Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Bull Cancer ; 2024 Jun 25.
Article in French | MEDLINE | ID: mdl-38926053

ABSTRACT

Treatment of pediatric high-risk acute myeloid leukemia (AML), defined either on molecular or cytogenetic features, relies on bone marrow transplant after cytologic remission. However, relapse remains the first post-transplant cause of mortality. In this 13th session of practice harmonization of the francophone society of bone marrow transplantation and cellular therapy (SFGM-TC), our group worked on recommendations regarding the management of post-transplant relapse in AML pediatric patients based on international literature, national survey and expert opinion. Overall, immunomodulation strategy relying on both measurable residual disease (MRD) and chimerism evaluation should be used for high-risk AML. In very high-risk (VHR) AML with a 5-year overall survival ≤30 %, a post-transplant maintenance should be proposed using either hypomethylating agents, combined with DLI whenever possible, or FLT3 tyrosine kinase inhibitors if this target is present on leukemia cells. In the pre-emptive or early relapse settings (< 6 months post-transplant), treatments combining DLI, Azacytidine and Venetoclax should be considered. Access to phase I/II trails for targeted therapies (menin, IDH or JAK inhibitors) should be discussed in each patient according to the underlying molecular abnormalities of the disease.

3.
Bull Cancer ; 111(5): 513-524, 2024 May.
Article in French | MEDLINE | ID: mdl-38503585

ABSTRACT

Neonatal acute myeloid leukemias (AML) occurred within the first 28 days of life and constitute only a small proportion of all AL. They are distinguished from leukemias of older children by their clinical presentation, which frequently includes cutaneous localizations ("blueberry muffin rash syndrome") and a leukocytosis above 50 ×109/L. This proliferation may be transient, causing a transient leukemoid reaction in a background of constitutional trisomy 21 ("Transient Abnormal Myelopoieseis" or TAM) or Infantile Myeloproliferative Disease in the absence of constitutional trisomy 21 ("Infantile Myeloproliferative Disease" or IMD). In cases of true neonatal AML, the prognosis of patients is poor. Overall survival is around 35 % in the largest historical series. This poor prognosis is mainly due to the period of onset of this pathology making the use of chemotherapy more limited and involving many considerations, both ethical and therapeutic. The objective of this work is to review this rare pathology by addressing the clinical, biological, therapeutic and ethical particularities of patients with true neonatal AML or transient leukemoid reactions occurring in a constitutional trisomy 21 (true TAM) or somatic background (IMD).


Subject(s)
Down Syndrome , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/diagnosis , Infant, Newborn , Down Syndrome/therapy , Prognosis , Leukemoid Reaction/therapy , Leukemoid Reaction/diagnosis , Myeloproliferative Disorders/therapy , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics
4.
Bull Cancer ; 110(4): 424-432, 2023 Apr.
Article in French | MEDLINE | ID: mdl-36870810

ABSTRACT

Elderly patients with acute myeloid leukemia, ineligible for intensive chemotherapy, have long had a very poor prognosis and have always represented one of the main patient populations included in early phase clinical research trials. In recent years, many molecules have shown very interesting efficacy, often targeted therapies whose indication is based on a specific mutation profile (gilteritinib, ivosidenib), or mutation-independent (venetoclax), but also drugs whose indication is based on a specific biomarker (tamibarotene) or on new generation immunotherapies targeting macrophages (magrolimab) or other immune effectors while targeting leukemic cells resulting in forced immunological synapse (flotetuzumab) or activation of lymphocyte effectors associated with inhibition of the AML cells' stem signature in their microenvironment (cusatuzumab sabatolimab). All of these new strategies are discussed in this review, as well as the challenges of this frail population, which has benefited in recent months from all the major advances in the field, questioning in a second phase the modification of practices in younger patients.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Aged , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Prognosis , Mutation , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Tumor Microenvironment
5.
Bull Cancer ; 110(2S): S108-S115, 2023 Feb.
Article in French | MEDLINE | ID: mdl-35791974

ABSTRACT

The role of allogeneic hematopoietic cell transplantation (allo-HCT) after CAR T- treatment cells in hematologic malignancies is currently controversial. Prolonged remissions after several years of follow-up suggest that there is a curative effect of CAR T-cells therapy, whereas allo-HCT was previously considered the only curative treatment in relapse situation. The aim of this harmonization workshop is to detail the existing data in the literature on the feasibility of allo-HCT after CAR T-cells and to propose to consider allograft in selected patients with B-acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL). In B-ALL, various intrinsic factors (inherent to the patient, to the disease, to the type of CAR T-cells) and especially various post CAR T-cells criteria (early expansion kinetics, residual disease at D28, early loss of B-cell aplasia) should lead to consider performing allo-HCT before the occurrence of a relapse. In DLBCL, although there are risk factors for relapse at diagnosis and prior to CAR T-cells therapy, response assessed by PET-CT at three months is critical and allo-HCT cannot currently be recommended in cases of complete or partial remission. In any case, if the age is appropriate for allogeneic transplantation, HLA typing should be performed before CAR T-cells treatment in order not to delay the allo-HCT project if needed.


Subject(s)
Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Positron Emission Tomography Computed Tomography , Hematopoietic Stem Cell Transplantation/adverse effects , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/transplantation , Recurrence
6.
Bull Cancer ; 110(2S): S30-S38, 2023 Feb.
Article in French | MEDLINE | ID: mdl-35562231

ABSTRACT

Management of acute lymphoblastic leukemia (ALL) patients in countries with limited resources depends on the means of prognostic stratification, available treatment and logistics. During the 12th annual harmonization workshops of the francophone Society of bone marrow transplantation and cellular therapy (SFGM-TC), a designated working group reviewed the literature in order to elaborate unified guidelines for allogeneic hematopoietic cell transplantation (Allo-HCT) in this disease. Conventional poor prognostic factors can be used to determine the indication of allo-HCT in first remission. Patients lacking a HLA-matched related donor can be allografted with a haploidentical donor allo-HCT if available. Chemotherapy based conditioning regimen can be used if TBI is not available, because the probability to find a radiotherapy department with the capacity for total body irradiation is low. For patients with Philadelphia chromosome positive (Phi+) ALL, post-transplantation tyrosine kinase inhibitors as a systematic maintenance strategy is recommended. Autologous HCT is optional for Phi+ ALL patients with negative minimal residual disease, who not eligible for allo-HCT. Patients with refractory/relapsed disease have a poor prognosis which highlights the importance of acquiring in the future new therapies such as: blinatumumab, inotuzumab, and CAR-T cells.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Developing Countries , Follow-Up Studies , Bone Marrow Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
8.
Bull Cancer ; 109(11): 1125-1131, 2022 Nov.
Article in French | MEDLINE | ID: mdl-35987855

ABSTRACT

Pegaspargase (Oncaspar®), a pegylated form of native Escherichia Coli-derived L-asparaginase is an essential component chemotherapy used in the treatment of acute lymphoblastic leukemia (ALL) in pediatric and adult patients. Its particular toxicity profile requires a specific management to improve safety and tolerability and optimize treatment outcome and therefore survival. Within the framework of workshops of practice harmonization of the French Society of Children and Adolescent Cancers, diagnostic and management of the most commonly occuring toxicities (excluding coagulation abnormalities) during Pegaspargase treatment were reviewed according to the analysis of published studies.


Subject(s)
Antineoplastic Agents , Blood Coagulation Disorders , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Humans , Adolescent , Child , Asparaginase/adverse effects , Polyethylene Glycols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Treatment Outcome , Antineoplastic Agents/adverse effects
9.
Rev Med Interne ; 43(11): 677-682, 2022 Nov.
Article in French | MEDLINE | ID: mdl-36041908

ABSTRACT

INTRODUCTION: Heterozygous germline mutations of GATA2 gene (guanine-adenine-thymine-adenine binding protein 2) are hereditary mutations that can be pathogenic, sometimes occurring sporadically, responsible for a florid clinical-biological picture, sometimes serious and quickly leading to the death. CASE REPORTS: We reported two women and one man with germline mutations in the GATA2 gene. The first patient, aged 19, initially presented with monocytopenia and chronic lymphedema of the four limbs, suggestive of Emberger syndrome. The second patient, 28-years-old, presented with a disseminated atypical mycobacterium (Mycobacterium kansasii) infection, raising suspicion of an immune deficiency such as MonoMAC syndrome (deficiency syndrome of dendritic cells, monocytes, B lymphocytes and NK cells). The last patient, 30-years-old, presented with pancytopenia, leading to the diagnosis of a family form of myelodysplastic syndromes and acute myeloid leukemia characterized by a mutation of the GATA2 gene. CONCLUSIONS: Each case illustrates a typical clinical presentation of GATA2 deficiency, although the evolution of these syndromes ultimately reveals a complex, heterogeneous and intricate picture of hematological, dermatological, infectious, pulmonary, ENT or oncological symptoms. Mutations in the GATA2 gene remain a diagnostic and therapeutic challenge for the internist, and require multidisciplinary management given the florid picture that can be of interest to all specialties. The clinical spectrum of these GATA2 mutations as well as the latest management recommendations from the recent litterature and the "GATA2 club" are described in this article.


Subject(s)
GATA2 Transcription Factor , Immunologic Deficiency Syndromes , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Adult , Female , Humans , Male , Adenine , GATA2 Transcription Factor/genetics , Immunologic Deficiency Syndromes/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics
10.
Bull Cancer ; 109(11): 1132-1143, 2022 Nov.
Article in French | MEDLINE | ID: mdl-35863954

ABSTRACT

Maintenance therapy is the last phase of treatment for acute lymphoblastic leukemia in children and adolescents. Although maintenance therapy is associated with toxicities and specific management issues, it is an essential phase of treatment that reduces the risk of relapse. The objective of this work is to propose a guide for the initiation, administration, and monitoring of maintenance therapy, and for the management of food, schooling, leisure, community life, risk of infection and links with family medicine.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Adolescent , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols , Recurrence
12.
Bull Cancer ; 108(10S): S40-S54, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34920807

ABSTRACT

The marketing authorization of tisagenlecleucel, a 2nd generation of CD19-directed CAR T-cells, containing the 4-1 BB co-stimulatory domain, in 2017 in USA and in 2018 in EU, has revolutionized the therapeutic strategy in advanced B-cell acute lymphoblastic leukemia (B-ALL) in children, adolescents and young adults (AYAs) with relapsed or refractory disease. This innovative treatment, based on a "living drug", has shown very impressive short-term responses. However, safety profile and complex logistics require high expertise centers and tight collaborations between addressing and treating centers. Current research is exploring the possibility to move to first line ALL with high-risk features and/or first high-risk relapse. More efficient CAR T-cells products, are still lacking to counteract the escape mechanisms already described. Moreover, to define the bridge-to-CAR time for each patient remains a challenge to obtain optimal disease burden allowing expansion and persistence of CAR T-cells. Also difficult is to identify patients who will benefit from further therapy after infusion, such as allogeneic HSCT or may be immuno-modulatory treatment. Finally, CAR T-cells directed against T-ALL are only in their beginning but require more complex engineering process to avoid T- cell immune-deficiency or fratricide.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy, Adoptive/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/immunology , Adolescent , Antigens, CD19/immunology , CD28 Antigens/immunology , Cell Engineering , Child , Clinical Trials as Topic , Cost of Illness , Humans , Immunomodulation , Immunotherapy, Adoptive/adverse effects , Leukemia, B-Cell/immunology , Leukemia, B-Cell/pathology , Leukemia, B-Cell/therapy , Lymphocyte Depletion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Tumor Escape/immunology , Young Adult
13.
Can Oncol Nurs J ; 31(3): 334-338, 2021.
Article in English | MEDLINE | ID: mdl-34395838

ABSTRACT

OBJECTIF: La présente étude a pour but d'examiner la réponse à un schéma chimiothérapeutique administré à des enfants atteints de LAL à cellules pré-B à risque élevé selon le protocole COG. MÉTHODE: L'étude transversale porte sur 55 enfants traités selon le protocole du groupe d'oncologie pédiatrique (mieux connu sous le nom de Children's Oncology Group ou COG), de septembre 2010 à février 2015, et évalue les résultats du schéma chimiothérapeutique. RÉSULTATS: Durant la première semaine suivant le traitement, le taux de rétablissement complet a été de 76,4 %. Les taux de survie après trois ans et cinq ans étaient respectivement de 85,5 % et de 81 %. Le taux de rechute après le premier épisode de rémission a été de 20 % et le taux de mortalité consécutif à cette rechute a été de 50 %. Trente pour cent de l'ensemble des décès ont eu lieu durant la période d'induction. Dans tous les cas, une septicémie en est la cause. CONCLUSION: Les résultats indiquent que le taux de survie a augmenté. Il est donc possible d'améliorer le taux de survie en optant pour le protocole COG et en contrôlant les infections chez les patients, et ce, sans égard au groupe de risque.

14.
Soins Pediatr Pueric ; 42(320): 35-40, 2021.
Article in French | MEDLINE | ID: mdl-34099236

ABSTRACT

Acute leukemias are the most common pediatric cancer. With a cure rate of about 80%, their treatment is based on a combination of cytotoxic chemotherapies whose intensity is adapted to prognostic factors. Sometimes, allogeneic hematopoietic stem cell transplantation is indicated. New therapeutic options are being developed, such as CAR T-cells.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans
15.
Bull Cancer ; 108(3): 295-303, 2021 Mar.
Article in French | MEDLINE | ID: mdl-33610284

ABSTRACT

Chimeric antigen receptor (CAR) T-cells are a new class of cancer treatments manufactured through autologous or allogeneic T cells genetic engineering to induce CAR expression directed against a membrane antigen present at the surface of malignant cells. In Europe, tisagenlecleucel (Kymriah™) has a marketing authorization for the treatment of relapsed/refractory B-cell acute lymphoblastic leukemia in children and young adults and for the relapsed/refractory diffuse large B-cell lymphoma (DLBCL). The marketing authorization for axicabtagene ciloleucel (Yescarta™) is the treatment of relapsed/refractory DLBCL and mediastinal B-cell lymphoma. Both products are "living drugs" and genetically modified autologous T cells directed against CD19 which is an antigen expressed throughout B lymphoid differentiation and on many B malignancies. This collaborative work - part of a series of expert works on the topic - aims to provide practical advice to assist collection facilities that procure the starting material i.e. blood mononuclear cells for autologous CAR T-cell manufacturing.


Subject(s)
Antigens, CD19/therapeutic use , Commerce , Consensus , Immunotherapy, Adoptive , Leukapheresis/methods , Receptors, Antigen, T-Cell/therapeutic use , Adolescent , Biological Products , Child , Genetic Engineering/methods , Humans , Leukemia, B-Cell/therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Mediastinal Neoplasms/therapy , T-Lymphocytes , Tissue and Organ Harvesting/methods , Young Adult
16.
Bull Cancer ; 108(7-8): 725-729, 2021.
Article in French | MEDLINE | ID: mdl-33423776

ABSTRACT

The chimeric antigen receptor T-cells are a new class of anticancer treatment consisting in genetically modifying autologous or allogenic T-cells to make express a CAR directed against a membrane tumor antigen. In Europe, tisagenlecleucel (KymriahTM) has a marketing authorization for the treatment of relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia in children and young adults and of R/R diffuse large B-cell lymphoma (DLBCL). The marketing authorization for axicabtagene ciloleucel (YescartaTM) is the treatment of DLBCL and primary R/R mediastinal B-cell lymphoma. The two products are autologous T-cells directed against CD19. This collaborative work, part of a series of expert opinion-based work, aims to give practical advice to help centers in selection of patients for commercially available CAR T-cell treatment.


Subject(s)
Antigens, CD19/therapeutic use , Immunotherapy, Adoptive , Patient Selection , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen , Biological Products , Child , Europe , Humans , Lymphoma, Large B-Cell, Diffuse/therapy , Mediastinal Neoplasms/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Substance-Related Disorders , Young Adult
17.
Bull Cancer ; 108(2): 187-197, 2021 Feb.
Article in French | MEDLINE | ID: mdl-32981690

ABSTRACT

For two decades, the prognostic of adult patients with ALL was improved based on pediatric-inspired protocols. These approaches based on less myelosuppressive drugs have led to improved response rates, decreased relapse rates, with a benefit in survival observed in patients aged up to 50-60-years-old. Therapeutic intensification came with a decrease in the use of allogeneic hematopoietic stem cell transplantation, with current indications mainly based on the level of measurable residual disease. Pediatric approaches are however limited in older patients or in patients with comorbidities, who are at greater risk to develop adverse effects especially to asparaginase. Future progresses will arise from personalized medicine including targeted therapy in some ALL oncogenic subgroups and immunotherapy. Monoclonal antibodies, bispecific antibodies, antibody drug conjugates and CAR-T cells have shown encouraging results in relapsed/refractory diseases. These strategies are now evaluated frontline in children and adults to further increase the quality of response, to limit the toxicity of treatments including allogeneic transplant. The objective of this review is to discuss the benefit and the limits of pediatric therapeutic strategies in adults and the perspectives offered by new approaches including immunotherapies.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Age Factors , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Asparaginase/adverse effects , Asparaginase/therapeutic use , Child , Humans , Immunoconjugates/therapeutic use , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Inotuzumab Ozogamicin/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Middle Aged , Molecular Targeted Therapy , Neoplasm, Residual , Pediatrics/methods , Precision Medicine/trends , Rituximab/therapeutic use , Young Adult
18.
Bull Cancer ; 107(12S): S193-S201, 2020 Dec.
Article in French | MEDLINE | ID: mdl-33187683

ABSTRACT

CAR T-cells are anti-cancer immunocellular therapy drugs that involve reprogramming the patient's T-cells using a transgene encoding a chimeric antigen receptor (CAR). Although CAR T-cells are cellular therapies, the organization for manufacturing and delivering these medicinal products is in many ways different from the one for hematopoietic cell grafts or donor lymphocyte infusions. The implementation of this innovative therapy is recent and requires close coordination between clinical teams, the therapeutic apheresis unit, the cell therapy unit, the pharmaceutical laboratory, and pharmacy. Apart from the regulatory texts, which are regularly modified, and the specific requirements of each pharmaceutical laboratory, there is currently no guide to help the centers initiating their activity and there is no specific indicator to assess the quality of the CAR T-cell activity in each center. The purpose of the current harmonization workshop is to clarify the regulatory prerequisites warranted for a center to have a CAR T-cell activity and to propose recommendations for implementing quality tools, in particular indicators, and allowing their sharing.


Subject(s)
Immunotherapy, Adoptive/standards , Quality Assurance, Health Care , Receptors, Chimeric Antigen , Accreditation , Congresses as Topic/organization & administration , France , Health Personnel/education , Humans , Immunotherapy, Adoptive/legislation & jurisprudence , Societies, Medical
20.
Bull Cancer ; 107(12S): S130-S139, 2020 Dec.
Article in French | MEDLINE | ID: mdl-32560899

ABSTRACT

Microtransplantation (MT) is based on injection of HLA-mismatched G-CSF mobilized hematopoietic stem cells, in combination with chemotherapy but without use of conditioning regimen nor immunosuppressive drugs. As a result, a transient microchimerism is induced without engraftment. Its efficacy relies both on host immune system stimulation (recipient versus tumor) and on a graft versus tumor effect. Data are scarce and concern mostly Asian patients with acute myeloid leukemia (AML) and high risk myelodysplastic syndrome (HR-MDS). In comparison to conventional treatment without MT, higher complete remission rates and longer disease free survival and overall survival have been reported. Safety seems acceptable. The most frequent adverse event is non-severe cytokine release syndrome. Risk of GVHD remains very low. Here, we summarize the published data and detail the practical aspects of the procedure. Current data are not strong enough to provide recommendations on indications. Nevertheless, it seems reasonable to propose MT to patients with AML or HR-MDS, regardless of age, presenting an indication for allogeneic stem cell transplantation but ineligible for it. MT is still under investigation and rather be proposed within clinical trials.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Age Factors , Antineoplastic Agents/therapeutic use , Chimerism , Combined Modality Therapy/methods , Cytokine Release Syndrome/etiology , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation/standards , Host vs Graft Reaction/immunology , Humans , Leukemia, Myeloid, Acute/ethnology , Progression-Free Survival , Societies, Medical
SELECTION OF CITATIONS
SEARCH DETAIL