Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
Leuk Lymphoma ; 63(7): 1566-1579, 2022 07.
Article En | MEDLINE | ID: mdl-35259043

Chimeric antigen receptors (CAR)-modified T cells are an emerging therapeutic tool for chronic lymphocytic leukemia (CLL). However, in patients with CLL, well-known T-cell defects and the inhibitory properties of the tumor microenvironment (TME) hinder the efficacy of CAR T cells. We explored a novel approach combining CARs with lenalidomide, an immunomodulatory drug that tempers the immunosuppressive activity of the CLL TME. T cells from patients with CLL were engineered to express a CAR specific for CD23, a promising target antigen. Lenalidomide maintained the in vitro effector functions of CD23.CAR+ T cells effector functions in terms of antigen-specific cytotoxicity, cytokine release and proliferation. Overall, lenalidomide preserved functional CAR T-CLL cell immune synapses. In a Rag2-/-γc-/--based xenograft model of CLL, we demonstrated that, when combined with low-dose lenalidomide, CD23.CAR+ T cells efficiently migrated to leukemic sites and delayed disease progression when compared to CD23.CAR+ T cells given with rhIL-2. These observations underline the therapeutic potential of this novel CAR-based combination strategy in CLL.


Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Interleukin Receptor Common gamma Subunit , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , T-Lymphocytes , Tumor Microenvironment
2.
J Clin Invest ; 130(11): 6021-6033, 2020 11 02.
Article En | MEDLINE | ID: mdl-32780725

BACKGROUNDChimeric antigen receptor (CAR) T cell immunotherapy has resulted in complete remission (CR) and durable response in highly refractory patients. However, logistical complexity and high costs of manufacturing autologous viral products limit CAR T cell availability.METHODSWe report the early results of a phase I/II trial in B cell acute lymphoblastic leukemia (B-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT) using donor-derived CD19 CAR T cells generated with the Sleeping Beauty (SB) transposon and differentiated into cytokine-induced killer (CIK) cells.RESULTSThe cellular product was produced successfully for all patients from the donor peripheral blood (PB) and consisted mostly of CD3+ lymphocytes with 43% CAR expression. Four pediatric and 9 adult patients were infused with a single dose of CAR T cells. Toxicities reported were 2 grade I and 1 grade II cytokine-release syndrome (CRS) cases at the highest dose in the absence of graft-versus-host disease (GVHD), neurotoxicity, or dose-limiting toxicities. Six out of 7 patients receiving the highest doses achieved CR and CR with incomplete blood count recovery (CRi) at day 28. Five out of 6 patients in CR were also minimal residual disease negative (MRD-). Robust expansion was achieved in the majority of the patients. CAR T cells were measurable by transgene copy PCR up to 10 months. Integration site analysis showed a positive safety profile and highly polyclonal repertoire in vitro and at early time points after infusion.CONCLUSIONSB-engineered CAR T cells expand and persist in pediatric and adult B-ALL patients relapsed after HSCT. Antileukemic activity was achieved without severe toxicities.TRIAL REGISTRATIONClinicalTrials.gov NCT03389035.FUNDINGThis study was supported by grants from the Fondazione AIRC per la Ricerca sul Cancro (AIRC); Cancer Research UK (CRUK); the Fundación Científica de la Asociación Española Contra el Cáncer (FC AECC); Ministero Della Salute; Fondazione Regionale per la Ricerca Biomedica (FRRB).


Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Allografts , Child , Child, Preschool , Female , Humans , Infant , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology
3.
Mol Ther ; 28(9): 1974-1986, 2020 09 02.
Article En | MEDLINE | ID: mdl-32526203

The successful implementation of chimeric antigen receptor (CAR)-T cell therapy in the clinical context of B cell malignancies has paved the way for further development in the more critical setting of acute myeloid leukemia (AML). Among the potentially targetable AML antigens, CD33 is insofar one of the main validated molecules. Here, we describe the feasibility of engineering cytokine-induced killer (CIK) cells with a CD33.CAR by using the latest optimized version of the non-viral Sleeping Beauty (SB) transposon system "SB100X-pT4." This offers the advantage of improving CAR expression on CIK cells, while reducing the amount of DNA transposase as compared to the previously employed "SB11-pT" version. SB-modified CD33.CAR-CIK cells exhibited significant antileukemic activity in vitro and in vivo in patient-derived AML xenograft models, reducing AML development when administered as an "early treatment" and delaying AML progression in mice with established disease. Notably, by exploiting an already optimized xenograft chemotherapy model that mimics human induction therapy in mice, we demonstrated for the first time that CD33.CAR-CIK cells are also effective toward chemotherapy resistant/residual AML cells, further supporting its future clinical development and implementation within the current standard regimens.


Cell Engineering/methods , Cell Transplantation/methods , Cytokine-Induced Killer Cells/immunology , Drug Resistance, Neoplasm , Genetic Therapy/methods , Heterografts , Immunotherapy, Adoptive/methods , Leukemia, Experimental/therapy , Leukemia, Myeloid, Acute/therapy , Receptors, Chimeric Antigen/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Animals , Feasibility Studies , Gene Transfer Techniques , Humans , Mice , Mice, Inbred NOD , Mice, SCID , THP-1 Cells , Transposases/genetics , Transposases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Hum Gene Ther ; 29(5): 602-613, 2018 05.
Article En | MEDLINE | ID: mdl-29641322

Infusion of patient-derived CD19-specific chimeric antigen receptor (CAR) T cells engineered by viral vectors achieved complete remission and durable response in relapsed and refractory (r/r) B-lineage neoplasms. Here, we expand on those findings by providing a preclinical evaluation of allogeneic non-viral cytokine-induced killer (CIK) cells transfected with the Sleeping Beauty (SB) transposon CD19CAR (CARCIK-CD19). Specifically, thanks to a large-scale 18-day manufacturing process, it was possible to achieve stable CD19CAR expression (62.425 ± 6.399%) and efficient T-cell expansion (23.36 ± 3.00-fold). Frozen/thawed CARCIK-CD19 remained fully functional both in vitro and in an established patient-derived xenograft (PDX) of MLL-ENL rearranged acute lymphoblastic leukemia (ALL). CARCIK-CD19 showed a dose-dependent antitumor response and prolonged persistence in a PDX, bearing the feature of a Philadelphia-like ALL with PAX5/AUTS2 translocation, and in a survival model of lymphoma, achieving complete eradication of disseminated tumors. Finally, the infusion of CARCIK-CD19 proved to be safe and well tolerated in a biodistribution and toxicity model. The infused cells persisted in the hematopoietic and post-injection perfused organs until the end of the study and consisted of CD8+, CD56+, and CAR+ T cells. Overall, these findings provide important implications for non-viral technology and the proof-of-concept that donor-derived CARCIK-CD19 are indeed effective against relapsed ALL, a possibility that will be tested in Phase I/II clinical trials after allogeneic hematopoietic stem-cell transplantation.


Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/therapeutic use , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/therapeutic use , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transfection , Xenograft Model Antitumor Assays
7.
J Autoimmun ; 85: 141-152, 2017 Dec.
Article En | MEDLINE | ID: mdl-28843422

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. Nowadays the survival rate is around 85%. Nevertheless, an urgent clinical need is still represented by primary refractory and relapsed patients who do not significantly benefit from standard approaches, including chemo-radiotherapy and hematopoietic stem cell transplantation (HSCT). For this reason, immunotherapy has so far represented a challenging novel treatment opportunity, including, as the most validated therapeutic options, cancer vaccines, donor-lymphocyte infusions and tumor-specific immune effector cells. More recently, unexpected positive clinical results in ALL have been achieved by application of gene-engineered chimeric antigen expressing (CAR) T cells. Several CAR designs across different trials have generated similar response rates, with Complete Response (CR) of 60-90% at 1 month and an Event-Free Survival (EFS) of 70% at 6 months. Relevant challenges anyway remain to be addressed, such as amelioration of technical, cost and feasibility aspects of cell and gene manipulation and the necessity to face the occurrence of relapse mechanisms. This review describes the state of the art of ALL immunotherapies, the novelties in terms of gene manipulation approaches and the problems emerged from early clinical studies. We describe and discuss the process of clinical translation, including the design of a cell manufacturing protocol, vector production and regulatory issues. Multiple antigen targeting and combination of CAR T cells with molecular targeted drugs have also been evaluated as latest strategies to prevail over immune-evasion.


Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Disease-Free Survival , Humans , Immunotherapy/methods
8.
Biol Blood Marrow Transplant ; 23(12): 2070-2078, 2017 Dec.
Article En | MEDLINE | ID: mdl-28712935

Seventy-four patients who relapsed after allogeneic stem cell transplantation were enrolled in a phase IIA study and treated with the sequential infusion of donor lymphocyte infusion (DLI) followed by cytokine-induced killer (CIK) cells. Seventy-three patients were available for the intention to treat analysis. At least 1 infusion of CIK cells was given to 59 patients, whereas 43 patients received the complete cell therapy planned (58%). Overall, 12 patients (16%) developed acute graft-versus-host disease (aGVHD) of grades I to II in 7 cases and grades III to IV in 5). In 8 of 12 cases, aGVHD developed during DLI treatment, leading to interruption of the cellular program in 3 patients, whereas in the remaining 5 cases aGVHD was controlled by steroids treatment, thus allowing the subsequent planned administration of CIK cells. Chronic GVHD (cGVHD) was observed in 11 patients (15%). A complete response was observed in 19 (26%), partial response in 3 (4%), stable disease in 8 (11%), early death in 2 (3%), and disease progression in 41 (56%). At 1 and 3 years, rates of progression-free survival were 31% and 29%, whereas rates of overall survival were 51% and 40%, respectively. By multivariate analysis, the type of relapse, the presence of cGVHD, and a short (<6 months) time from allogeneic hematopoietic stem cell transplantation to relapse were the significant predictors of survival. In conclusion, a low incidence of GVHD is observed after the sequential administration of DLI and CIK cells, and disease control can be achieved mostly after a cytogenetic or molecular relapse.


Cytokine-Induced Killer Cells/transplantation , Hematopoietic Stem Cell Transplantation/methods , Lymphocyte Transfusion/methods , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Humans , Lymphocyte Transfusion/adverse effects , Male , Middle Aged , Prognosis , Recurrence , Remission Induction , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Young Adult
9.
Mol Ther ; 25(8): 1933-1945, 2017 08 02.
Article En | MEDLINE | ID: mdl-28479045

Chimeric antigen receptor (CAR)-redirected T lymphocytes are a promising immunotherapeutic approach and object of pre-clinical evaluation for the treatment of acute myeloid leukemia (AML). We developed a CAR against CD123, overexpressed on AML blasts and leukemic stem cells. However, potential recognition of low CD123-positive healthy tissues, through the on-target, off-tumor effect, limits safe clinical employment of CAR-redirected T cells. Therefore, we evaluated the effect of context-dependent variables capable of modulating CAR T cell functional profiles, such as CAR binding affinity, CAR expression, and target antigen density. Computational structural biology tools allowed for the design of rational mutations in the anti-CD123 CAR antigen binding domain that altered CAR expression and CAR binding affinity without affecting the overall CAR design. We defined both lytic and activation antigen thresholds, with early cytotoxic activity unaffected by either CAR expression or CAR affinity tuning but later effector functions impaired by low CAR expression. Moreover, the anti-CD123 CAR safety profile was confirmed by lowering CAR binding affinity, corroborating CD123 is a good therapeutic target antigen. Overall, full dissection of these variables offers suitable anti-CD123 CAR design optimization for the treatment of AML.


Interleukin-3 Receptor alpha Subunit/chemistry , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , Binding Sites , Cytotoxicity, Immunologic , Gene Expression , Humans , Immunomodulation , Immunotherapy, Adoptive , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Models, Molecular , Molecular Conformation , Protein Binding , Receptors, Antigen, T-Cell/genetics , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Hum Gene Ther ; 28(3): 231-241, 2017 03.
Article En | MEDLINE | ID: mdl-27967241

Acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric high-risk patients, thus demanding advanced and personalized therapies. In this regard, different targeted immunotherapeutic approaches are available, ranging from naked monoclonal antibodies (mAb) to conjugated and multifunctional mAbs (i.e., BiTEs and DARTs). Recently, researchers have focused their attention on novel techniques of genetic manipulation specifically to redirect cytotoxic T cells endowed with chimeric antigen receptors (CARs) toward selected tumor associated antigens. So far, CAR T cells targeting the CD19 antigen expressed by B-cell origin hematological cancers have gained impressive clinical results, leading to the possibility of translating the CAR platform to treat other hematological malignancies such as AML. However, one of the main concerns in the field of AML CAR immunotherapy is the identification of an ideal target cell surface antigen, being highly expressed on tumor cells but minimally present on healthy tissues, together with the design of an anti-AML CAR appropriately balancing efficacy and safety profiles. The current review focuses mainly on AML target antigens and the related immunotherapeutic approaches developed so far, deeply dissecting methods of CAR T cell safety improvements, when designing novel CARs approaching human studies.


Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Leukemia, Myeloid, Acute/immunology
11.
Oncotarget ; 7(32): 51581-51597, 2016 08 09.
Article En | MEDLINE | ID: mdl-27323395

Chimeric antigen receptor (CAR)-modified T-cell adoptive immunotherapy is a remarkable therapeutic option proven effective in the treatment of hematological malignancies. In order to optimize cell manufacturing, we sought to develop a novel clinical-grade protocol to obtain CAR-modified cytokine-induced killer cells (CIKs) using the Sleeping Beauty (SB) transposon system. Administration of irradiated PBMCs overcame cell death of stimulating cells induced by non-viral transfection, enabling robust gene transfer together with efficient T-cell expansion. Upon single stimulation, we reached an average of 60% expression of CD123- and CD19- specific 3rd generation CARs (CD28/OX40/TCRzeta). Furthermore, modified cells displayed persistence of cell subsets with memory phenotype, specific and effective lytic activity against leukemic cell lines and primary blasts, cytokine secretion, and proliferation. Adoptive transfer of CD123.CAR or CD19.CAR lymphocytes led to a significant anti-tumor response against acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) disseminated diseases in NSG mice. Notably, we found no evidence of integration enrichment near cancer genes and transposase expression at the end of the differentiation. Taken all together, our findings describe a novel donor-derived non-viral CAR approach that may widen the repertoire of available methods for T cell-based immunotherapy.


Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Leukemia/pathology , Leukemia/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/transplantation , Transposases/genetics , Acute Disease , Adolescent , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cell Line, Tumor , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Infant , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/metabolism , Transposases/metabolism , Xenograft Model Antitumor Assays
12.
Cytotherapy ; 18(4): 481-6, 2016 Apr.
Article En | MEDLINE | ID: mdl-26971677

On November 10, 2014, the representatives of all six certified Good Manufacturing Practices (GMP) cell factories operating in the Lombardy Region of Italy convened a 1-day workshop in Milan titled "Management Models for the Development And Sustainability of Cell Factories: Public-Private Partnership?" The speakers and panelists addressed not only the many scientific, technological and cultural challenges faced by Lombardy Cell Factories, but also the potential impact of advanced therapy medicinal products (ATMPs) on public health and the role played by translational research in this process. Future perspectives for research and development (R&D) and manufacturing processes in the field of regenerative medicine were discussed as well. This report summarizes the most important issues raised by the workshop participants with particular emphasis on strengths and limitations of the R&D and manufacturing processes for innovative therapeutics in Lombardy and what can be improved in this context while maintaining GMP standards. The participants highlighted several strategies to translate patient-specific advanced therapeutics into scaled manufacturing products for clinical application. These included (i) the development of a synergistic interaction between public and private institutions, (ii) better integration with Italian regulatory agencies and (iii) the creation of a network among Lombardy cell factories and other Italian and European institutions.


Cell Culture Techniques , Cell Engineering , Laboratories/organization & administration , Models, Organizational , Therapies, Investigational , Biomedical Research/methods , Biomedical Research/organization & administration , Biomedical Research/standards , Biotechnology/organization & administration , Biotechnology/standards , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cell Engineering/methods , Cell Engineering/standards , Humans , Italy , Program Evaluation/standards , Quality Improvement , Therapies, Investigational/methods , Therapies, Investigational/standards
13.
Biomed Res Int ; 2015: 473159, 2015.
Article En | MEDLINE | ID: mdl-26495296

According to the European Medicine Agency (EMA) regulatory frameworks, Advanced Therapy Medicinal Products (ATMP) represent a new category of drugs in which the active ingredient consists of cells, genes, or tissues. ATMP-CD133 has been widely investigated in controlled clinical trials for cardiovascular diseases, making CD133(+) cells one of the most well characterized cell-derived drugs in this field. To ensure high quality and safety standards for clinical use, the manufacturing process must be accomplished in certified facilities following standard operative procedures (SOPs). In the present work, we report the fully compliant GMP-grade production of ATMP-CD133 which aims to address the treatment of chronic refractory ischemic heart failure. Starting from bone marrow (BM), ATMP-CD133 manufacturing output yielded a median of 6.66 × 10(6) of CD133(+) cells (range 2.85 × 10(6)-30.84 × 10(6)), with a viability ranged between 96,03% and 99,97% (median 99,87%) and a median purity of CD133(+) cells of 90,60% (range 81,40%-96,20%). Based on these results we defined our final release criteria for ATMP-CD133: purity ≥ 70%, viability ≥ 80%, cellularity between 1 and 12 × 10(6) cells, sterile, and endotoxin-free. The abovementioned criteria are currently applied in our Phase I clinical trial (RECARDIO Trial).


Antigens, CD/metabolism , Bone Marrow Cells/cytology , Bone Marrow Transplantation/standards , Cardiomyopathies/therapy , Glycoproteins/metabolism , Myocardial Ischemia/therapy , Peptides/metabolism , Stem Cell Transplantation/standards , AC133 Antigen , Animals , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Device Approval/standards , Europe , Guideline Adherence , Humans , Myocardial Ischemia/complications , Myocardial Ischemia/pathology , Practice Guidelines as Topic , Stem Cells
14.
Curr Opin Hematol ; 22(6): 497-502, 2015 Nov.
Article En | MEDLINE | ID: mdl-26390165

PURPOSE OF REVIEW: Allogeneic hematopoietic stem cell transplantation (HSCT) is still partially ineffective in curing high-risk hematological malignancies, with estimates of relapse rates ranging from 40 to 50%. The purpose of this review is to discuss the emerging therapeutic options for patients with relapsed disease following HSCT based on adoptive immunotherapy using donor-derived T cells genetically engineered to express CD19-specific chimeric antigen receptors (CARs). RECENT FINDINGS: Adoptive cell therapy (ACT) with CAR-modified T cells represents an attractive therapeutic option for further enhancing the graft-versus-leukemia effect. However, CAR-modified T cells are often obtained using apheresis products collected from the patient's own blood, a procedure that has hindered the application of CAR-based therapies into the clinic. Alternative approaches rely on CAR T cells derived from donors rather than the patient's own blood. Therefore, it appears that overcoming the practical limitation of allogeneic T cell-induced graft versus-host-disease is a key to providing access to CAR immunotherapy to all eligible patients. SUMMARY: Donor-derived CD19-CAR T cells may advance the field of CAR immunotherapy by controlling relapse in leukemic patients and improving the range of applications of ACT protocols.


Antigens, CD19/immunology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Immunotherapy , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tissue Donors , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Transplantation, Homologous
15.
World J Gastroenterol ; 21(14): 4379-84, 2015 Apr 14.
Article En | MEDLINE | ID: mdl-25892890

Mesenchymal stem cells (MSC) are cells of stromal origin which exhibit unlimited self-renewal capacity and pluripotency in vitro. It has recently been observed that MSC may also exert a profound immunosuppressive and anti-inflammatory effect both in vitro and in vivo with consequent potential use in autoimmune disorders. We present the case of a patient suffering from childhood-onset, multidrug resistant and steroid-dependent Crohn's disease who underwent systemic infusions of MSC, which led to a temporary reduction in CCR4, CCR7 and CXCR4 expression by T-cells, and a temporary decrease in switched memory B-cells, In addition, following MSC infusion, lower doses of steroids were needed to inhibit proliferation of the patient's peripheral blood mononuclear cells. Despite these changes, no significant clinical benefit was observed, and the patient required rescue therapy with infliximab and subsequent autologous hematopoietic stem cell transplantation. The results of biological and in vitro observations after MSC use and the clinical effects of infusion are discussed, and a brief description is provided of previous data on MSC-based therapy in autoimmune disorders.


Crohn Disease/surgery , Interferon-gamma/therapeutic use , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Adult , Cells, Cultured , Compassionate Use Trials , Crohn Disease/diagnosis , Crohn Disease/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppressive Agents/therapeutic use , Mesenchymal Stem Cells/immunology , Reoperation , Salvage Therapy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Failure
16.
Oncoimmunology ; 3: e28835, 2014.
Article En | MEDLINE | ID: mdl-25083319

Chimeric antigen receptor (CAR) modified T cells have emerged as powerful tools for controlling leukemias. We recently showed that anti-CD123 CAR-expressing cytokine-induced killer T cell treatment is an effective immunotherapeutic approach to eradicate Acute Myeloid Leukemia (AML) cells. Here, we discuss how this genetically modified cell-based strategy could be relevant to the field of AML therapeutics.

17.
Stem Cell Res Ther ; 5(1): 3, 2014 Jan 09.
Article En | MEDLINE | ID: mdl-24405828

INTRODUCTION: Despite having a proven immunosuppressive potential in vitro, human mesenchymal stromal cells (MSCs) are reported to display variable efficacy in vivo and, in fact, their proven benefit in the clinical practice is still limited and controversial. METHODS: The interplay between clinical grade MSCs and pre-activated donor lymphocytes or selected lymphocyte subsets was studied in vitro. The kinetics of MSC growth and viability was evaluated by adhesion-dependent changes of culture plate impedance and biochemically by a colorimetric assay. Activation of natural killer (NK) cells was assessed as well, using a flow cytometry assay. RESULTS: A strong inhibition of MSC growth was rapidly induced by the addition of pre-activated lymphocytes but not of resting lymphocytes. Inhibition seems not to be attributable to a single cell population, as similar results can be obtained by depleting NK cells or by using either selected CD4+ or CD8+ lymphocytes. In addition, conditioned medium (CM) from activated lymphocytes was able to inhibit MSC growth in a dose-dependent manner. Furthermore, licensing with IFN-γ partially protected MSCs from pre-activated lymphocytes but not from their CM. These results suggest an inhibitory role of lymphocyte-activation-derived substances. However, the identification of a single molecule responsible for MSC inhibition remained elusive, even if preliminary experiments showed that ATP and, to a lesser extent, TNF-α might play a role. CONCLUSIONS: These results suggest that survival of MSCs can be affected by soluble mediators released by activated lymphocytes. Thus it can be hypothesized that MSC immunosuppressive action in vivo could be impaired by ongoing immune activation through the release of inflammatory mediators.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Killer Cells, Natural/immunology , Lymphocyte Activation , Mesenchymal Stem Cells/physiology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Cytokines/analysis , Humans , Killer Cells, Natural/metabolism , Mesenchymal Stem Cells/drug effects
18.
Neuropharmacology ; 79: 119-26, 2014 Apr.
Article En | MEDLINE | ID: mdl-24246661

The need for immunosuppression after allo/xenogenic mesenchymal stromal cell (MSC) transplantation is debated. This study compared the long-term effects of human (h) bone marrow MSC transplant in immunocompetent or immunosuppressed traumatic brain injured (TBI) mice. C57Bl/6 male mice were subjected to TBI or sham surgery followed 24 h later by an intracerebroventricular infusion of phosphate buffer saline (PBS, control) or hMSC (150,000/5 µl). Immunocompetent and cyclosporin A immunosuppressed (CsA) mice were analyzed for gene expression at 72 h, functional deficits and histological analysis at five weeks. Gene expression analysis showed the effectiveness of immunosuppression (INFγ reduction in CsA treated groups), with no evidence of early rejection (no changes of MHCII and CD86 in all TBI groups) and selective induction of T-reg (increase of Foxp3) only in the TBI hMSC group. Five weeks after TBI, hMSC had comparable efficacy, with functional recovery (on both sensorimotor and cognitive deficits) and structural protection (contusion volume, vessel rescue effect, gliotic scar reduction, induction of neurogenesis) in immunosuppressed and immunocompetent mice. Therefore, long-term hMSC efficacy in TBI is not dependent on immunosuppressive treatment. These findings could have important clinical implication since immunosuppression in acute TBI patients may increase their risk of infection and not be tolerated.


Brain Injuries/physiopathology , Brain Injuries/therapy , Brain/physiopathology , Immunosuppression Therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Animals , Brain/drug effects , Brain/pathology , Brain Injuries/pathology , Cyclosporine/therapeutic use , Gene Expression/drug effects , Gene Expression/physiology , Graft Rejection/metabolism , Humans , Immunosuppressive Agents/therapeutic use , Male , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Neuropsychological Tests , Recovery of Function/drug effects , Recovery of Function/physiology , Treatment Outcome
19.
Biol Blood Marrow Transplant ; 20(3): 375-81, 2014 Mar.
Article En | MEDLINE | ID: mdl-24321746

This phase I multicenter study was aimed at assessing the feasibility and safety of intravenous administration of third party bone marrow-derived mesenchymal stromal cells (MSC) expanded in platelet lysate in 40 patients (15 children and 25 adults), experiencing steroid-resistant grade II to IV graft-versus-host disease (GVHD). Patients received a median of 3 MSC infusions after having failed conventional immunosuppressive therapy. A median cell dose of 1.5 × 10(6)/kg per infusion was administered. No acute toxicity was reported. Overall, 86 adverse events and serious adverse events were reported in the study, most of which (72.1%) were of infectious nature. Overall response rate, measured at 28 days after the last MSC injection, was 67.5%, with 27.5% complete response. The latter was significantly more frequent in patients exhibiting grade II GVHD as compared with higher grades (61.5% versus 11.1%, P = .002) and was borderline significant in children as compared with adults (46.7 versus 16.0%, P = .065). Overall survival at 1 and 2 years from the first MSC administration was 50.0% and 38.6%, with a median survival time of 1.1 years. In conclusion, MSC can be safely administered on top of conventional immunosuppression for steroid resistant GVHD treatment. Eudract Number 2008-007869-23, NCT01764100.


Antineoplastic Agents/therapeutic use , Graft vs Host Disease/therapy , Hematologic Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Adolescent , Adult , Aged , Child , Child, Preschool , Drug Resistance, Neoplasm , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Humans , Immunosuppressive Agents/therapeutic use , Infant , Male , Middle Aged , Remission Induction , Severity of Illness Index , Steroids/therapeutic use , Survival Analysis , Transplantation, Homologous
20.
Biologicals ; 41(6): 439-45, 2013 Nov.
Article En | MEDLINE | ID: mdl-24135082

Focal segmental glomerulosclerosis (FSGS) is the most frequent acquired renal condition resulting in end stage kidney disease in children. We describe a cell therapy treatment with human allogeneic bone marrow mesenchymal stem cells (MSC) in a 13-year-old patient developing recurrent FSGS after renal transplantation, which was not responding to conventional therapy. This treatment relied on the following measurements:clinical and laboratory evaluation of renal function, proteome array, biopsy, short tandem repeat assay. Before MSC treatment, the patient needed weekly plasmapheresis to achieve proteinuria-to-creatininuria ratio below 5. After three MSC infusions without adverse events, the patient has a stable renal function and the proteinuria target was reached without plasmapheresis. In addition, some circulating inflammatory factors decreased and their levels were still low after one year. This is the first report of an MSC treatment in an FSGS patient. Even though different factors may have contributed to the clinical results, after MSC infusion a stable reduction in the serum level of several inflammatory factors has been registered and the patient does not need anymore plasmapheresis to keep proteinuria under control. In addition, this encouraging single case let us identify some putative efficacy biomarkers that could be of clinical interest in chronic kidney diseases.


Glomerulosclerosis, Focal Segmental/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Adolescent , Cell Survival , Cells, Cultured , Flow Cytometry , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Immunophenotyping , Kidney Transplantation/adverse effects , Male , Mesenchymal Stem Cells/metabolism , Transplantation, Homologous , Treatment Outcome
...