1
|
Greiner J, Mohamed E, Fletcher DM, Schuler PJ, Schrezenmeier H, Götz M, Guinn BA. Immunotherapeutic Potential of Mutated NPM1 for the Treatment of Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:3443. [PMID: 39456538 PMCID: PMC11505958 DOI: 10.3390/cancers16203443] [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: 06/19/2024] [Revised: 09/15/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a malignant disease of the blood and bone marrow that is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Nucleophosmin 1 (NPM1) gene mutations are the most common genetic abnormality in AML, detectable in blast cells from about one-third of adults with AML. AML NPM1mut is recognized as a separate entity in the World Health Organization classification of AML. Clinical and survival data suggest that patients with this form of AML often have a more favorable prognosis, which may be due to the immunogenicity created by the mutations in the NPM1 protein. Consequently, AML with NPM1mut can be considered an immunogenic subtype of AML. However, the underlying mechanisms of this immunogenicity and associated favorable survival outcomes need to be further investigated. Immune checkpoint molecules, such as the programmed cell death-1 (PD-1) protein and its ligand, PD-L1, play important roles in leukemogenesis through their maintenance of an immunosuppressive tumor microenvironment. Preclinical trials have shown that the use of PD-1/PD-L1 checkpoint inhibitors in solid tumors and lymphoma work best in novel therapy combinations. Patients with AML NPM1mut may be better suited to immunogenic strategies that are based on the inhibition of the PD-1 immune checkpoint pathway than patients without this mutation, suggesting the genetic landscape of patients may also inform best practice for the use of PD-1 inhibitors.
Collapse
Affiliation(s)
- Jochen Greiner
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Internal Medicine, Diakonie Hospital Stuttgart, 70176 Stuttgart, Germany
| | - Eithar Mohamed
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| | - Daniel M. Fletcher
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| | - Patrick J. Schuler
- Department of Otorhinolaryngology, University Hospital Ulm, 89075 Ulm, Germany;
- Department of Oto-Rhino-Laryngology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm, 89073 Ulm, Germany;
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, 89081 Ulm, Germany
| | - Marlies Götz
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Internal Medicine, Diakonie Hospital Stuttgart, 70176 Stuttgart, Germany
| | - Barbara-ann Guinn
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; (E.M.); (D.M.F.)
| |
Collapse
|
2
|
Zhou W, Xu Z, Liu S, Lou X, Liu P, Xie H, Zhang S, Liu X, Zhuo B, Huang H. Landscape of clinical drug development of ADCs used for the pharmacotherapy of cancers: an overview of clinical trial registry data from 2002 to 2022. BMC Cancer 2024; 24:898. [PMID: 39060958 PMCID: PMC11282866 DOI: 10.1186/s12885-024-12652-5] [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: 01/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND To provide reference for clinical development of ADCs in the industry, we analyzed the landscape and characteristics of clinical trials about antibody-drug conjugates (ADCs). METHOD Clinical trials to study ADCs used for the pharmacotherapy of cancers initiated by the sponsor were searched in the Cite line Pharma Intelligence (Trialtrove database), and the landscape and characteristics of these clinical trials were analyzed from multiple perspectives, such as the number, phases, status, indications, and targets of the clinical trials. RESULT As of December 31, 2022, a total of 431 clinical trials have been initiated to study ADCs used for the pharmacotherapy of cancers, and the number of the last 10 years was 5.5 times as large as the first 11 years. These clinical trials involved 47 indications, including breast cancer, lymphoma (lymphoma, non-Hodgkin's and lymphoma, Hodgkin's), unspecified solid tumor, bladder cancer and lung cancer (lung, non-small cell cancer and lung, small cell cancer). As for each of these five indications, 50 + clinical trials have been carried out, accounting for as high as 48.50% (454/936). ADCs involve 38 targets, which are relatively concentrated. Among them, ERBB2 (HER2) and TNFRSF8 (CD30) involve in 100 + registered clinical trials, and TNFRSF17 (BCMA), NECTIN4 and CD19 in 10 + trials. The clinical trials for these five targets account for 79.02% (354/448) of the total number. Up to 93.97% (405/431) of these clinical trials explored the correlation between biomarkers and efficacy. Up to 45.91% (292/636) of Lots (lines of treatment) applied in the clinical trials were the second line. Until December 31, 2022, 54.52% (235/431) of the clinical trials have been completed or terminated. CONCLUSION ADCs are a hotspot of research and development in oncology clinical trials, but the indications, targets, phases, and Lot that have been registered are seemingly relatively concentrated at present. This study provides a comprehensive analysis which can assist researchers/developer quickly grasp relevant knowledge to assess a product and also providing new clues and ideas for future research.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China.
| | - Zhiyuan Xu
- Department of Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shu Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaohuan Lou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Pengcheng Liu
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Huali Xie
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shuiyan Zhang
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Xi Liu
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Baoshan Zhuo
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Hongbing Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
3
|
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: 2.5] [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
|
4
|
Immune-Based Therapeutic Strategies for Acute Myeloid Leukemia. Cancers (Basel) 2021; 14:cancers14010105. [PMID: 35008269 PMCID: PMC8744886 DOI: 10.3390/cancers14010105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary This review summarizes various therapeutic immune approaches representing their targets, the efficacy and toxicity in the treatment of acute myeloid leukemia. In particular, immune checkpoint inhibitors, bispecific T-cell engager antibodies and chimeric antigen receptor-T-cell approaches are highlighted. Abstract The development and design of immune-based strategies have become an increasingly important topic during the last few years in acute myeloid leukemia (AML), based on successful immunotherapies in solid cancer. The spectrum ranges from antibody drug conjugates, immune checkpoint inhibitors blocking programmed cell death protein 1 (PD1), cytotoxic T lymphocyte antigen 4 (CTLA4) or T cell immunoglobulin and mucin domain containing-3 (TIM3), to T-cell based monoclonal and bispecific T-cell engager antibodies, chimeric antigen receptor-T-cell (CAR-T) approaches and leukemia vaccines. Currently, there are many substances in development and multiple phase I/II studies are ongoing. These trials will help us to deepen our understanding of the pathogenesis of AML and facilitate the best immunotherapeutic strategy in AML. We discuss here the mode of action of immune-based therapies and provide an overview of the available data.
Collapse
|
5
|
Thol F. What to use to treat AML: the role of emerging therapies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:16-23. [PMID: 34889359 PMCID: PMC8791134 DOI: 10.1182/hematology.2021000309] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The development and approval of novel substances have resulted in substantial improvements in the treatment of acute myeloid leukemia (AML). In the current era of novel treatment options, genetic and molecular testing at the time of diagnosis and relapse becomes increasingly relevant. Midostaurin in combination with intensive chemotherapy is the standard of care as upfront therapy in younger AML patients with mutated fms-related tyrosine kinase 3 (FLT3). Gilteritinib, a second- generation FLT3 inhibitor, represents a key drug for relapsed/refractory (R/R) FLT3-mutated AML patients. Targeted therapy has also been developed for patients with mutated isocitrate dehydrogenase 1 (IDH1) and IDH2. The US Food and Drug Administration (FDA) approved ivosidenib as a monotherapy for newly diagnosed older adult IDH1-mutated patients and enasidenib for R/R IDH2-mutated AML patients. CPX-351, a liposomal formulation of daunorubicin and cytarabine, has become an important upfront treatment strategy for fit patients with therapy-related AML or AML with myelodysplasia-related changes that are generally challenging to treat. The antibody drug conjugate gemtuzumab ozogamicin was approved in combination with intensive therapy for patients with newly diagnosed (FDA/European Medicines Agency [EMA]) as well as R/R CD33+ AML. The combination of venetoclax, an oral selective B-cell leukemia/lymphoma-2 inhibitor, with hypomethylating agents or low-dose AraC (LDAC) has changed the treatment landscape and prognosis for older adult patients very favorably. The addition of glasdegib, a small-molecule hedgehog inhibitor, to LDAC is another example of novel options in older patients. Further substances have shown promising results in early clinical trials.
Collapse
Affiliation(s)
- Felicitas Thol
- Correspondence Felicitas Thol, Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str 1, 30625 Hannover, Germany; e-mail:
| |
Collapse
|
6
|
Liu Y, Wang S, Schubert ML, Lauk A, Yao H, Blank MF, Cui C, Janssen M, Schmidt C, Göllner S, Kleist C, Zhou F, Rahfeld JU, Sauer T, Schmitt M, Müller-Tidow C. CD33-directed immunotherapy with third-generation chimeric antigen receptor T cells and gemtuzumab ozogamicin in intact and CD33-edited acute myeloid leukemia and hematopoietic stem and progenitor cells. Int J Cancer 2021; 150:1141-1155. [PMID: 34766343 DOI: 10.1002/ijc.33865] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapies, such as chimeric antigen receptor (CAR) modified T cells and antibody-drug conjugates (ADCs), have revolutionized the treatment of cancer, especially of lymphoid malignancies. The application of targeted immunotherapy to patients with acute myeloid leukemia (AML) has been limited in particular by the lack of a tumor-specific target antigen. Gemtuzumab ozogamicin (GO), an ADC targeting CD33, is the only approved immunotherapeutic agent in AML. In our study, we introduce a CD33-directed third-generation CAR T-cell product (3G.CAR33-T) for the treatment of patients with AML. 3G.CAR33-T cells could be expanded up to the end-of-culture, that is, 17 days after transduction, and displayed significant cytokine secretion and robust cytotoxic activity when incubated with CD33-positive cells including cell lines, drug-resistant cells, primary blasts as well as normal hematopoietic stem and progenitor cells (HSPCs). When compared to second-generation CAR33-T cells, 3G.CAR33-T cells exhibited higher viability, increased proliferation and stronger cytotoxicity. Also, GO exerted strong antileukemia activity against CD33-positive AML cells. Upon genomic deletion of CD33 in HSPCs, 3G.CAR33-T cells and GO preferentially killed wildtype leukemia cells, while sparing CD33-deficient HSPCs. Our data provide evidence for the applicability of CD33-targeted immunotherapies in AML and its potential implementation in CD33 genome-edited stem cell transplantation approaches.
Collapse
Affiliation(s)
- Yi Liu
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL) and Heidelberg University Hospital, Heidelberg, Germany
| | - Sanmei Wang
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | | | - Annika Lauk
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hao Yao
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Chunhong Cui
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Maike Janssen
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Christina Schmidt
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefanie Göllner
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Fengbiao Zhou
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Tim Sauer
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Schmitt
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL) and Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Jaramillo S, Krisam J, Le Cornet L, Kratzmann M, Baumann L, Sauer T, Crysandt M, Rank A, Behringer D, Teichmann L, Görner M, Trappe RU, Röllig C, Krause S, Hanoun M, Hopfer O, Held G, Buske S, Fransecky L, Kayser S, Schliemann C, Schaefer-Eckart K, Al-Fareh Y, Schubert J, Geer T, Kaufmann M, Brecht A, Niemann D, Kieser M, Bornhäuser M, Platzbecker U, Serve H, Baldus CD, Müller-Tidow C, Schlenk RF. Rationale and design of the 2 by 2 factorial design GnG-trial: a randomized phase-III study to compare two schedules of gemtuzumab ozogamicin as adjunct to intensive induction therapy and to compare double-blinded intensive postremission therapy with or without glasdegib in older patients with newly diagnosed AML. Trials 2021; 22:765. [PMID: 34732236 PMCID: PMC8564967 DOI: 10.1186/s13063-021-05703-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/01/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Overall survival remains poor in older patients with acute myeloid leukemia (AML) with less than 10% being alive after 5 years. In recent studies, a significant improvement in event-free, relapse-free and overall survival was shown by adding gemtuzumab ozogamicin (GO), a humanized antibody-drug conjugate directed against CD33, to intensive induction therapy once or in a sequential dosing schedule. Glasdegib, the small-molecule inhibitor of smoothened (SMO), also showed improved overall survival in patients not eligible for intensive chemotherapy when combined with low-dose cytarabine compared to low-dose cytarabine alone. These findings warrant further investigations in the phase III GnG trial. METHODS/DESIGN This is a randomized phase III trial with measurable residual disease (MRD) after induction therapy and event-free survival (EFS) as primary endpoints. The two research questions are addressed in a 2 by 2 factorial design. Patients age 60 years and older are upfront randomized 1:1 in one of the two induction arms: GO administered to intensive induction therapy on days 1,4, and 7 versus GO administered once on day 1 (GO-147 versus GO-1), and double-blinded 1:1 in one of the subsequent treatment arms glasdegib vs. placebo as adjunct to consolidation therapy and as single-agent maintenance therapy for six months. Chemotherapy backbone for induction therapy consists of standard 7 + 3 schedule with cytarabine 200 mg/m2 continuously days 1 to 7, daunorubicin 60 mg/m2 days 1, 2, and 3 and high-dose cytarabine (1 g/m2, bi-daily, days 1, 2, and 3) for consolidation therapy. Addressing two primary endpoints, MRD-negativity after induction therapy and event-free survival (EFS), 252 evaluable patients are needed to reject each of the two null hypotheses at a two-sided significance level of 2.5% with a power of at least 85%. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL STATUS Protocol version: 1st version 20.10.2020, no amendments yet. Study initiation on February 16, 2021. First patient was recruited on April 1st. TRIAL REGISTRATION ClinicalTrials.gov NCT04093505 ; EudraCT 2019-003913-32. Registered on October 30, 2018.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| | - Johannes Krisam
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Lucian Le Cornet
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Markus Kratzmann
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Lukas Baumann
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Crysandt
- Department of Medicine IV, Aachen University Hospital, Aachen, Germany
| | - Andreas Rank
- Department of Medicine II, Augsburg University Hospital, Augsburg, Germany
| | - Dirk Behringer
- Department of Hematology, Oncology and Palliative Medicine, Augusta Hospital Bochum, Bochum, Germany
| | - Lino Teichmann
- Department of Medicine and Polyclinic III, Bonn University Hospital, Bonn, Germany
| | - Martin Görner
- Department of Hematology, Oncology and Palliative Medicine, Community Hospital Bielefeld, Bielefeld, Germany
| | - Ralf-Ulrich Trappe
- Department of Medicine II, Prot. Diaconal Hospital Bremen, Bremen, Germany
| | - Christoph Röllig
- Department of Internal Medicine I, TU Dresden University Hospital, Dresden, Germany
| | - Stefan Krause
- Department of Medicine V, Erlangen University Hospital, Erlangen, Germany
| | - Maher Hanoun
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Olaf Hopfer
- Department of Medicine I, Hospital Frankfurt (Oder), Frankfurt (Oder), Germany
| | - Gerhard Held
- Department of Internal Medicine I, Westpfalz Hospital Kaiserslautern, Kaiserslautern, Germany
| | - Sebastian Buske
- Department of Medicine II, Community Hospital Kiel, Kiel, Germany
| | - Lars Fransecky
- Department of Internal Medicine II, Schleswig-Holstein University Hospital Kiel, Kiel, Germany
| | - Sabine Kayser
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany.,Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | | | | | - Yousef Al-Fareh
- Department of Hematology and Oncology, St. Josef Brothers' Hospital Paderborn, Paderborn, Germany
| | - Jörg Schubert
- Department of Internal Medicine II, Elbland Hospital Riesa, Riesa, Germany
| | - Thomas Geer
- Department of Medicine II, Diaconal Hospital Schwäbisch-Hall, Schwäbisch Hall, Germany
| | - Martin Kaufmann
- Department of Hematology, Oncology and Palliative Medicine, Robert-Bosch Hospital Stuttgart, Stuttgart, Germany
| | - Arne Brecht
- Department of Internal Medicine II, Helios Dr. Horst Schmidt Hospital Wiesbaden, Wiesbaden, Germany
| | - Dirk Niemann
- Department of Internal Medicine, Hematology, Oncology and Palliative Medicine, Prot. Monastery Hospital St. Jakob Koblenz, Koblenz, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, TU Dresden University Hospital, Dresden, Germany
| | - Uwe Platzbecker
- Department of Medicine I - Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Hubert Serve
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Claudia D Baldus
- Department of Internal Medicine II, Schleswig-Holstein University Hospital Kiel, Kiel, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
8
|
Gottardi M, Simonetti G, Sperotto A, Nappi D, Ghelli Luserna di Rorà A, Padella A, Norata M, Giannini MB, Musuraca G, Lanza F, Cerchione C, Martinelli G. Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin. Cancers (Basel) 2021; 13:cancers13184566. [PMID: 34572794 PMCID: PMC8469571 DOI: 10.3390/cancers13184566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by genetic and clinical heterogeneity and high mortality. Despite the recent introduction of novel pharmaceutical agents in hemato-oncology, few advancements have been made in AML for decades. In the last years, the therapeutic options have rapidly changed, with the approval of innovative compounds that provide new opportunities, together with new challenges for clinicians: among them, on 1 September, 2017 the Food and Drug Administration granted approval for Gemtuzumab Ozogamicin (GO) in combination with daunorubicin and cytarabine for the treatment of adult patients affected by newly diagnosed CD33+ AML. Benefits of GO-based regimens were also reported in the pre- and post-transplantation settings. Moreover, several biomarkers of GO response have been suggested, including expression of CD33 and multidrug resistance genes, cytogenetic and molecular profiles, minimal residual disease and stemness signatures. Among them, elevated CD33 expression on blast cells and non-adverse cytogenetic or molecular risk represent largely validated predictors of good response.
Collapse
Affiliation(s)
- Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV, IRCCS, 31033 Padua, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Alessandra Sperotto
- Hematology and Transplant Center Unit, Dipartimento di Area Medica (DAME), Udine University Hospital, 33100 Udine, Italy
| | - Davide Nappi
- Department of Hematology and Cell Bone Marrow Transplantation (CBMT), Ospedale di Bolzano, 39100 Bolzano, Italy
| | - Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Antonella Padella
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Marianna Norata
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Maria Benedetta Giannini
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Gerardo Musuraca
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Francesco Lanza
- Hematology Unit & Romagna Transplant Network, Ravenna Hospital, 48121 Ravenna, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| |
Collapse
|
9
|
Jaramillo S, Schlenk RF. Post-Induction Treatment for Acute Myeloid Leukemia: Something Change? Curr Oncol Rep 2021; 23:109. [PMID: 34272619 PMCID: PMC8285306 DOI: 10.1007/s11912-021-01092-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Until recently, improvement in terms of survival for patients with acute myeloid leukemia (AML) was achieved mostly in younger patients with dose intensification of conventional chemotherapy and a broadening use of allogeneic hematopoietic cell transplantation (allo-HCT) whereas the results remained dismal and very stable in patients older than 60 years. The current review highlights the recent developments in standard intensive post-remission chemotherapy, evidence for the use of recently approved agents, and discusses the relevance of measurable residual disease (MRD) measurement in treatment adaptation. RECENT FINDINGS Current approvals of midostaurin, venetoclax, gemtuzumab ozogamicin, VYXEOS, ivosidenib, enasidenib, glasdegib, and CC-486 have changed the structure, aim, and schedule of consolidation therapy, and new, well-tolerated agents are being evaluated as maintenance therapies. Furthermore, MRD assessment has been implemented to guide the duration and type of consolidation and maintenance therapy as well as indicate the optimal timing of allo-HCT. Novel therapies have changed the structure and perspective of post-remission therapy in AML for both young and elderly patients. In addition, MRD assessment could guide the type, duration, and intensity of consolidation and maintenance therapy.
Collapse
Affiliation(s)
- Sonia Jaramillo
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Richard F. Schlenk
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- NCT-Trial Center, NCT Heidelberg, DKFZ and Heidelberg University Hospital, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Pollard JA, Guest E, Alonzo TA, Gerbing RB, Loken MR, Brodersen LE, Kolb EA, Aplenc R, Meshinchi S, Raimondi SC, Hirsch B, Gamis AS. Gemtuzumab Ozogamicin Improves Event-Free Survival and Reduces Relapse in Pediatric KMT2A-Rearranged AML: Results From the Phase III Children's Oncology Group Trial AAML0531. J Clin Oncol 2021; 39:3149-3160. [PMID: 34048275 DOI: 10.1200/jco.20.03048] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE We investigated the impact of the CD33-targeted agent gemtuzumab ozogamicin (GO) on survival in pediatric patients with KMT2A-rearranged (KMT2A-r) acute myeloid leukemia (AML) enrolled in the Children's Oncology Group trial AAML0531 (NCT01407757). METHODS Patients with KMT2A-r AML were identified and clinical characteristics described. Five-year overall survival (OS), event-free survival (EFS), disease-free survival (DFS), and relapse risk (RR) were determined overall and for higher-risk versus not high-risk translocation partners. GO's impact on response was determined and outcomes based on consolidation approach (hematopoietic stem cell transplant [HSCT] v chemotherapy) described. RESULTS Two hundred fifteen (21%) of 1,022 patients enrolled had KMT2A-r AML. Five-year EFS and OS from study entry were 38% and 58%, respectively. EFS was superior with GO treatment (EFS 48% with GO v 29% without, P = .003), although OS was comparable (63% v 53%, P = .054). For patients with KMT2A-r AML who achieved complete remission, GO was associated with lower RR (40% GO v 66% patients who did not receive GO [No-GO], P = .001) and improved 5-year DFS (GO 57% v No-GO 33%, P = .002). GO benefit was observed in both higher-risk and not high-risk KMT2A-r subsets. For patients who underwent HSCT, prior GO exposure was associated with decreased relapse (5-year RR: 28% GO and HSCT v 73% No-GO and HSCT, P = .006). In multivariable analysis, GO was independently associated with improved EFS, improved DFS, and reduced RR. CONCLUSION GO added to conventional chemotherapy improved outcomes for KMT2A-r AML; consolidation with HSCT may further enhance outcomes. Future clinical trials should study CD33-targeted agents in combination with HSCT for pediatric KMT2A-r AML.
Collapse
Affiliation(s)
- Jessica A Pollard
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | | | - Todd A Alonzo
- University of Southern California Keck School of Medicine, Los Angeles, CA.,Children's Oncology Group, Monrovia, CA
| | | | | | | | | | | | - Soheil Meshinchi
- Fred Hutchinson Cancer Research Center, Seattle, WA.,University of Washington School of Medicine, Seattle, WA
| | | | - Betsy Hirsch
- University of Minnesota Cancer Center, Minneapolis, MN
| | | |
Collapse
|
11
|
Mareque M, Montesinos P, Font P, Guinea JM, de la Fuente A, Soto J, Oyagüez I, Brockbank J, Iglesias T, Llinares J, Sierra J. Cost-Effectiveness Analysis of Gemtuzumab Ozogamicin for First-Line Treatment of Patients with Cd-33 Positive Acute Myeloid Leukaemia in Spain. CLINICOECONOMICS AND OUTCOMES RESEARCH 2021; 13:263-277. [PMID: 33911887 PMCID: PMC8075179 DOI: 10.2147/ceor.s302097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 11/23/2022] Open
Abstract
Objective To assess the incremental cost-utility ratio (ICUR) of gemtuzumab ozogamicin (GO) + standard of care (SOC) vs SOC alone for treatment of patients with de novo AML from a Spanish Health Service perspective. Methods A cohort state-transition model, with 12 health-states, was used to estimate the lifetime accumulated cost and benefits in terms of quality-adjusted-life-years (QALYs) in AML patients with favourable, intermediate, and unknown cytogenetic profiles. Patient profile was defined based on the ALFA-0701 trial. Therapeutic regimens were defined by 5 haematologists. SOC was assumed to be idarubicin and cytarabine, the combination most used in Spain. QALYs were estimated by applying utilities for the time spent by the cohort in each health-state and utility decrements associated with adverse events (AE). Total cost (€,2020) included drug-acquisition, hematologic stem-cell transplantation, disease management, AE management and end-of-life costs. Unit costs were derived from local databases. All parameters were validated by haematologist. Costs and outcomes were discounted (3%/year). Results Higher cost/patient (€177,618 vs €151,434) and greater QALYs (5,70 vs 4,62) were obtained with GO+SOC vs SOC. The ICUR was €24,203/QALY gained. Conclusion This simulation suggests that GO + SOC could be a cost-effective option for treatment of patients with de novo AML in first line.
Collapse
Affiliation(s)
- Maria Mareque
- Pharmacoeconomics & Outcomes Research Iberia (PORIB), Madrid, Spain
| | | | - Patricia Font
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | | | | | - Itziar Oyagüez
- Pharmacoeconomics & Outcomes Research Iberia (PORIB), Madrid, Spain
| | | | | | | | - Jorge Sierra
- Hospital Universitario de La Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
12
|
Zuo P. Capturing the Magic Bullet: Pharmacokinetic Principles and Modeling of Antibody-Drug Conjugates. AAPS JOURNAL 2020; 22:105. [PMID: 32767003 DOI: 10.1208/s12248-020-00475-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
Over the past two decades, antibody-drug conjugates (ADCs) have emerged as a promising class of drugs for cancer therapy and have expanded to nononcology fields such as inflammatory diseases, atherosclerosis, and bacteremia. Eight ADCs are currently approved by FDA for clinical applications, with more novel ADCs under clinical development. Compared with traditional chemotherapy, ADCs combine the target specificity of antibodies with chemotherapeutic capabilities of cytotoxic drugs. The benefits include reduced systemic toxicity and enhanced therapeutic index for patients. However, the heterogeneous structures of ADCs and their dynamic changes following administration create challenges in their development. The understanding of ADC pharmacokinetics (PK) is crucial for the optimization of clinical dosing regimens when translating from animal to human. In addition, it contributes to the optimization of dose selection and clinical monitoring with regard to safety and efficacy. This manuscript reviews the PK characteristics of ADCs and summarizes the diverse approaches for PK modeling that can be used to evaluate an ADC at the preclinical and clinical stages to support their successful development. Despite the numerous available options, fit-for-purpose modeling approaches for the PK and PD of ADCs should be critically planned and well-thought-out to adequately support the development of an ADC.
Collapse
Affiliation(s)
- Peiying Zuo
- Pharmacometrics US, Clinical Pharmacology & Exploratory Development, Astellas Pharma, Inc., USA, 1 Astellas Way, Northbrook, Illinois, 60062, USA.
| |
Collapse
|
13
|
Muhamad NA, Mohd Dali NS, Mohd Yacob A, Kassim MSA, Lodz NA, Abdul Wahid SF, Aris T. Effect and safety of gemtuzumab ozogamicin for the treatment of patients with acute myeloid leukaemia: a systematic review protocol. BMJ Open 2020; 10:e032503. [PMID: 32540885 PMCID: PMC7299015 DOI: 10.1136/bmjopen-2019-032503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Acute myeloid leukaemia (AML) is a type of cancer in which the bone marrow makes abnormal myeloblasts (a type of white blood cell), red blood cells or platelets. Gemtuzumab ozogamicin (GO) holds promise as a new agent that also could be efficacious in newly diagnosed AML with acceptable toxicity. This paper describes the design of a protocol to conduct a systematic review of published studies assessing GO for the treatment of AML. METHOD AND ANALYSIS We will conduct a systematic review of randomised controlled trials that investigate the effect and safety of GO for the treatment of patients with AML. We will search for any eligible articles from selected electronic databases. We will follow the Preferred Reporting Items for Systematic reviews and Meta-Analysis for study selection and reporting. We will use The Cochrane Handbook for Systematic Reviews of Interventions and Meta-Analysis as guidance to select eligible studies. All data will be extracted using a standardised data extraction form. ETHICS AND DISSEMINATION There was no patient involved in this study, therefore no ethical consideration is needed. The findings of this study will be disseminated in a peer-reviewed journal and any relevant conference presentation. PROSPERO REGISTRATION NUMBER CRD42019123286.
Collapse
Affiliation(s)
- Nor A Muhamad
- Sector for Evidence-Based, National Institutes of Health Malaysia, Shah Alam, Selangor, Malaysia
- Institute for Public Health, National Institutes of Health, Shah Alam, Selangor, Malaysia
| | - Nor S Mohd Dali
- Institute for Medical Research, National Institutes of Health, Ministry of Health, Shah Alam, Selangor, Malaysia
| | - Aliza Mohd Yacob
- Institute for Medical Research, National Institutes of Health, Ministry of Health, Shah Alam, Selangor, Malaysia
| | - Mohd S A Kassim
- Institute for Public Health, National Institutes of Health, Shah Alam, Selangor, Malaysia
| | - Noor A Lodz
- Institute for Public Health, National Institutes of Health, Shah Alam, Selangor, Malaysia
| | - S F Abdul Wahid
- Cell Therapy Centre, Pusat Perubatan Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Tahir Aris
- Institute for Medical Research, National Institutes of Health, Ministry of Health, Shah Alam, Selangor, Malaysia
| |
Collapse
|
14
|
Hibma J, Knight B. Population Pharmacokinetic Modeling of Gemtuzumab Ozogamicin in Adult Patients with Acute Myeloid Leukemia. Clin Pharmacokinet 2020; 58:335-347. [PMID: 30062662 DOI: 10.1007/s40262-018-0699-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Gemtuzumab ozogamicin is an antibody-drug conjugate composed of the anti-CD33 monoclonal antibody hP67.6 covalently linked to N-acetyl-gamma-calicheamicin dimethylhydrazide, a potent cytotoxic antibiotic. The aim of this study was to characterize the population pharmacokinetics of gemtuzumab ozogamicin, represented by total hP67.6 antibody and unconjugated calicheamicin, in adult patients with acute myeloid leukemia to support drug dosing strategies and explore intrinsic and extrinsic factors that may influence exposure. Pharmacokinetic data from seven previous phase I and II studies in adult patients with relapsed, refractory, or de novo acute myeloid leukemia were integrated and analyzed using nonlinear mixed-effects modeling. METHODS The pharmacokinetics of total hP67.6 antibody was described in 407 patients (5643 concentrations) who received gemtuzumab ozogamicin doses ranging from 0.25 to 9 mg/m2 using a two-compartment model with linear and time-dependent clearance components. The pharmacokinetics of unconjugated calicheamicin was characterized in 338 patients (4281 concentrations) using a two-compartment model with an input rate of formation dependent on the amount of hP67.6 eliminated. No statistically significant baseline covariates (sex, albumin, bone marrow, and peripheral blast percentage) demonstrated a clinically meaningful impact. RESULTS AND CONCLUSION Total hP67.6 antibody disposition did not appear altered in patients with mild or moderate renal disease or hepatic impairment. Gemtuzumab ozogamicin was approved for the treatment of acute myeloid leukemia by the US Food and Drug Administration in September 2017. The model-based simulations described here provided a pharmacokinetic rationale for the approved dosing regimen of 3 mg/m2 on days 1, 4, and 7, and served as the basis for all exposure-response modeling included in the recent Biologics License Application submission. Clinical trials identifiers: 0903A1-101-US; 0903A1-103-JA; 0903B1-201-US/CA (NCT00003131); 0903B1-202-EU; 0903B1-203-US/EU (NCT00003673); 0903B1-205-US/EU/AU (NCT00037596); and 0903B1-206-US/EU/AU (NCT00037583).
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/therapeutic use
- Calicheamicins/blood
- Calicheamicins/pharmacokinetics
- Drug Design
- Female
- Gemtuzumab/administration & dosage
- Gemtuzumab/pharmacokinetics
- Gemtuzumab/therapeutic use
- Humans
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/ethnology
- Male
- Middle Aged
- Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
- Sialic Acid Binding Ig-like Lectin 3/blood
- Sialic Acid Binding Ig-like Lectin 3/pharmacokinetics
- United States
- United States Food and Drug Administration
- Young Adult
Collapse
Affiliation(s)
- Jennifer Hibma
- Pfizer Inc., 10555 Science Center Drive, 2405, San Diego, CA, 92121, USA.
| | - Beverly Knight
- Pfizer Inc., 10555 Science Center Drive, 2405, San Diego, CA, 92121, USA
| |
Collapse
|
15
|
Mahalleh M, Shabani M, Rayzan E, Rezaei N. Reinforcing the primary immunotherapy modulators against acute leukemia; monoclonal antibodies in AML. Immunotherapy 2019; 11:1583-1600. [PMID: 31841068 DOI: 10.2217/imt-2019-0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent therapeutic advances in cancer treatment recruit immune system potentiation against malignant cells. Numerous ongoing clinical trials on immunotherapy methods, either monotherapy or combination therapy, are investigating the impeding factors on the way of acute myeloid leukemia (AML) treatment. Due to the genetic diversity in AML progenitors, combining various strategies is more likely to be useful for improving patient outcomes. This review describes the details of applying monoclonal antibodies against AML, focusing on CD33, CD123, FLT3, CD45 and CD66 targeting. Furthermore, it clarifies the importance of immunotoxins, bispecific antibodies, chimeric antigen receptor (CAR)-T cells and T cell receptor-modified cells as reinforcing agents for monoclonal antibodies.
Collapse
Affiliation(s)
- Mehrdad Mahalleh
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsima Shabani
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Huang X, Lin H, Huang F, Xie Y, Wong KH, Chen X, Wu D, Lu A, Yang Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019; 17:1559325819887048. [PMID: 31853234 PMCID: PMC6906351 DOI: 10.1177/1559325819887048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy, which is commonly
associated with high incidence and mortality among adult patients. The standard
induction regimen for AML has been substantially unchanged over the past 40
years, for which novel nanomedicines have represented a promising strategy in
AML therapies. Despite developments of multiple nanoparticles formulated with
drugs or genes, less there is not much information available about approaches in
AML is available. This review presents an overview of nanomedicines currently
being evaluated in AML. First, it briefly summarized conventional chemotherapies
in use. Second, nanomedicines presently ongoing in clinical trials or
preclinical researches were classified and described, with illustrative examples
from recent literatures. Finally, limitations and potential safety issues
concerns in clinical translation of AML treatment were discussed as well.
Collapse
Affiliation(s)
- Xiao Huang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Feng Huang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Dongyue Wu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
17
|
[What is recommended in the treatment of acute myeloid leukemia?]. Internist (Berl) 2019; 60:1240-1250. [PMID: 31690995 DOI: 10.1007/s00108-019-00696-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by a malignant transformation and proliferation of myeloid progenitor cells that cause a replacement of normal hematopoiesis. Diagnostic workup for AML includes cytogenetic analysis and mutational screening covering frequently mutated genes in AML. The genetic analysis is required for risk stratification and treatment decisions. Very recently, three novel drugs have been approved for patients who can be intensively treated: a tyrosine kinase inhibitor (midostaurin) for patients with FLT3 mutations, a liposomal formulation of chemotherapy (CPX) for patients with features of secondary AML, and a CD33 antibody-drug conjugate (gemtuzumab-ozogamicin) for AML with CD33 expression. Allogeneic stem cell transplantation remains an important treatment strategy for patients with intermediate- or high-risk AML and for patients with relapsed AML. For elderly patients who cannot undergo intensive treatment, demethylating agents are the treatment of choice. The aim is to prolong life expectancy with acceptable quality of life. In recent clinical trials, novel drugs have shown promising results in this patient population. Some of these drugs have already been approved in the US. Among these drugs are the Bcl‑2 inhibitor venetoclax, which is already approved in Germany for chronic lymphatic leukemia, as well as IDH1/IDH2 inhibitors (the latter for patients with IDH1/IDH2 mutated AML). Acute promyelocytic leukemia represents a special type of AML that should be treated with a combination of all-trans retinoic acid and arsenic trioxide leading to excellent outcome.
Collapse
|
18
|
Hofmann S, Schubert ML, Wang L, He B, Neuber B, Dreger P, Müller-Tidow C, Schmitt M. Chimeric Antigen Receptor (CAR) T Cell Therapy in Acute Myeloid Leukemia (AML). J Clin Med 2019; 8:jcm8020200. [PMID: 30736352 PMCID: PMC6406805 DOI: 10.3390/jcm8020200] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 02/07/2023] Open
Abstract
Despite high response rates after initial chemotherapy in patients with acute myeloid leukemia (AML), relapses occur frequently, resulting in a five-year-survival by <30% of the patients. Hitherto, allogeneic hemotopoietic stem cell transplantation (allo-HSCT) is the best curative treatment option in intermediate and high risk AML. It is the proof-of-concept for T cell-based immunotherapies in AML based on the graft-versus-leukemia (GvL)-effect, but it also bears the risk of graft-versus-host disease. CD19-targeting therapies employing chimeric antigen receptor (CAR) T cells are a breakthrough in cancer therapy. A similar approach for myeloid malignancies is highly desirable. This article gives an overview on the state-of-the art of preclinical and clinical studies on suitable target antigens for CAR T cell therapy in AML patients.
Collapse
Affiliation(s)
- Susanne Hofmann
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Maria-Luisa Schubert
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Lei Wang
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Bailin He
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Brigitte Neuber
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Peter Dreger
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| | - Carsten Müller-Tidow
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| | - Michael Schmitt
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Schlenk RF, Jaramillo S, Müller-Tidow C. What's new in consolidation therapy in AML? Semin Hematol 2018; 56:96-101. [PMID: 30926097 DOI: 10.1053/j.seminhematol.2018.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/17/2022]
Abstract
Intensive induction chemotherapy followed by postremission treatment with either high-dose cytarabine-based regimens, autologous or allogeneic hematopoietic stem cell transplantation is still recognized as the main road toward cure in acute myeloid leukemia (AML). Pretreatment risk classification remains a key determinant of type and intensity of post-remission therapy. Still, high-dose cytarabine-based consolidation therapy is a cornerstone of postremission therapy with some recent adjustments regarding dosage and schedule. Current approvals of midostaurin, gemtuzumab ozogamicin, CPX-351, and ivosidenib as well as enasidenib comprise induction as well as consolidation therapy. In recent years measurable residual disease assessment is increasingly used to dynamically fine tune treatment during postremission treatment.
Collapse
Affiliation(s)
- Richard F Schlenk
- NCT-Trial Center, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany; Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany.
| | - Sonia Jaramillo
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology, and Rheumatology at Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Single-cell analysis identifies a CD33 + subset of human cord blood cells with high regenerative potential. Nat Cell Biol 2018; 20:710-720. [PMID: 29802403 DOI: 10.1038/s41556-018-0104-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 04/13/2018] [Indexed: 12/15/2022]
Abstract
Elucidation of the identity and diversity of mechanisms that sustain long-term human blood cell production remains an important challenge. Previous studies indicate that, in adult mice, this property is vested in cells identified uniquely by their ability to clonally regenerate detectable, albeit highly variable levels and types, of mature blood cells in serially transplanted recipients. From a multi-parameter analysis of the molecular features of very primitive human cord blood cells that display long-term cell outputs in vitro and in immunodeficient mice, we identified a prospectively separable CD33+CD34+CD38-CD45RA-CD90+CD49f+ phenotype with serially transplantable, but diverse, cell output profiles. Single-cell measurements of the mitogenic response, and the transcriptional, DNA methylation and 40-protein content of this and closely related phenotypes revealed subtle but consistent differences both within and between each subset. These results suggest that multiple regulatory mechanisms combine to maintain different cell output activities of human blood cell precursors with high regenerative potential.
Collapse
|
21
|
CD64-directed microtubule associated protein tau kills leukemic blasts ex vivo. Oncotarget 2018; 7:67166-67174. [PMID: 27564103 PMCID: PMC5341865 DOI: 10.18632/oncotarget.11568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/11/2016] [Indexed: 01/04/2023] Open
Abstract
Fc gamma receptor I (FcγRI, CD64) is a well-known target antigen for passive immunotherapy against acute myeloid leukemia and chronic myelomonocytic leukemia. We recently reported the preclinical immunotherapeutic potential of microtubule associated protein tau (MAP) against a variety of cancer types including breast carcinoma and Hodgkin's lymphoma. Here we demonstrate that the CD64-directed human cytolytic fusion protein H22(scFv)-MAP kills ex vivo 15–50% of CD64+ leukemic blasts derived from seven myeloid leukemia patients. Furthermore, in contrast to the nonspecific cytostatic agent paclitaxel, H22(scFv)-MAP showed no cytotoxicity towards healthy CD64+ PBMC-derived cells and macrophages. The targeted delivery of this microtubule stabilizing agent therefore offers a promising new strategy for specific treatment of CD64+ leukemia.
Collapse
|
22
|
Lucas AT, Price LSL, Schorzman AN, Storrie M, Piscitelli JA, Razo J, Zamboni WC. Factors Affecting the Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:E10. [PMID: 31544862 PMCID: PMC6698819 DOI: 10.3390/antib7010010] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lauren S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mallory Storrie
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | | | - Juan Razo
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
23
|
Kayser S, Levis MJ. Advances in targeted therapy for acute myeloid leukaemia. Br J Haematol 2018; 180:484-500. [PMID: 29193012 PMCID: PMC5801209 DOI: 10.1111/bjh.15032] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022]
Abstract
In the past few years, research in the underlying pathogenic mechanisms of acute myeloid leukaemia (AML) has led to remarkable advances in our understanding of the disease. Cytogenetic and molecular aberrations are the most important factors in determining response to chemotherapy as well as long-term outcome, but beyond prognostication are potential therapeutic targets. Our increased understanding of the pathogenesis of AML, facilitated by next-generation sequencing, has spurred the development of new compounds in the treatment of AML, particularly the creation of small molecules that target the disease on a molecular level. Various new agents, such as tyrosine kinase inhibitors, immune checkpoint inhibitors, monoclonal or bispecific T-cell engager antibodies, metabolic and pro-apoptotic agents are currently investigated within clinical trials. The highest response rates are often achieved when new molecularly targeted therapies are combined with standard chemotherapy. Presented here is an overview of novel therapies currently being evaluated in AML.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Haematology/Oncology, German Cancer Research Centre (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mark J. Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
24
|
Wagh A, Song H, Zeng M, Tao L, Das TK. Challenges and new frontiers in analytical characterization of antibody-drug conjugates. MAbs 2018; 10:222-243. [PMID: 29293399 DOI: 10.1080/19420862.2017.1412025] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a growing class of biotherapeutics in which a potent small molecule is linked to an antibody. ADCs are highly complex and structurally heterogeneous, typically containing numerous product-related species. One of the most impactful steps in ADC development is the identification of critical quality attributes to determine product characteristics that may affect safety and efficacy. However, due to the additional complexity of ADCs relative to the parent antibodies, establishing a solid understanding of the major quality attributes and determining their criticality are a major undertaking in ADC development. Here, we review the development challenges, especially for reliable detection of quality attributes, citing literature and new data from our laboratories, highlight recent improvements in major analytical techniques for ADC characterization and control, and discuss newer techniques, such as two-dimensional liquid chromatography, that have potential to be included in analytical control strategies.
Collapse
Affiliation(s)
- Anil Wagh
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Hangtian Song
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Ming Zeng
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Li Tao
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Tapan K Das
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| |
Collapse
|
25
|
O’Sullivan JA, Carroll DJ, Bochner BS. Glycobiology of Eosinophilic Inflammation: Contributions of Siglecs, Glycans, and Other Glycan-Binding Proteins. Front Med (Lausanne) 2017; 4:116. [PMID: 28824909 PMCID: PMC5539825 DOI: 10.3389/fmed.2017.00116] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/10/2017] [Indexed: 12/31/2022] Open
Abstract
The historical focus on protein-protein interactions in biological systems, at the expense of attention given to interactions between other classes of molecules, has overlooked important and clinically relevant processes and points of potential clinical intervention. For example, the significance of protein-carbohydrate interactions, especially in the regulation of immune responses, has recently received greater recognition and appreciation. This review discusses several ways by which cell-surface lectin-glycan interactions can modulate eosinophil function, particularly at the levels of eosinophil recruitment and survival, and how such interactions can be exploited therapeutically. A primary focus is on discoveries concerning Siglec-8, a glycan-binding protein selectively expressed on human eosinophils, and its closest functional paralog in the mouse, Siglec-F. Recent advances in the synthesis of polymeric ligands, the identification of physiological ligands for Siglec-8 and Siglec-F in the airway, and the determination of the basis of glycan ligand discrimination of Siglec-8 are discussed. Important similarities and differences between these siglecs are outlined. Eosinophil expression of additional glycan-binding proteins or their glycan ligands, including interactions involving members of the selectin, galectin, and siglec families, is summarized. The roles of these molecules in eosinophil recruitment, survival, and inflammation are described. Finally, the modulation of these interactions and potential therapeutic exploitation of glycan-binding proteins and their ligands to ameliorate eosinophil-associated diseases are considered.
Collapse
Affiliation(s)
- Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Daniela J. Carroll
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
26
|
Lucas AT, Price LS, Schorzman A, Zamboni WC. Complex effects of tumor microenvironment on the tumor disposition of carrier-mediated agents. Nanomedicine (Lond) 2017; 12:2021-2042. [PMID: 28745129 DOI: 10.2217/nnm-2017-0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Major advances in carrier-mediated agents, including nanoparticle, conjugates and antibody-drug conjugates, have created revolutionary drug delivery systems in cancer over the past two decades. While these agents provide several advantages, such as greater duration of exposure and solubility, compared with their small-molecule counterparts, there is substantial variability in delivery of these agents to tissues and especially tumors. This review provides an overview of tumor microenvironment factors that affect the pharmacokinetics and pharmacodynamics of carrier-mediated agents observed in preclinical models and patients.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren Sl Price
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Allison Schorzman
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C Zamboni
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
27
|
Lamba JK, Chauhan L, Shin M, Loken MR, Pollard JA, Wang YC, Ries RE, Aplenc R, Hirsch BA, Raimondi SC, Walter RB, Bernstein ID, Gamis AS, Alonzo TA, Meshinchi S. CD33 Splicing Polymorphism Determines Gemtuzumab Ozogamicin Response in De Novo Acute Myeloid Leukemia: Report From Randomized Phase III Children's Oncology Group Trial AAML0531. J Clin Oncol 2017. [PMID: 28644774 DOI: 10.1200/jco.2016.71.2513] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Gemtuzumab ozogamicin (GO), a CD33-targeted immunoconjugate, is a re-emerging therapy for acute myeloid leukemia (AML). CD33 single nucleotide polymorphism rs12459419 C>T in the splice enhancer region regulates the expression of an alternatively spliced CD33 isoform lacking exon2 (D2-CD33), thus eliminating the CD33 IgV domain, which is the antibody-binding site for GO, as well as diagnostic immunophenotypic panels. We aimed to determine the impact of the genotype of this splicing polymorphism in patients with AML treated with GO-containing chemotherapy. Patients and Methods CD33 splicing single nucleotide polymorphism was evaluated in newly diagnosed patients with AML randomly assigned to receive standard five-course chemotherapy alone (No-GO arm, n = 408) or chemotherapy with the addition of two doses of GO once during induction and once during intensification (GO arm, n = 408) as per the Children's Oncology Group AAML0531 trial. Results The rs12459419 genotype was CC in 415 patients (51%), CT in 316 patients (39%), and TT in 85 patients (10%), with a minor allele frequency of 30%. The T allele was significantly associated with higher levels of D2-CD33 transcript ( P < 1.0E-6) and with lower diagnostic leukemic cell surface CD33 intensity ( P < 1.0E-6). Patients with the CC genotype had significantly lower relapse risk in the GO arm than in the No-GO arm (26% v 49%; P < .001). However, in patients with the CT or TT genotype, exposure to GO did not influence relapse risk (39% v 40%; P = .85). Disease-free survival was higher in patients with the CC genotype in the GO arm than in the No-GO arm (65% v 46%, respectively; P = .004), but this benefit of GO addition was not seen in patients with the CT or TT genotype. Conclusion Our results suggest that patients with the CC genotype for rs12459419 have a substantial response to GO, making this a potential biomarker for the selection of patients with a likelihood of significant response to GO.
Collapse
Affiliation(s)
- Jatinder K Lamba
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Lata Chauhan
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Miyoung Shin
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Michael R Loken
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Jessica A Pollard
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Yi-Cheng Wang
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Rhonda E Ries
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Richard Aplenc
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Betsy A Hirsch
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Susana C Raimondi
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Roland B Walter
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Irwin D Bernstein
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Alan S Gamis
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Todd A Alonzo
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Soheil Meshinchi
- Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children's Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| |
Collapse
|
28
|
Gemtuzumab ozogamicin in acute myeloid leukemia. Leukemia 2017; 31:1855-1868. [PMID: 28607471 DOI: 10.1038/leu.2017.187] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/26/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
CD33 is variably expressed on leukemia blasts in almost all patients with acute myeloid leukemia (AML) and possibly leukemia stem cells in some. Efforts to target CD33 therapeutically have focused on gemtuzumab ozogamicin (GO; Mylotarg), an antibody-drug conjugate delivering a DNA-damaging calicheamicin derivative. GO is most effective in acute promyelocytic leukemia but induces remissions in other AML types and received accelerated approval in the US in 2000. However, because a large follow-up study showed no survival improvement and increased early deaths the drug manufacturer voluntarily withdrew the US New Drug Application in 2010. More recently, a meta-analysis of data from several trials reported better survival in adults with favorable- and intermediate-risk cytogenetics but not adverse-risk AML randomized to receive GO along with intensive induction chemotherapy. As a result, GO is being re-evaluated by regulatory agencies. Responses to GO are diverse and predictive biological response markers are needed. Besides cytogenetic risk, ATP-binding cassette transporter activity and possibly CD33 display on AML blasts may predict response, but established clinical assays and prospective validation are lacking. Single-nucleotide polymorphisms in CD33 may also be predictive, most notably rs12459419 where the minor T-allele leads to decreased display of full-length CD33 and preferential translation of a splice variant not recognized by GO. Data from retrospective analyses suggest only patients with the rs12459419 CC genotype may benefit from GO therapy but confirmation is needed. Most important may be markers for AML cell sensitivity to calicheamicin, which varies over 100 000-fold, but useful assays are unavailable. Novel CD33-targeted drugs may overcome some of GO's limitations but it is currently unknown whether such drugs will be more effective in patients benefitting from GO and/or improve outcomes in patients not benefitting from GO, and what the supportive care requirements will be to enable their safe use.
Collapse
|
29
|
Impact of salvage regimens on response and overall survival in acute myeloid leukemia with induction failure. Leukemia 2017; 31:1306-1313. [PMID: 28138160 DOI: 10.1038/leu.2017.23] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/26/2016] [Accepted: 01/05/2017] [Indexed: 12/22/2022]
Abstract
We evaluated the impact of salvage regimens and allogeneic hematopoietic cell transplantation (allo-HCT) in acute myeloid leukemia (AML) with induction failure. Between 1993 and 2009, 3324 patients with newly diagnosed AML were enrolled in 5 prospective treatment trials of the German-Austrian AML Study Group. After first induction therapy with idarubicin, cytarabine and etoposide (ICE), 845 patients had refractory disease. In addition, 180 patients, although responding to first induction, relapsed after second induction therapy. Of the 1025 patients with induction failure, 875 (median age 55 years) received intensive salvage therapy: 7+3-based (n=59), high-dose cytarabine combined with mitoxantrone (HAM; n=150), with all-trans retinoic acid (A; A-HAM) (n=247), with gemtuzumab ozogamicin and A (GO; GO-A-HAM) (n=140), other intensive regimens (n=165), experimental treatment (n=27) and direct allo-HCT (n=87). In patients receiving intensive salvage chemotherapy (n=761), response (complete remission/complete remission with incomplete hematological recovery (CR/CRi)) was associated with GO-A-HAM treatment (odds ratio (OR), 1.93; P=0.002), high-risk cytogenetics (OR, 0.62; P=0.006) and age (OR for a 10-year difference, 0.75; P<0.0001). Better survival probabilities were seen in an extended Cox regression model with time-dependent covariables in patients responding to salvage therapy (P<0.0001) and having the possibility to perform an allo-HCT (P<0.0001). FLT3 internal tandem duplication, mutated IDH1 and adverse cytogenetics were unfavorable factors for survival.
Collapse
|
30
|
Frontline treatment of acute myeloid leukemia in adults. Crit Rev Oncol Hematol 2016; 110:20-34. [PMID: 28109402 DOI: 10.1016/j.critrevonc.2016.12.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 11/01/2016] [Accepted: 12/07/2016] [Indexed: 01/13/2023] Open
Abstract
Recent years have highlighted significant progress in understanding the underlying genetic and epigenetic signatures of acute myeloid leukemia(AML). Most importantly, novel chemotherapy and targeted strategies have led to improved outcomes in selected genetic subsets. AML is a remarkably heterogeneous disease, and individualized therapies for disease-specific characteristics (considering patients' age, cytogenetics, and mutations) could yield better outcomes. Compared with the historical 5-to 10-year survival rate of 10%, the survival of patients who undergo modern treatment approaches reaches up to 40-50%, and for specific subsets, the improvements are even more dramatic; for example, in acute promyelocytic leukemia, the use of all-trans retinoic acid and arsenic trioxide improved survival from 30 to 40% up to 80 to 90%. Similar progress has been documented in core-binding-factor-AML, with an increase in survival from 30% to 80% upon the use of high-dose cytarabine/fludarabine/granulocyte colony-stimulating factor combination regimens. AML treatment was also recently influenced by the discovery of the superiority of regimens with higher dose Ara-C and nucleoside analogues compared with the "7+3"regimen, with about a 20% improvement in overall survival. Despite these significant differences, most centers continue to use the "7+3" regimen, and greater awareness will improve the outcome. The discovery of targetable molecular abnormalities and recent studies of targeted therapies (gemtuzumab ozagomycin, FLT3 inhibitors, isocitrate dehydrogenase inhibitors, and epigenetic therapies), future use of checkpoint inhibitors and other immune therapies such as chimeric antigen receptor T-cells, and maintenance strategies based on the minimal residual disease evaluation represent novel, exciting clinical leads aimed to improve AML outcomes in the near future.
Collapse
|
31
|
Affiliation(s)
- M B Agarwal
- Department of Haematology, Bombay Hospital Institute of Medical Sciences, Mumbai, Maharashtra, India
| |
Collapse
|
32
|
Hütter-Krönke ML, Benner A, Döhner K, Krauter J, Weber D, Moessner M, Köhne CH, Horst HA, Schmidt-Wolf IGH, Rummel M, Götze K, Koller E, Petzer AL, Salwender H, Fiedler W, Kirchen H, Haase D, Kremers S, Theobald M, Matzdorff AC, Ganser A, Döhner H, Schlenk RF. Salvage therapy with high-dose cytarabine and mitoxantrone in combination with all-trans retinoic acid and gemtuzumab ozogamicin in acute myeloid leukemia refractory to first induction therapy. Haematologica 2016; 101:839-45. [PMID: 27036160 DOI: 10.3324/haematol.2015.141622] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 03/24/2016] [Indexed: 12/29/2022] Open
Abstract
Outcome of patients with primary refractory acute myeloid leukemia remains unsatisfactory. We conducted a prospective phase II clinical trial with gemtuzumab ozogamicin (3 mg/m(2) intravenously on day 1), all-trans retinoic acid (45 mg/m(2) orally on days 4-6 and 15 mg/m(2) orally on days 7-28), high-dose cytarabine (3 g/m(2)/12 h intravenously on days 1-3) and mitoxantrone (12 mg/m(2) intravenously on days 2-3) in 93 patients aged 18-60 years refractory to one cycle of induction therapy. Primary end point of the study was response to therapy; secondary end points included evaluation of toxicities, in particular, rate of sinusoidal obstruction syndrome after allogeneic hematopoietic cell transplantation. Complete remission or complete remission with incomplete blood count recovery was achieved in 47 (51%) and partial remission in 10 (11%) patients resulting in an overall response rate of 61.5%; 33 (35.5%) patients had refractory disease and 3 patients (3%) died. Allogeneic hematopoietic cell transplantation was performed in 71 (76%) patients; 6 of the 71 (8.5%) patients developed moderate or severe sinusoidal obstruction syndrome after transplantation. Four-year overall survival rate was 32% (95% confidence interval 24%-43%). Patients responding to salvage therapy and undergoing allogeneic hematopoietic cell transplantation (n=51) had a 4-year survival rate of 49% (95% confidence intervaI 37%-64%). Patients with fms-like tyrosine kinase internal tandem duplication positive acute myeloid leukemia had a poor outcome despite transplantation. In conclusion, the described regimen is an effective and tolerable salvage therapy for patients who are primary refractory to one cycle of conventional intensive induction therapy. (clinicaltrials.gov identifier: 00143975).
Collapse
Affiliation(s)
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital Ulm, Germany
| | - Jürgen Krauter
- Department of Oncology and Hematology, Klinikum Braunschweig, Germany Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Hospital Ulm, Germany
| | - Margit Moessner
- Department of Internal Medicine III, University Hospital Ulm, Germany
| | | | - Heinz A Horst
- Department of Internal Medicine II, University Hospital Schleswig-Holstein Campus Kiel, Germany
| | | | - Mathias Rummel
- Department of Hematology/Oncology, University-hospital Giessen, Germany
| | - Katharina Götze
- Department of Internal Medicine III, Technical University of Munich, Germany
| | - Elisabeth Koller
- Department of Hematology/Oncology, Hanuschkrankenhaus, Wien, Austria
| | - Andreas L Petzer
- Department of Medical Oncology and Hematology, Krankenhaus der Barmherzigen Schwestern, Linz, Austria
| | - Hans Salwender
- Department of Hematology/Oncology, Asklepios Klinik Altona, Hamburg, Germany
| | - Walter Fiedler
- Department of Internal Medicine II, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heinz Kirchen
- Department of Hematology/Oncology, Krankenhaus der Barmherzigen Brüder, Trier, Germany
| | - Detlef Haase
- Department of Hematology and Oncology, Georg-August-University Hospital of Göttingen, Germany
| | - Stephan Kremers
- Department of Hematology/Oncology, Caritas-Krankenhaus, Lebach, Germany
| | - Matthias Theobald
- Department of Medicine III, Johannes Gutenberg-University Mainz, Germany
| | - Axel C Matzdorff
- Department of Hematology/Oncology, Caritas-Krankenhaus, Saarbrücken, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital Ulm, Germany
| | - Richard F Schlenk
- Department of Internal Medicine III, University Hospital Ulm, Germany
| |
Collapse
|
33
|
Pollard JA, Loken M, Gerbing RB, Raimondi SC, Hirsch BA, Aplenc R, Bernstein ID, Gamis AS, Alonzo TA, Meshinchi S. CD33 Expression and Its Association With Gemtuzumab Ozogamicin Response: Results From the Randomized Phase III Children's Oncology Group Trial AAML0531. J Clin Oncol 2016; 34:747-55. [PMID: 26786921 DOI: 10.1200/jco.2015.62.6846] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE CD33 is variably expressed on acute myeloid leukemia (AML) blasts and is targeted by gemtuzumab ozogamicin (GO). GO has shown benefit in both adult and pediatric AML trials, yet limited data exist about whether GO response correlates with CD33 expression level. PATIENTS AND METHODS CD33 expression levels were prospectively quantified by multidimensional flow cytometry in 825 patients enrolled in Children's Oncology Group AAML0531 and correlated with response to GO. RESULTS Patients with low CD33 expression (lowest quartile of expression [Q1]) had no benefit with the addition of GO to conventional chemotherapy (relapse risk [RR]: GO 36% v No-GO 34%, P = .731; event-free survival [EFS]: GO 53% v No-GO 58%, P = .456). However, patients with higher CD33 expression (Q2 to Q4) had significantly reduced RR (GO 32% v No-GO 49%, P < .001) and improved EFS (GO 53% v No-GO 41%, P = .005). This differential effect was observed in all risk groups. Specifically, low-risk (LR), intermediate-risk (IR), and high-risk (HR) patients with low CD33 expression had similar outcomes regardless of GO exposure, whereas the addition of GO to conventional chemotherapy resulted in a significant decrease in RR and disease-free survival (DFS) for patients with higher CD33 expression (LR RR, GO 13% v No-GO 35%, P = .001; LR DFS, GO 79% v No-GO 59%, P = .007; IR RR, GO 44% v No-GO 57%, P = .044; IR DFS, GO 51% v No-GO 40%, P = .078; HR RR, GO 40% v No-GO 73%, P = .016; HR DFS, GO 47% v No-GO 28%, P = .135). CONCLUSION We demonstrate that GO lacks clinical benefit in patients with low CD33 expression but significantly reduces RR and improves EFS in patients with high CD33 expression, which suggests a role for CD33-targeted therapeutics in subsets of pediatric AML.
Collapse
Affiliation(s)
- Jessica A Pollard
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO.
| | - Michael Loken
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Robert B Gerbing
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Susana C Raimondi
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Betsy A Hirsch
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Richard Aplenc
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Irwin D Bernstein
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Alan S Gamis
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Todd A Alonzo
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Soheil Meshinchi
- Jessica A. Pollard, Maine Medical Center, Portland, ME; and Tufts University, Boston, MA; Michael Loken, Hematologics; Irwin D. Bernstein and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Irwin D. Bernstein and Soheil Meshinchi, University of Washington, Seattle, WA; Robert B. Gerbing and Todd A. Alonzo, Children's Oncology Group, Arcadia; Todd A. Alonzo, Keck School of Medicine of University of Southern California, Los Angeles, CA; Susana C. Raimondi, St Jude Children's Research Hospital, Memphis, TN; Betsy Hirsch, University of Minnesota Cancer Center, Minneapolis, MN; Richard Aplenc, Children's Hospital of Philadelphia, Philadelphia, PA; and Alan S. Gamis, Children's Mercy Hospitals and Clinics, Kansas City, MO
| |
Collapse
|
34
|
Heiblig M, Elhamri M, Michallet M, Thomas X. Adoptive immunotherapy for acute leukemia: New insights in chimeric antigen receptors. World J Stem Cells 2015; 7:1022-1038. [PMID: 26328018 PMCID: PMC4550626 DOI: 10.4252/wjsc.v7.i7.1022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/28/2014] [Accepted: 06/19/2015] [Indexed: 02/06/2023] Open
Abstract
Relapses remain a major concern in acute leukemia. It is well known that leukemia stem cells (LSCs) hide in hematopoietic niches and escape to the immune system surveillance through the outgrowth of poorly immunogenic tumor-cell variants and the suppression of the active immune response. Despite the introduction of new reagents and new therapeutic approaches, no treatment strategies have been able to definitively eradicate LSCs. However, recent adoptive immunotherapy in cancer is expected to revolutionize our way to fight against this disease, by redirecting the immune system in order to eliminate relapse issues. Initially described at the onset of the 90’s, chimeric antigen receptors (CARs) are recombinant receptors transferred in various T cell subsets, providing specific antigens binding in a non-major histocompatibility complex restricted manner, and effective on a large variety of human leukocyte antigen-divers cell populations. Once transferred, engineered T cells act like an expanding “living drug” specifically targeting the tumor-associated antigen, and ensure long-term anti-tumor memory. Over the last decades, substantial improvements have been made in CARs design. CAR T cells have finally reached the clinical practice and first clinical trials have shown promising results. In acute lymphoblastic leukemia, high rate of complete and prolonged clinical responses have been observed after anti-CD19 CAR T cell therapy, with specific but manageable adverse events. In this review, our goal was to describe CAR structures and functions, and to summarize recent data regarding pre-clinical studies and clinical trials in acute leukemia.
Collapse
|
35
|
Abstract
Induction followed by post-remission therapy including intensive chemotherapy with high-dose cytarabine, autologous and allogeneic hematopoietic stem cell transplantation is recognized as the main road towards cure in acute myeloid leukemia. In recent years, also a renaissance of maintenance therapy after completion of intensive consolidation has been observed with the introduction of kinase inhibitors and demethylating agents in clinical trials. Greater insight into the genetic background of the disease fostered the extension of disease classification and pretreatment risk-categorization by gene mutations. In addition, the pre-treatment risk-defining parameters have been supplemented by markers evaluated at distinct time points during treatment and follow up. In this context, minimal residual disease assessment is increasingly used to dynamically fine tune treatment recommendations. Currently, the gold standard to counterbalance a higher risk of relapse by treatment strategies based on hematopoietic stem cell transplantation with grafts from matched related or unrelated donors is still valuable, whereas autologous hematopoietic stem cell transplantation showed promising results especially in patients categorized as low-risk. Nonetheless, more targeted approaches including kinase inhibitors and demethylating agents in combination with or sequentially before or after intensive chemotherapy are currently in clinical evaluation and may lead to more genotype- instead of purely risk-adapted treatment strategies.
Collapse
Affiliation(s)
- Richard F Schlenk
- Department of Internal Medicine III, University Hospital Ulm, Germany
| |
Collapse
|
36
|
Visintin A, Knowlton K, Tyminski E, Lin CI, Zheng X, Marquette K, Jain S, Tchistiakova L, Li D, O'Donnell CJ, Maderna A, Cao X, Dunn R, Snyder WB, Abraham AK, Leal M, Shetty S, Barry A, Zawel L, Coyle AJ, Dvorak HF, Jaminet SC. Novel Anti-TM4SF1 Antibody-Drug Conjugates with Activity against Tumor Cells and Tumor Vasculature. Mol Cancer Ther 2015; 14:1868-76. [PMID: 26089370 DOI: 10.1158/1535-7163.mct-15-0188] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/07/2015] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) represent a promising therapeutic modality for managing cancer. Here, we report a novel humanized ADC that targets the tetraspanin-like protein TM4SF1. TM4SF1 is highly expressed on the plasma membranes of many human cancer cells and also on the endothelial cells lining tumor blood vessels. TM4SF1 is internalized upon interaction with antibodies. We hypothesized that an ADC against TM4SF1 would inhibit cancer growth directly by killing cancer cells and indirectly by attacking the tumor vasculature. We generated a humanized anti-human TM4SF1 monoclonal antibody, v1.10, and armed it with an auristatin cytotoxic agent LP2 (chemical name mc-3377). v1.10-LP2 selectively killed cultured human tumor cell lines and human endothelial cells that express TM4SF1. Acting as a single agent, v1.10-LP2 induced complete regression of several TM4SF1-expressing tumor xenografts in nude mice, including non-small cell lung cancer and pancreas, prostate, and colon cancers. As v1.10 did not react with mouse TM4SF1, it could not target the mouse tumor vasculature. Therefore, we generated a surrogate anti-mouse TM4SF1 antibody, 2A7A, and conjugated it to LP2. At 3 mpk, 2A7A-LP2 regressed several tumor xenografts without noticeable toxicity. Combination therapy with v1.10-LP2 and 2A7A-LP2 together was more effective than either ADC alone. These data provide proof-of-concept that TM4SF1-targeting ADCs have potential as anticancer agents with dual action against tumor cells and the tumor vasculature. Such agents could offer exceptional therapeutic value and warrant further investigation. Mol Cancer Ther; 14(8); 1868-76. ©2015 AACR.
Collapse
Affiliation(s)
- Alberto Visintin
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Kelly Knowlton
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Edyta Tyminski
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Chi-Iou Lin
- The Center for Vascular Biology Research and the Departments of Pathology, Beth Israel Deaconess Medical Center (BIDMC) and Harvard Medical School, Boston, Massachusetts
| | - Xiang Zheng
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Kimberly Marquette
- Pfizer Inc., Global Biotherapeutic Technologies (GBT), Cambridge, Massachusetts
| | - Sadhana Jain
- Pfizer Inc., Global Biotherapeutic Technologies (GBT), Cambridge, Massachusetts
| | | | - Dan Li
- The Center for Vascular Biology Research and the Departments of Pathology, Beth Israel Deaconess Medical Center (BIDMC) and Harvard Medical School, Boston, Massachusetts
| | | | - Andreas Maderna
- Pfizer Inc., Worldwide Medicinal Chemistry, Groton, Connecticut
| | - Xianjun Cao
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), San Diego, California
| | - Robert Dunn
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), San Diego, California
| | - William B Snyder
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), San Diego, California
| | - Anson K Abraham
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Mauricio Leal
- Pfizer Inc., Pharmacokinetics, Dynamics and Metabolism (PDM), Pearl River, New York
| | - Shoba Shetty
- Pfizer Inc., Drug Safety R&D, Investigative Toxicology, Groton, Connecticut
| | - Anthony Barry
- Pfizer Inc., Biotherapeutics Pharmaceutical Sciences, Andover, Massachusetts
| | - Leigh Zawel
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Anthony J Coyle
- Pfizer Inc., Centers for Therapeutic Innovation (CTI), Boston, Massachusetts
| | - Harold F Dvorak
- The Center for Vascular Biology Research and the Departments of Pathology, Beth Israel Deaconess Medical Center (BIDMC) and Harvard Medical School, Boston, Massachusetts.
| | - Shou-Ching Jaminet
- The Center for Vascular Biology Research and the Departments of Pathology, Beth Israel Deaconess Medical Center (BIDMC) and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
37
|
Ramos NR, Mo CC, Karp JE, Hourigan CS. Current Approaches in the Treatment of Relapsed and Refractory Acute Myeloid Leukemia. J Clin Med 2015; 4:665-95. [PMID: 25932335 PMCID: PMC4412468 DOI: 10.3390/jcm4040665] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/20/2015] [Indexed: 01/07/2023] Open
Abstract
The limited sensitivity of the historical treatment response criteria for acute myeloid leukemia (AML) has resulted in a different paradigm for treatment compared with most other cancers presenting with widely disseminated disease. Initial cytotoxic induction chemotherapy is often able to reduce tumor burden to a level sufficient to meet the current criteria for "complete" remission. Nevertheless, most AML patients ultimately die from their disease, most commonly as clinically evident relapsed AML. Despite a variety of available salvage therapy options, prognosis in patients with relapsed or refractory AML is generally poor. In this review, we outline the commonly utilized salvage cytotoxic therapy interventions and then highlight novel investigational efforts currently in clinical trials using both pathway-targeted agents and immunotherapy based approaches. We conclude that there is no current standard of care for adult relapsed or refractory AML other than offering referral to an appropriate clinical trial.
Collapse
Affiliation(s)
- Nestor R. Ramos
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1583, USA; E-Mail:
- Department of Hematology-Oncology, John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA; E-Mail:
| | - Clifton C. Mo
- Department of Hematology-Oncology, John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA; E-Mail:
| | - Judith E. Karp
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; E-Mail:
| | - Christopher S. Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1583, USA; E-Mail:
| |
Collapse
|