1
|
Cluzeau T, Guolo F, Chiche E, Minetto P, Rahme R, Bertoli S, Fianchi L, Micol JB, Gottardi M, Peterlin P, Galimberti S, Thomas X, Rizzuto G, Legrand O, Rondoni M, Raffoux E, Bertani G, Caulier A, D'Argenio M, Bonmati C, Billio A, Lejeune C, Scappini B, Pigneux A, Zappasodi P, Recher C, Grimaldi F, Ades L, Lemoli RM. Long-term real-world evidence of CPX-351 of high-risk patients with AML identified high rate of negative MRD and prolonged OS. Blood Adv 2025; 9:752-758. [PMID: 39454204 DOI: 10.1182/bloodadvances.2024014279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/27/2024] Open
Abstract
ABSTRACT CPX-351 has been approved for patients with therapy-related acute myeloid leukemia (t-AML) or AML with myelodysplasia-related changes (MRC-AML). No extensive data are available on measurable residual disease (MRD) and long-term clinical outcome using CPX-351 in AML in real life. We retrospectively collected data from 168 patients in 36 centers in France and Italy who had received 1 or 2 cycles of induction with CPX-351. All patients were aged >18 years and had newly diagnosed, untreated t-AML or MRC-AML. With a median follow-up of 3 years, the median overall survival (OS) was 13.3 months. The median OS was 20.4 months vs 12.9 months for patients with MRD below or above 10-3, respectively (P = .006). In a multivariate analysis, only MRD >10-3 was associated with a poorer OS (hazard ratio, 2.6; 95% confidence interval, 1.2-5.5; P = .013). We also observed a trend toward a better median OS in patients who underwent hematopoietic stem cell transplantation with MRD <10-3 (not reached vs 26.0 months; P = .06). Achievement of MRD negativity contributed to the improvement of OS in the overall population and, maybe, in patients receiving transplant. These data provide the rationale for the 2 ongoing studies evaluating CPX-351 vs 7+3 in non-MRC-AML and non-t-AML using MRD as the primary end point for ALFA-2101 phase 2 clinical trial and event-free survival for AMLSG 30-18 phase 3 clinical trial.
Collapse
Affiliation(s)
- Thomas Cluzeau
- Hematology Department, Cote d'Azur University, Centre Hospitalier Universitaire Nice, Nice, France
- Hematology Department, Nice University Hospital, Nice, France
- Hematology Department Cote d'Azur University, Mediterranean Center for Molecular Medicine, Nice, France
| | - Fabio Guolo
- Department of Internal Medicine, Clinic of Hematology, University of Genoa, Genoa, Italy
- Department of Hematology and Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Edmond Chiche
- Hematology Department, Cote d'Azur University, Centre Hospitalier Universitaire Nice, Nice, France
| | - Paola Minetto
- Department of Internal Medicine, Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Ramy Rahme
- Senior Hematology Department, Hospital Saint-Louis, Université Paris Cité, INSERM U944, Paris, France
| | - Sarah Bertoli
- Hematology Department, Institut universitaire du cancer de Toulouse-Oncopole, Centre Hospitalier Universitaire Toulouse, Toulouse, France
| | - Luana Fianchi
- Hematology Department Istituto di Ematologia, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | | | - Michele Gottardi
- Hematology Department, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Oncologico Veneto, Castelfranco Veneto, Italy
| | - Pierre Peterlin
- Hematology Department, Centre Hospitalier Universitaire Nantes, Nantes, France
| | - Sara Galimberti
- Ospedaliera Universitaria Ematologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Xavier Thomas
- Hematology Department, Hopitaux civils de Lyon, Centre Hospitalier Universitaire Lyon, Lyon, France
| | - Giuliana Rizzuto
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera Universitaria Papa Giovanni XXIII, Bergamo, Italy
| | | | - Michela Rondoni
- Unità Operativa di Cancerologia di Ematologia, Azienda Unità Sanitaria Locale della Romagna, Ravenna, Italy
| | | | - Giambattista Bertani
- Hematology Department, S.C. Ematologia, ASST Grande Ospedale Metropolitano, Niguarda Ca' Granda Milano, Milan, Italy
| | - Alexis Caulier
- Department of Hematology, Centre Hospitalier Universitaire Amiens, Amiens, France
| | | | - Caroline Bonmati
- Hematology Department, centre hospitalier régional universitaire Nancy, Nancy, France
| | - Atto Billio
- Hematology Department, Ospedale di Bolzano, Bolzano, Italy
| | - Caroline Lejeune
- Hematology Department, L'institut de Cancérologie de la Loire, centre hospitalier universitaire Saint-Etienne, Saint-Etienne, France
| | - Barbara Scappini
- Dipartimento di Oncologia-SODc Ematologia, Azienda Ospedaliero - Universitaria Careggi, Florence, Italy
| | - Arnaud Pigneux
- Hematology Department, centre hospitalier universitaire Bordeaux, Bordeaux, France
| | - Patrizia Zappasodi
- Hematology Department, Clinica Ematologica, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Christan Recher
- Hematology Department, Institut universitaire du cancer de Toulouse-Oncopole, Centre Hospitalier Universitaire Toulouse, Toulouse, France
| | - Francesco Grimaldi
- Dipartimento di Medicina Clinica e Chirurgia, Azienda Ospedaliera Universitaria Federico II di Napoli, Naples, Italy
| | - Lionel Ades
- Senior Hematology Department, Hospital Saint-Louis, Université Paris Cité, INSERM U944, Paris, France
| | - Roberto M Lemoli
- Department of Internal Medicine, Clinic of Hematology, University of Genoa, Genoa, Italy
- Department of Hematology and Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
2
|
Rodríguez-Arbolí E, Rodríguez-Veiga R, Soria-Saldise E, Bergua JM, Caballero-Velázquez T, Arnán M, Vives S, Serrano J, Bernal T, Martínez-Sánchez P, Tormo M, Rodríguez-Medina C, Herrera-Puente P, Lavilla-Rubira E, Boluda B, Acuña-Cruz E, Cano I, Cáceres S, Ballesteros J, Falantes J, Martínez-Cuadrón D, Pérez-Simón JA, Montesinos P. A phase 2, multicenter, clinical trial of CPX-351 in older patients with secondary or high-risk acute myeloid leukemia: PETHEMA-LAMVYX. Cancer 2025; 131:e35618. [PMID: 39476204 DOI: 10.1002/cncr.35618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND LAMVYX was a multicenter, single-arm, phase 2 trial designed to validate the safety and efficacy of CPX-351 in patients aged 60-75 years with newly diagnosed, secondary acute myeloid leukemia and to generate evidence on key issues not addressed in the preceding regulatory pivotal trial. METHODS The primary end point of the study was the complete remission (CR)/CR with incomplete hematologic recovery (CRi) rate after induction. Eligible patients were recommended to undergo allogeneic hematopoietic stem cell transplantation after the first consolidation cycle. Alternatively, patients could undergo up to six maintenance cycles with CPX-351. RESULTS Twenty-nine patients (49%; 95% exact confidence interval [CI], 37%-62%) patients achieved a CR/CRi after one or two cycles of induction, with a measurable residual disease negativity rate of 67% as assessed by centralized, multiparameter flow cytometry. Among patients who had serial next-generation sequencing analyses available, clearance of somatic mutations that were present at diagnosis was achieved in 7 (35%). The median follow-up among survivors was 16.8 months (range, 8.7-24.3 months). The median event-free survival was 3.0 months (95% CI, 1.4-7.3 months), and the median overall survival was 7.4 months (95% CI, 3.7-12.7 months). In landmark analyses at day +100 from diagnosis, the 1-year overall and event-free survival rate among patients who underwent allogeneic hematopoietic stem cell transplantation was 70% (95% CI, 47%-100%) and 70% (95% CI, 47%-100%), respectively. The corresponding values were 89% (95% CI, 71%-100%) and 44% (95% CI, 21%-92%), respectively, for patients who entered the maintenance phase. No significant longitudinal changes were observed in severity index or quality-of-life visual analog scale scores. CONCLUSIONS The current data provide novel insights that might inform the clinical positioning and optimal use of CPX-351, complementing previous results (ClinicalTrials.gov identifier NCT04230239).
Collapse
Affiliation(s)
- Eduardo Rodríguez-Arbolí
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS/CSIC), University of Seville, Seville, Spain
| | | | - Elena Soria-Saldise
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS/CSIC), University of Seville, Seville, Spain
| | - Juan M Bergua
- Department of Hematology, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Teresa Caballero-Velázquez
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS/CSIC), University of Seville, Seville, Spain
| | - Montserrat Arnán
- Department of Hematology, Institut Català d'Oncología, Hospital Duran i Reynals, Barcelona, Spain
| | - Susana Vives
- Department of Hematology, Hospital Germans Trias i Pujol-Institut Catalá d'Oncologia, Badalona, Spain
| | - Josefina Serrano
- Department of Hematology, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba, Universidad de Córdoba, Cordoba, Spain
| | - Teresa Bernal
- Department of Hematology, Hospital Universitario Central Asturias, Instituto Universitario del Principado de Asturias, Instituto de Oncología del Principado de Asturias, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Oviedo, Spain
| | | | - Mar Tormo
- Department of Hematology, Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria del Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Carlos Rodríguez-Medina
- Department of Hematology, Hospital Universitario Dr Negrín, Las Palmas de Gran Canaria, Spain
| | | | | | - Blanca Boluda
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Evelyn Acuña-Cruz
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Isabel Cano
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Sara Cáceres
- Department of Hematology, Hospital San Pedro de Alcántara, Cáceres, Spain
| | | | - José Falantes
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS/CSIC), University of Seville, Seville, Spain
| | | | - José A Pérez-Simón
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS/CSIC), University of Seville, Seville, Spain
| | - Pau Montesinos
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| |
Collapse
|
3
|
Riva C, Minetto P, Chies M, Vernarecci C, Colombo N, Rosellini S, Parodi A, Tedone E, Carminati E, Nurra C, Puglisi F, Frello M, Maio E, Ferro B, Zecchetti G, Fugazza G, Nozza P, Cea M, Lemoli RM, Guolo F. Comparing Outcomes Between CPX-351 and Fludarabine-Based Induction in Secondary Acute Myeloid Leukemia in the Real-World Setting: The Prognostic Role of Measurable Residual Disease. Hematol Oncol 2025; 43:e70005. [PMID: 39635952 DOI: 10.1002/hon.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
Secondary acute myeloid leukemia (s-AML) is associated with inferior outcomes with conventional chemotherapy, and fludarabine combinations (FLAG-Ida) have been tested to improve results. More recently, CPX-351 resulted superior to conventional 3 + 7 in s-AML patients. In the UK NCRI AML19 trial, AML patients were randomized to receive either FLAG-Ida or CPX-351. Subgroup analysis revealed better overall survival (OS) with CPX-351 in patients with MDS-related gene mutations. Unfortunately, minimal residual disease (MRD) was evaluated only in a minority of patients. The aim of this study was to further disclose the mechanisms of higher efficacy of CPX-351 in s-AML, with a focus on MRD. We analyzed 183 consecutive s-AML elderly patients (median age 69, range 60-77) treated with CPX-351 (n = 82) or receiving FLAG-Ida (n = 101). Complete remission (CR) rate and MRD negativity probability were higher among patients receiving CPX-351 (MRD negative CR rate of 40/64, 62.5%, compared to 25/55, 45% in patients who received FLAG-Ida, p < 0.05). Extra-hematological toxicity was lower in CPX-351 arm, and 30 days mortality was 3.6% and 8% in patients receiving CPX-351 or FLAG-Ida, respectively. Notably, 21/64 (32.8%) CR patients treated with CPX 351 underwent allogeneic stem cell transplantation (HSCT), compared to 5/55 with FLAG-Ida (9%, p < 0.05). Overall, CPX-351 treatment resulted in higher OS (median OS 17.7 vs. 11.2 months with FLAG-Ida, p < 0.05). The better outcome of CPX-351 compared to FLAG-Ida in our cohort may be explained by a greater probability of MRD negativity, alongside with an improved tolerance, enabling more s-AML patients to undergo HSCT.
Collapse
Affiliation(s)
- Carola Riva
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Paola Minetto
- Dipartimento di Oncologia ed Ematologia (DipOE), IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Chies
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Chiara Vernarecci
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Nicoletta Colombo
- Dipartimento di Anatomia Patologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sara Rosellini
- Dipartimento di Anatomia Patologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessia Parodi
- Dipartimento di Anatomia Patologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Elisabetta Tedone
- Dipartimento di Anatomia Patologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Enrico Carminati
- Dipartimento di Oncologia ed Ematologia (DipOE), IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Clara Nurra
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Francesco Puglisi
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Michela Frello
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Elena Maio
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Beatrice Ferro
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Giada Zecchetti
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Giuseppina Fugazza
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
| | - Paolo Nozza
- Dipartimento di Anatomia Patologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Cea
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
- Dipartimento di Oncologia ed Ematologia (DipOE), IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberto Massimo Lemoli
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
- Dipartimento di Oncologia ed Ematologia (DipOE), IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Guolo
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
- Dipartimento di Oncologia ed Ematologia (DipOE), IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
4
|
Uy GL, Pullarkat V, Baratam P, Stuart RK, Walter RB, Winer ES, Wang Q, Faderl S, Chakravarthy D, Menno D, Cheung RS, Lin TL. Lower-intensity CPX-351 plus venetoclax induction for adults with newly diagnosed AML unfit for intensive chemotherapy. Blood Adv 2024; 8:6248-6256. [PMID: 39418644 PMCID: PMC11699081 DOI: 10.1182/bloodadvances.2024013687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Preclinical data suggest a rationale for combining CPX-351, a dual-drug liposomal encapsulation of daunorubicin and cytarabine, with venetoclax, a B-cell lymphoma-2 inhibitor. This phase 1b study evaluated lower-intensity CPX-351 combined with venetoclax in adults with acute myeloid leukemia (AML) considered unfit/ineligible for intensive chemotherapy. In a dose-exploration phase using a 3+3 design, patients received stepwise dosing of CPX-351 IV on days 1 and 3 plus venetoclax 400 mg orally on days 2 to 21 per cycle to determine the recommended phase 2 dose (RP2D) for this combination. During the expansion phase, additional patients received CPX-351 plus venetoclax at the identified RP2D. The primary end points were the RP2D and safety of CPX-351 combined with venetoclax. Secondary end points included preliminary efficacy and pharmacokinetics. Overall, 35 patients were enrolled in the study. A RP2D of CPX-351 30 units/m2 (daunorubicin 13.2 mg/m2 and cytarabine 30 mg/m2) plus venetoclax 400 mg was established. The safety profile of the combination was consistent with the known safety profiles of CPX-351 and venetoclax. Complete remission (CR)/CR with incomplete hematologic recovery (CRi) was achieved by 17 of 35 patients (49%), all after cycle 1; of these, 14 were negative for measurable residual disease. CR was achieved by 1 of 8 patients (13%) with a mutation in TP53, and CR/CRi was achieved by 15 of 26 patients (58%) with wild-type TP53. This study highlights that lower-intensity therapy of CPX-351 plus venetoclax as induction therapy provides a well-tolerated treatment option in adults with AML deemed unfit for intensive chemotherapy. This trial was registered at www.ClinicalTrials.gov as #NCT04038437.
Collapse
Affiliation(s)
- Geoffrey L. Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Vinod Pullarkat
- Division of Leukemia, Department of Hematology & Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Praneeth Baratam
- Hematology/Oncology Division, Department of Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Robert K. Stuart
- Hematology/Oncology Division, Department of Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Roland B. Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Eric S. Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Qi Wang
- Jazz Pharmaceuticals, Philadelphia, PA
| | | | | | | | | | - Tara L. Lin
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
5
|
Shahzad M, Amin MK, Daver NG, Shah MV, Hiwase D, Arber DA, Kharfan-Dabaja MA, Badar T. What have we learned about TP53-mutated acute myeloid leukemia? Blood Cancer J 2024; 14:202. [PMID: 39562552 PMCID: PMC11576745 DOI: 10.1038/s41408-024-01186-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
TP53 is a tumor suppressor gene frequently mutated in human cancers and is generally associated with poor outcomes. TP53 mutations are found in approximately 5% to 10% of patients with de novo acute myeloid leukemia (AML), more frequently observed in elderly patients and those with therapy-related AML. Despite recent advances in molecular profiling and the emergence of targeted therapies, TP53-mutated AML remains a challenge to treat. Current treatment strategies, including conventional chemotherapy, hypomethylating agents, and venetoclax-based therapies, have shown limited efficacy in TP53-mutated AML, with low response rates and poor overall survival. Allogeneic hematopoietic stem cell transplantation is a potentially curative option; however, its efficacy in TP53-mutated AML depends on comorbid conditions and disease status at transplantation. Novel therapeutic modalities, including immune-based therapies, did show promise in early-phase studies but did not translate into effective therapies in randomized controlled trials. This review provides a comprehensive overview of TP53 mutations in AML, outcomes based on allelic burden, clinical implications, and therapeutic challenges.
Collapse
Affiliation(s)
- Moazzam Shahzad
- Division of Hematology and Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Muhammad Kashif Amin
- Division of Hematologic Malignancies & Cellular Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Devendra Hiwase
- Department of Hematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Talha Badar
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
6
|
Prajapati SK, Kumari N, Bhowmik D, Gupta R. Recent advancements in biomarkers, therapeutics, and associated challenges in acute myeloid leukemia. Ann Hematol 2024; 103:4375-4400. [PMID: 39198271 DOI: 10.1007/s00277-024-05963-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Acute myeloid leukemia (AML) is a common type of leukemia that has a high mortality rate. The reasons for high mortality in patients with AML are therapeutic resistance, limited ability to predict duration of response, and likelihood of cancer relapse. Biomarkers, such as leukemic stem cell biomarkers, circulatory biomarkers, measurable residual disease biomarkers, and molecular biomarkers, are used for prognosis, diagnosis, and targeted killing to selectively eliminate AML cells. They also play an indispensable role in providing therapeutic resistance to patients with AML. Therefore, targeting these biomarkers will improve the outcome of AML patients. However, identifying biomarkers that can differentiate between treatment-responsive and non-responsive AML patients remains a challenge. This review discusses recent advancements in AML biomarkers, promising therapeutics, and associated challenges in the treatment of AML.
Collapse
Affiliation(s)
- Suresh Kumar Prajapati
- Research and Development Cell, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, India
| | - Neha Kumari
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India
| | - Doulat Bhowmik
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India
| | - Reeshu Gupta
- Research and Development Cell, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, India.
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India.
| |
Collapse
|
7
|
Mehta P, Campbell V, Maddox J, Floisand Y, Kalakonda AJM, O'Nions J, Coats T, Nagumantry S, Hodgson K, Whitmill R, Amott I, Flynn G, Taussig D, Zhao R, Cunningham N, Roset M, Cuadras D, Medalla G, Kuter H, Park S, Legg A, Khan AB. CREST-UK: Real-world effectiveness, safety and outpatient delivery of CPX-351 for first-line treatment of newly diagnosed therapy-related AML and AML with myelodysplasia-related changes in the UK. Br J Haematol 2024; 205:1326-1336. [PMID: 38977430 DOI: 10.1111/bjh.19622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
Favourable outcomes with CPX-351 versus conventional 7 + 3 were demonstrated in the pivotal phase III trial in adults aged 60-75 years with newly diagnosed, highrisk/secondary acute myeloid leukaemia (AML). As a complement to the clinical trial and to address important data gaps, the CPX-351 Real-World Effectiveness and SafeTy (CREST-UK; NCT05169307) study evaluated the use of CPX-351 in routine clinical practice in the UK, in 147 patients with newly diagnosed therapy-related AML or AML with myelodysplasia-related changes. Best response of complete remission or complete remission with incomplete platelet or neutrophil recovery was achieved by 53% of evaluable patients. Kaplan-Meier median overall survival (OS) was 12.8 months (95% confidence interval 9.2-15.3). Fifty (34%) patients proceeded to haematopoietic cell transplantation (HCT); median OS landmarked from the HCT date was not reached. There were no new safety concerns with CPX-351 identified in CREST-UK. Patients treated with CPX-351 in the outpatient setting spent an average of 24.4, 16.7, 28.2, and 27.7 fewer days on the ward compared with inpatients during first induction, second induction, first consolidation, and second consolidation, respectively. The results from CREST-UK provide valuable insights into the effectiveness, safety, and outpatient delivery of CPX-351 in routine clinical practice in the UK.
Collapse
Affiliation(s)
- Priyanka Mehta
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - Jamie Maddox
- South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| | - Yngvar Floisand
- Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Anita J M Kalakonda
- Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
- NIHR CRN North West Coast, Liverpool, UK
| | - Jenny O'Nions
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Thomas Coats
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | | | | | | | - Ian Amott
- University Hospitals Derby & Burton, NHS Foundation Trust, Derby, UK
| | - Gillian Flynn
- Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | - David Taussig
- Haemato-Oncology Department, Royal Marsden Hospital, UK
- Institute of Cancer Research, London, UK
| | - Rui Zhao
- Torbay and South Devon NHS Foundation Trust, UK
| | | | | | | | | | | | | | | | - Anjum B Khan
- Leeds Teaching Hospitals NHS Foundation Trust, Leeds, UK
| |
Collapse
|
8
|
Travaglini S, Marinoni M, Visconte V, Guarnera L. Therapy-Related Myeloid Neoplasm: Biology and Mechanistic Aspects of Malignant Progression. Biomedicines 2024; 12:1054. [PMID: 38791019 PMCID: PMC11118122 DOI: 10.3390/biomedicines12051054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) arise after a documented history of chemo/radiotherapy as treatment for an unrelated condition and account for 10-20% of myelodysplastic syndromes and acute myeloid leukemia. T-MN are characterized by a specific genetic signature, aggressive features and dismal prognosis. The nomenclature and the subsets of these conditions have changed frequently over time, and despite the fact that, in the last classification, they lost their autonomous entity status and became disease qualifiers, the recognition of this feature remains of major importance. Furthermore, in recent years, extensive studies focusing on clonal hematopoiesis and germline variants shed light on the mechanisms of positive pressure underpinning the rise of driver gene mutations in t-MN. In this manuscript, we aim to review the evolution of defining criteria and characteristics of t-MN from a clinical and biological perspective, the advances in mechanistic aspects of malignant progression and the challenges in prevention and management.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimiliano Marinoni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
9
|
Sackstein P, Williams A, Zemel R, Marks JA, Renteria AS, Rivero G. Transplant Eligible and Ineligible Elderly Patients with AML-A Genomic Approach and Next Generation Questions. Biomedicines 2024; 12:975. [PMID: 38790937 PMCID: PMC11117792 DOI: 10.3390/biomedicines12050975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024] Open
Abstract
The management of elderly patients diagnosed with acute myelogenous leukemia (AML) is complicated by high relapse risk and comorbidities that often preclude access to allogeneic hematopoietic cellular transplantation (allo-HCT). In recent years, fast-paced FDA drug approval has reshaped the therapeutic landscape, with modest, albeit promising improvement in survival. Still, AML outcomes in elderly patients remain unacceptably unfavorable highlighting the need for better understanding of disease biology and tailored strategies. In this review, we discuss recent modifications suggested by European Leukemia Network 2022 (ELN-2022) risk stratification and review recent aging cell biology advances with the discussion of four AML cases. While an older age, >60 years, does not constitute an absolute contraindication for allo-HCT, the careful patient selection based on a detailed and multidisciplinary risk stratification cannot be overemphasized.
Collapse
Affiliation(s)
- Paul Sackstein
- Lombardi Cancer Institute, School of Medicine, Georgetown University, Washington, DC 20007, USA; (P.S.); (R.Z.); (J.A.M.)
| | - Alexis Williams
- Department of Medicine, New York University, New York, NY 10016, USA;
| | - Rachel Zemel
- Lombardi Cancer Institute, School of Medicine, Georgetown University, Washington, DC 20007, USA; (P.S.); (R.Z.); (J.A.M.)
| | - Jennifer A. Marks
- Lombardi Cancer Institute, School of Medicine, Georgetown University, Washington, DC 20007, USA; (P.S.); (R.Z.); (J.A.M.)
| | - Anne S. Renteria
- Lombardi Cancer Institute, School of Medicine, Georgetown University, Washington, DC 20007, USA; (P.S.); (R.Z.); (J.A.M.)
| | - Gustavo Rivero
- Lombardi Cancer Institute, School of Medicine, Georgetown University, Washington, DC 20007, USA; (P.S.); (R.Z.); (J.A.M.)
| |
Collapse
|
10
|
Bono R, Sapienza G, Tringali S, Rotolo C, Patti C, Mulè A, Calafiore V, Santoro A, Castagna L. Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia. Cells 2024; 13:755. [PMID: 38727291 PMCID: PMC11083056 DOI: 10.3390/cells13090755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Refractory acute myeloid leukaemia is very difficult to treat and represents an unmet clinical need. In recent years, new drugs and combinations of drugs have been tested in this category, with encouraging results. However, all treated patients relapsed and died from the disease. The only curative option is allogeneic transplantation through a graft from a healthy donor immune system. Using myeloablative conditioning regimens, the median overall survival regimens is 19%. Several so-called sequential induction chemotherapies followed by allogeneic transplantation conditioned by reduced intensity regimens have been developed, improving the overall survival to 25-57%. In the allogeneic transplantation field, continuous improvements in practices, particularly regarding graft versus host disease prevention, infection prevention, and treatment, have allowed us to observe improvements in survival rates. This is true mainly for patients in complete remission before transplantation and less so for refractory patients. However, full myeloablative regimens are toxic and carry a high risk of treatment-related mortality. In this review, we describe the results obtained with the different modalities used in more recent retrospective and prospective studies. Based on these findings, we speculate how allogeneic stem cell transplantation could be modified to maximise its therapeutic effect on refractory acute myeloid leukaemia.
Collapse
Affiliation(s)
- Roberto Bono
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (R.B.); (G.S.); (S.T.); (C.R.)
| | - Giuseppe Sapienza
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (R.B.); (G.S.); (S.T.); (C.R.)
| | - Stefania Tringali
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (R.B.); (G.S.); (S.T.); (C.R.)
| | - Cristina Rotolo
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (R.B.); (G.S.); (S.T.); (C.R.)
| | - Caterina Patti
- Onco-Hematology Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (C.P.); (A.M.); (V.C.)
| | - Antonino Mulè
- Onco-Hematology Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (C.P.); (A.M.); (V.C.)
| | - Valeria Calafiore
- Onco-Hematology Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (C.P.); (A.M.); (V.C.)
| | - Alessandra Santoro
- Onco-Hematology and Cell Manipulation Laboratory Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy;
| | - Luca Castagna
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy; (R.B.); (G.S.); (S.T.); (C.R.)
| |
Collapse
|
11
|
Buffet R. How lipid coating soothes the gut in AML therapy. Blood 2024; 143:1559-1561. [PMID: 38635249 DOI: 10.1182/blood.2024023908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
|
12
|
Renga G, Nunzi E, Stincardini C, Pariano M, Puccetti M, Pieraccini G, Di Serio C, Fraziano M, Poerio N, Oikonomou V, Mosci P, Garaci E, Fianchi L, Pagano L, Romani L. CPX-351 exploits the gut microbiota to promote mucosal barrier function, colonization resistance, and immune homeostasis. Blood 2024; 143:1628-1645. [PMID: 38227935 DOI: 10.1182/blood.2023021380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
ABSTRACT CPX-351, a liposomal combination of cytarabine plus daunorubicin, has been approved for the treatment of adults with newly diagnosed, therapy-related acute myeloid leukemia (AML) or AML with myelodysplasia-related changes, because it improves survival and outcome of patients who received hematopoietic stem cell transplant compared with the continuous infusion of cytarabine plus daunorubicin (referred to as "7 + 3" combination). Because gut dysbiosis occurring in patients with AML during induction chemotherapy heavily affects the subsequent phases of therapy, we have assessed whether the superior activity of CPX-351 vs "7 + 3" combination in the real-life setting implicates an action on and by the intestinal microbiota. To this purpose, we have evaluated the impact of CPX-351 and "7 + 3" combination on mucosal barrier function, gut microbial composition and function, and antifungal colonization resistance in preclinical models of intestinal damage in vitro and in vivo and fecal microbiota transplantation. We found that CPX-351, at variance with "7 + 3" combination, protected from gut dysbiosis, mucosal damage, and gut morbidity while increasing antifungal resistance. Mechanistically, the protective effect of CPX-351 occurred through pathways involving both the host and the intestinal microbiota, namely via the activation of the aryl hydrocarbon receptor-interleukin-22 (IL-22)-IL-10 host pathway and the production of immunomodulatory metabolites by anaerobes. This study reveals how the gut microbiota may contribute to the good safety profile, with a low infection-related mortality, of CPX-351 and highlights how a better understanding of the host-microbiota dialogue may contribute to pave the way for precision medicine in AML.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | | | - Claudia Di Serio
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Maurizio Fraziano
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Noemi Poerio
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | | | - Paolo Mosci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luana Fianchi
- Division of Hematology, Policlinico Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Livio Pagano
- Division of Hematology, Policlinico Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- San Raffaele Sulmona, Sulmona, Italy
| |
Collapse
|
13
|
Santoro N, Salutari P, Di Ianni M, Marra A. Precision Medicine Approaches in Acute Myeloid Leukemia with Adverse Genetics. Int J Mol Sci 2024; 25:4259. [PMID: 38673842 PMCID: PMC11050344 DOI: 10.3390/ijms25084259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The treatment of acute myeloid leukemia (AML) with adverse genetics remains unsatisfactory, with very low response rates to standard chemotherapy and shorter durations of remission commonly observed in these patients. The complex biology of AML with adverse genetics is continuously evolving. Herein, we discuss recent advances in the field focusing on the contribution of molecular drivers of leukemia biogenesis and evolution and on the alterations of the immune system that can be exploited with immune-based therapeutic strategies. We focus on the biological rationales for combining targeted therapy and immunotherapy, which are currently being investigated in ongoing trials, and could hopefully ameliorate the poor outcomes of patients affected by AML with adverse genetics.
Collapse
Affiliation(s)
- Nicole Santoro
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
| | - Prassede Salutari
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
| | - Mauro Di Ianni
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
- Department of Medicine and Science of Aging, “G.D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Andrea Marra
- Laboratory of Molecular Medicine and Biotechnology, Department of Medicine, University Campus Bio-Medico of Rome, 00128 Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00196 Rome, Italy
| |
Collapse
|
14
|
Palmieri R, Billio A, Ferrara F, Galimberti S, Lemoli RM, Todisco E, Moretti F, Venditti A. Literature review and expert opinion on the treatment of high-risk acute myeloid leukemia in patients who are eligible for intensive chemotherapy. Front Oncol 2024; 14:1367393. [PMID: 38444680 PMCID: PMC10912626 DOI: 10.3389/fonc.2024.1367393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024] Open
Abstract
In patients with Acute Myeloid Leukemia (AML), the assessment of disease risk plays a central role in the era of personalized medicine. Indeed, integrating baseline clinical and biological features on a case-by-case basis is not only essential to select which treatment would likely result in a higher probability of achieving complete remission, but also to dynamically customize any subsequent therapeutic intervention. For young high-risk patients with low comorbidities burden and in good general conditions (also called "fit" patients), intensive chemotherapy followed by allogeneic stem cell transplantation still represents the backbone of any therapeutic program. However, with the approval of novel promising agents in both the induction/consolidation and the maintenance setting, the algorithms for the management of AML patients considered eligible for intensive chemotherapy are in constant evolution. In this view, we selected burning issues regarding the identification and management of high-risk AML, aiming to provide practical advice to facilitate their daily clinical management in patients considered eligible for intensive chemotherapy.
Collapse
Affiliation(s)
- Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | - Atto Billio
- Division of Hematology and Bone Marrow Transplant (BMT), Hospital S. Maurizio, Bolzano, Italy
| | | | - Sara Galimberti
- Hematology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto M. Lemoli
- Cattedra di Ematologia, Dipartimento di Medicina Interna (DiMI), Università di Genova, Genova, Italy
- Clinica Ematologica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Martino, Genova, Italy
| | - Elisabetta Todisco
- Struttura Complessa (SC) Ematologia, Ospedale Busto Arsizio, Azienda Socio Sanitaria Territoriale (ASST) Valle Olona, Varese, Italy
| | - Federico Moretti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| |
Collapse
|
15
|
Stafylidis C, Vlachopoulou D, Kontandreopoulou CN, Diamantopoulos PΤ. Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia. J Clin Med 2024; 13:1082. [PMID: 38398394 PMCID: PMC10889132 DOI: 10.3390/jcm13041082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Acute myeloid leukemia (AML) remains a challenging hematologic malignancy. The presence of TP53 mutations in AML poses a therapeutic challenge, considering that standard treatments face significant setbacks in achieving meaningful responses. There is a pressing need for the development of innovative treatment modalities to overcome resistance to conventional treatments attributable to the unique biology of TP53-mutated (TP53mut) AML. This review underscores the role of TP53 mutations in AML, examines the current landscape of treatment options, and highlights novel therapeutic approaches, including targeted therapies, combination regimens, and emerging immunotherapies, as well as agents being explored in preclinical studies according to their potential to address the unique hurdles posed by TP53mut AML.
Collapse
Affiliation(s)
| | | | | | - Panagiotis Τ. Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.S.); (D.V.); (C.-N.K.)
| |
Collapse
|
16
|
Martínez-Cuadrón D, Megías-Vericat JE, Gil C, Bernal T, Tormo M, Martínez-Sánchez P, Rodríguez-Medina C, Serrano J, Herrera P, Simón JAP, Sayas MJ, Bergua J, Lavilla-Rubira E, Amigo ML, Benavente C, Lorenzo JLL, Pérez-Encinas MM, Vidriales MB, Colorado M, De Rueda B, García-Boyero R, Marini S, García-Suárez J, López-Pavía M, Gómez-Roncero MI, Noriega V, López A, Labrador J, Cabello A, Sossa C, Algarra L, Stevenazzi M, Solana-Altabella A, Boluda B, Montesinos P. Outcomes after intensive chemotherapy for secondary and myeloid-related changes acute myeloid leukemia patients aged 60 to 75 years old: a retrospective analysis from the PETHEMA registry. Haematologica 2024; 109:115-128. [PMID: 37199127 PMCID: PMC10772508 DOI: 10.3324/haematol.2022.282506] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
Treatment options for patients with secondary acute myeloid leukemia (sAML) and AML with myeloid-related changes (AMLMRC) aged 60 to 75 years are scarce and unsuitable. A pivotal trial showed that CPX-351 improved complete remission with/without incomplete recovery (CR/CRi) and overall survival (OS) as compared with standard "3+7" regimens. We retrospectively analyze outcomes of 765 patients with sAML and AML-MRC aged 60 to 75 years treated with intensive chemotherapy, reported to the PETHEMA registry before CPX-351 became available. The CR/CRi rate was 48%, median OS was 7.6 months (95% confidence interval [CI]: 6.7-8.5) and event-free survival (EFS) 2.7 months (95% CI: 2-3.3), without differences between intensive chemotherapy regimens and AML type. Multivariate analyses identified age ≥70 years, Eastern Cooperative Oncology Group performance status ≥1 as independent adverse prognostic factors for CR/CRi and OS, while favorable/intermediate cytogenetic risk and NPM1 were favorable prognostic factors. Patients receiving allogeneic stem cell transplant (HSCT), autologous HSCT, and those who completed more consolidation cycles showed improved OS. This large study suggests that classical intensive chemotherapy could lead to similar CR/CRi rates with slightly shorter median OS than CPX-351.
Collapse
Affiliation(s)
- David Martínez-Cuadrón
- Hospital Universitari i Politècnic La Fe, Valencia, Spain; Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid.
| | - Juan E Megías-Vericat
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe, Valencia
| | - Cristina Gil
- Hospital General Universitario de Alicante, Alicante
| | - Teresa Bernal
- Hospital Universitario Central de Asturias, Asturias, Instituto Universitario de Oncología del Principado de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, IUOPA, ISPA
| | - Mar Tormo
- Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, Valencia
| | | | | | | | | | - José A Pérez Simón
- Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS / CISC), Universidad de Sevilla, Sevilla
| | | | | | | | | | | | | | | | | | | | | | | | - Sandra Marini
- Centro Hospitalar e Universitário de Coimbra, EPE, Coimbra
| | | | | | | | | | | | | | - Ana Cabello
- Hospital Universitario Nuestra Señora de Candelaria, Tenerife
| | - Claudia Sossa
- FOSCAL, Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga, Bucaramanga
| | | | | | | | - Blanca Boluda
- Hospital Universitari i Politècnic La Fe, Valencia, Spain; Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia
| | - Pau Montesinos
- Hospital Universitari i Politècnic La Fe, Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid
| |
Collapse
|
17
|
Röllig C. Improving long-term outcomes with intensive induction chemotherapy for patients with AML. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:175-185. [PMID: 38066853 PMCID: PMC10727094 DOI: 10.1182/hematology.2023000504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Intensive chemotherapy in combination with allogeneic hematopoietic cell transplantation and supportive care can induce long-term remissions in around 50% of acute myeloid leukemia patients eligible for intensive treatment. Several treatment optimization trials helped to refine schedule and dosing of the historic "7 + 3" combination. Together with the addition of novel agents, increased efficacy and tolerability led to improved long-term outcomes. Unsatisfactory outcomes in fit elderly patients and unfavorable genetic subgroups have raised the question of whether less-intensive venetoclax-based approaches may be beneficial as an alternative. Although tempting and worth exploring, this issue will remain controversial until the results of randomized comparisons appear. To date, intensive chemotherapy remains the only evident curative treatment option for long-term disease eradication in a fixed treatment time. With the advent of more novel agents and advances in minimal residual disease (MRD) detection and maintenance approaches, the face of intensive treatment could change in many ways. Several are being explored in clinical trials, such as (1) combinations of more than 1 novel agent with the intensive backbone, (2) head-to-head comparisons of novel agents, (3) replacement or dose reduction of cytotoxic components such as anthracyclines, and (4) MRD-guided escalation and de-escalation strategies. The combination of intensive treatment with individualized tailored innovative strategies will most certainly reduce treatment-related toxicities and increase the chances for long-term remission in the future.
Collapse
Affiliation(s)
- Christoph Röllig
- Department of Internal Medicine I, University Hospital TU Dresden, Dresden, Germany
| |
Collapse
|
18
|
Andrews C, Pullarkat V, Recher C. CPX-351 in FLT3-mutated acute myeloid leukemia. Front Oncol 2023; 13:1271722. [PMID: 38044999 PMCID: PMC10691756 DOI: 10.3389/fonc.2023.1271722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
CPX-351, a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a 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 a pivotal phase III trial, CPX-351 significantly improved overall survival compared with standard-of-care 7 + 3 chemotherapy (7 days cytarabine; 3 days daunorubicin) in adults aged 60-75 years with newly diagnosed high-risk or secondary AML (median = 9.56 months vs. 5.95 months; hazard ratio = 0.69; 95% confidence interval = 0.52-0.90; p = 0.003). Approximately 30% of patients with newly diagnosed AML have mutations in the FLT3 gene, which may be associated with poor outcomes. Here, we review the current in vitro, clinical, and real-world evidence on the use of CPX-351 in patients with AML and mutations in FLT3. Additionally, we review preliminary data from clinical trials and patient case reports that suggest the combination of CPX-351 with FLT3 inhibitors may represent another treatment option for patients with FLT3 mutation-positive AML.
Collapse
Affiliation(s)
- Claire Andrews
- Department of Haematology, St Vincent’s University Hospital, Dublin, Ireland
| | - Vinod Pullarkat
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Christian Recher
- Service d’Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
19
|
Benedetti E, Traverso G, Pucci G, Morganti R, Bramanti E, Lippolis P, Susini MC, Mazzantini E, Giubbolini R, Mavilia F, Capochiani E, Neri E, Arena C, Cerri F, De Simone L, Valentini K, Stella SM, Ricchiuto V, Bruno B, Galimberti S. Impact of different chemotherapy regimens on intestinal mucosal injury assessed with bedside ultrasound: a study in 213 AML patients. Front Oncol 2023; 13:1272072. [PMID: 38023169 PMCID: PMC10646482 DOI: 10.3389/fonc.2023.1272072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Neutropenic enterocolitis (NEC) is a life-threatening complication reported in patients with acute myeloid leukemia (AML) following chemotherapy (CHT). Intensive induction and consolidation CHT may damage intestinal mucosa leading to a NEC episode (NECe). NEC reported mortality may be up to 30-60%. Early US-guided bed-side diagnosis and prompt treatment may substantially improve the survival. An emerging worldwide concern is the intestinal colonization by multi-drug-resistant bacteria especially when patients are exposed to chemotherapy regimens potentially correlated to mucosal damage. Methods In our study we prospectively enrolled all AML patients admitted in our leukemia unit to receive intensive induction and consolidation chemotherapy and experiencing chemotherapy-induced-neutropenia (CHTN). Results and discussion Overall, we enrolled N=213 patients from 2007 to March 2023. We recorded N=465 CHTN, and N=42 NECe (9.0% incidence). The aim of our study was to assess which chemotherapy regimens are more associated with NEC. We found that ALM1310, followed by 7 + 3 (daunorubicin), 7 + 3 (idarubicin), 5 + 3 + 3 (cytarabine, etoposide, idarubicin), and AML1310 (consolidation) were associated with a statistically higher incidence of NEC. We did not detect NEC episodes in patients treated with CPX-351, 5 + 2 (cytarabine, idarubicine), and high-dose cytarabine. Thus, we found that cytarabine could determine mucosal damage when associated with an anthracycline but not if delivered either alone or as dual-drug liposomal encapsulation of daunorubicin/cytarabine. We also describe NEC mortality, symptoms at diagnosis, intestinal sites involvement, and prognostic significance of bowel wall thickening.
Collapse
Affiliation(s)
- Edoardo Benedetti
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Ginevra Traverso
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Giulia Pucci
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Riccardo Morganti
- Section of Statistics, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Emilia Bramanti
- Institute of Chemistry of Organo Metallic Compounds (ICCOM), Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Piero Lippolis
- General and Peritoneal Surgery Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Maria Chiara Susini
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Elisa Mazzantini
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | | | - Fabrizio Mavilia
- Hematology Operative Unit (UO), Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | | | - Emanuele Neri
- Radiology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Arena
- Radiology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesca Cerri
- Radiology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luigi De Simone
- Anesthesia and Maternal-Infantile Resuscitation Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Katia Valentini
- Anesthesia and Maternal-Infantile Resuscitation Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | | | - Vittorio Ricchiuto
- Dipartimento di Tecnologie Sanitarie ESTAR, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Sara Galimberti
- Hematology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
20
|
Jain AG, Ball S, Aguirre LE, Tobon KA, Chan O, Padron E, Kuykendall A, Komrokji R, Sallman D, Lancet JE, Sweet K. Utilization of Serial Next-Generation Sequencing Among Patients Receiving CPX-351 for Newly Diagnosed Acute Myeloid Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e315-e322. [PMID: 37558530 DOI: 10.1016/j.clml.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND The phase III trial that led to the approval of CPX-351 for treating secondary acute myeloid leukemia (sAML) in 2017 did not study the effect of specific mutations on outcomes. METHODS This retrospective study was done to evaluate the effect of next-generation sequencing (NGS) results at the time of best response and before allogeneic stem cell transplant (alloSCT) in patients treated with CPX-351 as frontline therapy for sAML between 2017 and 2021. RESULTS The most common mutations seen were DNMT3A (n = 17, 29.8%), SRSF2 (n = 13, 22.8%), RUNX1 (n = 13, 22.8%), TET2 (n = 9, 15.8%), ASXL1 (n = 9, 15.8%), and BCOR (n = 9, 15.8%). Median OS (mOS) for the entire cohort was 47 months. Though 64.7% patients cleared the DNMT3A mutation, only 44.4% and 22.2% of patients cleared the TET2 and ASXL1 mutations, respectively. The mOS for patients who cleared their mutations vs. for those who did not was not significantly longer (46 vs. 30 months; P = .991). The relapse-free survival (RFS) for patients who cleared mutations was numerically longer compared to those who had persistent mutations; however, this did not reach statistical significance (44 months vs. 26 months; P = .786). CONCLUSION This is the first study reporting NGS at best response and before alloSCT and its effect on OS and RFS. We found that OS and RFS were numerically longer among patients who cleared mutations; however, this did not reach statistical significance. In addition, alloSCT led to improved RFS irrespective of mutational clearance.
Collapse
Affiliation(s)
- Akriti G Jain
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Department of Internal Medicine, University of South Florida, Tampa, FL
| | - Somedeb Ball
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Department of Internal Medicine, University of South Florida, Tampa, FL
| | - Luis E Aguirre
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Department of Internal Medicine, University of South Florida, Tampa, FL
| | - Katherine A Tobon
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Onyee Chan
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Eric Padron
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Andrew Kuykendall
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Rami Komrokji
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - David Sallman
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jeffrey E Lancet
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kendra Sweet
- Division of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| |
Collapse
|
21
|
Rosenquist R, Bernard E, Erkers T, Scott DW, Itzykson R, Rousselot P, Soulier J, Hutchings M, Östling P, Cavelier L, Fioretos T, Smedby KE. Novel precision medicine approaches and treatment strategies in hematological malignancies. J Intern Med 2023; 294:413-436. [PMID: 37424223 DOI: 10.1111/joim.13697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Genetic testing has been applied for decades in clinical routine diagnostics of hematological malignancies to improve disease (sub)classification, prognostication, patient management, and survival. In recent classifications of hematological malignancies, disease subtypes are defined by key recurrent genetic alterations detected by conventional methods (i.e., cytogenetics, fluorescence in situ hybridization, and targeted sequencing). Hematological malignancies were also one of the first disease areas in which targeted therapies were introduced, the prime example being BCR::ABL1 inhibitors, followed by an increasing number of targeted inhibitors hitting the Achilles' heel of each disease, resulting in a clear patient benefit. Owing to the technical advances in high-throughput sequencing, we can now apply broad genomic tests, including comprehensive gene panels or whole-genome and whole-transcriptome sequencing, to identify clinically important diagnostic, prognostic, and predictive markers. In this review, we give examples of how precision diagnostics has been implemented to guide treatment selection and improve survival in myeloid (myelodysplastic syndromes and acute myeloid leukemia) and lymphoid malignancies (acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and chronic lymphocytic leukemia). We discuss the relevance and potential of monitoring measurable residual disease using ultra-sensitive techniques to assess therapy response and detect early relapses. Finally, we bring up the promising avenue of functional precision medicine, combining ex vivo drug screening with various omics technologies, to provide novel treatment options for patients with advanced disease. Although we are only in the beginning of the field of precision hematology, we foresee rapid development with new types of diagnostics and treatment strategies becoming available to the benefit of our patients.
Collapse
Affiliation(s)
- Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Elsa Bernard
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
- PRISM Center for Personalized Medicine, Gustave Roussy, Villejuif, France
| | - Tom Erkers
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Stockholm, Sweden
| | - David W Scott
- BC Cancer's Centre for Lymphoid Cancer, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Rousselot
- Department of Hematology, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Jean Soulier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, Paris, France
- Hématologie Biologique, APHP, Hôpital Saint-Louis, Paris, France
| | - Martin Hutchings
- Department of Haematology and Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Päivi Östling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Stockholm, Sweden
| | - Lucia Cavelier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Thoas Fioretos
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Karin E Smedby
- Department of Hematology, Karolinska University Hospital, Solna, Stockholm, Sweden
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Legg A, Lambova A, Broe A, Levy J, Medalla G. Real-World Experience With CPX-351 Treatment for Acute Myeloid Leukemia in England: An Analysis From the National Cancer Registration and Analysis Service. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e323-e330. [PMID: 37544809 DOI: 10.1016/j.clml.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND CPX-351 demonstrated improved overall survival (OS) versus conventional 3 + 7 daunorubicin/cytarabine chemotherapy in a registrational phase III study in older patients with newly diagnosed, high-risk secondary acute myeloid leukemia (AML). This retrospective, population-based cohort study aimed to describe and compare the characteristics and survival outcomes of younger (<60 years) versus older (≥60 years) patients with AML treated with CPX-351 in England. PATIENTS AND METHODS The study included adults aged ≥18 years diagnosed with AML in England between January 2013 and March 2022, and treated with CPX-351 in routine clinical practice (patients who received CPX-351 in a clinical trial were excluded). Patient records were sourced from the population-level cancer analysis system database available through the National Cancer Registration and Analysis Service. RESULTS Of 353 included patients, 104 (29.5%) were <60 years. With a median follow-up of 10.9 months from diagnosis, the estimated median OS was 12.9 months overall, 17.3 months for adults <60 years and 11.7 months for those ≥60 years. All-cause mortality by Day 30 from diagnosis was 6% overall, 4% for adults <60 years and 6% for those ≥60 years. Hematopoietic cell transplantation (HCT) was received by 54% of adults <60 years and 38% of those ≥60 years after CPX-351, with median OS landmarked from the HCT date not yet reached for either age subgroup. CONCLUSION This study provides real-world survival outcomes data suggesting that CPX-351 is an effective treatment for both younger (<60 years) and older (≥60 years) adults with AML.
Collapse
|
23
|
Othman J, Wilhelm-Benartzi C, Dillon R, Knapper S, Freeman SD, Batten LM, Canham J, Hinson EL, Wych J, Betteridge S, Villiers W, Kleeman M, Gilkes A, Potter N, Overgaard UM, Mehta P, Kottaridis P, Cavenagh J, Hemmaway C, Arnold C, Dennis M, Russell NH. A randomized comparison of CPX-351 and FLAG-Ida in adverse karyotype AML and high-risk MDS: the UK NCRI AML19 trial. Blood Adv 2023; 7:4539-4549. [PMID: 37171402 PMCID: PMC10425682 DOI: 10.1182/bloodadvances.2023010276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023] Open
Abstract
Liposomal daunorubicin and cytarabine (CPX-351) improved overall survival (OS) compared with 7+3 chemotherapy in older patients with secondary acute myeloid leukemia (AML); to date, there have been no randomized studies in younger patients. The high-risk cohort of the UK NCRI AML19 trial (ISRCTN78449203) compared CPX-351 with FLAG-Ida in younger adults with newly diagnosed adverse cytogenetic AML or high-risk myelodysplastic syndromes (MDS). A total of 189 patients were randomized (median age, 56 years). Per clinical criteria, 49% of patients had de novo AML, 20% had secondary AML, and 30% had high-risk MDS. MDS-related cytogenetics were present in 73% of the patients, with a complex karyotype in 49%. TP53 was the most common mutated gene, in 43%. Myelodysplasia-related gene mutations were present in 75 (44%) patients. The overall response rate (CR + CRi) after course 2 was 64% and 76% for CPX-351 and FLAG-Ida, respectively. There was no difference in OS (13.3 months vs 11.4 months) or event-free survival in multivariable analysis. However, relapse-free survival was significantly longer with CPX-351 (median 22.1 vs 8.35 months). There was no difference between the treatment arms in patients with clinically defined secondary AML or those with MDS-related cytogenetic abnormalities; however, an exploratory subgroup of patients with MDS-related gene mutations had significantly longer OS with CPX-351 (median 38.4 vs 16.3 months). In conclusion, the OS of younger patients with adverse risk AML/MDS was not significantly different between CPX-351 and FLAG-Ida.
Collapse
Affiliation(s)
- Jad Othman
- Department of Medical and Molecular Genetics, Kings College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Richard Dillon
- Department of Medical and Molecular Genetics, Kings College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Steve Knapper
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sylvie D. Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Leona M. Batten
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Joanna Canham
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Emily L. Hinson
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Julie Wych
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Sophie Betteridge
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - William Villiers
- Department of Medical and Molecular Genetics, Kings College London, London, United Kingdom
| | - Michelle Kleeman
- Genomics Facility, NIHR Biomedical Research Centre at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Amanda Gilkes
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Nicola Potter
- Department of Medical and Molecular Genetics, Kings College London, London, United Kingdom
| | | | - Priyanka Mehta
- Bristol Haematology and Oncology Centre, University Hospitals of Bristol and Weston NHS Trust, Bristol, United Kingdom
| | | | - Jamie Cavenagh
- Department of Haemato-Oncology, Barts Health NHS Trust, St Bartholomew’s Hospital, London, United Kingdom
| | - Claire Hemmaway
- Department of Haematology, Auckland Hospital, Auckland, New Zealand
| | - Claire Arnold
- Clinical Haematology, Belfast City Hospital, Belfast, Northern Ireland
| | - Mike Dennis
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Nigel H. Russell
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - UK National Cancer Research Institute Acute Myeloid Leukaemia Working Group
- Department of Medical and Molecular Genetics, Kings College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
- School of Medicine, Cardiff University, Cardiff, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Genomics Facility, NIHR Biomedical Research Centre at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, United Kingdom
- Copenhagen University Hospital, Copenhagen, Denmark
- Bristol Haematology and Oncology Centre, University Hospitals of Bristol and Weston NHS Trust, Bristol, United Kingdom
- Department of Haematology, University College Hospital, London, United Kingdom
- Department of Haemato-Oncology, Barts Health NHS Trust, St Bartholomew’s Hospital, London, United Kingdom
- Department of Haematology, Auckland Hospital, Auckland, New Zealand
- Clinical Haematology, Belfast City Hospital, Belfast, Northern Ireland
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
24
|
Boscaro E, Urbino I, Catania FM, Arrigo G, Secreto C, Olivi M, D'Ardia S, Frairia C, Giai V, Freilone R, Ferrero D, Audisio E, Cerrano M. Modern Risk Stratification of Acute Myeloid Leukemia in 2023: Integrating Established and Emerging Prognostic Factors. Cancers (Basel) 2023; 15:3512. [PMID: 37444622 DOI: 10.3390/cancers15133512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
An accurate estimation of AML prognosis is complex since it depends on patient-related factors, AML manifestations at diagnosis, and disease genetics. Furthermore, the depth of response, evaluated using the level of MRD, has been established as a strong prognostic factor in several AML subgroups. In recent years, this rapidly evolving field has made the prognostic evaluation of AML more challenging. Traditional prognostic factors, established in cohorts of patients treated with standard intensive chemotherapy, are becoming less accurate as new effective therapies are emerging. The widespread availability of next-generation sequencing platforms has improved our knowledge of AML biology and, consequently, the recent ELN 2022 recommendations significantly expanded the role of new gene mutations. However, the impact of rare co-mutational patterns remains to be fully disclosed, and large international consortia such as the HARMONY project will hopefully be instrumental to this aim. Moreover, accumulating evidence suggests that clonal architecture plays a significant prognostic role. The integration of clinical, cytogenetic, and molecular factors is essential, but hierarchical methods are reaching their limit. Thus, innovative approaches are being extensively explored, including those based on "knowledge banks". Indeed, more robust prognostic estimations can be obtained by matching each patient's genomic and clinical data with the ones derived from very large cohorts, but further improvements are needed.
Collapse
Affiliation(s)
- Eleonora Boscaro
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Irene Urbino
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Federica Maria Catania
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Giulia Arrigo
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Carolina Secreto
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Matteo Olivi
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Stefano D'Ardia
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Chiara Frairia
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Valentina Giai
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Roberto Freilone
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Dario Ferrero
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy
| | - Ernesta Audisio
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Marco Cerrano
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
25
|
Fianchi L, Guolo F, Marchesi F, Cattaneo C, Gottardi M, Restuccia F, Candoni A, Ortu La Barbera E, Fazzi R, Pasciolla C, Finizio O, Fracchiolla N, Delia M, Lessi F, Dargenio M, Bonuomo V, Del Principe MI, Zappasodi P, Picardi M, Basilico C, Piedimonte M, Minetto P, Giordano A, Chiusolo P, Prezioso L, Buquicchio C, Melillo LMA, Zama D, Farina F, Mancini V, Terrenato I, Rondoni M, Urbino I, Tumbarello M, Busca A, Pagano L. Multicenter Observational Retrospective Study on Febrile Events in Patients with Acute Myeloid Leukemia Treated with Cpx-351 in "Real-Life": The SEIFEM Experience. Cancers (Basel) 2023; 15:3457. [PMID: 37444567 DOI: 10.3390/cancers15133457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
In the present study, we aimed to evaluate the absolute risk of infection in the real-life setting of AML patients treated with CPX-351. The study included all patients with AML from 30 Italian hematology centers of the SEIFEM group who received CPX-351 from July 2018 to June 2021. There were 200 patients included. Overall, 336 CPX-351 courses were counted: all 200 patients received the first induction cycle, 18 patients (5%) received a second CPX-351 induction, while 86 patients (26%) proceeded with the first CPX-351 consolidation cycle, and 32 patients (10%) received a second CPX-351 consolidation. A total of 249 febrile events were recorded: 193 during the first or second induction, and 56 after the first or second consolidation. After the diagnostic work-up, 92 events (37%) were classified as febrile neutropenia of unknown origin (FUO), 118 (47%) were classifiable as microbiologically documented infections, and 39 (17%) were classifiable as clinically documented infections. The overall 30-day mortality rate was 14% (28/200). The attributable mortality-infection rate was 6% (15/249). A lack of response to the CPX-351 treatment was the only factor significantly associated with mortality in the multivariate analysis [p-value: 0.004, OR 0.05, 95% CI 0.01-0.39]. Our study confirms the good safety profile of CPX-351 in a real-life setting, with an incidence of infectious complications comparable to that of the pivotal studies; despite prolonged neutropenia, the incidence of fungal infections was low, as was infection-related mortality.
Collapse
Affiliation(s)
- Luana Fianchi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Fabio Guolo
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | | | - Chiara Cattaneo
- SC Ematologia e Dipartimento di Oncologia Clinica, A.O. Spedali Civili, 25123 Brescia, Italy
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology, IOV-IRCCS, 31033 Padua, Italy
| | | | - Anna Candoni
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Università degli Studi di Modena e Reggio Emilia, 41125 Modena, Italy
| | | | - Rita Fazzi
- Hematology Unit-A.O.U.P. Ospedale Santa Chiara, 56126 Pisa, Italy
| | - Crescenza Pasciolla
- Haematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy
| | | | - Nicola Fracchiolla
- Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico di Milano, 20122 Milano, Italy
| | - Mario Delia
- Hematology and Transplant Unit, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Federica Lessi
- Ematologia e Immunologia, Clinica Azienda Ospedaliera di Padova, 35128 Padova, Italy
| | | | | | - Maria Ilaria Del Principe
- Dipartimento di Biomedicina e Prevenzione, Università degli studi di Roma "Tor Vergata", 00133 Roma, Italy
| | | | | | - Claudia Basilico
- Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy
| | | | - Paola Minetto
- Ematologia e Trapianto, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Antonio Giordano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Patrizia Chiusolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | | | | | | | - Daniele Zama
- Policlinico Sant'Orsola Malpighi, 40138 Bologna, Italy
| | - Francesca Farina
- U.O. Ematologia e Trapianto Midollo, Dipartimento di Oncologia, Istituto Scientifico San Raffaele, 20132 Milano, Italy
| | - Valentina Mancini
- Divisione di Ematologia, Ospedale Niguarda Milano, 20162 Milano, Italy
| | - Irene Terrenato
- UOSD Clinical Trial Center e Biostatistica e Bioinformatica, IRCCS Istituto Nazionale Tumori Regina Elena, 00128 Roma, Italy
| | - Michela Rondoni
- U.O.C. di Ematologia, Azienda Unità Sanitaria Locale della Romagna, 48124 Ravenna, Italy
| | - Irene Urbino
- SC Ematologia, Ospedale AOU Città Della Salute e della Scienza, 10126 Torino, Italy
| | - Mario Tumbarello
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Alessandro Busca
- SC Ematologia, Ospedale AOU Città Della Salute e della Scienza, 10126 Torino, Italy
| | - Livio Pagano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
26
|
Bernal T, Moreno AF, de LaIglesia A, Benavente C, García‐Noblejas A, Belmonte DG, Riaza R, Salamero O, Foncillas MA, Roldán A, Concepción VN, González LL, Bergua Burgués JM, Lorente de Uña S, Rodríguez‐Macías G, de la Fuente Burguera A, García Pérez MJ, López‐Lorenzo JL, Martínez P, Aláez C, Callejas M, Martínez‐Chamorro C, Roca JR, Barciela LA, Mena Durán AV, Gómez Correcha K, Lavilla Rubira E, Amigo ML, Vall‐llovera F, Garrido A, García‐Fortes M, de Miguel Llorente D, Leonardo AA, Cervero C, Jordá RC, Pérez‐Encinas MM, Zarzuela MP, Figuera A, Rad G, Martínez‐Cuadrón D, Montesinos P. Clinical outcomes after CPX-351 in patients with high-risk acute myeloid leukemia: A comparison with a matched cohort from the Spanish PETHEMA registry. Cancer Med 2023; 12:14892-14901. [PMID: 37212507 PMCID: PMC10417130 DOI: 10.1002/cam4.6120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND CPX-351 is approved for the treatment of therapy related acute myeloid leukemia (t-AML) and AML with myelodysplastic related changes (MRC-AML). The benefits of this treatment over standard chemotherapy has not been addressed in well matched cohorts of real-life patients. METHODS Retrospective analysis of AML patients treated with CPX-351 as per routine practice. A propensity score matching (PSM) was used to compare their main outcomes with those observed in a matched cohort among 765 historical patients receiving intensive chemotherapy (IC), all of them reported to the PETHEMA epidemiologic registry. RESULTS Median age of 79 patients treated with CPX-351 was 67 years old (interquartile range 62-71), 53 were MRC-AML. The complete remission (CR) rate or CR without recovery (CRi) after 1 or 2 cycles of CPX-351 was 52%, 60-days mortality 18%, measurable residual disease <0.1% in 54% (12 out of 22) of them. Stem cell transplant (SCT) was performed in 27 patients (34%), median OS was 10.3 months, and 3-year relapse incidence was 50%. Using PSM, we obtained two comparable cohorts treated with CPX-351 (n = 52) or IC (n = 99), without significant differences in CR/CRi (60% vs. 54%) and median OS (10.3 months vs. 9.1 months), although more patients were bridged to SCT in the CPX-351 group (35% vs. 12%). The results were confirmed when only 3 + 7 patients were included in the historical cohort. In multivariable analyses, SCT was associated with better OS (HR 0.33 95% CI: 0.18-0.59), p < 0.001. CONCLUSION Larger post-authorization studies may provide evidence of the clinical benefits of CPX-351 for AML in the real-life setting.
Collapse
Affiliation(s)
- Teresa Bernal
- Hospital Universitario Central AsturiasOviedoSpain
- Instituto de Oncología del Principado de Asturias (IUOPA), Instituto de Investigación del Principado de Asturias (ISPA)Spain
| | - Ainhoa Fernández Moreno
- Hospital Universitario Central AsturiasOviedoSpain
- Instituto de Oncología del Principado de Asturias (IUOPA), Instituto de Investigación del Principado de Asturias (ISPA)Spain
| | | | | | | | | | | | | | | | - Alicia Roldán
- Departamento de MedicinaHospital Infanta Sofía San Sebastián de los Reyes, Universidad EuropeaMadridSpain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ana Garrido
- Hospital de la Santa Creu i Sant PauBarcelonaSpain
| | | | | | | | | | - Rosa Coll Jordá
- ICO Girona, Hospital Universitario Dr. Josep TruetaGironaSpain
| | | | | | | | - Guillermo Rad
- Instituto de Oncología del Principado de Asturias (IUOPA), Instituto de Investigación del Principado de Asturias (ISPA)Spain
| | | | | |
Collapse
|
27
|
Zhao D, Zarif M, Zhou Q, Capo-Chichi JM, Schuh A, Minden MD, Atenafu EG, Kumar R, Chang H. TP53 Mutations in AML Patients Are Associated with Dismal Clinical Outcome Irrespective of Frontline Induction Regimen and Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2023; 15:3210. [PMID: 37370821 DOI: 10.3390/cancers15123210] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
TP53 mutations are associated with extremely poor outcomes in acute myeloid leukemia (AML). The outcomes of patients with TP53-mutated (TP53MUT) AML after different frontline treatment modalities are not well established. Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative procedure for AML; however, long-term outcomes among patients with TP53MUT AML after allo-HCT are dismal, and the benefit of allo-HCT remains controversial. We sought to evaluate the outcomes of patients with TP53MUT AML after treatment with different frontline induction therapies and allo-HCT. A total of 113 patients with TP53MUT AML were retrospectively evaluated. Patients with TP53MUT AML who received intensive or azacitidine-venetoclax induction had higher complete remission rates compared to patients treated with other hypomethylating-agent-based induction regimens. However, OS and EFS were not significantly different among the induction regimen groups. Allo-HCT was associated with improved OS and EFS among patients with TP53MUT AML; however, allo-HCT was not significantly associated with improved OS or EFS in time-dependent or landmark analysis. While the outcomes of all patients were generally poor irrespective of therapeutic strategy, transplanted patients with lower TP53MUT variant allele frequency (VAF) at the time of diagnosis had superior outcomes compared to transplanted patients with higher TP53 VAF. Our study provides further evidence that the current standards of care for AML confer limited therapeutic benefit to patients with TP53 mutations.
Collapse
Affiliation(s)
- Davidson Zhao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Hematology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Mojgan Zarif
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Hematology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Qianghua Zhou
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Hematology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - José-Mario Capo-Chichi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Hematology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Andre Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Mark D Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Rajat Kumar
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Hong Chang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Hematology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
28
|
Daver NG, Iqbal S, Huang J, Renard C, Lin J, Pan Y, Williamson M, Ramsingh G. Clinical characteristics and overall survival among acute myeloid leukemia patients with TP53 gene mutation or chromosome 17p deletion. Am J Hematol 2023. [PMID: 37139921 DOI: 10.1002/ajh.26941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023]
Abstract
Approximately 5% to 15% of acute myeloid leukemia (AML) patients have TP53 gene mutations (TP53m), which are associated with very poor outcomes. Adults (≥18 years) with a new AML diagnosis were included from a nationwide, de-identified, real-world database. Patients receiving first-line therapy were divided into three cohorts: venetoclax (VEN) + hypomethylating agents (HMAs; Cohort A), intensive chemotherapy (Cohort B), or HMA without VEN (Cohort C). A total of 370 newly diagnosed AML patients with TP53m (n = 124), chromosome 17p deletion (n = 166), or both (n = 80) were included. The median age was 72 years (range, 24-84); most were male (59%) and White (69%). Baseline bone marrow (BM) blasts were ≤30%, 31%-50%, and >50% in 41%, 24%, and 29% of patients in Cohorts A, B, and C, respectively. BM remission (<5% blasts) with first-line therapy was reported in 54% of patients (115/215) overall, and 67% (38/57), 62% (68/110), and 19% (9/48) for respective cohorts (median BM remission duration: 6.3, 6.9, and 5.4 months). Median overall survival (95% CI) was 7.4 months (6.0-8.8) for Cohort A, 9.4 months (7.2-10.4) for Cohort B, and 5.9 months (4.3-7.5) for Cohort C. There were no differences in survival by treatment type after adjusting for the effects of relevant covariates (Cohort A vs. C adjusted hazard ratio [aHR] = 0.9; 95% CI, 0.7-1.3; Cohort A vs. B aHR = 1.0; 95% CI, 0.7-1.5; and Cohort C vs. B aHR = 1.1; 95% CI, 0.8-1.6). Patients with TP53m AML have dismal outcomes with current therapies, demonstrating the high unmet need for improved treatments.
Collapse
Affiliation(s)
- Naval G Daver
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shahed Iqbal
- Gilead Sciences, Inc., Foster City, California, USA
| | - Julie Huang
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Joyce Lin
- Gilead Sciences, Inc., Foster City, California, USA
| | - Yang Pan
- Gilead Sciences, Inc., Foster City, California, USA
| | | | | |
Collapse
|
29
|
Lemoli RM, Montesinos P, Jain A. Real-world experience with CPX-351 in high-risk acute myeloid leukemia. Crit Rev Oncol Hematol 2023; 185:103984. [PMID: 37028531 DOI: 10.1016/j.critrevonc.2023.103984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
CPX-351, a dual-drug liposomal encapsulation of daunorubicin/cytarabine, was approved for newly diagnosed therapy-related acute myeloid leukemia (AML) and AML with myelodysplasia-related changes in adults in 2017 (US; updated to patients aged ≥1 year in 2021) and 2018 (EU/UK) based on improved survival and remission and comparable safety versus 7+3 chemotherapy in a randomized trial in older adults. Real-world studies have since evaluated CPX-351 in routine practice across several countries and addressed important data gaps (e.g., use in younger adults, measurable residual disease negativity, outcomes by mutation). This review discusses real-world studies of CPX-351 as AML treatment, with the aim of helping prescribers make informed treatment decisions.
Collapse
Affiliation(s)
- Roberto M Lemoli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Clinic of Hematology, University of Genoa, Genoa, Italy.
| | - Pau Montesinos
- Hospital Universitari i Politècnic La Fe, València, Spain
| | - Akriti Jain
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
30
|
Daver NG, Iqbal S, Renard C, Chan RJ, Hasegawa K, Hu H, Tse P, Yan J, Zoratti MJ, Xie F, Ramsingh G. Treatment outcomes for newly diagnosed, treatment-naïve TP53-mutated acute myeloid leukemia: a systematic review and meta-analysis. J Hematol Oncol 2023; 16:19. [PMID: 36879351 PMCID: PMC9990239 DOI: 10.1186/s13045-023-01417-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/26/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND TP53 mutations, which are present in 5% to 10% of patients with acute myeloid leukemia (AML), are associated with treatment resistance and poor outcomes. First-line therapies for TP53-mutated (TP53m) AML consist of intensive chemotherapy (IC), hypomethylating agents (HMA), or venetoclax combined with HMA (VEN + HMA). METHODS We conducted a systematic review and meta-analysis to describe and compare treatment outcomes in newly diagnosed treatment-naïve patients with TP53m AML. Randomized controlled trials, single-arm trials, prospective observational studies, and retrospective studies were included that reported on complete remission (CR), CR with incomplete hematologic recovery (CRi), overall survival (OS), event-free survival (EFS), duration of response (DoR), and overall response rate (ORR) among patients with TP53m AML receiving first-line treatment with IC, HMA, or VEN + HMA. RESULTS Searches of EMBASE and MEDLINE identified 3006 abstracts, and 17 publications describing 12 studies met the inclusion criteria. Random-effects models were used to pool response rates, and time-related outcomes were analyzed with the median of medians method. IC was associated with the greatest CR rate of 43%, and CR rates were 33% for VEN + HMA and 13% for HMA. Rates of CR/CRi were comparable for IC (46%) and VEN + HMA (49%) but were lower for HMA (13%). Median OS was uniformly poor across treatments: IC, 6.5 months; VEN + HMA, 6.2 months; and HMA, 6.1 months. For IC, the EFS estimate was 3.7 months; EFS was not reported for VEN + HMA or HMA. The ORR was 41% for IC, 65% for VEN + HMA, and 47% for HMA. DoR was 3.5 months for IC, 5.0 months for VEN + HMA, and was not reported for HMA. CONCLUSIONS Despite improved responses seen with IC and VEN + HMA compared to HMA, survival was uniformly poor, and clinical benefits were limited across all treatments for patients with newly diagnosed, treatment-naïve TP53m AML, demonstrating a significant need for improved treatment for this difficult-to-treat population.
Collapse
Affiliation(s)
- Naval G Daver
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Shahed Iqbal
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, 94404, USA
| | - Camille Renard
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, 94404, USA
| | - Rebecca J Chan
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, 94404, USA
| | - Ken Hasegawa
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, 94404, USA
| | - Hao Hu
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, 94404, USA
| | - Preston Tse
- McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Jiajun Yan
- McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | | | - Feng Xie
- McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | | |
Collapse
|
31
|
Ball S, Loghavi S, Zeidan AM. TP53-altered higher-risk myelodysplastic syndromes/neoplasms and acute myeloid leukemia: a distinct genetic entity with unique unmet needs. Leuk Lymphoma 2023; 64:540-550. [PMID: 36323304 DOI: 10.1080/10428194.2022.2136969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Pathogenic alterations of TP53 are an independent poor prognostic factor in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Clinical course of TP53- altered myeloid neoplasms is dictated by genetic characteristics, such as TP53 allelic state and variant allele frequency (VAF), and not the blast count. Hence, it was recently proposed that MDS (with increased blasts) and AML with TP53 alterations may be best classified as a single molecular disease entity, TP53-mutated higher-risk (HR)-MDS/AML. TP53 mutations drive resistance to intensive chemotherapies and less intensive hypomethylating agents (HMA). Novel combinations incorporating BCL2 inhibitor venetoclax improve response rates for TP53-mutated subgroup, but the survival is not improved. Early clinical studies combining HMA with investigational agents demonstrated activity in TP53-mutated HR-MDS/AML, but updated results with larger samples, longer follow-up, or randomized trials were less impressive to date. Future research should focus on finding novel, potentially disease-modifying therapies to improve outcomes in patients with TP53-mutated HR-MDS/AML.
Collapse
Affiliation(s)
- Somedeb Ball
- Division of Hematology and Medical Oncology, University of South Florida/H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|
32
|
Fever of Unknown Origin and Multidrug Resistant Organism Colonization in AML Patients. Mediterr J Hematol Infect Dis 2023; 15:e2023013. [PMID: 36660358 PMCID: PMC9833311 DOI: 10.4084/mjhid.2023.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/31/2022] [Indexed: 01/03/2023] Open
Abstract
Background Colonization by multidrug-resistant organisms (MDRO) is a frequent complication in hematologic departments, which puts patients at risk of life-threatening bacterial sepsis. Fever of unknown origin (FUO) is a condition related to the delivery of chemotherapy in hematologic malignancies, in which the use of antibiotics is debated. The incidence, risk factors, and influence on the outcome of these conditions in patients with acute myeloid leukemia (AML) are not clearly defined. Methods We retrospectively analyzed 132 consecutive admissions of non-promyelocytic AML patients at the Hematology Unit of the University Tor Vergata in Rome between June 2019 and February 2022. MDRO swab-based screening was performed in all patients on the day of admission and once weekly after that. FUO was defined as fever with no evidence of infection. Results Of 132 consecutive hospitalizations (69 AML patients), MDRO colonization was observed in 35 cases (26%) and resulted independently related to a previous MDRO colonization (p=0.001) and length of hospitalization (p=0.03). The colonization persistence rate in subsequent admissions was 64%. MDRO-related bloodstream infection was observed in 8 patients (23%) and correlated with grade III/IV mucositis (p=0.008) and length of hospitalization (p=0.02). FUO occurred in 68 cases (51%) and correlated with an absolute neutrophilic count <500μ/L at admission (0.04). Conclusion In our experience, MDRO colonization is a frequent and difficult-to-eradicate condition that can arise at all stages of treatment. Prompt discharge of patients as soon as clinical conditions allow could limit the spread of MDRO. In addition, the appropriate use of antibiotics, especially in the case of FUO, and the contraction of hospitalization length, when feasible, are measures to tackle the further spread of MDRO.
Collapse
|
33
|
Clinical and Cytogenetic Characterization of Early and Late Relapses in Patients Allografted for Myeloid Neoplasms with a Myelodysplastic Component. Cancers (Basel) 2022; 14:cancers14246244. [PMID: 36551729 PMCID: PMC9776604 DOI: 10.3390/cancers14246244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
An improved understanding of relapse kinetics is required to optimize detection and treatment strategies for the post-transplant relapse of myeloid neoplasms. Therefore, we retrospectively analyzed data from 91 patients allografted for MDS (n = 54), AML-MRC (n = 29) and chronic myelomonocytic leukemia (CMML, n = 8), who relapsed after transplant. Patients with early (<12 months, n = 56) and late relapse (>12 months, n = 35) were compared regarding patient-, disease- and transplant-related factors, including karyotype analyses at diagnosis and relapse. After a median follow-up of 17.4 months after relapse, late relapses showed improved outcomes compared with early relapses (2-yr OS 67% vs. 32%, p = 0.0048). Comparing frequency of distinct patient-, disease- and transplant-related factors among early and late relapses, complex karyotype (p = 0.0004) and unfavorable disease risk at diagnosis (p = 0.0008) as well as clonal evolution at relapse (p = 0.03) were more common in early than in late relapses. Furthermore, patients receiving transplant without prior cytoreduction or in complete remission were more frequently present in the group of late relapses. These data suggest that cytogenetics rather than disease burden at diagnosis and transplant-related factors determine the timepoint of post-transplant relapse and that upfront transplantation may be favored in order to delay relapse.
Collapse
|
34
|
Pollyea DA, Pratz KW, Wei AH, Pullarkat V, Jonas BA, Recher C, Babu S, Schuh AC, Dail M, Sun Y, Potluri J, Chyla B, DiNardo CD. Outcomes in Patients with Poor-Risk Cytogenetics with or without TP53 Mutations Treated with Venetoclax and Azacitidine. Clin Cancer Res 2022; 28:5272-5279. [PMID: 36007102 PMCID: PMC9751752 DOI: 10.1158/1078-0432.ccr-22-1183] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE To evaluate efficacy and safety of venetoclax + azacitidine in treatment-naïve patients with acute myeloid leukemia harboring poor-risk cytogenetics and TP53mut or TP53wt. PATIENTS AND METHODS We analyzed data from a phase III study (NCT02993523) comparing venetoclax (400 mg orally days 1-28) + azacitidine (75 mg/m2 days 1-7) or placebo + azacitidine, and from a phase Ib study (NCT02203773) of venetoclax + azacitidine. Patients were ineligible for intensive therapy. TP53 status was analyzed centrally; cytogenetic studies were performed locally. RESULTS Patients (n = 127) with poor-risk cytogenetics receiving venetoclax + azacitidine (TP53wt = 50; TP53mut = 54) were compared with patients with poor-risk cytogenetics (n = 56) receiving azacitidine alone (TP53wt = 22; TP53mut = 18).For poor-risk cytogenetics + TP53wt patients, venetoclax + azacitidine versus azacitidine alone resulted in composite remission rates (CRc) of 70% versus 23%, median duration of remission (DoR) of 18.4 versus 8.5 months, and median overall survival (OS) of 23.4 versus 11.3 months, respectively. Outcomes with venetoclax + azacitidine were comparable with similarly treated patients with intermediate-risk cytogenetics and TP53wt.For poor-risk cytogenetics + TP53mut patients, venetoclax + azacitidine versus azacitidine alone resulted in CRc of 41% versus 17%, median DoR of 6.5 versus 6.7 months, and median OS of 5.2 versus 4.9 months, respectively.For poor-risk cytogenetics + TP53mut patients, predominant grade ≥3 adverse events (AE) for venetoclax + azacitidine versus azacitidine were febrile neutropenia (55%/39%), thrombocytopenia (28%/28%), neutropenia (26%/17%), anemia (13%/6%), and pneumonia (28%/33%). AEs were comparable between TP53mut and TP53wt patients. CONCLUSIONS In poor-risk cytogenetics + TP53mut patients, venetoclax + azacitidine improved remission rates but not DoR or OS compared with azacitidine alone. However, in poor-risk cytogenetics + TP53wt patients, venetoclax + azacitidine resulted in higher remission rates and longer DoR and OS than azacitidine alone, with outcomes comparable with similarly treated patients with intermediate-risk cytogenetics. Toxicities were similar in TP53mut and TP53wt patients. See related commentary by Green and Zeidner, p. 5235.
Collapse
Affiliation(s)
- Daniel A. Pollyea
- Division of Hematology, School of Medicine, University of Colorado, Aurora, Colorado.,Corresponding Author: Daniel A. Pollyea, University of Colorado, 1665 Aurora Court, Mail Stop F754, Aurora, CO 80045. E-mail:
| | - Keith W. Pratz
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew H. Wei
- Department of Clinical Haematology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation and Gehr Family Center for Leukemia Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Brian A. Jonas
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis School of Medicine, Sacramento, California
| | | | - Sunil Babu
- Fort Wayne Medical Oncology and Hematology, Fort Wayne, Indiana
| | - Andre C. Schuh
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Yan Sun
- AbbVie Inc., North Chicago, Illinois
| | | | | | - Courtney D. DiNardo
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
35
|
Zhao D, Eladl E, Zarif M, Capo‐Chichi J, Schuh A, Atenafu E, Minden M, Chang H. Molecular characterization of
AML‐MRC
reveals
TP53
mutation as an adverse prognostic factor irrespective of
MRC
‐defining criteria,
TP53
allelic state, or
TP53
variant allele frequency. Cancer Med 2022; 12:6511-6522. [PMID: 36394085 PMCID: PMC10067127 DOI: 10.1002/cam4.5421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/23/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) generally confers poor prognosis, however, patient outcomes are heterogeneous. The impact of TP53 allelic state and variant allele frequency (VAF) in AML-MRC remains poorly defined. METHODS We retrospectively evaluated 266 AML-MRC patients who had NGS testing at our institution from 2014 to 2020 and analyzed their clinical outcomes based on clinicopathological features. RESULTS TP53 mutations were associated with cytogenetic abnormalities in 5q, 7q, 17p, and complex karyotype. Prognostic evaluation of TP53MUT AML-MRC revealed no difference in outcome between TP53 double/multi-hit state and single-hit state. Patients with high TP53MUT variant allele frequency (VAF) had inferior outcomes compared to patients with low TP53MUT VAF. When compared to TP53WT patients, TP53MUT patients had inferior outcomes regardless of MRC-defining criteria, TP53 allelic state, or VAF. TP53 mutations and elevated serum LDH were independent predictors for inferior OS and EFS, while PHF6 mutations and transplantation were independent predictors for favorable OS and EFS. NRAS mutation was an independent predictor for favorable EFS. CONCLUSIONS Our study suggests that TP53MUT AML-MRC defines a very-high-risk subentity of AML in which novel therapies should be explored.
Collapse
Affiliation(s)
- Davidson Zhao
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| | - Entsar Eladl
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
- Department of Pathology, Faculty of Medicine Mansoura University Mansoura Egypt
| | - Mojgan Zarif
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| | - José‐Mario Capo‐Chichi
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| | - Andre Schuh
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| | - Eshetu Atenafu
- Department of Biostatistics University Health Network Toronto Ontario Canada
| | - Mark Minden
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| | - Hong Chang
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Department of Hematology and Medical Oncology University Health Network Toronto Ontario Canada
| |
Collapse
|
36
|
Ambinder AJ, DeZern AE. Navigating the contested borders between myelodysplastic syndrome and acute myeloid leukemia. Front Oncol 2022; 12:1033534. [PMID: 36387170 PMCID: PMC9650616 DOI: 10.3389/fonc.2022.1033534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 10/23/2023] Open
Abstract
Myelodysplastic syndrome and acute myeloid leukemia are heterogeneous myeloid neoplasms which arise from the accumulation of mutations in a myeloid stem cell or progenitor that confer survival or growth advantages. These disease processes are formally differentiated by clinical, laboratory, and morphological presentations, especially with regard to the preponderance of blasts in the peripheral blood or bone marrow (AML); however, they are closely associated through their shared lineage as well as their existence on a spectrum with some cases of MDS displaying increased blasts, a feature that reflects more AML-like behavior, and the propensity for MDS to transform into AML. It is increasingly recognized that the distinctions between these two entities result from the divergent patterns of genetic alterations that drive each of them. Mutations in genes related to chromatin-remodeling and the spliceosome are seen in both MDS and AML arising out of antecedent MDS, while mutations in genes related to signaling pathways such as RAS or FLT3 are more typically seen in AML or otherwise are a harbinger of transformation. In this review, we focus on the insights into the biological and genetic distinctions and similarities between MDS and AML that are now used to refine clinical prognostication, guide disease management, and to inform development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Amy E. DeZern
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
37
|
Huerga-Domínguez S, Villar S, Prósper F, Alfonso-Piérola A. Updates on the Management of Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:4756. [PMID: 36230677 PMCID: PMC9563665 DOI: 10.3390/cancers14194756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Acute myeloid leukemia is a heterogeneous disease defined by a large spectrum of genetic aberrations that are potential therapeutic targets. New targeted therapies have changed the landscape for a disease with poor outcomes. They are more effective than standard chemotherapy with a good safety profile. For "fit patients" in first-line, the combination of gemtuzumab ozogamicin or midostaurin with intensive chemotherapy or Vyxeos is now considered the "standard of care" for selected patients. On the other hand, for "unfit patients", azacitidine-venetoclax has been consolidated as a frontline treatment, while other combinations with magrolimab or ivosidenib are in development. Nevertheless, global survival results, especially in relapsed or refractory patients, remain unfavorable. New immunotherapies or targeted therapies, such as Menin inhibitors or sabatolimab, represent an opportunity in this situation. Future directions will probably come from combinations of different targeted therapies ("triplets") and maintenance strategies guided by measurable residual disease.
Collapse
Affiliation(s)
| | | | | | - Ana Alfonso-Piérola
- Hematology and Hemotherapy Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
38
|
How I Treat TP53-Mutated Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cancers (Basel) 2022; 14:cancers14184519. [PMID: 36139679 PMCID: PMC9496940 DOI: 10.3390/cancers14184519] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/19/2022] Open
Abstract
TP53-mutated acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are among the myeloid malignancies with the poorest prognosis. In this review, we analyze the prognosis of these two diseases, focussing particularly on the extent of the mono or biallelic mutation status of TP53 mutation, which is largely correlated with cytogenetic complexity. We discuss the possible/potential improvement in outcome based on recent results obtained with new drugs (especially eprenetapopt and magrolimab). We also focus on the impact of allogeneic hematopoietic stem cell transplantation (aHSCT) including post aHSCT treatment.
Collapse
|
39
|
Uy GL, Newell LF, Lin TL, Goldberg SL, Wieduwilt MJ, Ryan RJ, Faderl S, Lancet JE. Transplant outcomes after CPX-351 vs 7 + 3 in older adults with newly diagnosed high-risk and/or secondary AML. Blood Adv 2022; 6:4989-4993. [PMID: 35443022 PMCID: PMC9631647 DOI: 10.1182/bloodadvances.2021006468] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Geoffrey L. Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Laura F. Newell
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR
| | - Tara L. Lin
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Stuart L. Goldberg
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | | | - Robert J. Ryan
- Department of Biostatistics, Jazz Pharmaceuticals, Philadelphia, PA
| | - Stefan Faderl
- Department of Clinical Development, Jazz Pharmaceuticals, Palo Alto, CA; and
| | - Jeffrey E. Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
40
|
Alotaibi S, Niederwieser D, Ahmed SO, Sanz J, Mohty M, Aljurf M. Current Status of CPX-351 Therapy in Acute Myeloid Leukemia and Myelodysplastic Syndrome. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:575-580. [PMID: 35418351 DOI: 10.1016/j.clml.2022.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 06/14/2023]
Abstract
Acute myeloid leukemia (AML) treatment landscape had evolved over the last decades with better understanding of the disease genomics and the use of the targeted therapy, despite this treatment evolution, 7 + 3 remains the mainstay treatment for most AML cases. Many attempts had been made to improve the treatment outcome with 7 + 3 like manipulating the doses or the duration, but with no significant change in the outcome. In 2017 FDA approved CPX-351,a liposomal formulation of cytarabine and daunorubicin at a fixed 5:1 molar ratio, for the treatment of adults with newly diagnosed AML with myelodysplasia-related changes and therapy-related AML (t-AML). Since the approval, many trials were conducted or still ongoing in assessing the role of CPX-351 in treating different patient populations, AML subcategories or when combined with different agents. In this review, we will summarize the current role of CPX-351 in treating this largely heterogeneous disease.
Collapse
Affiliation(s)
- Shaykhah Alotaibi
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA.
| | | | - Syed Osman Ahmed
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA
| | - Jaime Sanz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, Paris, France
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA
| |
Collapse
|
41
|
Molica M, Perrone S, Mazzone C, Cesini L, Canichella M, de Fabritiis P. CPX-351: An Old Scheme with a New Formulation in the Treatment of High-Risk AML. Cancers (Basel) 2022; 14:cancers14122843. [PMID: 35740508 PMCID: PMC9221356 DOI: 10.3390/cancers14122843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Secondary AML (s-AML) including therapy-related acute myeloid leukemia (t-AML) and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) represent approximately one quarter of all AML cases. These AML subcategories are predominantly associated with advanced age and present a specific biologic profile including adverse genetics and a multidrug resistance phenotype, which often determine dramatically poor outcomes after conventional chemotherapy. In 2017, the FDA approved CPX-351, a liposomal formulation of cytarabine and daunorubicin at a fixed 5:1 molar ratio, for the treatment of adults with newly diagnosed t-AML and MRC-AML. Since the approval, many trials have been conducted or are still ongoing in order to assess the role of CPX-351 as frontline treatment in different AML subcategories, as a potential bridge to transplant or in combination with target therapies. In this review, we will discuss the current role of CPX-351 in treating these high-risk AML, focusing on how its use may potentially change the treatment paradigms of AML. Abstract Therapy-related acute myeloid leukemia (t-AML) and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) represent aggressive diseases characterized by a dismal prognosis if compared with de novo acute myeloid leukemia, especially in older patients. In these AML subsets, standard chemotherapy regimens produce poor response rates and unsatisfactory outcomes. Historically, conventional approaches consisted of an anthracycline combined with continuous infusion of cytarabine for 7 days, the “3+7” regimen. Several attempts have been conducted to ameliorate this combination regimen but inconsistent improvements in response rates and no significant changes in overall survival have been observed, until the recent introduction of targeted molecules. A liposomal formulation of traditional chemotherapy agents cytarabine and daunorubicin, termed CPX-351, enhances pharmacodynamics and synergistic effects through the maintenance of the optimal 5:1 molar ratio, which extends the treatment’s half-life and increases the bone marrow tropism of the drug. The use of CPX-351 in newly diagnosed AML-MRC and t-AML patients aged 60–75 years has demonstrated superior remission rates compared to conventional chemotherapy and improvements in event-free and overall survival. Recently, published data from a 5-year follow-up highlighted evidence that CPX-351 has the ability to produce and contribute to long-term remission and survival in older patients with newly diagnosed high-risk/secondary AML. Future perspectives include evaluation of dose intensification with CPX-351 in high-risk settings, combining this agent with targeted therapies, and better understanding the mechanism of improved responses in t-AML and AML-MRC. In this review, we will examine the role of CPX-351 inside the new AML therapeutic scenario and how its employment could potentially modify the treatment algorithm of high-risk and elderly patients with AML
Collapse
Affiliation(s)
- Matteo Molica
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy; (C.M.); (L.C.); (M.C.); (P.d.F.)
- Correspondence:
| | - Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, 04100 Latina, Italy;
| | - Carla Mazzone
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy; (C.M.); (L.C.); (M.C.); (P.d.F.)
| | - Laura Cesini
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy; (C.M.); (L.C.); (M.C.); (P.d.F.)
| | - Martina Canichella
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy; (C.M.); (L.C.); (M.C.); (P.d.F.)
| | - Paolo de Fabritiis
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, 00144 Rome, Italy; (C.M.); (L.C.); (M.C.); (P.d.F.)
- Department of Biomedicina and Prevenzione, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
42
|
Shallis RM, Bewersdorf JP, Stahl MF, Halene S, Zeidan AM. Are We Moving the Needle for Patients with TP53-Mutated Acute Myeloid Leukemia? Cancers (Basel) 2022; 14:2434. [PMID: 35626039 PMCID: PMC9140008 DOI: 10.3390/cancers14102434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
The currently available therapeutic options for patients with TP53-mutated acute myeloid leukemia (AML) are insufficient, as they translate to a median overall of only 6-9 months, and less than 10% of patients undergoing the most aggressive treatments, such as intensive induction therapy and allogeneic hematopoietic stem cell transplantation, will be cured. The lack of clear differences in outcomes with different treatments precludes the designation of a standard of care. Recently, there has been growing attention on this critical area of need by way of better understanding the biology of TP53 alterations and the disparities in outcomes among patients in this molecular subgroup, reflected in the development and testing of agents with novel mechanisms of action. Promising preclinical and efficacy data exist for therapies that are directed at the p53 protein rendered dysfunctional via mutation or that inhibit the CD47/SIRPα axis or other immune checkpoints such as TIM-3. In this review, we discuss recently attractive and emerging therapeutic agents, their preclinical rationale and the available clinical data as a monotherapy or in combination with the currently accepted backbones in frontline and relapsed/refractory settings for patients with TP53-mutated AML.
Collapse
Affiliation(s)
- Rory M. Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| | - Jan P. Bewersdorf
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maximilian F. Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| |
Collapse
|
43
|
Real-world effectiveness of CPX-351 vs venetoclax and azacitidine in acute myeloid leukemia. Blood Adv 2022; 6:3997-4005. [PMID: 35507945 PMCID: PMC9278286 DOI: 10.1182/bloodadvances.2022007265] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/26/2022] [Indexed: 11/20/2022] Open
Abstract
CPX-351 and venetoclax and azacitidine (ven/aza) are both indicated as initial therapy for acute myeloid leukemia (AML) in older adults. In the absence of prospective randomized comparisons of these regimens, we used retrospective observational data to evaluate various outcomes for patients with newly diagnosed AML receiving either CPX-351 (n=217) or ven/aza (n=439). This study used both a nationwide electronic health record (EHR)-derived de-identified database and the University of Pennsylvania EHR. Our study includes 217 patients who received CPX-351 and 439 who received venetoclax/azacitidine. Ven/aza patients were older, more likely to be treated in the community, and more likely to have a diagnosis of de novo AML. Other baseline covariates were not statistically significantly different between the groups. Median overall survival (OS) for all patients was 12 months and did not differ based upon therapy (13 months for CPX-351 versus 11 months for ven/aza, HR 0.88, 95% CI 0.71-1.08, p = 0.22). Overall survival was similar across multiple sensitivity analyses. Regarding safety outcomes, early mortality was similar (10% vs. 13% at 60 days). However, documented infections were higher with CPX-351 as were rates of febrile neutropenia. Hospital length of stay, including any admission prior to next cycle of therapy, was more than twice as long for CPX-351. In this large multi-center real word dataset, there was no statistically significant difference in OS. Prospective randomized studies with careful attention to side effects, quality of life, and impact on transplant outcomes are needed in these populations.
Collapse
|
44
|
Granowicz EM, Jonas BA. Targeting TP53-Mutated Acute Myeloid Leukemia: Research and Clinical Developments. Onco Targets Ther 2022; 15:423-436. [PMID: 35479302 PMCID: PMC9037178 DOI: 10.2147/ott.s265637] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
Abstract
TP53 is a key tumor suppressor gene that plays an important role in regulating apoptosis, senescence, and DNA damage repair in response to cellular stress. Although somewhat rare, TP53-mutated AML has been identified as an important molecular subgroup with a prognosis that is arguably the worst of any. Survival beyond one year is rare after induction chemotherapy with or without consolidative allogeneic stem cell transplant. Although response rates have been improved with hypomethylating agents, outcomes remain particularly poor due to short response duration. Improvements in our understanding of AML genetics and biology have led to a surge in novel treatment options, though the clinical applicability of these agents in TP53-mutated disease remains largely unknown. This review will focus on the epidemiology, molecular characteristics, and clinical significance of TP53 mutations in AML as well as emerging treatment options that are currently being studied.
Collapse
Affiliation(s)
- Eric M Granowicz
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Brian A Jonas
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
45
|
Nguyen-Khac F, Bidet A, Daudignon A, Lafage-Pochitaloff M, Ameye G, Bilhou-Nabéra C, Chapiro E, Collonge-Rame MA, Cuccuini W, Douet-Guilbert N, Eclache V, Luquet I, Michaux L, Nadal N, Penther D, Quilichini B, Terre C, Lefebvre C, Troadec MB, Véronèse L. The complex karyotype in hematological malignancies: a comprehensive overview by the Francophone Group of Hematological Cytogenetics (GFCH). Leukemia 2022; 36:1451-1466. [DOI: 10.1038/s41375-022-01561-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
|
46
|
Refining AML Treatment: The Role of Genetics in Response and Resistance Evaluation to New Agents. Cancers (Basel) 2022; 14:cancers14071689. [PMID: 35406464 PMCID: PMC8996853 DOI: 10.3390/cancers14071689] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is an aggressive cancer of the hematopoietic system. At present, we know that AML is heterogeneous and varies from one patient to another, often characterized by specific changes in the DNA (mutations). Likewise, we know that the mutational landscape of the disease predicts its response to certain therapies and that it can change under the influence of therapy. Since 2017, the number of potential drugs intended to treat AML has substantially increased and so has our knowledge about the role of certain mutations in the prediction of disease response, relapse and resistance. In this article, we review the current state of knowledge of genetic aberrations with respect to clinical decision making. Abstract The number of treatment options for acute myeloid leukemia (AML) has greatly increased since 2017. This development is paralleled by the broad implantation of genetic profiling as an integral part of clinical studies, enabling us to characterize mutation–response, mutation–non-response, or mutation–relapse patterns. The aim of this review is to provide a concise overview of the current state of knowledge with respect to newly approved AML treatment options and the association of response, relapse and resistance with genetic alterations. Specifically, we will highlight current genetic data regarding FLT3 inhibitors, IDH inhibitors, hypomethylating agents (HMA), the BCL-2 inhibitor venetoclax (VEN), the anti-CD33 antibody conjugate gemtuzumab ozogamicin (GO) and the liposomal dual drug CPX-351.
Collapse
|
47
|
Bewersdorf JP, Patel KK, Goshua G, Shallis RM, Podoltsev NA, Huntington SF, Zeidan AM. Cost-effectiveness of liposomal cytarabine/daunorubicin in patients with newly diagnosed acute myeloid leukemia. Blood 2022; 139:1766-1770. [PMID: 35298594 PMCID: PMC8931513 DOI: 10.1182/blood.2021014401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY; and
| | - Kishan K Patel
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - George Goshua
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Nikolai A Podoltsev
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Scott F Huntington
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, and
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| |
Collapse
|
48
|
Cluzeau T, Lemoli RM, McCloskey J, Cooper T. Measurable Residual Disease in High-Risk Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:cancers14051278. [PMID: 35267586 PMCID: PMC8909238 DOI: 10.3390/cancers14051278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Assessment of measurable residual disease (MRD) identifies small numbers of acute myeloid leukemia (AML) cells that may remain after initiating treatment. The achievement of MRD negativity (no detectable AML cells remaining) typically predicts better outcomes for patients with AML. Some patients with AML have disease characteristics that put them at a higher risk of treatment failure or relapse; while outcomes for patients with high-risk AML are historically poor with traditional chemotherapy regimens, newer chemotherapy formulations (i.e., CPX-351) and targeted therapies may be more effective in achieving MRD negativity in these patients. Currently, there is no agreement on the best method for determining whether a patient has achieved MRD negativity, and the use of several different methods makes it difficult to compare outcomes across studies. Despite these challenges, regular monitoring of patients for the achievement of MRD negativity will become increasingly important in the routine management of patients with high-risk AML. Abstract Mounting evidence suggests measurable residual disease (MRD) assessments are prognostic in acute myeloid leukemia (AML). High-risk AML encompasses a subset of AML with poor response to therapy and prognosis, with features such as therapy-related AML, an antecedent hematologic disorder, extramedullary disease (in adults), and selected mutations and cytogenetic abnormalities. Historically, few patients with high-risk AML achieved deep and durable remission with conventional chemotherapy; however, newer agents might be more effective in achieving MRD-negative remission. CPX-351 (dual-drug liposomal encapsulation of daunorubicin/cytarabine at a synergistic ratio) demonstrated MRD-negativity rates of 36–64% across retrospective studies in adults with newly diagnosed high-risk AML and 84% in pediatric patients with first-relapse AML. Venetoclax (BCL2 inhibitor) demonstrated MRD-negativity rates of 33–53% in combination with hypomethylating agents for high-risk subgroups in studies of older adults with newly diagnosed AML who were ineligible for intensive therapy and 65% in combination with chemotherapy in pediatric patients with relapsed/refractory AML. However, there is no consensus on optimal MRD methodology in AML, and the use of different techniques, sample sources, sensitivity thresholds, and the timing of assessments limit comparisons across studies. Robust MRD analyses are needed in future clinical studies, and MRD monitoring should become a routine aspect of AML management.
Collapse
Affiliation(s)
- Thomas Cluzeau
- Service d’hématologie, Université Cote d’Azur, CHU de Nice, 06200 Nice, France
- Correspondence: ; Tel.: +33-492035841; Fax: +33-492035895
| | - Roberto M. Lemoli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
- Clinic of Hematology, Department of Internal Medicine (DIMI), University of Genoa, 16132 Genoa, Italy
| | - James McCloskey
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| | - Todd Cooper
- Division of Hematology/Oncology, Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA 98105, USA;
| |
Collapse
|
49
|
Voso MT, Ferrara F, Galimberti S, Rambaldi A, Venditti A. Diagnostic Workup of Acute Myeloid Leukemia: What Is Really Necessary? An Italian Survey. Front Oncol 2022; 12:828072. [PMID: 35251997 PMCID: PMC8893956 DOI: 10.3389/fonc.2022.828072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/26/2022] [Indexed: 11/23/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a wide variety of clinical presentations, morphological features, and immunophenotypes. The diagnostic approaches to AML that are adopted in Italy have been explored using an online Delphi-based process to expand the global discussion on mandatory tests for the correct diagnosis and, consequently, for optimal management of AML in clinical practice. The final results of the panel of Italian hematologists involved in this work highlight the importance of genetic evaluation for classification and risk stratification and firmly establish that karyotyping, fluorescence in situ hybridization in cases with non-evaluable karyotype, and molecular tests must be performed in every case of AML, regardless of age. Obtaining clinically relevant genetic data at diagnosis is the basis for the success of patient-tailored therapy. The Italian specialists also confirm the role of multidisciplinary diagnostics for AML, now mandatory and expected to become more important in the future context of “precision” medicine.
Collapse
Affiliation(s)
- Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- *Correspondence: Maria Teresa Voso,
| | | | - Sara Galimberti
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Alessandro Rambaldi
- Department of Oncology-Hematology, University of Milan, Milan, Italy
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
50
|
Price K, Cao Z, Lipkin C, Profant D, Robinson S. Comparison of Hospital Length of Stay and Supportive Care Utilization Between Patients Treated with CPX-351 and 7+3 for Therapy-Related Acute Myeloid Leukemia or Acute Myeloid Leukemia with Myelodysplasia-Related Changes. Clinicoecon Outcomes Res 2022; 14:21-34. [PMID: 35035224 PMCID: PMC8754465 DOI: 10.2147/ceor.s342303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/22/2021] [Indexed: 12/02/2022]
Abstract
Purpose CPX-351 is dual-drug liposomal encapsulation of daunorubicin and cytarabine at a fixed synergistic 1:5 molar ratio. This study determined current real-world use of CPX-351 versus conventional 7+3 (cytarabine+daunorubicin) therapy and evaluated hospital length of stay (LOS) and supportive care utilization in t-AML and AML-MRC. Patients and Methods This retrospective, observational study utilized the Premier Healthcare Database and included patients who were aged ≥18 years with t-AML or AML-MRC and treated with CPX-351 or 7+3 between August 1, 2017 and February 28, 2019. All patients treated with 7+3 were required to be eligible for CPX-351 based on its FDA-approved indication. Outcome variables were annualized and adjusted for patient, hospital, and clinical confounding factors. The primary outcome was inpatient LOS. Secondary outcomes included use of blood products and use of anti-infectives. Results The study included 195 qualifying patients treated with CPX-351 and 160 patients treated with 7+3 who were eligible for CPX-351. Approximately one-third of the patients treated with CPX-351 were administered therapy in a hospital-based outpatient setting, and all patients treated with 7+3 received it in the inpatient setting. The regression-adjusted annualized inpatient LOS was shorter with CPX-351 than 7+3 (mean of 183.7 vs 197.1 days, p<0.001). The difference in mean-adjusted LOS was most pronounced for t-AML, with a mean-adjusted LOS of 168.9 versus 192.5 days for CPX-351 versus 7+3, respectively (nominal p<0.001). Supportive care utilization, including the number of administrations of red blood cells, the number of administrations of platelets, and the number of days on anti-infectives, was similar between treatment groups. Conclusion CPX-351 was associated with a shorter inpatient LOS than 7+3. Supportive care use, including blood products and anti-infectives, was similar for CPX-351 and 7+3. These findings suggest CPX-351 conveys resource advantages over 7+3 in patients with newly diagnosed t-AML and AML-MRC.
Collapse
Affiliation(s)
| | - Zhun Cao
- Premier Inc., Charlotte, NC, USA
| | | | | | | |
Collapse
|