Azenkot, Selina (Qiuying) Liu, Jason Gilbert, Edna Cheung, Kimberly Lau, Daniel A Pollyea, Elie Traer, Brian A Jonas, Tian Y Zhang, and Gabriel N Mannis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
- Full Text
- View/download PDF
11. NCCN Guidelines Insights: Acute Myeloid Leukemia, Version 2.2021
- Author
-
Daniel A. Pollyea, Matthew J. Wieduwilt, Pankit Vachhani, Ivana Gojo, Dinesh S. Rao, Alice S. Mims, Dale L. Bixby, Jessica K. Altman, Alexander E. Perl, Jeffrey E. Lancet, Reza Nejati, Paul J. Shami, Richard Stone, Aric C. Hall, Mary Elizabeth Percival, Guido Marcucci, Kristina M. Gregory, Frederick R. Appelbaum, Jadee L. Neff, Gabriel N. Mannis, Meagan A. Jacoby, Farhad Ravandi-Kashani, Marcos de Lima, Rebecca L. Olin, James M. Foran, Martin S. Tallman, Stephen A. Strickland, Amir T. Fathi, Kendra Sweet, Amanda Przespolewski, Michael Gary Martin, Thomas Prebet, Vijaya Raj Bhatt, and Ndiya Ogba
- Subjects
Acute promyelocytic leukemia ,medicine.medical_specialty ,Myeloid ,business.industry ,Venetoclax ,MEDLINE ,Myeloid leukemia ,medicine.disease ,Clinical trial ,Leukemia ,chemistry.chemical_compound ,medicine.anatomical_structure ,Oncology ,chemistry ,hemic and lymphatic diseases ,medicine ,Treatment strategy ,Intensive care medicine ,business - Abstract
The NCCN Guidelines for Acute Myeloid Leukemia (AML) provide recommendations for the diagnosis and treatment of adults with AML based on clinical trials that have led to significant improvements in treatment, or have yielded new information regarding factors with prognostic importance, and are intended to aid physicians with clinical decision-making. These NCCN Guidelines Insights focus on recent select updates to the NCCN Guidelines, including familial genetic alterations in AML, postinduction or postremission treatment strategies in low-risk acute promyelocytic leukemia or favorable-risk AML, principles surrounding the use of venetoclax-based therapies, and considerations for patients who prefer not to receive blood transfusions during treatment.
- Published
- 2021
- Full Text
- View/download PDF
12. Hematopoietic Cell Transplantation in the Treatment of Newly Diagnosed Adult Acute Myeloid Leukemia: An Evidence-Based Review from the American Society of Transplantation and Cellular Therapy
- Author
-
Yoshihiro Inamoto, Michael R. Grunwald, Stefan O. Ciurea, Bhagirathbhai Dholaria, Cesar Rodriguez, Tania Jain, Miguel-Angel Perales, Betul Oran, Betty K. Hamilton, Firas El Chaer, Navneet S. Majhail, Martin S. Tallman, Mahmoud Aljurf, Mohamed A. Kharfan-Dabaja, Bipin N. Savani, Hien D. Liu, Arnon Nagler, Gabriel N. Mannis, Mehdi Hamadani, Zachariah DeFilipp, Gordon L. Phillips, Nina Shah, Shahrukh K. Hashmi, Paul A. Carpenter, Al Fadel AlShaibani, Frederick R. Appelbaum, Noa G. Holtzman, Steven M. Devine, and Mohamad Mohty
- Subjects
Oncology ,medicine.medical_specialty ,Context (language use) ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Maintenance therapy ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Immunology and Allergy ,Transplantation ,business.industry ,Adult Acute Myeloid Leukemia ,Cell Biology ,Hematology ,medicine.disease ,surgical procedures, operative ,Systematic review ,Graft-versus-host disease ,030220 oncology & carcinogenesis ,Cord blood ,Molecular Medicine ,business ,030215 immunology - Abstract
The role of hematopoietic cell transplantation (HCT) in the management of newly diagnosed adult acute myeloid leukemia (AML) is reviewed and critically evaluated in this evidence-based review. An AML expert panel, consisting of both transplant and nontransplant experts, was invited to develop clinically relevant frequently asked questions covering disease- and HCT-related topics. A systematic literature review was conducted to generate core recommendations that were graded based on the quality and strength of underlying evidence based on the standardized criteria established by the American Society of Transplantation and Cellular Therapy Steering Committee for evidence-based reviews. Allogeneic HCT offers a survival benefit in patients with intermediate- and high-risk AML and is currently a part of standard clinical care. We recommend the preferential use of myeloablative conditioning in eligible patients. A haploidentical related donor marrow graft is preferred over a cord blood unit in the absence of a fully HLA-matched donor. The evolving role of allogeneic HCT in the context of measurable residual disease monitoring and recent therapeutic advances in AML with regards to maintenance therapy after HCT are also discussed.
- Published
- 2021
- Full Text
- View/download PDF
13. Patterns and Predictors of Functional Decline after Allogeneic Hematopoietic Cell Transplantation in Older Adults
- Author
-
Li-Wen Huang, Ying Sheng, Charalambos Andreadis, Aaron C. Logan, Gabriel N. Mannis, Catherine C. Smith, Karin M.L. Gaensler, Thomas G. Martin, Lloyd E. Damon, Chiung-Yu Huang, and Rebecca L. Olin
- Subjects
Aging ,Regenerative Medicine ,7.1 Individual care needs ,Clinical Research ,Neoplasms ,Activities of Daily Living ,Immunology and Allergy ,Humans ,Prospective Studies ,Aged ,Transplantation ,Rehabilitation ,Hematopoietic Stem Cell Transplantation ,Functional status ,Cell Biology ,Hematology ,Middle Aged ,Allogeneic transplant ,Geriatric assessment ,Physical Rehabilitation ,Good Health and Well Being ,Older adults ,Quality of Life ,Molecular Medicine ,Mental health ,Management of diseases and conditions - Abstract
As allogeneic hematopoietic cell transplantation (alloHCT) is increasingly offered to older adults, geriatric assessment (GA) has been identified as a useful tool for predicting outcomes, particularly functional status. However, very few studies have examined the longitudinal change in GA measures in the post-alloHCT period. The objectives of this study were to describe the longitudinal change in GA and quality of life (QoL) measures after alloHCT and to identify predictors of greater functional decline post-transplantation. In this single-center prospective cohort study, patients age ≥50 years scheduled for alloHCT completed a cancer-specific GA and the Functional Assessment of Cancer Therapy-Bone Marrow Transplant (FACT-BMT) survey at baseline prior to alloHCT and then at 3, 6, and 12 months post-transplantation. Changes in GA and QoL measures at each post-transplantation time point (3, 6, and 12 months) compared to baseline were analyzed using paired t-tests. Exploration of potential predictors of greater post-transplantation functional decline, as measured by instrumental activities of daily living (IADL) and the Medical Outcomes Study Physical Health scale (MOS-PH), were examined using linear regression and the chi-square 2-sample test of proportions. Mean functional status generally exhibited a pattern of decline at 3 to 6 months post-alloHCT, with recovery to near baseline by 12 months. Mean mental health and emotional QoL were lowest at baseline and improved at all post-transplantation time points. Differences in baseline clinical characteristics were not associated with any differences in functional trajectories. Differences in baseline GA measures-patient-rated Karnofsky Performance Status, IADL, MOS-PH, Timed-Up-and-Go, Blessed Orientation-Memory-Concentration test, and Mental Health Inventory 5-also did not predict greater functional decline at 3 months. Patients whose IADL was improved or maintained at 3 months generally maintained their functional status at 6 and 12 months. Similarly, most patients who had an IADL decline at 3 months still had a functional decline at 6 months, although a proportion did have functional recovery by 12 months. Compared with patients who had improved/maintained IADL at 3 months, those with a decline in IADL at 3 months were significantly more likely to have persistent functional decline at 6 months (P < .0001) and 12 months (P=.02). In older alloHCT recipients, mean functional status declines short term after alloHCT with the possibility of recovery by 6 to 12 months, whereas mean mental and emotional health improve post-alloHCT. Functional decline at 3 months post-alloHCT is associated with persistent functional decline at 12 months.
- Published
- 2022
14. Hepatic veno-occlusive disease in allogeneic stem cell transplant recipients with prior exposure to gemtuzumab ozogamicin or inotuzumab ozogamicin
- Author
-
Lori Muffly, Abdullah Ladha, and Gabriel N. Mannis
- Subjects
Cancer Research ,medicine.medical_specialty ,Hepatic veno-occlusive disease ,Gemtuzumab ozogamicin ,CD33 ,Hepatic Veno-Occlusive Disease ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Calicheamicin ,medicine ,Humans ,Inotuzumab Ozogamicin ,Inotuzumab ozogamicin ,business.industry ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Hematology ,Jaundice ,medicine.disease ,Gemtuzumab ,Leukemia ,surgical procedures, operative ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,medicine.symptom ,business ,Stem Cell Transplantation ,030215 immunology ,medicine.drug - Abstract
Hepatic veno-occlusive disease (VOD/sinusoidal obstructive syndrome) represents a constellation of clinical findings including right upper quadrant pain, jaundice, hepatomegaly, and ascites. In the post-hematopoietic stem cell transplant (SCT) setting, the reported incidence has been 10-15%, with severe VOD historically resulting in high mortality rates. Novel agents including calicheamicin conjugated with CD33 (gemtuzumab ozogamicin; GO) and CD22 (inotuzumab ozogamicin; InO) are increasingly used for the treatment of acute myeloid leukemia and acute lymphoblastic leukemia, respectively. Both GO and InO are highly active, but also have unique hepatotoxicity profiles, including a higher risk of VOD in recipients of SCT. Introduction of GO and InO into pre-SCT leukemia management adds additional complexity to SCT patient selection and toxicity monitoring. In this article, we describe and review the risks and management associated with VOD in SCT recipients exposed to GO and InO.
- Published
- 2020
- Full Text
- View/download PDF
15. Outcomes of allogeneic transplantation after hypomethylating agents with venetoclax in acute myeloid leukemia
- Author
-
Vanessa E. Kennedy, Gavin Hui, Tali Azenkot, Daria Gaut, Matthew J. Wieduwilt, Caspian Oliai, Brian A. Jonas, Varun Mittal, Aaron C. Logan, Lori S. Muffly, and Gabriel N. Mannis
- Subjects
Leukemia, Myeloid, Acute ,Sulfonamides ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Transplantation, Homologous ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic - Published
- 2022
16. Measurable residual disease status and FLT3 inhibitor therapy in patients with FLT3-ITD mutated AML following allogeneic hematopoietic cell transplantation
- Author
-
Emily C. Liang, Rong Lu, Gabriel N. Mannis, Connie Chen, and Lori Muffly
- Subjects
Oncology ,medicine.medical_specialty ,Disease status ,Neoplasm, Residual ,Immunology ,Disease ,Biochemistry ,Article ,fluids and secretions ,Maintenance therapy ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Protein Kinase Inhibitors ,Transplantation ,Hematopoietic cell ,business.industry ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,hemic and immune systems ,Cell Biology ,Hematology ,Prognosis ,body regions ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,embryonic structures ,FLT3 Inhibitor ,business ,Tyrosine kinase - Abstract
BACKGROUND: Measurable residual disease (MRD) is associated with increased risk of relapse and death in acute myeloid leukemia (AML), even after allogeneic hematopoietic cell transplantation (HCT). Recently, next-generation sequencing (NGS) has emerged as a sensitive and specific method to detect MRD from both bone marrow and peripheral blood. NGS methods are particularly useful for detecting internal tandem duplication mutations of the FMS-like tyrosine kinase 3 gene (FLT3-ITD), which define a subgroup of AML patients who may be at higher risk for relapse and who often undergo consolidative allogeneic HCT. The SORMAIN trial recently reported a highly significant relapse-free survival benefit for the first generation FLT3 inhibitor sorafenib as post-HCT maintenance, particularly among patients with detectable MRD following HCT. In 2019 our program initiated prospective FLT3-ITD monitoring by NGS using a commercially available assay (Invivoscribe, Inc., San Diego, CA) of the peripheral blood or bone marrow throughout the first three months following HCT for all patients with FLT3-ITD mutated AML undergoing HCT. We encouraged but did not mandate FLT3 inhibitor use, regardless of MRD status. In this study, we describe the clinical impact of sensitive FLT3 MRD testing early after HCT, maintenance FLT3 inhibitor use in a non-clinical trial setting, and the ability of early post-HCT FLT3 inhibitor therapy to modify transplant outcomes. RESULTS: Thirty-six adults with FLT3-ITD mutated AML underwent HCT at Stanford University between April 2019 and August 2020 and were included in this observational study (Table 1). These patients were not enrolled in clinical trials evaluating FLT3 inhibitor therapy after HCT. Twenty-nine (81%) received pre-HCT FLT3 inhibitors, and 78% were in first complete remission (CR1) at HCT. Median follow up after HCT was 14.0 months (range 4.6-25.7). Two patients were excluded from subsequent analyses due to early graft failure and receipt of azacitadine/enasidenib maintenance, respectively. Of the evaluable cohort, 9 (26%) experienced clinical relapse at a median of 4.2 months after HCT (range 1.8-6.1), and 10 (29%) died, 5 from transplant-related mortality and 5 from AML (Figure 1A and B). Of the 34 patients evaluable for post-HCT MRD analysis, 10 (29%) had detectable MRD within the first three months while 24 (71%) remained MRD-negative (Figure 2A and B). Although there was a trend towards inferior progression-free survival (PFS) for patients with early post-HCT MRD (p = 0.13, Figure 3A), OS was not significantly impacted by MRD (p = 0.35, Figure 3B). Among the four patients with early post-HCT MRD negativity who ultimately relapsed, only one was FLT3-negative at time of relapse. Twenty-four patients (71%) received FLT3 inhibitor maintenance, initiated a median of 2.8 months after transplant (range 0.7-17.1), and the median duration was 10.6 months (range 1.7-23.7). The most common FLT3 inhibitor used was gilteritinib (n = 15), followed by midostaurin (n = 9), and sorafenib (n = 5). Among the 10 patients (29%) who did not receive post-transplant maintenance, reasons included graft-versus-host disease (GVHD) (n = 4), clinician decision (n = 3), cytopenias (n = 2), and thrombotic microangiopathy (n = 1). The use of maintenance FLT3 inhibitors led to a significantly superior PFS and OS (Figure 3C and D). Interestingly, the use of early FLT3 inhibitors post-HCT augmented PFS and OS in both MRD-negative and MRD-positive patients (Figure 3E and F), and effectively prevented clinical relapse in four patients with post-HCT MRD positivity (Figure 2A). CONCLUSION: In this cohort study, we evaluated the clinical utility of sensitive post-transplant MRD monitoring and maintenance FLT3 inhibitors on transplant outcomes. We demonstrate that clinical relapse can be prevented even in patients with post-HCT MRD through the incorporation of early FLT3 inhibitors. This finding is in keeping with the results from the SORMAIN trial. We also found that real-time sensitive MRD results allowed clinicians to begin early initiation of FLT3 inhibitor therapy, which positively affected outcomes. Figure 1 Figure 1. Disclosures Mannis: AbbVie, Agios, Astellas Pharma, Bristol Myers Squibb, Genentech, MacroGenics, Pfizer, and Stemline: Consultancy; Astex, Forty Seven Inc/Gilead, Glycomimetics, and Jazz Pharmaceuticals: Research Funding. Muffly: Adaptive: Honoraria, Other: fees for non-CME/CE services: , Research Funding; Astellas, Jasper, Adaptive, Baxalta: Research Funding; Pfizer, Amgen, Jazz, Medexus, Pfizer: Consultancy.
- Published
- 2021
17. Inhibition of MET Signaling with Ficlatuzumab in Combination with Chemotherapy in Refractory AML: Clinical Outcomes and High-Dimensional Analysis
- Author
-
Karin M.L. Gaensler, Charalambos Andreadis, Ravi K. Patel, Michael Flanders, Chun Jimmie Ye, Arjun A. Rao, Rebecca L. Olin, Vivian Weinberg, Bradley W. Blaser, Tommy Jiang, Frank McCormick, David P. Carbone, Pamela N. Munster, Aaron C Logan, Victoria E. Wang, Gregory K. Behbehani, Lloyd E. Damon, Blythe Durbin-Johnson, Matthew L. Settles, Peter H. Sayre, Gabriel N. Mannis, Thomas G. Martin, Gabriela K. Fragiadakis, and Emma McMahon
- Subjects
Myeloid ,Oncology ,Proteomics ,medicine.medical_specialty ,Childhood Leukemia ,Pediatric Cancer ,medicine.medical_treatment ,Clinical Trials and Supportive Activities ,Population ,Acute ,Antibodies ,Article ,Rare Diseases ,Clinical Research ,single cell RNA sequencing ,Internal medicine ,Ficlatuzumab ,Monoclonal ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Prospective Studies ,education ,Adverse effect ,Cancer ,Pediatric ,ficlatuzumab ,education.field_of_study ,Chemotherapy ,Leukemia ,refractory AML ,business.industry ,Evaluation of treatments and therapeutic interventions ,Myeloid leukemia ,Antibodies, Monoclonal ,Hematology ,General Medicine ,medicine.disease ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,6.1 Pharmaceuticals ,MET ,Cytarabine ,CyTOF ,business ,Febrile neutropenia ,medicine.drug - Abstract
Acute myeloid leukemia (AML) patients refractory to induction therapy or relapsed within 1 year have poor outcomes. Autocrine production of hepatocyte growth factor by myeloid blasts drives leukemogenesis in preclinical models. A phase Ib trial evaluated ficlatuzumab, a first-in-class anti-HGF antibody, in combination with cytarabine in this high-risk population. Dose-limiting toxicities were not observed, and 20 mg/kg was established as the recommended phase II dose. The most frequent treatment-related adverse event was febrile neutropenia. Among 17 evaluable patients, the overall response rate was 53%, all complete remissions. Phospho-proteomic mass cytometry showed potent on-target suppression of p-MET after ficlatuzumab treatment and that attenuation of p-S6 was associated with clinical response. Multiplexed single-cell RNA sequencing using prospectively acquired patient specimens identified IFN response genes as adverse predictive factors. The ficlatuzumab and cytarabine combination is well tolerated, with favorable efficacy. High-dimensional analyses at single-cell resolution represent promising approaches for identifying biomarkers of response and mechanisms of resistance in prospective clinical studies. Significance: This study demonstrates a favorable safety profile and promising clinical activity of ficlatuzumab and cytarabine in high-risk AML, thus supporting further investigation of this combination in a randomized trial. It also shows the utility of a novel application using multiplexed single-cell analyses to detect on-target activity and identify biomarkers of response. This article is highlighted in the In This Issue feature, p. 403
- Published
- 2021
18. V-FAST: A Phase 1b Master Trial to Investigate CPX-351 Combined with Various Targeted Agents in Patients with Previously Untreated Acute Myeloid Leukemia
- Author
-
Harry P. Erba, Gabriel N. Mannis, Heng Zou, Mark J. Levis, Ronald S. Cheung, Vinod Pullarkat, Stefan Faderl, and Tara L. Lin
- Subjects
medicine.medical_specialty ,Combination therapy ,business.industry ,Venetoclax ,Immunology ,Phases of clinical research ,Cell Biology ,Hematology ,Enasidenib ,Biochemistry ,Transplantation ,Regimen ,chemistry.chemical_compound ,chemistry ,Family medicine ,medicine ,Clinical endpoint ,Midostaurin ,business ,health care economics and organizations - Abstract
Introduction: CPX-351 (Vyxeos®; daunorubicin and cytarabine liposome for injection), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar drug ratio, has been approved by the US FDA and EMA for the treatment of adults with newly diagnosed therapy-related AML or AML with myelodysplasia-related changes. A randomized phase 3 study that evaluated patients 60 to 75 years of age with newly diagnosed high-risk/secondary AML provided the basis for approval and demonstrated that induction followed by consolidation with CPX-351 significantly improved overall survival and remission rates versus conventional 7+3, with a comparable safety profile. Several targeted therapies have demonstrated efficacy when added to cytotoxic therapy for the treatment of patients with previously untreated AML, including venetoclax (Venclexta®/Venclyxto®; a B-cell leukemia/lymphoma-2 [BCL-2] inhibitor), midostaurin (Rydapt®; a FMS-related tyrosine kinase 3 [FLT3] inhibitor), and enasidenib (Idhifa®; an isocitrate dehydrogenase 2 [IDH2] inhibitor). Preclinical data indicate synergistic activity may be achieved with CPX-351 in combination with venetoclax or midostaurin. Results of these studies suggest a rationale for the combination of targeted therapies using CPX-351 as a chemotherapy backbone. Study Design and Methods: V-FAST (Vyxeos - First Phase Assessment with Targeted Agents) is an open-label, multicenter, multi-arm, nonrandomized, phase 1b master trial (ClinicalTrials.gov #NCT04075747) to evaluate CPX-351 in combination with targeted agents (venetoclax, midostaurin, or enasidenib) in adults with previously untreated AML who are considered fit for intensive chemotherapy. As V-FAST is a master trial, it is designed to permit the expedited incorporation of CPX-351 combinations with other targeted agents into the study as it proceeds, thus ensuring a timely investigation of novel combinations moving forward. Key eligibility criteria are shown in Table 1. The primary study endpoints are to establish the recommended phase 2 dose (RP2D) and safety of each combination regimen. Secondary endpoints include remission rates (via morphologic assessment), bone marrow measurable residual disease status, and pharmacokinetics. Exploratory endpoints include duration of remission, overall survival, event-free survival, and the proportion of patients proceeding to hematopoietic cell transplantation. Patients will be monitored for safety until 1 month following the end of treatment and survival for up to 2 years following the first administration of treatment. Cytogenetic and/or molecular testing will determine which treatment arm each patient is assigned to (ie, no FLT3 or IDH2 mutations: CPX-351 plus venetoclax; FLT3 mutation: CPX-351 plus midostaurin; IDH2 mutation: CPX-351 plus enasidenib). For each combination, a dose-exploration phase (standard 3+3 design; up to 12 patients/combination) will employ dose de-escalation or escalation of CPX-351 and/or the targeted agent based on the occurrence of dose-limiting toxicities to determine a RP2D and evaluate the safety of the combination. The initial cohort for each arm will be treated with the dosing described in Table 2. In the subsequent expansion phase for each combination, an additional 20 patients will be treated to confirm the RP2D, further evaluate safety, and provide an initial assessment of efficacy. In both study phases, CPX-351 will be administered intravenously by 90-minute infusion on Days 1, 3, and 5; targeted therapies will be administered per a standard route for each agent. Patients may receive 1 to 2 induction cycles of combination therapy. Those achieving remission may receive up to 2 consolidation cycles with CPX-351 plus the targeted agent received during induction; hematopoietic stem cell transplantation is permitted in place of or following chemotherapy consolidation at the discretion of the treating physician. This study is ongoing and actively enrolling patients. Disclosures Lin: Celgene: Research Funding; Aptevo: Research Funding; Abbvie: Research Funding; Astellas Pharma: Research Funding; Ono Pharmaceutical: Research Funding; Mateon Therapeutics: Research Funding; Prescient Therapeutics: Research Funding; Pfizer: Research Funding; Celyad: Research Funding; Genetech-Roche: Research Funding; Gilead Sciences: Research Funding; Incyte: Research Funding; Trovagene: Research Funding; Tolero Pharmaceuticals: Research Funding; Seattle Genetics: Research Funding; Bio-Path Holdings: Research Funding; Jazz: Research Funding. Mannis:AbbVie, Agios, Bristol-Myers Squibb, Genentech: Consultancy; Glycomimetics, Forty Seven, Inc, Jazz Pharmaceuticals: Research Funding. Erba:Glycomimetics: Other: member of Scientific Steering Committee; AbbVie, Daiichi Sankyo, Forma, ImmunoGen, Jazz Pharmaceuticals, MacroGenics, Novartis, PTC: Research Funding; AbbVie, Agios, Celgene, Incyte, Jazz Pharmaceuticals, and Novartis: Speakers Bureau; AbbVie, Agios, Amgen, Astellas, Celgene, Daiichi Sankyo, Glycomimetics, ImmunoGen, Incyte, Jazz Pharmaceuticals, MacroGenics, Novartis, and Pfizer: Consultancy; Celgene: Other: chair of the Scientific Steering Committee; Covance (AbbVie): Other: chair of the Independent Review Committee. Levis:Daiichi-Sankyo: Honoraria; FujiFilm: Honoraria, Research Funding; Astellas: Honoraria, Research Funding; Amgen: Honoraria; Menarini: Honoraria. Zou:Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Faderl:Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Cheung:Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Pullarkat:Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; AbbVie, Inc.: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Genetech: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Dova: Consultancy, Honoraria; Servier: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. OffLabel Disclosure: This study will explore CPX-351 in combination with targeted agents for the treatment of adults with AML.
- Published
- 2020
- Full Text
- View/download PDF
19. Venetoclax monotherapy for cutaneous blastic plasmacytoid dendritic cell neoplasm
- Author
-
Irena T. Tan, Meghan M. Dickman, Danielle F Atibalentja, Kerri E. Rieger, Gabriel N. Mannis, and Matthew Schwede
- Subjects
medicine.medical_specialty ,Hematology ,business.industry ,Venetoclax ,General Medicine ,Blastic plasmacytoid dendritic cell neoplasm ,chemistry.chemical_compound ,Bridged Bicyclo Compounds ,chemistry ,Internal medicine ,Cancer research ,Medicine ,business - Published
- 2020
- Full Text
- View/download PDF
20. Hypomethylating agents super-responders: challenging the dogma of long-term remission for acute myeloid leukemia
- Author
-
Michael Ozga, Will Pulley, Joseph Maakaron, Alice S. Mims, Gabriel N. Mannis, Joshua F. Zeidner, and Matthew C. Foster
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Hematology ,business.industry ,Treatment outcome ,Myeloid leukemia ,General Medicine ,medicine.disease ,Remission induction ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,medicine ,Molecular diagnostic techniques ,Long term remission ,business - Published
- 2020
- Full Text
- View/download PDF
21. Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology
- Author
-
Steven Coutre, Alexander E. Perl, Richard Stone, Vijaya Raj Bhatt, Deniz Peker, Ndiya Ogba, Keith W. Pratz, Frederick R. Appelbaum, Martin S. Tallman, Michael Gary Martin, Thomas W. LeBlanc, Alice S. Mims, Dale L. Bixby, Aric C. Hall, Paul J. Shami, Jeffrey E. Lancet, Marcos de Lima, Stephen A. Strickland, Thomas Prebet, Melanie Fiorella, Meagan A. Jacoby, Lydia J. Hammond, Margaret R. O'Donnell, Guido Marcucci, Jessica K. Altman, Rebecca L. Olin, Daniel A. Pollyea, Gabriel N. Mannis, Matthew J. Wieduwilt, Farhad Ravandi, Eunice S. Wang, Kristina M. Gregory, James M. Foran, and Amir T. Fathi
- Subjects
0301 basic medicine ,Graft vs Host Disease ,Medical Oncology ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,HLA Antigens ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Transplantation, Homologous ,Medicine ,Aged ,business.industry ,Histocompatibility Testing ,Remission Induction ,Age Factors ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Middle Aged ,United States ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Cancer research ,business - Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia among adults and accounts for the largest number of annual deaths due to leukemias in the United States. Recent advances have resulted in an expansion of treatment options for AML, especially concerning targeted therapies and low-intensity regimens. This portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for AML focuses on the management of AML and provides recommendations on the workup, diagnostic evaluation and treatment options for younger (age
- Published
- 2019
- Full Text
- View/download PDF
22. Low‐dose lenalidomide maintenance after induction therapy in older patients with primary central nervous system lymphoma
- Author
-
Jimmy Hwang, Gabriel N. Mannis, James L. Rubenstein, Huimin Geng, and Khoan Vu
- Subjects
Male ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Article ,Central Nervous System Neoplasms ,Older patients ,Internal medicine ,Induction therapy ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Lenalidomide ,Aged ,business.industry ,Lymphoma, Non-Hodgkin ,Low dose ,Primary central nervous system lymphoma ,Induction Chemotherapy ,Hematology ,Immunotherapy ,Immunosenescence ,medicine.disease ,Lymphoma ,Treatment Outcome ,business ,medicine.drug - Published
- 2019
- Full Text
- View/download PDF
23. V-FAST master trial: Preliminary results of treatment with CPX-351 plus midostaurin in adults with newly diagnosed FLT3-mutated acute myeloid leukemia
- Author
-
James K. McCloskey, Vinod A. Pullarkat, Gabriel N. Mannis, Tara L. Lin, Stephen Anthony Strickland, Amir Tahmasb Fathi, Harry Paul Erba, Stefan Faderl, Divya Chakravarthy, Yana Lutska, Vijayalakshmi Chandrasekaran, Ronald Cheung, and Mark J. Levis
- Subjects
Cancer Research ,Oncology - Abstract
7043 Background: CPX-351 (US: Vyxeos; Europe: Vyxeos liposomal), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio, is approved for newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes in patients aged ≥1 year in the US and in adults in Europe. In a phase 3 study in older adults with newly diagnosed, high-risk/secondary AML, CPX-351 significantly improved overall survival and remission rates versus conventional 7+3, with a comparable safety profile. Preclinical data suggest CPX-351 may have synergistic activity with targeted agents, including the FLT3 inhibitor midostaurin (MID). Herein, we report preliminary results for the cohort of adults treated with CPX-351 + MID in the V-FAST (Vyxeos – First Phase Assessment with Targeted Agents) trial. Methods: V-FAST is an open-label, multicenter, multiarm, nonrandomized, phase 1b master trial (NCT04075747) to evaluate the safety and preliminary efficacy of CPX-351 combined with targeted agents (midostaurin, venetoclax, enasidenib). Eligible adults in the CPX-351 + MID cohort were aged 18 to 75 years, had newly diagnosed AML with a FLT3 internal tandem duplication (ITD) or tyrosine kinase domain (TKD) mutation, were fit for intensive chemotherapy, and had an ECOG performance status of 0 to 2. The dose-exploration phase (3+3 design) determined a recommended phase 2 dose of CPX-351 100 units/m2 (daunorubicin 44 mg/m2 + cytarabine 100 mg/m2) on Days 1, 3, and 5 + MID 50 mg BID on Days 8 to 21. There were no dose-limiting toxicities, and additional patients were enrolled in the expansion phase at this dose. Results: A total of 23 patients received CPX-351 + MID and had sufficient data to be included in the analysis (cutoff date: 1/20/2022). Patient baseline characteristics are shown in the Table. Treatment-emergent adverse events (TEAEs) in ≥40% of patients included febrile neutropenia (78%), nausea (65%), increased alanine aminotransferase (57%), leukopenia (57%), thrombocytopenia (57%), headache (43%), and hyponatremia (43%). All patients experienced a grade 3/4 TEAE, primarily hematologic events. Nonhematologic grade 3/4 TEAEs in ≥2 patients included pneumonia (17%), lung infection (13%), and hyperglycemia (9%). There were no grade 5 TEAEs and no deaths on or before Day 60. Complete remission was achieved by 18/22 (82%) evaluable patients after the first induction cycle. Conclusions: Preliminary results from the V-FAST trial suggest CPX-351 + MID is feasible, with a manageable safety profile and promising remission rates in adults with newly diagnosed AML who have a FLT3 mutation. Clinical trial information: NCT04075747. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
24. Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in frontline TP53m AML patients: Phase 1b results
- Author
-
Naval Guastad Daver, Paresh Vyas, Suman Kambhampati, Monzr M. Al Malki, Richard A. Larson, Adam Steven Asch, Gabriel N. Mannis, Wanxing Chai-Ho, Tiffany N. Tanaka, Terrence J. Bradley, Deepa Jeyakumar, Eunice S. Wang, Guan Xing, Mark Chao, Giridharan Ramsingh, Camille Renard, Indu Lal, Joshua F. Zeidner, and David Andrew Sallman
- Subjects
Cancer Research ,Oncology - Abstract
7020 Background: Magrolimab is a monoclonal antibody blocking CD47, a “don’t eat me” signal overexpressed on cancer cells such as acute myeloid leukemia (AML). This blockade induces phagocytosis of tumor cells and is synergistic with azacitidine (AZA) via upregulation of “eat me” signals. We report data from a Phase 1b trial of magrolimab+AZA in frontline TP53-mutant ( TP53m) AML. Methods: Patients (pts) with frontline AML not suitable for intensive chemotherapy received IV magrolimab starting with a priming dose (1 mg/kg) followed by ramp-up to 30 mg/kg QW or Q2W as maintenance dose. AZA 75 mg/m2 was given IV or SC on Days 1–7 of each 28-day cycle. Primary endpoints were safety/tolerability and complete remission (CR) rate by ELN 2017 criteria. Results: 72 TP53m AML pts were treated (Table). Common all-grade TEAEs were constipation (52.8%), diarrhea (47.2%), febrile neutropenia (45.8%), nausea (43.1%), fatigue (37.5%), decreased appetite (37.5%), thrombocytopenia (31.9%), peripheral edema (30.6%), and cough (30.6%). Most common Grade 3+ TEAEs were febrile neutropenia (37.5%), anemia (29.2%; Grade 3, 26.4%; Grade 4, 2.8%), thrombocytopenia (29.2%), pneumonia (26.4%), and neutropenia (20.8%). Objective response rate (ORR) by intent-to-treat was 48.6% (33.3% CR, 8.3% CR with incomplete hematologic recovery [CRi] / CR with partial hematologic recovery [CRh], 1.4% morphologic leukemia-free state [MLFS], 5.6% partial response). Stable disease was reported in 16.7%, progressive disease (PD) in 5.6%. 30- and 60-day mortalities were 8.3% and 18.1%, respectively. Response assessment was unavailable in 4.2% who discontinued due to AEs and 6.9% due to other, prior to the C3D1 assessment. Median time to CR/CRi was 2.2 months (mos; range 1.7–7.2) and to CR was 3.0 mos (range 1.8–9.6). 45.2% (14/31) of evaluable CR/CRi/CRh/MLFS pts achieved negative MRD by flow cytometry (investigator reported). Of 24 CR patients, 8 had a longitudinal TP53 VAF assessment, and 5/8 (63%) had VAF decreased to ≤5%. Treatment was stopped due to SCT in 9 pts (12.5%), PD 26 (36.1%), death 8 (11.1%), AE 13 (18.1%), and other 14 (19.4%). Median durations of CR and CR/CRi were 7.7 mos (95% CI: 4.7, 10.9) and 8.7 mos (95% CI: 5.3, 10.9), respectively. Median overall survival (OS) for the 72 pts was 10.8 mos (95% CI: 6.8, 12.8) with median follow up 8.3 mos. Conclusions: In high-risk frontline TP53m AML pts unsuitable for intensive chemotherapy, magrolimab+AZA showed durable responses and encouraging OS in a single-arm study. A Phase 3 trial in TP53m AML (ENHANCE-2; NCT04778397) of this combination vs standard of care is ongoing. Clinical trial information: NCT03248479. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
25. Embedded outpatient palliative care for hematologic malignancies: Referral patterns and health care utilization
- Author
-
Mazie Tsang, Kara E. Bischoff, Kelly L. Schoenbeck, Kim Berry, David O'Riordan, Bita Fakhri, Sandy Wai Kuan Wong, Nina Shah, Eve Cohen, Nancy Shepard Lopez, Gabriel N. Mannis, and Michael W. Rabow
- Subjects
Cancer Research ,Oncology - Abstract
12117 Background: Patients with hematologic malignancies are less likely to receive outpatient palliative care (OPC) compared to patients with other cancer types. Little is known about the characteristics or health care utilization of patients with hematologic malignancies who are co-managed by OPC. In this study, we evaluated referral patterns and health care utilization of patients with hematologic malignancies who were seen in an embedded OPC clinic. Methods: We conducted a retrospective cohort study of patients who established care with an embedded OPC nurse practitioner from 3/2016 – 5/2020 at a quaternary academic medical center. We obtained information about patients’ demographics, clinical characteristics, and reasons for referral to OPC from the electronic health record. Information about costs and health care utilization were provided by our finance team. For patients who were followed by OPC for at least 6 months, we used two-tailed t-tests to compare the number of hospitalizations and emergency department (ED) visits, as well as total costs, for the 6 months before and the 6 months after initiating OPC. This was approved by the UCSF IRB. Results: A total of 120 patients received OPC. Median age was 59 years (range 24-89), 48% were female, and 64% were Non-Hispanic White. Myeloma was the most common cancer (n = 50/120, 41.7%), followed by aggressive lymphoma (n = 21/120, 17.5%), and acute myeloid leukemia (n = 18/120, 15%). The primary reason for referral was for symptom management, such as pain (60%, n = 72/120), mood symptoms (12.5%, n = 15/120), and fatigue (7.5%, n = 9/120). Ten percent (n = 12/120) were referred for goals of care conversations prior to stem cell transplant (SCT). An advance directive was on file for 29% (n = 35/120) of patients, of which 34% (n = 12/35) were completed after OPC enrollment. Of the 38 patients who died, the median time from PC enrollment to death was 15.3 months, and 39% died on hospice. For the 65 patients who were followed by OPC for at least 6 months, the total number of inpatient hospitalizations, excluding SCT, went from 0.82 to 0.54 (p = 0.11) per person in the 6 months before compared to the 6 months after initiating OPC. ED visits went from 0.28 to 0.18 (p = 0.33). The total direct cost of inpatient hospitalizations, excluding SCT, decreased from $43,428 to $13,226 (p = 0.01), and the cost of ED visits went from $640 to $297 (p = 0.32) per person. Conclusions: There is an important role for embedded OPC for patients with hematologic malignancies, long before the end-of-life period, to manage symptoms and support decision-making. OPC is associated with a trend towards lower health care utilization and decreased hospitalization costs. Prospective studies are warranted to further explore the impact of OPC on symptoms and patient/caregiver experience, as well as to clarify how OPC impacts health care utilization.
- Published
- 2022
- Full Text
- View/download PDF
26. AML-065: Measurable Residual Disease Status and FLT3 Inhibitor Therapy in Patients with FLT3-ITD-Mutated AML Following Allogeneic Hematopoietic Cell Transplantation
- Author
-
Gabriel N. Mannis, Rong Lu, Lori Muffly, Emily C. Liang, and Connie Chen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Azacitidine ,Context (language use) ,Hematology ,Disease ,Enasidenib ,body regions ,Transplantation ,medicine.anatomical_structure ,Maintenance therapy ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Bone marrow ,FLT3 Inhibitor ,business ,medicine.drug - Abstract
Context: In 2019, our transplant program initiated prospective FLT3-ITD measurable residual disease (MRD) monitoring by next-generation sequencing (NGS) of the peripheral blood or bone marrow throughout the first three months following HCT for all patients with FLT3-ITD-mutated AML undergoing HCT. We encouraged FLT3 inhibitor use as post-HCT maintenance. Objective: To evaluate the impact of post-HCT FLT3 MRD testing and FLT3 inhibitor use in a non-clinical trial setting. Design: Observational study of adults with FLT3-ITD-mutated AML who underwent allogeneic HCT at Stanford University between April 2019 and August 2020. Setting: Tertiary referral center. Patients or Other Participants: Thirty-six adults with FLT3-ITD-mutated AML are included in this study. Two patients were excluded from analyses due to early graft failure and receipt of azacitidine/enasidenib maintenance therapy. Interventions: Twenty-four patients (71%) received an FLT3 inhibitor after HCT, initiated a median of 2.8 months after transplant (range: 0.7–17.1). The median duration was 6.8 months (range: 1.1–18.8). Main Outcome Measures: Progression-free survival (PFS) and overall survival (OS). Results: Ten patients (29%) had detectable MRD within the first three months following HCT, while 24 (71%) patients remained MRD-negative. Although there was a trend toward inferior PFS for patients with early post-HCT MRD (p = 0.12), OS was not significantly impacted by MRD (p = 0.66). FLT3 inhibitor therapy improved PFS and OS, particularly in MRD-negative patients (maintenance vs no maintenance: PFS not reached vs 5.52 months, p = 0.002; OS not reached vs 5.85 months, p = 0.006). Conclusions: Clinical relapse can be prevented, even in patients with post-HCT MRD, by using early FLT3 inhibitor maintenance therapy, in keeping with the results from the SORMAIN trial. Real-time sensitive MRD testing allows clinicians to begin early initiation of FLT3 inhibitor therapy. This work was partially supported by the National Institutes of Health Grant UL1-TR003142 and Grant P30-CA124435.
- Published
- 2021
- Full Text
- View/download PDF
27. AML-084: Feasibility Study Integrating Electronic Patient-Reported Outcomes (PROs) and Palliative Care for High-Risk Acute Myeloid Leukemia (AML) Patients
- Author
-
Joshua Fronk, Irena Tan, Tian Zhang, Kavitha Ramchandran, Ji Hyun Choi, Gabriel N. Mannis, and Kristen M. Cunanan
- Subjects
Cancer Research ,medicine.medical_specialty ,Palliative care ,Referral ,business.industry ,Psychological intervention ,Cancer ,Context (language use) ,Hematology ,Subspecialty ,medicine.disease ,Hospital Anxiety and Depression Scale ,Quality of life (healthcare) ,Oncology ,Emergency medicine ,medicine ,business - Abstract
Context: Despite recent lower-intensity therapies that have improved outcomes for older AML patients, AML remains associated with poor prognosis as well as high symptom burden. While the benefits of early palliative care and electronic PROs have been well-described in oncology patients, neither have been well-studied in AML, and they have never been studied in combination. Objective: To evaluate the feasibility of a virtually mediated supportive care model utilizing both electronic PROs and palliative care for patients with AML being treated with lower-intensity therapy (NCT04885127). Design: This will be a prospective, single-arm, non-blinded trial with a year of enrollment and six months of follow-up. Setting: All patients will be enrolled at Stanford Comprehensive Cancer Center. Patients or Other Participants: We plan on enrolling 40 patients who are on first-line lower-intensity therapy for AML, defined as therapy that allows patients to receive care exclusively in the outpatient setting. Interventions: Patients who are enrolled will be referred to the subspecialty palliative care clinic for virtual consultation and monthly follow-up visits for 6 months. Patients will also be instructed on the use of Noona, a digital application that allows real-time, remote monitoring of PROs. Patients will be requested to fill out a Noona symptom questionnaire prior to each palliative care visit and will be prompted to maintain a weekly symptom diary, which will be available to both their primary leukemia and palliative care providers. They will also fill out health-related quality of life surveys, including Functional Assessment of Cancer Therapy – Leukemia (FACT-Leu), Hospital Anxiety and Depression Scale (HADS), Patient Health Questionnaire-9 (PHQ-9), and Satisfaction with Decision-Making Scale, at baseline, three, and six months. Main Outcome Measures: The primary outcome to be measured at completion of the study will be the feasibility of palliative care referral, defined as completion of the initial virtual consultation and at least 50% of the six scheduled monthly follow-up visits. Secondary outcomes will include the proportion of patients who complete the digital symptom questionnaires and the proportion of patients who complete their health-related quality of life surveys. Scores obtained from surveys will be used to generate hypotheses for future work.
- Published
- 2021
- Full Text
- View/download PDF
28. Improved outcomes of octogenarians and nonagenarians with acute myeloid leukemia in the era of novel therapies
- Author
-
Ritika Dutta, Mark Y. Jeng, Gabriel N. Mannis, Irena T. Tan, and Tian Y. Zhang
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,MEDLINE ,Myeloid leukemia ,Hematology ,business - Published
- 2020
- Full Text
- View/download PDF
29. Epstein-Barr virus-positive lymphoproliferative disorder manifesting as pulmonary disease in a patient with acute myeloid leukemia: a case report
- Author
-
Susanna Y. Miao, Gabriel N. Mannis, Ritika Dutta, Parveen Shiraz, Dora Y. Ho, Tian Y. Zhang, Paul Phan, and Sebastian Fernandez-Pol
- Subjects
Lung Diseases ,Male ,medicine.medical_specialty ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Salvage therapy ,Lymphoproliferative disorders ,lcsh:Medicine ,Case Report ,Lung biopsy ,Gastroenterology ,Post-transplant lymphoproliferative disorder ,Epstein–Barr virus ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,Biopsy ,medicine ,Humans ,Aged ,Acute myeloid leukemia ,medicine.diagnostic_test ,business.industry ,lcsh:R ,Hematopoietic Stem Cell Transplantation ,General Medicine ,medicine.disease ,Lymphoproliferative Disorders ,Pneumonia ,Leukemia, Myeloid, Acute ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Differential diagnosis ,Hematopoietic stem cell transplant ,business ,Progressive disease ,030215 immunology - Abstract
Background Patients with lymphoproliferative disorders following hematopoietic stem cell transplant (HSCT) most commonly present with fever and lymphadenopathy within the first 5 months of transplant. Pulmonary post-transplant lymphoproliferative disorder (PTLD) is a particularly aggressive and rapidly progressive disease, with high morbidity and mortality. There are a very limited number of reported pulmonary PTLD cases following HSCT in patients with acute myeloid leukemia (AML). Early diagnosis and detection of pulmonary PTLD is critical given its high lethality. However, variable clinical presentations and nonspecific radiographic findings make pulmonary PTLD difficult to distinguish from other more common causes of pulmonary disease in AML patients. Case presentation Here, we describe a 68-year-old Caucasian man who presented for salvage induction therapy following relapse of his AML after a haploidentical allogeneic HSCT 10 months earlier. He developed recurrent fevers, dry cough, and hypoxemia, with chest computed tomography (CT) showing bibasilar consolidations and increased nodularity without increased lymphadenopathy. His symptoms initially improved with antibiotic and antifungal therapy, but his follow-up chest CT showed progression of disease despite symptomatic improvement. Epstein–Barr virus (EBV) was detected in his blood by polymerase chain reaction (PCR), and a lung biopsy revealed monomorphic PTLD with B cells positive for EBV. Unfortunately, the patient’s condition rapidly deteriorated, and he passed away prior to treatment initiation. Conclusions To our knowledge, this is the first reported case of an AML patient developing pulmonary PTLD relatively late in his post-transplant course in the setting of relapsed disease and salvage therapy. Pulmonary PTLD, a rare but highly lethal disorder, can imitate the symptoms and radiographic findings of pneumonia, a common diagnosis in immunocompromised AML patients. This case illustrates the importance of considering pulmonary PTLD in the differential diagnosis for pulmonary disease in AML patients with a history of HSCT, especially in the setting of progressive radiographic findings despite broad antibacterial and antifungal therapy. Further, our case demonstrates the importance of biopsy and uninterrupted EBV DNA monitoring in the definitive diagnosis of PTLD, given nonspecific symptomatology and radiographic findings.
- Published
- 2020
30. Venetoclax and hypomethylating agent therapy in high risk myelodysplastic syndromes: a retrospective evaluation of a real-world experience
- Author
-
Ritika Dutta, Shyam A. Patel, Tian Zhang, Ravindra Majeti, Armon Azizi, Gabriel N. Mannis, Asiri Ediriwickrema, William Shomali, David J. Iberri, Bruno C. Medeiros, Jason Gotlib, and Peter L. Greenberg
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Azacitidine ,Decitabine ,Context (language use) ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Retrospective Studies ,Sulfonamides ,Venetoclax ,business.industry ,Myelodysplastic syndromes ,Retrospective cohort study ,Hematology ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Clinical trial ,Treatment Outcome ,chemistry ,Hypomethylating agent ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,business ,030215 immunology ,medicine.drug - Abstract
Treatment with hypomethylating agents (HMAs) azacitidine or decitabine is the current standard of care for high risk myelodysplastic syndromes (MDSs) but is associated with low rates of response. The limited number of treatment options for patients with high risk MDS highlights a need for new therapeutic options. Venetoclax is an inhibitor of the BCL-2 protein which, when combined with an HMA, has shown high response rates in unfit and previously untreated acute myeloid leukemia. We performed a retrospective study of high risk MDS patients receiving combination HMA plus venetoclax in order to determine their effectiveness in this context. We show that in our cohort, the combination results in high response rates but is associated with a high frequency of myelosuppression. These data highlight the efficacy of combination HMA plus venetoclax in high risk MDS, warranting further prospective evaluation in clinical trials.
- Published
- 2020
31. Serial comprehensive geriatric and quality of life assessments in adults age ≥ 50 years undergoing autologous hematopoietic cell transplantation
- Author
-
Gabriel N. Mannis, Mariam T. Nawas, Thomas G. Martin, Rebecca L. Olin, Weiyun Z. Ai, Lawrence D. Kaplan, Jeffrey L. Wolf, Aaron C Logan, Ying Sheng, Lloyd E. Damon, Chiung Yu Huang, and Charalambos Andreadis
- Subjects
medicine.medical_specialty ,Activities of daily living ,Transplantation, Autologous ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Internal medicine ,Activities of Daily Living ,medicine ,Autologous transplantation ,Humans ,030212 general & internal medicine ,Geriatric Assessment ,Aged ,Hematopoietic cell ,business.industry ,Hematopoietic Stem Cell Transplantation ,Geriatric assessment ,Natural history ,Transplantation ,Oncology ,030220 oncology & carcinogenesis ,Quality of Life ,Functional status ,Geriatrics and Gerontology ,business - Abstract
Objectives We sought to examine the natural history of geriatric assessment (GA) and quality of life (QOL) domains among adults age ≥ 50 years undergoing autologous hematopoietic cell transplantation (autoHCT). Materials and Methods A QOL tool and cancer-specific GA were completed before autoHCT in patients ≥50 years, and at 100 days, six months, and one year post-transplant. Results One hundred eighty-four patients completed the pre-transplant QOL/GA assessment, 169 (92%) completed the 100-day assessment, 162 (88%) completed the six-month assessment, and 145 (79%) completed the twelve-month assessment. Functional status, as measured by instrumental activities of daily living (IADL), decreased from baseline to day 101 (mean change −0.42 points, 95% CI, −0.75 to −0.09, p = 0.01) but returned to baseline by one year. Physical function as measured by Medical Outcomes Study-Physical Health (MOS-PH) increased by mean of 3.27 points (95% CI, −0.02 to 6.56, p = 0.05) by one year. Physician-rated KPS improved by one year, but patient-rated KPS did not. No QOL metric deteriorated from baseline. Baseline factors predictive of IADL and MOS-PH as measured over time included comorbidities and disease status at transplant. IADL and MOS-PH as measured over time were not significantly associated with age. Conclusions AutoHCT for adults age ≥ 50 years resulted in an initial decrease in functional status, with subsequent improvement back to baseline by one year. Physical health and QOL measures were improved or unchanged over time. AutoHCT is well tolerated in well selected older patients, using patient reported geriatric metrics as outcomes.
- Published
- 2020
32. Functional Status as Measured by Geriatric Assessment Predicts Inferior Survival in Older Allogeneic Hematopoietic Cell Transplantation Recipients
- Author
-
Charalambos Andreadis, Gabriel N. Mannis, Chiung Yu Huang, Thomas G. Martin, Lloyd E. Damon, Aaron C Logan, Catherine C. Smith, Karin M.L. Gaensler, Rebecca L. Olin, Li-Wen Huang, Michael A. Steinman, and Ying Sheng
- Subjects
Male ,Myeloid ,medicine.medical_specialty ,Aging ,Activities of daily living ,Allogeneic transplantation ,Clinical Sciences ,Immunology ,Acute ,Article ,Disease-Free Survival ,Clinical Research ,Internal medicine ,Activities of Daily Living ,Medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Geriatric Assessment ,Aged ,Cancer ,Transplantation ,Leukemia ,business.industry ,Prevention ,Hazard ratio ,Rehabilitation ,Hematopoietic Stem Cell Transplantation ,Geriatric assessment ,Functional status ,Hematology ,Middle Aged ,Allografts ,Confidence interval ,Survival Rate ,Leukemia, Myeloid, Acute ,Good Health and Well Being ,Female ,business - Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) has been increasingly offered to older adults with hematologic malignancies. However, optimal methods to determine fitness for alloHCT have yet to be defined. We evaluated the ability of a comprehensive geriatric assessment (CGA) to predict post-alloHCT outcomes in a single-center prospective cohort study of patients age 50 years and older. Outcomes included overall survival (OS), progression-free survival (PFS), and nonrelapse mortality (NRM). A total of 148 patients were included, with a median age of 62 years (range, 50 to 76 years). In multivariate regression analysis, several CGA measures of functional status were predictive of post-alloHCT outcomes, after adjusting for traditional prognostic factors. Any deficit in instrumental activities of daily living (IADL) was associated with inferior OS (hazard ratio [HR], 1.81, 95% confidence interval [CI], 1.07 to 3.08; P = .03) and PFS (HR, 1.85; 95% CI, 1.15 to 2.99; P = .01). A Medical Outcomes Study Physical Health scale (MOS-PH) score
- Published
- 2020
33. Preliminary Results By Age Group of Treatment with CPX-351 Plus Venetoclax in Adults with Newly Diagnosed AML: Subgroup Analysis of the V-FAST Phase 1b Master Trial
- Author
-
Vinod Pullarkat, Mark J. Levis, Gabriel N. Mannis, Tara L. Lin, Stefan Faderl, Divya Chakravarthy, Vijayalakshmi Chandrasekaran, Ronald S. Cheung, and Harry P. Erba
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
- Full Text
- View/download PDF
34. Correction to: Measurable residual disease status and FLT3 inhibitor therapy in patients with FLT3-ITD mutated AML following allogeneic hematopoietic cell transplantation
- Author
-
Emily C. Liang, Connie Chen, Rong Lu, Gabriel N. Mannis, and Lori Muffly
- Subjects
Transplantation ,Hematology - Published
- 2022
- Full Text
- View/download PDF
35. Venetoclax in Combination with Decitabine for Relapsed T-Cell Acute Lymphoblastic Leukemia after Allogeneic Hematopoietic Cell Transplant
- Author
-
Gabriel N. Mannis, Leena T. Rahmat, Sonam Prakash, Anna Nguyen, Haifaa Abdulhaq, and Aaron C Logan
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Pediatric Cancer ,Childhood Leukemia ,T cell ,Salvage therapy ,Decitabine ,Disease ,03 medical and health sciences ,chemistry.chemical_compound ,Therapeutic approach ,Rare Diseases ,0302 clinical medicine ,Refractory ,Clinical Research ,Internal medicine ,medicine ,Cancer ,Pediatric ,Transplantation ,lcsh:RC633-647.5 ,Venetoclax ,business.industry ,Hematology ,lcsh:Diseases of the blood and blood-forming organs ,General Medicine ,Stem Cell Research ,medicine.disease ,Lymphoma ,Orphan Drug ,Good Health and Well Being ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Stem Cell Research - Nonembryonic - Non-Human ,business ,medicine.drug - Abstract
Long-term disease-free survival in adults with T-cell acute lymphoblastic leukemia (T-ALL) remains poor, particularly after relapse, with few available salvage options. Preclinical data suggest that inhibition of the antiapoptotic protein BCL-2 (B-cell lymphoma 2) either alone or in combination with other agents, may be a unique therapeutic approach for the treatment of T-ALL. We present a case of a young male with T-ALL, relapsed after allogeneic hematopoietic stem cell transplant, who achieved a second complete remission following salvage therapy with combined venetoclax and decitabine. Assessment of measurable residual disease by next generation sequencing showed no evidence of residual disease of a sensitivity of 1 × 10−6. While the combination of venetoclax and hypomethylating agents has shown promise in the treatment of relapsed/refractory AML, and to our knowledge, this is the first report of this combination demonstrating clinical activity in relapsed/refractory T-ALL.
- Published
- 2018
- Full Text
- View/download PDF
36. Durable Remissions with Ivosidenib inIDH1-Mutated Relapsed or Refractory AML
- Author
-
Courtney D. DiNardo, Hua Liu, Gail J. Roboz, Hongfang Wang, Martha Arellano, Samuel V. Agresta, Vickie Zhang, Ronan T. Swords, Martin S. Tallman, Geoffrey L. Uy, William B. Donnellan, David Dai, Stephanie M. Kapsalis, Christophe Willekens, Jessica K. Altman, James M. Foran, A. Pigneux, Meredith Goldwasser, Sung Choe, Gabriel N. Mannis, Eyal C. Attar, James L. Slack, Amir T. Fathi, Harry P. Erba, Bin Wu, Bin Fan, Robert K. Stuart, S. de Botton, Hagop M. Kantarjian, Richard Stone, Mikkael A. Sekeres, Katharine E. Yen, Hua Yang, Elie Traer, Eytan M. Stein, Robert H. Collins, Alice S. Mims, Daniel A. Pollyea, Anthony S. Stein, and Gabrielle T. Prince
- Subjects
Male ,0301 basic medicine ,Myeloid ,Pyridines ,Administration, Oral ,Cell Count ,Gastroenterology ,Hemoglobins ,0302 clinical medicine ,Recurrence ,hemic and lymphatic diseases ,Leukocytosis ,Enzyme Inhibitors ,Aged, 80 and over ,education.field_of_study ,Remission Induction ,Myeloid leukemia ,General Medicine ,Middle Aged ,Isocitrate Dehydrogenase ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Adult ,medicine.medical_specialty ,Adolescent ,Population ,Glycine ,Enasidenib ,Young Adult ,03 medical and health sciences ,Refractory ,Internal medicine ,medicine ,Humans ,education ,Survival rate ,Aged ,Dose-Response Relationship, Drug ,business.industry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,business ,Follow-Up Studies - Abstract
Mutations in the gene encoding isocitrate dehydrogenase 1 ( IDH1) occur in 6 to 10% of patients with acute myeloid leukemia (AML). Ivosidenib (AG-120) is an oral, targeted, small-molecule inhibitor of mutant IDH1.We conducted a phase 1 dose-escalation and dose-expansion study of ivosidenib monotherapy in IDH1-mutated AML. Safety and efficacy were assessed in all treated patients. The primary efficacy population included patients with relapsed or refractory AML receiving 500 mg of ivosidenib daily with at least 6 months of follow-up.Overall, 258 patients received ivosidenib and had safety outcomes assessed. Among patients with relapsed or refractory AML (179 patients), treatment-related adverse events of grade 3 or higher that occurred in at least 3 patients were prolongation of the QT interval (in 7.8% of the patients), the IDH differentiation syndrome (in 3.9%), anemia (in 2.2%), thrombocytopenia or a decrease in the platelet count (in 3.4%), and leukocytosis (in 1.7%). In the primary efficacy population (125 patients), the rate of complete remission or complete remission with partial hematologic recovery was 30.4% (95% confidence interval [CI], 22.5 to 39.3), the rate of complete remission was 21.6% (95% CI, 14.7 to 29.8), and the overall response rate was 41.6% (95% CI, 32.9 to 50.8). The median durations of these responses were 8.2 months (95% CI, 5.5 to 12.0), 9.3 months (95% CI, 5.6 to 18.3), and 6.5 months (95% CI, 4.6 to 9.3), respectively. Transfusion independence was attained in 29 of 84 patients (35%), and patients who had a response had fewer infections and febrile neutropenia episodes than those who did not have a response. Among 34 patients who had a complete remission or complete remission with partial hematologic recovery, 7 (21%) had no residual detectable IDH1 mutations on digital polymerase-chain-reaction assay. No preexisting co-occurring single gene mutation predicted clinical response or resistance to treatment.In patients with advanced IDH1-mutated relapsed or refractory AML, ivosidenib at a dose of 500 mg daily was associated with a low frequency of grade 3 or higher treatment-related adverse events and with transfusion independence, durable remissions, and molecular remissions in some patients with complete remission. (Funded by Agios Pharmaceuticals; ClinicalTrials.gov number, NCT02074839 .).
- Published
- 2018
- Full Text
- View/download PDF
37. A Phase 1 Study Evaluating ASTX727 (decitabine and cedazuridine) and Venetoclax Combination Therapy in Newly Diagnosed AML Patients Unfit for Intensive Induction Chemotherapy
- Author
-
Lixia Zhu, Danna Chan, Michael R. Savona, Aram Oganesian, Jacqueline Dillingham, Harold N. Keer, Casey O'Connell, Courtney D. DiNardo, Gail J. Roboz, Gabriel N. Mannis, Olatoyosi Odenike, Kim-Hien Dao, Prieya Wason, and Elizabeth A. Griffiths
- Subjects
Oncology ,medicine.medical_specialty ,Combination therapy ,Venetoclax ,business.industry ,Immunology ,Decitabine ,Induction chemotherapy ,Cell Biology ,Hematology ,Newly diagnosed ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Introduction: The combination of a DNA methyltransferase inhibitor (DNMTi; parenteral azacitidine or decitabine) with the BCL2 inhibitor venetoclax is a newly established standard-of-care regimen for patients with newly diagnosed acute myeloid leukemia (AML) ineligible to receive intensive induction chemotherapy (DiNardo et al, 2020). Replacing the parenteral DNMTi with an oral DNMTi with equivalent exposure may provide the benefit of reducing patient and caregiver burden of chronic parenteral therapy, and may help responding patients stay on treatment longer. ASTX727 (a fixed-dose combination of decitabine 35 mg and cedazuridine 100 mg) is an oral DNMTi that provides equivalent exposure to its parenteral DNMTi at standard dosing (Savona et al, 2020) and is under evaluation in combination with venetoclax as an all-oral regimen. Methods: This is an ongoing Phase 1 study being conducted at 7 US medical centers (ClinicalTrials.gov NCT04657081). Newly diagnosed AML patients 75 years or older, or with comorbidities that preclude use of intensive induction chemotherapy are eligible. The primary objective is to evaluate the effect of ASTX727 on the PK of venetoclax. Key secondary objectives are to evaluate the effect of venetoclax on the PK of ASTX727, and to determine the safety and efficacy profile for the combination. For Cycle 2 and beyond, ASTX727 is administered orally daily on days 1-5 and venetoclax 400 mg is administered orally daily on days 1-28 of 28-day cycles. For Cycle 1, ASTX727 is given in the same dose schedule while venetoclax is given as a ramp-up on days 1 and 2 according to the venetoclax US prescribing information (USPI); therefore, the PK studies are conducted during Cycle 2. Delay of subsequent cycles and venetoclax dose modifications for hematologic toxicities and anti-fungal concomitant medications follow the venetoclax USPI. Response assessments are evaluated using the 2017 ELN criteria (Döhner et al, 2017) and the CRh criterion (complete response [CR] with partial hematologic recovery) defined as those patients achieving marrow CR criteria but not peripheral blood count criteria and demonstrating an absolute neutrophil count >500/μL and platelet >50,000/μL (Kantarjian et al, 2017). Results: At the data cut-off date of August 1, 2021, 15 AML patients have enrolled and received study treatment. Median age is 78 years (range 66 - 84) and 9 (60%) are males. Of the 12 patients with data, 2 (17%), 6 (50%), and 4 (33%) patients are in the favorable, intermediate, and adverse risk ELN categories, respectively. Of the 15 dosed patients, 6 (40%) are diagnosed with AML with myelodysplasia-related changes. The median duration of exposure is 2 cycles (range 1-5) and 1.7 months (range 0.8-5.6). The most common adverse events (AEs) of Grade 2 or higher occurring in ≥10% of patients include neutropenia (5, 33%), febrile neutropenia (2, 13%), anemia (2, 13%), thrombocytopenia (2, 13%), vomiting (2, 13%), pneumonia (2, 13%), peripheral edema (2, 13%), hypertension (2, 13%) and vascular access complication (2, 13%). There are 17 serious AEs experienced in 7 patients; a grade 3 pneumonia and a grade 3 dysphagia are the only serious AEs assessed as related to ASTX727 and/or venetoclax and both events occurred in the same patient. Both AEs were part of the patient's medical history. Two deaths have occurred to date: one patient due to rapidly progressive disease during Cycle 2 and one patient who achieved a best response of CRh transitioned to hospice due to progressive multiple myeloma. Of 9 patients with response assessments and evaluable data, 3 (33%) achieved CR and 4 (44%) achieved CRh as the best response for a composite CR+CRh rate of 78%. Preliminary PK data available from 9 patients show venetoclax exposures are not affected by coadministration of ASTX727and are similar to historical data. Exposures of decitabine and cedazuridine are consistent with the range seen in previous studies. Updated PK, safety, and efficacy data will be provided in December 2021. Conclusions: A preliminary analysis of ASTX727 and venetoclax combination therapy in patients with newly diagnosed AML unfit for intensive induction chemotherapy demonstrate expected PK data, and a similar safety and efficacy profile to the approved combination therapy of a DNMTi and venetoclax. These preliminary data suggest that an all-oral regimen of a DNMTi in combination with venetoclax is feasible and should be investigated further. Disclosures Mannis: AbbVie, Agios, Astellas Pharma, Bristol Myers Squibb, Genentech, MacroGenics, Pfizer, and Stemline: Consultancy; Astex, Forty Seven Inc/Gilead, Glycomimetics, and Jazz Pharmaceuticals: Research Funding. Griffiths: Novartis: Honoraria; Apellis Pharmaceuticals: Research Funding; Alexion Pharmaceuticals: Consultancy, Research Funding; Boston Biomedical: Consultancy; Genentech: Research Funding; Abbvie: Consultancy, Honoraria; Takeda Oncology: Consultancy, Honoraria; Astex Pharmaceuticals: Honoraria, Research Funding; Taiho Oncology: Consultancy, Honoraria; Celgene/Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding. Savona: Geron: Consultancy, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; CTI: Consultancy, Membership on an entity's Board of Directors or advisory committees; BMS-Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; NOVARTIS: Consultancy, Membership on an entity's Board of Directors or advisory committees; Ryvu: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sierra Oncology: Consultancy, Membership on an entity's Board of Directors or advisory committees; Taiho: Consultancy, Membership on an entity's Board of Directors or advisory committees; TG Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; ALX Oncology: Research Funding; Astex: Research Funding; Incyte: Research Funding. Odenike: Celgene, Incyte, AstraZeneca, Astex, NS Pharma, AbbVie, Gilead, Janssen, Oncotherapy, Agios, CTI/Baxalta, Aprea: Research Funding; AbbVie, Celgene, Impact Biomedicines, Novartis, Taiho Oncology, Takeda: Consultancy. Roboz: MEI Pharma - IDMC Chair: Consultancy; Amgen: Consultancy; Jazz: Consultancy; Celgene: Consultancy; Agios: Consultancy; Janssen: Consultancy; Astex: Consultancy; Daiichi Sankyo: Consultancy; Janssen: Research Funding; AstraZeneca: Consultancy; Blueprint Medicines: Consultancy; Bayer: Consultancy; Actinium: Consultancy; AbbVie: Consultancy; Jasper Therapeutics: Consultancy; Helsinn: Consultancy; Glaxo SmithKline: Consultancy; Mesoblast: Consultancy; Bristol Myers Squibb: Consultancy; Novartis: Consultancy; Astellas: Consultancy; Otsuka: Consultancy; Pfizer: Consultancy; Roche/Genentech: Consultancy. Dillingham: Astex Pharmaceuticals, Inc.: Current Employment. Wason: Astex Pharmaceuticals, Inc.: Current Employment. Zhu: Astex Pharmaceuticals, Inc.: Current Employment. Chan: Astex Pharmaceuticals, Inc.: Current Employment. Keer: Astex Pharmaceuticals, Inc.: Current Employment. Oganesian: Astex Pharmaceuticals, Inc.: Current Employment. Dao: Astex Pharmaceuticals, Inc.: Current Employment. DiNardo: AbbVie: Consultancy, Research Funding; Agios/Servier: Consultancy, Honoraria, Research Funding; Novartis: Honoraria; Takeda: Honoraria; Notable Labs: Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees; ImmuneOnc: Honoraria, Research Funding; GlaxoSmithKline: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Research Funding; Foghorn: Honoraria, Research Funding; Forma: Honoraria, Research Funding; Celgene, a Bristol Myers Squibb company: Honoraria, Research Funding. OffLabel Disclosure: 1. Inqovi (35 mg decitabine and 100 mg cedazuridine) is a prescription medicine approved in the United States to treat adults with myelodysplastic syndromes and chronic myelomonocytic leukemia. In this study, Inqovi is referred as ASTX727 due to the off-label investigational use in combination with venetoclax in adults with newly diagnosed acute myeloid leukemia 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy. 2. Venclexta is a prescription medicine approved in the United States for: 1) adults with chronic lymphocytic leukemia or small lymphocytic lymphoma; 2) adults with newly diagnosed acute myeloid leukemia 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy, in combination with azacitidine, decitabine, or low-dose cytarabine. In this study, Venclexta is referred as venetoclax due to the off-label investigational use in combination with ASTX727 in adults with newly diagnosed acute myeloid leukemia 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy.
- Published
- 2021
- Full Text
- View/download PDF
38. Preliminary Results By Age Group of Treatment with CPX-351 Plus Venetoclax in Adults with Newly Diagnosed AML: Subgroup Analysis of the V-FAST Phase 1b Master Trial
- Author
-
Mark J. Levis, Vijayalakshmi Chandrasekaran, Divya Chakravarthy, Vinod Pullarkat, Gabriel N. Mannis, Harry P. Erba, Stefan Faderl, Tara L. Lin, Ronald S. Cheung, and Stephen A. Strickland
- Subjects
medicine.medical_specialty ,Group (mathematics) ,Venetoclax ,business.industry ,Immunology ,Subgroup analysis ,Cell Biology ,Hematology ,Newly diagnosed ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,business - Abstract
Introduction: CPX-351 (United States: Vyxeos ®; Europe: Vyxeos ® Liposomal), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio, is approved for the treatment of newly diagnosed therapy-related acute myeloid leukemia (AML) or AML with myelodysplasia-related changes in adults and pediatric patients aged ≥1 year in the United States and in adults in the European Union. In a phase 3 study in adults aged 60 to 75 years with newly diagnosed high-risk/secondary AML, after 5 years of follow-up CPX-351 significantly improved median overall survival and remission rates versus conventional 7+3 cytarabine/daunorubicin, with a comparable safety profile. Preclinical data suggest CPX-351 may have synergistic activity with targeted agents, including the BCL-2 inhibitor venetoclax (VEN). Interim results from the V-FAST (Vyxeos - First Phase Assessment with Targeted Agents) study have demonstrated the safety and preliminary efficacy of CPX-351 in combination with VEN or midostaurin. Herein, we report the preliminary results of a subgroup analysis of adults with newly diagnosed AML treated with CPX-351 + VEN, based on age, to provide insights into the tolerability of this novel combination in younger versus older adults with AML. Methods: V-FAST is an ongoing, open-label, multicenter, multi-arm, nonrandomized, phase 1b master trial (NCT04075747) to evaluate the safety and preliminary efficacy of CPX-351 combined with targeted agents (venetoclax, midostaurin, enasidenib). Each combination arm had a dose-exploration phase (3+3 design; n ≤12) and a subsequent expansion phase (n = 20) to determine the recommended phase 2 dose (RP2D), safety, and initial efficacy of that treatment combination. Eligible patients included adults aged 18 to ≤75 years with newly diagnosed AML who were deemed fit for intensive chemotherapy and had an ECOG performance status of 0 to 2. Patients assigned to treatment with CPX-351 + VEN were to have wild type FLT3 and IDH2 based on molecular testing. The RP2D of this treatment arm was determined to be dose level 1, in which patients received CPX-351 100 units/m 2 (daunorubicin 44 mg/m 2 + cytarabine 100 mg/m 2) on Days 1, 3, and 5 + VEN 400 mg on Days 1 to 14 of the first induction. At this dose level, 1 of 6 patients in the dose-exploration phase experienced 2 dose-limiting toxicities (grade 4 neutropenia [Day 16] and grade 4 thrombocytopenia [Day 18]) that extended beyond 49 days; no dose adjustments were required and the study enrolled an additional 21 patients at this dose level. For this subgroup analysis, patients were grouped by age: 18 to ≤59 years (younger) and 60 to ≤75 years (older). Results: A total of 21 patients received CPX-351 + VEN and had sufficient data to be included in the analysis, comprising 14 patients aged 18 to ≤59 years and 7 patients aged 60 to ≤75 years. Patient baseline characteristics are shown in Table 1. The most common TEAEs of any grade in younger adults were neutropenia, febrile neutropenia, and constipation; the most common TEAEs in older adults were febrile neutropenia, thrombocytopenia, and nausea (Table 2). The most common grade ≥3 TEAEs in both age groups included febrile neutropenia, thrombocytopenia, neutropenia, and leukopenia (Table 2). TEAEs categorized as serious events included febrile neutropenia (n = 3 [younger]; n = 1 [older]), sepsis (n = 2 [younger]), ischemic stroke (n = 1 [older]), skin infection (n = 1 [older]), and AML (n = 1 [younger]). Six evaluable patients in the younger age group and 1 in the older age group died during the study due to primary causes of relapse/progression (n = 4), adverse event (n = 2), and other (n = 1; sepsis and multiorgan failure after early termination from treatment). Median platelet and neutrophil recovery times were similar between age groups (Table 3) and consistent with the previously reported safety profile of CPX-351 monotherapy. Complete remission (CR) or CR with incomplete platelet or neutrophil recovery was achieved by 6/14 (43%) evaluable younger adults and 4/6 (67%) evaluable older adults, including 5/14 (36%) and 3/6 (50%) who achieved CR. Conclusions: This analysis of preliminary results by age group from the V-FAST study supports the conclusion that the combination of CPX-351 + VEN is equally feasible with a manageable safety profile in both younger and older adult patients with newly diagnosed AML. Promising remission rates were also reported for both age groups. Figure 1 Figure 1. Disclosures Pullarkat: Amgen, Dova, and Novartis: Consultancy, Honoraria; AbbVie, Amgen, Genentech, Jazz Pharmaceuticals, Novartis, Pfizer, and Servier: Membership on an entity's Board of Directors or advisory committees. Levis: Astellas and FujiFilm: Research Funding; Jazz: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Amgen, Astellas Pharma, Daiichi-Sankyo, FujiFilm, and Menarini: Honoraria; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Mannis: AbbVie, Agios, Astellas Pharma, Bristol Myers Squibb, Genentech, MacroGenics, Pfizer, and Stemline: Consultancy; Astex, Forty Seven Inc/Gilead, Glycomimetics, and Jazz Pharmaceuticals: Research Funding. Strickland: Sunesis: Research Funding; AbbVie, ArcherDx, Genentech, Incyte, Kura Oncology, Novartis, Pfizer, and Syros: Consultancy. Lin: AbbVie, Aptevo Therapeutics, Astellas Pharma, Bio-Path Holdings, Celgene, Celyad, Genentech-Roche, Gilead Sciences, Incyte, Jazz Pharmaceuticals, Novartis, Ono Pharmaceutical, Pfizer, Prescient Therapeutics, Seattle Genetics, Tolero, Trovagene: Research Funding. Faderl: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Chakravarthy: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Chandrasekaran: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Cheung: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Erba: AbbVie, Daiichi Sankyo, Forma, ImmunoGen, Jazz Pharmaceuticals, MacroGenics, Novartis, and PTC: Research Funding; AbbVie, Agios, Celgene, Incyte, Jazz Pharmaceuticals, and Novartis: Speakers Bureau; AbbVie, Agios, Amgen, Astellas Pharma, Celgene, Daiichi Sankyo, Glycomimetics, ImmunoGen, Incyte, Jazz Pharmaceuticals, MacroGenics, Novartis, and Pfizer: Consultancy; Glycomimetics: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Covance (AbbVie): Other: Independent Review Committee . OffLabel Disclosure: combination of CPX-351 [Vyxeos] and venetoclax in adults with previously untreated AML
- Published
- 2021
- Full Text
- View/download PDF
39. Heterogeneous Definitions of Secondary Acute Myeloid Leukemia (AML) Yield Distinct Outcomes in Response to First-Line Treatment with Hypomethylating Agents (HMA) and Venetoclax (Ven)
- Author
-
Paul Phan, Raymond Yin, Gabriel N. Mannis, Irena Tan, Matthew Schwede, and Tian Y. Zhang
- Subjects
Oncology ,medicine.medical_specialty ,Yield (engineering) ,Venetoclax ,business.industry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,First line treatment ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Ven ,Medicine ,Secondary Acute Myeloid Leukemia ,business - Abstract
Background: The combination of HMA and venetoclax is now standard of care for patients with AML who are not candidates for intensive chemotherapy. Elderly patients are more likely to have secondary AML (sAML), although the presence of an antecedent hematologic malignancy is often not apparent by history. Lindsley et al (Blood, 2015) showed that a somatic mutation in SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, or STAG2 is >95% specific for sAML and associated with worse outcomes. While outcomes with HMA/ven in patients meeting standard criteria for sAML have recently been reported (Pullarkat, ASCO 2021), we set out to conduct a real-world analysis of sAML patients receiving HMA/ven, including those with a secondary mutation profile (SMP) as described by Lindsley et al. We hypothesized that-when treated with HMA/ven-outcomes of patients with SMP may be most similar to those with de novo AML. Methods: Patients diagnosed with AML at Stanford Cancer Institute from 4/2017-3/2021 and treated with front-line HMA/ven were retrospectively reviewed. These included patients previously treated with HMA monotherapy for an antecedent hematologic malignancy and those who had previously received ≤ 3 cycles of HMA monotherapy for AML. Responses were classified per the modified International Working Group response criteria. Overall survival (OS) was assessed for all patients, and for patients who had a complete response (CR) or CR with incomplete hematologic recovery (CRi), duration of response (DoR) was also assessed. Statistical analyses were performed in R using the logrank test, with hazard ratios (HR) computed using the Cox proportional hazards model. For multivariate analyses, p-values for a specific variable were calculated using Cox proportional hazards regression. Results: 82 patients met criteria for inclusion; 78 had valid response assessments and 49 (62.8%) had achieved a CR or CRi at first response assessment. Median age was 72 years, with 3 patients younger than 60. 62 patients were male, median ECOG performance status (PS) was 1, median Charlson Comorbidity Index (CCI) was 6, median time to death or end of follow-up from the start of treatment was 366 days, and 58% of patients had adverse risk AML per ELN guidelines. Fig 1a demonstrates demographics for de novo, sAML (excluding SMP), and patients with SMP AML. 13 patients met criteria for AML-MRC, 23 patients had prior history of antecedent hematologic malignancy (18 with MDS or CMML, 5 with MDS/MPN overlap or MPN), 12 had tAML, and 20 patients possessed a SMP and did not meet criteria for the other three categories of sAML. 14 patients with de novo AML were characterized by the absence of any of the above factors. Patients with de novo AML were less likely to have adverse risk disease (29% vs. 64% in others) and had lower CCI scores (mean 5.1 vs. 6.2) but had no significant differences in age, gender, follow-up time, or PS. There was no statistically significant difference in rates of CR/CRi between the different subgroups or the different types of sAML; 69% of patients with de novo AML, 79% of SMP patients, and 57% of patients with other types of sAML achieved a CR or CRi. However, SMP patients had response durations and OS patterns similar to patients with de novo AML (Fig 1b and 1c), and when grouped with de novo patients, both DoR (HR = 3.5, p = 0.047, Fig 1d) and OS (HR = 2.1, p = 0.042, Fig 1e) were significantly longer than those of the sAML patients. Neither DoR nor OS were significantly longer when the SMP patients were grouped with sAML patients (respectively: HR = 3.3, p = 0.22, Fig 1f; HR = 1.5, p = 0.37, Fig 1g). In multivariate Cox proportional regression adjusting for age, ELN risk category, CCI, and PS, worse OS for sAML patients was maintained relative to the SMP and de novo patients (HR 2.9, p = 0.036), although the difference in DoR was no longer significant (HR 4.4, p= 0.10). Conclusions: Patients meeting standard definitions of sAML had worse outcomes than those with de novo AML when treated with HMA/ven in a retrospective, real-world analysis. Although a secondary mutation profile as described by Lindsley et al may be helpful in identifying patients with sAML, when treated with HMA/ven, patients with this profile have outcomes that align more closely with those of patients with de novo AML. Figure 1 Figure 1. Disclosures Mannis: Astex, Forty Seven Inc/Gilead, Glycomimetics, and Jazz Pharmaceuticals: Research Funding; AbbVie, Agios, Astellas Pharma, Bristol Myers Squibb, Genentech, MacroGenics, Pfizer, and Stemline: Consultancy.
- Published
- 2021
- Full Text
- View/download PDF
40. AML-196: The First-in-Class Anti-CD47 Antibody Magrolimab in Combination with Azacitidine Is Well Tolerated and Effective in AML Patients: Phase 1b Results
- Author
-
Naval Daver, Joshua F. Zeidner, Paresh Vyas, Ming Lin, Daniel J. Lee, Joanna Van Elk, David A. Sallman, Andrew Whiteley, Rami S. Komrokji, Guillermo Garcia-Manero, Deepa Jeyakumar, Jens-Peter Volkmer, Gabriel N. Mannis, Mark P. Chao, Tiffany N. Tanaka, Adam S. Asch, Richard A. Larson, Chris H. Takimoto, Roy Louis Maute, Monzr M. Al Malki, William B. Donnellan, Wanxing Chai-Ho, Guido G. Marcucci, and Suman Kambhampati
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Anemia ,Nausea ,Azacitidine ,Context (language use) ,Hematology ,Neutropenia ,medicine.disease ,Gastroenterology ,Leukemia ,Regimen ,Oncology ,Internal medicine ,medicine ,medicine.symptom ,business ,neoplasms ,Progressive disease ,medicine.drug - Abstract
Context: Magrolimab, an anti-CD47 antibody, induces tumor phagocytosis and eliminates leukemia stem cells. Azacitidine (AZA) synergizes with magrolimab by inducing prophagocytic “eat me” signals. Magrolimab + AZA is clinically effective in acute myeloid leukemia (AML) and myelodysplastic syndrome. Objective: To report Phase 1b data of magrolimab + AZA in untreated AML. Design: This is an open-label, nonrandomized, Phase 1b, interventional study. Interventions: Patients received a magrolimab priming/intrapatient dose-escalation regimen (1–30 mg/kg intravenous weekly followed by 30 mg/kg every 2 weeks in cycle 3 and beyond) and AZA (75 mg/m2 on days 1–7 on a 28-day cycle). Patients or Other Participants: Treatment-naive AML patients unfit for intensive chemotherapy. Main Outcomes Measures: Outcome measures included the percentage of patients with adverse events (AEs), objective response (OR), time to response, and duration of response. Results: Fifty-two patients were treated with magrolimab + AZA. Overall, 64% had poor-risk cytogenetics, and 65% had TP53 mutations. Magrolimab + AZA was well tolerated with a safety profile similar to AZA monotherapy. Treatment-related AEs (≥15% of patients) were anemia (31%), fatigue (19%), blood bilirubin increase (19%), neutropenia (19%), thrombocytopenia (17%), and nausea (15%). On-target anemia was generally transient and reversible, and no immune-related AEs associated with magrolimab were observed. Of 34 patients evaluable for efficacy, 22 (65%) achieved an OR, 15 (44%) achieved complete response (CR), 4 (12%) with CR with incomplete count recovery (CRi), 1 (3%) with partial response, 2 (6%) with morphological leukemia-free state (MLFS), 11 (32%) with stable disease (SD), and 1 (3%) with progressive disease (PD). Time to response was 2.04 months. In TP53-mutant patients, 15/21 (71%) achieved an OR, 10 (48%) achieved a CR, 4 (19%) with CRi, 1 (5%) with MLFS, 5 (24%) with SD, and 1 (5%) with PD. The median overall survival for TP53-mutant patients (n=34) and TP53–wild-type patients (n=16) was 12.9 and 18.9 months, respectively, with a median follow-up of 4 and 12 months, respectively. Conclusions: Magrolimab + AZA was well tolerated, with efficacy in both TP53-mutant and TP53–wild-type AML patients. A Phase 3 trial evaluating magrolimab + AZA in untreated TP53-mutant AML patients is planned (NCT04778397).
- Published
- 2021
- Full Text
- View/download PDF
41. Poster: AML-065: Measurable Residual Disease Status and FLT3 Inhibitor Therapy in Patients with FLT3-ITD-Mutated AML Following Allogeneic Hematopoietic Cell Transplantation
- Author
-
Emily C. Liang, Connie Chen, Rong Lu, Gabriel N. Mannis, and Lori Muffly
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
- Full Text
- View/download PDF
42. Poster: AML-084: Feasibility Study Integrating Electronic Patient-Reported Outcomes (PROs) and Palliative Care for High-Risk Acute Myeloid Leukemia (AML) Patients
- Author
-
Irena Tan, Ji Hyun Choi, Kristen Cunanan, Joshua Fronk, Tian Zhang, Gabriel N. Mannis, and Kavitha Ramchandran
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
- Full Text
- View/download PDF
43. Acute myeloid leukemia with t(14;21) involving RUNX1 and SYNE2: A novel favorable-risk translocation?
- Author
-
Zhongxia Qi, Boris C. Bastian, James P. Grenert, Iwei Yeh, Jingwei Yu, Gabriel N. Mannis, Scott C. Kogan, Jessica Van Ziffle, and Nicole Foley
- Subjects
Male ,Myeloid ,0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Chromosomal translocation ,cytogenetics ,Fusion gene ,chemistry.chemical_compound ,0302 clinical medicine ,Bone Marrow ,Risk Factors ,hemic and lymphatic diseases ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,next generation sequencing ,Pediatric ,Genome ,Leukemia ,Microfilament Proteins ,Nuclear Proteins ,Myeloid leukemia ,Hematology ,RUNX1 ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Core Binding Factor Alpha 2 Subunit ,Human ,Pediatric Research Initiative ,medicine.medical_specialty ,Pediatric Cancer ,Childhood Leukemia ,Oncology and Carcinogenesis ,Translocation ,Nerve Tissue Proteins ,Acute ,Biology ,Chromosomes ,03 medical and health sciences ,Rare Diseases ,Genetic ,FISH ,Genetics ,medicine ,Humans ,Oncology & Carcinogenesis ,Molecular Biology ,Aged ,Chemotherapy ,Acute myeloid leukemia ,Base Sequence ,Pair 14 ,Cytogenetics ,Chromosome ,030104 developmental biology ,chemistry ,Immunology ,Cancer research ,Pair 21 ,Chromosome 21 - Abstract
In acute myeloid leukemia (AML), a translocation between chromosomes 8q22 and 21q22 leads to the RUNX1-RUNXT1 fusion gene which, in the absence of a concomitant KIT mutation, generally portends a more favorable prognosis. Translocations at 21q22, other than those involving 8q22, are uncommon, and the specific prognostic and therapeutic implications are accordingly limited by the small number of reported cases. In this report, we describe the case of a 67-year-old gentleman who presented with AML harboring t(14;21)(q23;q22). Subsequent molecular analysis revealed mutations in RUNX1, ASXL1, and SF3B1, with translocation breakpoints identified within SYNE2 on chromosome 14 and RUNX1 on chromosome 21. The functional consequence of the DNA fusion between SYNE2 and RUNX1 is unclear. Nonetheless, despite several adverse risk factors associated with this patient's AML, he achieved a long-lasting remission with standard chemotherapy alone, potentially suggestive of a novel favorable-risk translocation in AML involving 21q22.
- Published
- 2017
- Full Text
- View/download PDF
44. Regression of methotrexate-resistant AIDS-related primary central nervous system lymphoma with lenalidomide plus combination anti-retroviral therapy
- Author
-
Gabriel N. Mannis, John-Paul J. Yu, Chia-Ching Wang, James L. Rubenstein, and Neel K. Gupta
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Drug resistance ,Article ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Acquired immunodeficiency syndrome (AIDS) ,Antiretroviral Therapy, Highly Active ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Drug Resistance, Viral ,medicine ,Humans ,General hospital ,Lenalidomide ,Lymphoma, AIDS-Related ,Acquired Immunodeficiency Syndrome ,business.industry ,Primary central nervous system lymphoma ,Hematology ,medicine.disease ,Thalidomide ,Lymphoma ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,Methotrexate ,Antiretroviral medication ,business ,030215 immunology ,medicine.drug - Abstract
A 33-year-old woman with a 2-year history of documented human immunodeficiency virus (HIV) infection presented to the emergency room at San Francisco General Hospital with a new-onset seizure disor...
- Published
- 2017
- Full Text
- View/download PDF
45. Routine Use of Gemtuzumab Ozogamicin in 7+3-Based Inductions for All 'Non-Adverse' Risk AML
- Author
-
Abdullah Ladha, Michaela Liedtke, Lori Muffly, Gavin Hui, Steven Coutre, Caroline Berube, Gabriel N. Mannis, Edna Cheung, Tian Y. Zhang, and Jason Gotlib
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,medicine.drug_class ,Gemtuzumab ozogamicin ,Immunology ,Monoclonal antibody ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Calicheamicin ,medicine ,Humans ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Gemtuzumab ,body regions ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Aminoglycosides ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,business ,030215 immunology ,Conjugate ,medicine.drug - Abstract
Introduction: The addition of gemtuzumab ozogamicin (GO) to 7+3 chemotherapy for newly diagnosed acute myeloid leukemia (AML) has been shown to significantly improve event-free survival (EFS) for cytogenetically favorable-risk AML, with marginal benefit for intermediate-risk AML, and no benefit for cytogenetically adverse-risk AML. Of note, with the exception of mutated FLT3-ITD, little is known about the impact of GO in ELN 2017-defined genotypically adverse-risk AML, and a recent randomized trial found no EFS benefit for 7+3+GO in patients (pts) with genotypically favorable-risk, NPM1-mutated AML. Since 2017, our institution incorporated GO into 7+3-based inductions for all "non-adverse" risk AML pts, as defined by wild-type FLT3 and no abnormalities on rapid FISH analysis for del(5q)/monosomy 5, del(7q)/monosomy 7, and del(20q). We report our experience treating all pts with "non-adverse" risk AML-as defined by this algorithm-with 7+3+GO. Methods: An institutional database was queried in order to identify all pts ≥18 years old who received 7+3-based chemotherapy for newly diagnosed AML between 2017 and 2020; pts who received the FDA-approved fractionated dose of GO were included in the analysis. Data collection included demographic variables, karyotype/FISH, targeted PCR analyses, and multigene NGS panels for AML-related mutations including, but not limited to, mutations in FLT3, NPM1, CEBPA, TP53, RUNX1, and ASXL1. Outcome data included response to induction, relapse, and death, as well as hematopoietic cell transplant (HCT) rates, conditioning regimens, and post-transplant complications. Results: Between January 2017 and July 2020, 96 pts received 7+3-based induction at our institution. Of these, 29 (30%) received 7+3 in combination with GO. Median age at diagnosis was 46 years (range 23-66), with 17 (59%) males. Sixteen (55%) pts had ELN favorable-risk AML (5 [31%] by cytogenetics and 11 [69%] by genotype), 6 (21%) pts had ELN intermediate-risk AML, and 7 (24%) pts had ELN adverse-risk AML (4 [57%] by cytogenetics and 3 [43%] by genotype). Median time from diagnosis to start of induction was 4 days (range 0-43). For cytogenetically adverse-risk pts, median time from diagnostic bone marrow biopsy to receipt of adverse karyotype results was 8 days (7-14). Median time from start of induction to receipt of multigene NGS results for all pts was 15 days (3-32). Overall, 22 (76%) pts achieved remission. All genotypically adverse-risk pts (1 with mutated TP53 and 2 with mutated RUNX1) were refractory to induction, while 3 of 4 (75%) cytogenetically adverse-risk pts (1 with t(6;9), 1 with monosomy 7, and 2 with 11q23 abnormalities) achieved remission. Eight of the 29 (28%) pts proceeded to HCT, including 4 adverse-risk pts. Of the adverse-risk pts, all received myeloablative conditioning prior to HCT and 3 (75%) developed veno-occlusive disease (VOD), with 2 (50%) requiring defibrotide therapy. In favorable/intermediate-risk pts, 4 (18%) proceeded to HCT (2 intermediate-risk pts in first remission and 2 favorable-risk pts in second remission). Of these, 2 (50%) received myeloablative conditioning and 1 (25%) developed VOD. At last follow-up, 23 of 29 pts (79%) remained alive, with a median overall survival not reached (range 1-29 months) and a median EFS of 20 months (9-31). The percentage of ELN favorable-, intermediate-, and adverse-risk pts who remained event-free at last follow-up was 75%, 33%, and 43%, respectively. Discussion: This single-center, retrospective cohort describes the outcomes of pts with "non-adverse" risk AML who received induction chemotherapy with 7+3+GO according to a pre-defined algorithm. Using this algorithm, 30% of all pts receiving 7+3-based inductions received GO. Of these, nearly 25% were ultimately found to have adverse-risk AML as defined by ELN 2017 criteria, largely driven by long turn-around times for karyotyping and NGS multigene panel results. No patient with genotypically adverse-risk AML by ELN criteria responded to induction chemotherapy, and 75% of cytogenetically adverse-risk pts who proceeded to HCT developed VOD. Routine use of 7+3+GO induction outside of the context of cytogenetically favorable-risk AML remains controversial, and further study is needed to define the role of GO, particularly for pts with ELN genotypically adverse-risk AML. Table Disclosures Gotlib: Blueprint Medicines Corporation: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Chair of the Response Adjudication Committee and Research Funding, Research Funding; Deciphera: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: co-chair of the Study Steering Committee and Research Funding. Liedtke:Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria; GSK: Membership on an entity's Board of Directors or advisory committees; Adaptive: Membership on an entity's Board of Directors or advisory committees; Caelum: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Muffly:Adaptive: Research Funding; Amgen: Consultancy; Servier: Research Funding. Mannis:AbbVie, Agios, Bristol-Myers Squibb, Genentech: Consultancy; Glycomimetics, Forty Seven, Inc, Jazz Pharmaceuticals: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
46. The Value of an Embedded Outpatient Palliative Care Program in Malignant Hematology: Concurrent Care and the Impact on Health Care Utilization
- Author
-
Nancy Shepard Lopez, Michael W. Rabow, Mazie Tsang, Kara Bischoff, Eve Cohen, Kelly L. Schoenbeck, Gabriel N. Mannis, and Kim Berry
- Subjects
Advance care planning ,Pediatrics ,medicine.medical_specialty ,Palliative care ,Referral ,business.industry ,Immunology ,Retrospective cohort study ,Cell Biology ,Hematology ,Emergency department ,Biochemistry ,Indirect costs ,Statistical significance ,Health care ,Medicine ,business - Abstract
Background: Patients with hematologic malignancies are less likely to be referred to palliative care (PC) compared to those with solid malignancies. There is little that is known about the healthcare utilization of patients with hematologic malignancies who receive outpatient PC concurrently with cancer-directed therapy. The aim of this study was to describe the demographic and clinical characteristics and health care utilization of patients with hematologic malignancies who received longitudinal, concurrent outpatient PC. Methods: We conducted a single-center, retrospective cohort study of all patients with hematologic malignancies who received embedded outpatient PC at the malignant hematology clinic between April 1, 2017 and December 31, 2018. Patients were referred to PC by their primary oncologist or hematology nurse practitioner (NP). Patients referred to PC were seen by a PC NP with expertise in hematologic malignancies. Follow-up visits with the outpatient PC NP were scheduled as needed. Demographics, clinical characteristics, and reasons for referral were extracted from the electronic health record. For patients who were followed by PC for at least 6 months, the number of hospitalizations and emergency department (ED) visits, as well as inpatient costs were compared from the 6 months prior to enrollment in PC to the 6 months after enrollment in PC. Approval for this study was obtained from the UCSF IRB. Results: Overall, 80 patients who were seen in the malignant hematology clinic were referred for embedded PC during our study period. Of these patients, 45% (n= 36) were female. Median age was 57 years (range: 27-89). Common primary diagnoses were myeloma (47.3%, n= 38), lymphoma (18.8%, n= 15), and acute myeloid leukemia (15%, n= 12). The most frequent reasons for referral to PC were pain (32.7%, n= 34), fatigue (16.3%, n= 17), and mood disorders (11.5%, n= 12). There were 9 referrals (8.7%) for advance care planning prior to bone marrow transplant (BMT). One quarter of the patients (n= 20) were referred to PC for two or more reasons. Patients were followed in the PC clinic for a median of 3.18 months (mean 8.03 months, range 0 - 36 months, SD 9.6 months) and patients had a median of 3 PC visits (mean 5, range 1-27, SD 6). The median overall survival of the patient population was 36.5 months (SD 35.8 months). There were 31 patients who were followed in the PC clinic for at least 6 months and were included in our healthcare utilization analyses. The total number of hospital encounters decreased from 1.48 inpatient admissions/ED visits in the 6-months before enrollment in PC to 0.71 per patient in the 6-months after enrollment in PC (p=0.04). Total inpatient direct costs per patient were $52,250.65 in the 6 months before PC enrollment and $30,360.90 in the 6 months after enrollment (p=0.18). The cost of inpatient medical hospitalizations went from $23,457.68 to $4,621.26 per patient (p=0.05), the cost of procedure-based hospitalizations, which included hospitalization for BMT, went from $27,667.84 to $25,434.48 (p=0.90), and the cost of ED visits went from $1,125.13 to $305.16 (p=0.25). Conclusion: Our study suggests that outpatient PC has an important role for patients with hematologic malignancies far upstream of the end-of-life period. Patients were referred for a wide variety of reasons, including management of various symptoms and advance care planning prior to BMT. There was a statistically significant and substantial decrease in the number of hospital and ED encounters per patient after enrollment in PC. There was also a trend towards overall cost savings, although this did not reach statistical significance. Further research is warranted to explore the effects of outpatient PC co-management on symptoms, rates of advance care planning, and patient/family experience for patients with hematologic malignancies. Disclosures Schoenbeck: American Society of Hematology: Research Funding. Mannis:Glycomimetics, Forty Seven, Inc, Jazz Pharmaceuticals: Research Funding; AbbVie, Agios, Bristol-Myers Squibb, Genentech: Consultancy.
- Published
- 2020
- Full Text
- View/download PDF
47. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia
- Author
-
Gail J. Roboz, Justin M. Watts, Denice Hickman, Vickie Zhang, Daniel A. Pollyea, Courtney D. DiNardo, Stephanie M. Kapsalis, Hagop M. Kantarjian, Anthony S. Stein, Martha Arellano, Gabrielle T. Prince, Alice S. Mims, Amir T. Fathi, Eytan M. Stein, Hua Liu, Samuel V. Agresta, Katharine E. Yen, Martin S. Tallman, Harry P. Erba, Will Donnellan, David Dai, Stéphane de Botton, Richard Stone, Sung Choe, Bin Fan, Bin Wu, Hongfang Wang, Geoffrey L. Uy, Eyal C. Attar, Jessica K. Altman, and Gabriel N. Mannis
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Myeloid ,Blood transfusion ,Nausea ,Pyridines ,medicine.medical_treatment ,Immunology ,Glycine ,Plenary Paper ,Biochemistry ,Gastroenterology ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Blood Transfusion ,Adverse effect ,Aged ,Aged, 80 and over ,business.industry ,Remission Induction ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Survival Analysis ,Isocitrate Dehydrogenase ,Discontinuation ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Hypomethylating agent ,030220 oncology & carcinogenesis ,Mutation ,Female ,medicine.symptom ,business - Abstract
Ivosidenib (AG-120) is an oral, targeted agent that suppresses production of the oncometabolite 2-hydroxyglutarate via inhibition of the mutant isocitrate dehydrogenase 1 (IDH1; mIDH1) enzyme. From a phase 1 study of 258 patients with IDH1-mutant hematologic malignancies, we report results for 34 patients with newly diagnosed acute myeloid leukemia (AML) ineligible for standard therapy who received 500 mg ivosidenib daily. Median age was 76.5 years, 26 patients (76%) had secondary AML, and 16 (47%) had received ≥1 hypomethylating agent for an antecedent hematologic disorder. The most common all-grade adverse events were diarrhea (n = 18; 53%), fatigue (n = 16; 47%), nausea (n = 13; 38%), and decreased appetite (n = 12; 35%). Differentiation syndrome was reported in 6 patients (18%) (grade ≥3 in 3 [9%]) and did not require treatment discontinuation. Complete remission (CR) plus CR with partial hematologic recovery (CRh) rate was 42.4% (95% confidence interval [CI], 25.5% to 60.8%); CR 30.3% (95% CI, 15.6% to 48.7%). Median durations of CR+CRh and CR were not reached, with 95% CI lower bounds of 4.6 and 4.2 months, respectively; 61.5% and 77.8% of patients remained in remission at 1 year. With median follow-up of 23.5 months (range, 0.6-40.9 months), median overall survival was 12.6 months (95% CI, 4.5-25.7). Of 21 transfusion-dependent patients (63.6%) at baseline, 9 (42.9%) became transfusion independent. IDH1 mutation clearance was seen in 9/14 patients achieving CR+CRh (5/10 CR; 4/4 CRh). Ivosidenib monotherapy was well-tolerated and induced durable remissions and transfusion independence in patients with newly diagnosed AML. This trial was registered at www.clinicaltrials.gov as #{"type":"clinical-trial","attrs":{"text":"NCT02074839","term_id":"NCT02074839"}}NCT02074839.
- Published
- 2019
48. Allogeneic HSCT for adult-onset leukoencephalopathy with spheroids and pigmented glia
- Author
-
Keith Van Haren, Christopher P. Hess, Ariele L. Greenfield, Matthew J. Barkovich, Jeffrey M. Gelfand, Bryce A. Mendelsohn, and Gabriel N. Mannis
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Fluid-attenuated inversion recovery ,Colony stimulating factor 1 receptor ,Leukoencephalopathy ,03 medical and health sciences ,0302 clinical medicine ,Leukoencephalopathies ,Internal medicine ,medicine ,Humans ,business.industry ,Parkinsonism ,Leukodystrophy ,Brain ,Middle Aged ,medicine.disease ,Transplantation ,030104 developmental biology ,Graft-versus-host disease ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Mutation ,Disease Progression ,Female ,Neurology (clinical) ,Microglia ,business ,Cognition Disorders ,Neuroglia ,030217 neurology & neurosurgery - Abstract
Adult-onset leukoencephalopathy with spheroids and pigmented glia (ALSP) is an autosomal dominant leukoencephalopathy caused by mutations in colony stimulating factor 1 receptor (CSF1R). Here we report clinical and imaging outcomes following allogeneic haematopoietic stem cell transplantation (HSCT) in two patients with ALSP at the University of California, San Francisco between January 2016 and December 2017. Patient 1 proceeded to transplantation at age 53 with a haplo-identical sibling donor. Patient 2, whose sister and mother had died of the disease, proceeded to transplantation at age 49 with a 12/12 human leukocyte antigen-matched unrelated donor. Both patients received reduced intensity conditioning regimens. At 28 and 26 months post-HSCT, respectively, both patients were alive, without evidence of graft-versus-host disease, with major infection at 1 year in one and new-onset seizures in the other. In both cases, neurological worsening continued post-HSCT; however, the progression in cognitive deficits, overall functional status and gait impairment gradually stabilized. There was continued progression of parkinsonism in both patients. On brain MRI, within 1 year there was stabilization of T2/FLAIR abnormalities, and after 2 years there was complete resolution of abnormal multifocal reduced diffusion. In summary, after >2 years of follow-up, allogeneic HSCT in ALSP led to interval resolution of diffusion MRI abnormalities, stabilization of T2/FLAIR MRI abnormalities, and partial clinical stabilization, supportive of treatment response. Allogeneic HSCT may be beneficial in ALSP by providing a supply of bone marrow-derived brain-engrafting myeloid cells with donor wild-type CSF1R to repopulate the microglial niche.
- Published
- 2019
49. Event free survival in adults with relapsed ALL who underwent front-line therapy with CALGB 10403
- Author
-
Michaela Liedtke, Gabriel N. Mannis, Ilana R. Yurkiewicz, Lori Muffly, Suravi Raychaudhuri, S. E. Coutre, and Bruno C. Medeiros
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Regimen ,business.industry ,Internal medicine ,Event free survival ,medicine ,Improved survival ,Front line ,Young adult ,business - Abstract
e19005 Background: CALGB 10403 is a pediatric-inspired ALL regimen that has recently been shown to have improved survival rates in adolescents and young adults with ALL when compared to historical outcomes with traditional adult ALL regimens (Stock et. al, 2019). Methods: This is a retrospective cohort study of ALL patients who received induction CALGB 10403 at Stanford University (both on and off trial), achieved CR1, and subsequently relapsed. Primary outcome of interest was event free survival from time of diagnosis. Events were defined as relapse or death. Secondary outcomes were overall survival and event free survival from first relapse. Patients were censored at time of last clinical follow up. Results: 25 patients met inclusion criteria and received front-line CALGB 10403 from April 2010 to September 2018. At the time of initial diagnosis median age was 30 years (range 18 – 39 years). 68% of patients were male. 48% of patients were overweight and 40% were obese. 76% of patients had precursor B cell ALL while 24% had T cell ALL. 12% had CNS disease at diagnosis. 36% of patients had WBC greater than 30k. 12% of patients had CRLF2 rearrangement. 12% of patients were MRD positive after first induction. 20% of patients received rituximab. Median event free survival time from diagnosis was 20 months (range 3 – 79 months) and median overall survival time was 53 months. Blinatumomab was the most common salvage therapy after 1st relapse, followed by inotuzumab. 15 patients (60%) achieved CR2, of which 4 (27%) were MRD positive after 2nd induction. 15 patients (60%) went to HSCT. Of the patients who achieved CR2, 8 relapsed for a second time. Median event free survival time after first relapse was 23 months. Survival 1 year after relapse was 60%. 11 of the 25 patients were alive at last follow up. Median follow up time of survivors was 6 years. Conclusions: This is a descriptive retrospective cohort study of adult patients in a real world setting who received CALGB 10403 induction and subsequently relapsed. Compared to other studies of relapsed ALL patients who were induced with traditional chemotherapy (Fielding et. al, 2007), survival 1 year after relapse was much higher (60% vs. 22%). As CALGB 10403 becomes an increasingly common induction regimen for AYA and adults with ALL, further outcomes study is required.[Table: see text]
- Published
- 2021
- Full Text
- View/download PDF
50. Preliminary results of V-FAST, a phase 1b master trial to investigate CPX-351 combined with targeted agents in newly diagnosed AML
- Author
-
Vinod Pullarkat, Stefan Faderl, Tara L. Lin, Vijayalakshmi Chandrasekaran, Ronald S. Cheung, Stephen A. Strickland, Divya Chakravarthy, Mark J. Levis, Harry P. Erba, and Gabriel N. Mannis
- Subjects
Drug ,Cancer Research ,Liposome ,Daunorubicin ,business.industry ,media_common.quotation_subject ,Newly diagnosed ,Pharmacology ,Oncology ,Cytarabine ,medicine ,business ,media_common ,medicine.drug - Abstract
7026 Background: CPX-351 (US: Vyxeos; EU: Vyxeos Liposomal), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar drug ratio, is approved by the US FDA and EMA for adults with newly diagnosed t-AML or AML with myelodysplasia-related changes. Preclinical data suggest CPX-351 may exert synergistic activity when combined with agents such as the BCL-2 inhibitor venetoclax (VEN) or FLT3 inhibitor midostaurin (MIDO). Methods: V-FAST (Vyxeos – First Phase Assessment With Targeted Agents) is an open-label, multicenter, phase 1b master trial (NCT04075747) to evaluate safety and establish the recommended phase 2 dose (RP2D) of CPX-351 combined with targeted agents in patients (pts) aged 18-75 y with untreated AML who are fit for intensive chemotherapy. The study includes a dose-exploration phase (3+3 design) and subsequent expansion phase. Pts received CPX-351 (dose level 1 for first induction [DL1]: 100 units/m2 on Days 1, 3, and 5) plus VEN (Arm A; DL1: 400 mg on Days 1-14), MIDO (Arm B; DL1: 50 mg BID on Days 8-21), or the IDH2 inhibitor enasidenib ([ENA] Arm C; DL1: 100 mg on Days 8-28) based on mutation testing. Results: Among 21 pts with available data enrolled by 11/06/20 (24 pts enrolled total; data cut-off: 01/19/21), the median age was 54 y (range: 35, 69). In Arm A (n = 17), 11 (65%) pts had de novo AML, 5 (29%) had an antecedent hematologic disorder (2 [12%] had myelofibrosis), and 2 (12%) had t-AML; 12 (71%) had adverse-risk AML; and 6 (35%) had mutated TP53. In Arms B (n = 3) and C (n = 1), all pts had intermediate-risk de novo AML. DL1 was the RP2D in Arms A and B; the RP2D in Arm C is still under investigation. In Arm A, 1/6 pts in the dose-exploration phase had 2 dose-limiting toxicities (DLTs) of grade 4 neutropenia and thrombocytopenia that extended beyond 49 days; no DLTs have occurred for Arms B and C. The combinations exhibited manageable safety profiles (Table). Of pts with available response data, complete remission (CR) or CR with incomplete platelet or neutrophil recovery was achieved by 6/14 (43%) pts in Arm A, including 4 (29%) with CR. All pts in Arms B and C achieved CR. Conclusions: These preliminary results suggest CPX-351 can be combined with VEN and MIDO with manageable toxicities in newly diagnosed AML pts, with DL1 determined to be the RP2D. The study is ongoing and actively enrolling pts; updated results will be presented at the meeting. Clinical trial information: NCT04075747. [Table: see text]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.