Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Blood Adv ; 8(2): 453-467, 2024 01 23.
Article in English | MEDLINE | ID: mdl-37903325

ABSTRACT

ABSTRACT: More than half of the patients treated with CD19-targeted chimeric antigen receptor (CAR) T-cell immunotherapy for large B-cell lymphoma (LBCL) do not achieve durable remission, which may be partly due to PD-1/PD-L1-associated CAR T-cell dysfunction. We report data from a phase 1 clinical trial (NCT02706405), in which adults with LBCL were treated with autologous CD19 CAR T cells (JCAR014) combined with escalating doses of the anti-PD-L1 monoclonal antibody, durvalumab, starting either before or after CAR T-cell infusion. The addition of durvalumab to JCAR014 was safe and not associated with increased autoimmune or immune effector cell-associated toxicities. Patients who started durvalumab before JCAR014 infusion had later onset and shorter duration of cytokine release syndrome and inferior efficacy, which was associated with slower accumulation of CAR T cells and lower concentrations of inflammatory cytokines in the blood. Initiation of durvalumab before JCAR014 infusion resulted in an early increase in soluble PD-L1 (sPD-L1) levels that coincided with the timing of maximal CAR T-cell accumulation in the blood. In vitro, sPD-L1 induced dose-dependent suppression of CAR T-cell effector function, which could contribute to inferior efficacy observed in patients who received durvalumab before JCAR014. Despite the lack of efficacy improvement and similar CAR T-cell kinetics early after infusion, ongoing durvalumab therapy after JCAR014 was associated with re-expansion of CAR T cells in the blood, late regression of CD19+ and CD19- tumors, and enhanced duration of response. Our results indicate that the timing of initiation of PD-L1 blockade is a key variable that affects outcomes after CD19 CAR T-cell immunotherapy for adults with LBCL.


Subject(s)
Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Adult , Humans , B7-H1 Antigen , Cytokine Release Syndrome/etiology , Immunotherapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/etiology
2.
Eur J Neurol ; 30(10): 3016-3031, 2023 10.
Article in English | MEDLINE | ID: mdl-37515394

ABSTRACT

BACKGROUND AND PURPOSE: Transcranial direct current stimulation (tDCS) has been shown to improve signs of consciousness in a subset of patients with disorders of consciousness (DoC). However, no multicentre study confirmed its efficacy when applied during rehabilitation. In this randomized controlled double-blind study, the effects of tDCS whilst patients were in rehabilitation were tested at the group level and according to their diagnosis and aetiology to better target DoC patients who might repond to tDCS. METHODS: Patients received 2 mA tDCS or sham applied over the left prefrontal cortex for 4 weeks. Behavioural assessments were performed weekly and up to 3 months' follow-up. Analyses were conducted at the group and subgroup levels based on the diagnosis (minimally conscious state [MCS] and unresponsive wakefulness syndrome) and the aetiology (traumatic or non-traumatic). Interim analyses were planned to continue or stop the trial. RESULTS: The trial was stopped for futility when 62 patients from 10 centres were enrolled (44 ± 14 years, 37 ± 24.5 weeks post-injury, 18 women, 32 MCS, 39 non-traumatic). Whilst, at the group level, no treatment effect was found, the subgroup analyses at 3 months' follow-up revealed a significant improvement for patients in MCS and with traumatic aetiology. CONCLUSIONS: Transcranial direct current stimulation during rehabilitation does not seem to enhance patients' recovery. However, diagnosis and aetiology appear to be important factors leading to a response to the treatment. These findings bring novel insights into possible cortical plasticity changes in DoC patients given these differential results according to the subgroups of patients.


Subject(s)
Transcranial Direct Current Stimulation , Humans , Female , Transcranial Direct Current Stimulation/methods , Treatment Outcome , Consciousness Disorders/therapy , Consciousness Disorders/diagnosis , Prefrontal Cortex , Persistent Vegetative State/therapy , Persistent Vegetative State/diagnosis
3.
Blood Adv ; 7(11): 2479-2493, 2023 06 13.
Article in English | MEDLINE | ID: mdl-36332004

ABSTRACT

Chimeric antigen receptor (CAR)-modified T-cell therapies targeting CD19 represent a new treatment option for patients with relapsed/refractory (R/R) B-cell malignancies. However, CAR T-cell therapy fails to elicit durable responses in a significant fraction of patients. Limited in vivo proliferation and survival of infused CAR T cells are key causes of failure. In a phase 1/2 clinical trial of CD19 CAR T cells for B-cell malignancies (#NCT01865617), low serum interleukin 15 (IL-15) concentration after CAR T-cell infusion was associated with inferior CAR T-cell kinetics. IL-15 supports T-cell proliferation and survival, and therefore, supplementation with IL-15 may enhance CAR T-cell therapy. However, the clinical use of native IL-15 is challenging because of its unfavorable pharmacokinetic (PK) and toxicity. NKTR-255 is a polymer-conjugated IL-15 that engages the entire IL-15 receptor complex (IL-15Rα/IL-2Rßγ) and exhibits reduced clearance, providing sustained pharmacodynamic (PD) responses. We investigated the PK and immune cell PDs in nonhuman primates treated with NKTR-255 and found that NKTR-255 enhanced the in vivo proliferation of T cells and natural killer cells. In vitro, NKTR-255 induced dose-dependent proliferation and accumulation of human CD19 CAR T cells, especially at low target cell abundance. In vivo studies in lymphoma-bearing immunodeficient mice demonstrated enhanced antitumor efficacy of human CD19 CAR T cells. In contrast to mice treated with CAR T cells alone, those that received CAR T cells and NKTR-255 had markedly higher CAR T-cell counts in the blood and marrow that were sustained after tumor clearance, without evidence of persistent proliferation or ongoing activation/exhaustion as assessed by Ki-67 and inhibitory receptor coexpression. These data support an ongoing phase 1 clinical trial of combined therapy with CD19 CAR T cells and NKTR-255 for R/R B-cell malignancies.


Subject(s)
Interleukin-15 , Receptors, Antigen, T-Cell , Humans , Animals , Mice , Neoplasm Recurrence, Local , T-Lymphocytes , Immunotherapy , Antigens, CD19
4.
Eur J Neurol ; 29(2): 390-399, 2022 02.
Article in English | MEDLINE | ID: mdl-34657359

ABSTRACT

BACKGROUND AND PURPOSE: Patients with prolonged disorders of consciousness (pDoC) have a high mortality rate due to medical complications. Because an accurate prognosis is essential for decision-making on patients' management, we analysed data from an international multicentre prospective cohort study to evaluate 2-year mortality rate and bedside predictors of mortality. METHODS: We enrolled adult patients in prolonged vegetative state/unresponsive wakefulness syndrome (VS/UWS) or minimally conscious state (MCS) after traumatic and nontraumatic brain injury within 3 months postinjury. At enrolment, we collected demographic (age, sex), anamnestic (aetiology, time postinjury), clinical (Coma Recovery Scale-Revised [CRS-R], Disability Rating Scale, Nociception Coma Scale-Revised), and neurophysiologic (electroencephalogram [EEG], somatosensory evoked and event-related potentials) data. Patients were followed up to gather data on mortality up to 24 months postinjury. RESULTS: Among 143 traumatic (n = 55) and nontraumatic (n = 88) patients (VS/UWS, n = 68, 19 females; MCS, n = 75, 22 females), 41 (28.7%) died within 24 months postinjury. Mortality rate was higher in VS/UWS (42.6%) than in MCS (16%; p < 0.001). Multivariate regression in VS/UWS showed that significant predictors of mortality were older age and lower CRS-R total score, whereas in MCS female sex and absence of alpha rhythm on EEG at study entry were significant predictors. CONCLUSIONS: This study demonstrated that a feasible multimodal assessment in the postacute phase can help clinicians to identify patients with pDoC at higher risk of mortality within 24 months after brain injury. This evidence can help clinicians and patients' families to navigate the complex clinical decision-making process and promote an international standardization of prognostic procedures for patients with pDoC.


Subject(s)
Brain Injuries , Consciousness , Adult , Brain Injuries/complications , Consciousness/physiology , Consciousness Disorders , Female , Humans , Persistent Vegetative State , Prognosis , Prospective Studies , Risk Factors
5.
Front Immunol ; 12: 780442, 2021.
Article in English | MEDLINE | ID: mdl-34899742

ABSTRACT

A range of emerging therapeutic approaches for the treatment of cancer aim to induce or augment endogenous T cell responses. Chimeric antigen receptor (CAR) T cell therapy (CTT) is one such approach that utilises the patient's own T cells, engineered ex vivo to target cell surface antigens, to eliminate haematological malignancies. Despite mediating high rates of responses in some clinical trials, this approach can be limited by dysfunctional T cells if they are present at high frequencies either in the starting material from the patient or the CAR T cell product. The fitness of an individual's T cells, driven by age, chronic infection, disease burden and cancer treatment, is therefore likely to be a crucial limiting factor of CTT. Currently, T cell dysfunction and its impact on CTT is not specifically quantified when patients are considering the therapy. Here, we review our current understanding of T cell fitness for CTT, how fitness may be impacted by age, chronic infection, malignancy, and treatment. Finally, we explore options to specifically tailor clinical decision-making and the CTT protocol for patients with more extensive dysfunction to improve treatment efficacy. A greater understanding of T cell fitness throughout a patient's treatment course could ultimately be used to identify patients likely to achieve favourable CTT outcomes and improve methods for T cell collection and CTT delivery.


Subject(s)
Genetic Therapy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/transplantation , Animals , Clinical Decision-Making , Genetic Therapy/adverse effects , Health Status , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Humans , Immunotherapy, Adoptive/adverse effects , Patient Selection , Phenotype , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Risk Assessment , Risk Factors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
6.
Clin Cancer Res ; 27(20): 5718-5730, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34380639

ABSTRACT

PURPOSE: We previously identified mesothelin (MSLN) as highly expressed in a significant fraction of acute myeloid leukemia (AML) but entirely silent in normal hematopoiesis, providing a promising antigen for immunotherapeutic targeting that avoids hematopoietic toxicity. Given that T cells genetically modified to express chimeric antigen receptors (CAR) are effective at eradicating relapsed/refractory acute lymphocytic leukemia, we developed MSLN-directed CAR T cells for preclinical evaluation in AML. EXPERIMENTAL DESIGN: The variable light (VL) and heavy (VH) sequences from the MSLN-targeting SS1P immunotoxin were used to construct the single-chain variable fragment of the standard CAR containing 41-BB costimulatory and CD3Zeta stimulatory domains. The preclinical efficacy of MSLN CAR T cells was evaluated against AML cell lines and patient samples expressing various levels of MSLN in vitro and in vivo. RESULTS: We demonstrate that MSLN is expressed on the cell surface of AML blasts and leukemic stem cell-enriched CD34+CD38- subset, but not on normal hematopoietic stem and progenitor cells (HSPC). We further establish that MSLN CAR T cells are highly effective in eliminating MSLN-positive AML cells in cell line- and patient-derived xenograft models. Importantly, MSLN CAR T cells can target and eradicate CD34+CD38- cells without impacting the viability of normal HSPCs. Finally, we show that CAR T-cell functionality can be improved by inhibition of the ADAM17 metalloprotease that promotes shedding of MSLN. CONCLUSIONS: These findings demonstrate that MSLN is a viable target for CAR T-cell therapy in AML and that inhibiting MSLN shedding is a promising approach to improve CAR T-cell efficacy.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Mesothelin/antagonists & inhibitors , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/therapeutic use , Adolescent , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Infant , Male
7.
BioDrugs ; 35(3): 281-302, 2021 May.
Article in English | MEDLINE | ID: mdl-33826079

ABSTRACT

Chimeric antigen receptor (CAR)-T cell therapy has shown impressive results in chemorefractory B cell malignancies, raising the possibilities of using this immunotherapeutic modality for other devastating hematologic malignancies, such as acute myeloid leukemia (AML). AML is an aggressive hematologic malignancy which, like B cell malignancies, poses several challenges for clinical translation of successful immunotherapy. The antigenic heterogeneity of AML results in a list of potential targets that CAR-T cells could be directed towards, each with advantages and disadvantages. In this review, we provide an up-to-date report of outcomes and adverse effects from published and presented clinical trials of CAR-T cell therapy for AML and provide the preclinical rationale underlying these studies and antigen selection. Comparison across trials is difficult, yet themes emerge with respect to appropriate antigen selection and association of adverse effects with outcomes. We highlight currently active clinical trials and the potential improvements and caveats with these novel approaches. Key hurdles to the successful introduction of CAR-T cell therapy for the treatment of AML include the effect of antigenic heterogeneity and trade-offs between therapy specificity and sensitivity; on-target off-tumor toxicities; the AML tumor microenvironment; and practical considerations for future trials that should be addressed to enable successful CAR-T cell therapy for AML.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Humans , Immunotherapy , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/therapy , Receptors, Chimeric Antigen/genetics , Tumor Microenvironment
8.
Immunometabolism ; 3(1)2021.
Article in English | MEDLINE | ID: mdl-33552594

ABSTRACT

Immune reconstitution following allogeneic hematopoietic stem cell transplantation (allo-HSCT) sets the stage for the goal of a successful transplant-the prevention of disease relapse without graft versus host disease (GVHD) and opportunistic infection. In both epidemiologic studies and in controlled animal studies, it is known that the gut microbiome (GM) can profoundly influence normal innate and adaptive immune development and can be altered by microbial transfer and antibiotics. Following allo-HSCT the GM has been shown to influence clinical outcomes but published associations between the GM and immune reconstitution post-allo-HSCT are lacking. In this viewpoint we propose that the extensive knowledge garnered from studying normal immune development can serve as a framework for studying immune development post-allo-HSCT. We summarize existing studies addressing the effect of the GM on immune ontogeny and draw associations with immune reconstitution and the GM post-allo-HSCT.

9.
Leukemia ; 35(9): 2496-2507, 2021 09.
Article in English | MEDLINE | ID: mdl-33589747

ABSTRACT

There is increasing interest in targeting CD33 in malignant and non-malignant disorders. In acute myeloid leukemia, longer survival with the CD33 antibody-drug conjugate gemtuzumab ozogamicin (GO) validates this strategy. Still, GO benefits only some patients, prompting efforts to develop more potent CD33-directed therapeutics. As one limitation, CD33 antibodies typically recognize the membrane-distal V-set domain. Using various artificial CD33 proteins, in which this domain was differentially positioned within the extracellular portion of the molecule, we tested whether targeting membrane-proximal epitopes enhances the effector functions of CD33 antibody-based therapeutics. Consistent with this idea, a CD33V-set/CD3 bispecific antibody (BsAb) and CD33V-set-directed chimeric antigen receptor (CAR)-modified T cells elicited substantially greater cytotoxicity against cells expressing a CD33 variant lacking the entire C2-set domain than cells expressing full-length CD33, whereas cytotoxic effects induced by GO were independent of the position of the V-set domain. We therefore raised murine and human antibodies against the C2-set domain of human CD33 and identified antibodies that bound CD33 regardless of the presence/absence of the V-set domain ("CD33PAN antibodies"). These antibodies internalized when bound to CD33 and, as CD33PAN/CD3 BsAb, had potent cytolytic effects against CD33+ cells. Together, our data provide the rationale for further development of CD33PAN antibody-based therapeutics.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Gemtuzumab/chemistry , Immunoconjugates/pharmacology , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/biosynthesis , Antineoplastic Agents, Immunological/chemistry , Humans , Immunoconjugates/chemistry , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Cells, Cultured
10.
Blood ; 137(3): 323-335, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32967009

ABSTRACT

CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T-cell therapy has shown significant efficacy for relapsed or refractory (R/R) B-cell malignancies. Yet, CD19 CAR T cells fail to induce durable responses in most patients. Second infusions of CD19 CAR T cells (CART2) have been considered as a possible approach to improve outcomes. We analyzed data from 44 patients with R/R B-cell malignancies (acute lymphoblastic leukemia [ALL], n = 14; chronic lymphocytic leukemia [CLL], n = 9; non-Hodgkin lymphoma [NHL], n = 21) who received CART2 on a phase 1/2 trial (NCT01865617) at our institution. Despite a CART2 dose increase in 82% of patients, we observed a low incidence of severe toxicity after CART2 (grade ≥3 cytokine release syndrome, 9%; grade ≥3 neurotoxicity, 11%). After CART2, complete response (CR) was achieved in 22% of CLL, 19% of NHL, and 21% of ALL patients. The median durations of response after CART2 in CLL, NHL, and ALL patients were 33, 6, and 4 months, respectively. Addition of fludarabine to cyclophosphamide-based lymphodepletion before the first CAR T-cell infusion (CART1) and an increase in the CART2 dose compared with CART1 were independently associated with higher overall response rates and longer progression-free survival after CART2. We observed durable CAR T-cell persistence after CART2 in patients who received cyclophosphamide and fludarabine (Cy-Flu) lymphodepletion before CART1 and a higher CART2 compared with CART1 cell dose. The identification of 2 modifiable pretreatment factors independently associated with better outcomes after CART2 suggests strategies to improve in vivo CAR T-cell kinetics and responses after repeat CAR T-cell infusions, and has implications for the design of trials of novel CAR T-cell products after failure of prior CAR T-cell immunotherapies.


Subject(s)
Antigens, CD19/metabolism , Immunotherapy, Adoptive , Leukemia, B-Cell/therapy , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphoma, Non-Hodgkin/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Aged , Cell Proliferation , Cyclophosphamide/therapeutic use , Cytokine Release Syndrome/complications , Female , Humans , Leukemia, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphoma, Non-Hodgkin/immunology , Male , Middle Aged , Multivariate Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Progression-Free Survival , T-Lymphocytes/immunology , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
11.
Neurosci Lett ; 736: 135293, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32771601

ABSTRACT

Previous evidence suggests that changes in spontaneous eye blink rate (EBR) in human adults might reflect the amount of attentional demand (i.e. cognitive load) during cognitive tasks. However, the actual direction of this relation is uncertain, since most studies investigated the role of cognitive load on EBR by employing visual tasks only. Here we aimed at elucidating the relationship between EBR and cognitive load in non-visual tasks. Sixteen healthy participants performed two auditory oddball tasks, i.e. passive listening to auditory tones versus active counting of target tones; each oddball task was immediately followed by a rest phase. Throughout the oddball tasks we assessed EBR and recorded the P300 on ERPs as an electrophysiological measure of attention. The results showed that participants' EBR increased during the active task compared to the respective rest phase. Amplitude and latency of the P300 too differed between passive and active tasks, but changes in EBR and P300 features were not correlated with each other. Our findings demonstrated that an increase in cognitive load is associated with an increase in EBR in cognitive tasks not involving visual attention. These findings are consistent with previous evidence suggesting shared neurobiological bases between attention and EBR.


Subject(s)
Attention/physiology , Auditory Perception/physiology , Blinking/physiology , Cognition/physiology , Event-Related Potentials, P300/physiology , Acoustic Stimulation , Adult , Brain/physiology , Electroencephalography , Female , Humans , Male , Young Adult
12.
Neurology ; 95(11): e1488-e1499, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32661102

ABSTRACT

OBJECTIVE: This international multicenter, prospective, observational study aimed at identifying predictors of short-term clinical outcome in patients with prolonged disorders of consciousness (DoC) due to acquired severe brain injury. METHODS: Patients in vegetative state/unresponsive wakefulness syndrome (VS/UWS) or in minimally conscious state (MCS) were enrolled within 3 months from their brain injury in 12 specialized medical institutions. Demographic, anamnestic, clinical, and neurophysiologic data were collected at study entry. Patients were then followed up for assessing the primary outcome, that is, clinical diagnosis according to standardized criteria at 6 months postinjury. RESULTS: We enrolled 147 patients (44 women; mean age 49.4 [95% confidence interval 46.1-52.6] years; VS/UWS 71, MCS 76; traumatic 55, vascular 56, anoxic 36; mean time postinjury 59.6 [55.4-63.6] days). The 6-month follow-up was complete for 143 patients (VS/UWS 70; MCS 73). With respect to study entry, the clinical diagnosis improved in 72 patients (VS/UWS 27; MCS 45). Younger age, shorter time postinjury, higher Coma Recovery Scale-Revised total score, and presence of EEG reactivity to eye opening at study entry predicted better outcome, whereas etiology, clinical diagnosis, Disability Rating Scale score, EEG background activity, acoustic reactivity, and P300 on event-related potentials were not associated with outcome. CONCLUSIONS: Multimodal assessment could identify patients with higher likelihood of clinical improvement in order to help clinicians, families, and funding sources with various aspects of decision-making. This multicenter, international study aims to stimulate further research that drives international consensus regarding standardization of prognostic procedures for patients with DoC.


Subject(s)
Brain Injuries/diagnosis , Brain Injuries/physiopathology , Consciousness Disorders/diagnosis , Consciousness Disorders/physiopathology , Adult , Brain Injuries/complications , Consciousness Disorders/etiology , Electroencephalography/trends , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Prognosis , Prospective Studies , Time Factors , Treatment Outcome
13.
Bone Marrow Transplant ; 55(9): 1706-1715, 2020 09.
Article in English | MEDLINE | ID: mdl-32474570

ABSTRACT

In the United States the increasing number of Food and Drug Administration (FDA)-approved, innovative, and potentially effective commercial cancer therapies pose a significant financial burden on public and private payers. Chimeric antigen receptor (CAR) T cells are prototypical of this challenge. In 2017 and 2018, tisagenlecleucel (Kymriah, Novartis) and axicabtagene ciloleucel (Yescarta, Kite) were approved by the FDA for use after showing groundbreaking results in relapsed/refractory B-cell malignancies. In 2020 and 2021, four further submissions to the FDA are expected for CAR T-cell therapies for indolent and aggressive B-cell malignancies and plasma cell myeloma. Yet, with marketed prices of over $350,000 per infusion for the two FDA-approved therapies and similar price tags expected for the coming products, serious concerns are raised over value and affordability. In this review we summarize recent, peer-reviewed cost-effectiveness studies of tisagenlecleucel and axicabtagene ciloleucel in the United States; discuss key issues concerning the health plan budget impact of CAR T-cell therapy; and review policy, payment and scientific approaches that may improve the value and affordability of CAR T-cell therapy.


Subject(s)
Immunotherapy, Adoptive , Neoplasm Recurrence, Local , Cost-Benefit Analysis , Humans , Receptors, Antigen, T-Cell , United States , United States Food and Drug Administration
14.
J Clin Med ; 9(5)2020 May 04.
Article in English | MEDLINE | ID: mdl-32375368

ABSTRACT

The data from patients with severe brain injuries show complex brain functions. Due to the difficulties associated with these complex data, computational modeling is an especially useful tool to examine the structure-function relationship in these populations. By using computational modeling for patients with a disorder of consciousness (DoC), not only we can understand the changes of information transfer, but we also can test changes to different states of consciousness by hypothetically changing the anatomical structure. The generalized Ising model (GIM), which specializes in using structural connectivity to simulate functional connectivity, has been proven to effectively capture the relationship between anatomical structures and the spontaneous fluctuations of healthy controls (HCs). In the present study we implemented the GIM in 25 HCs as well as in 13 DoC patients diagnosed at three different states of consciousness. Simulated data were analyzed and the criticality and dimensionality were calculated for both groups; together, those values capture the level of information transfer in the brain. Ratifying previous studies, criticality was observed in simulations of HCs. We were also able to observe criticality for DoC patients, concluding that the GIM is generalizable for DoC patients. Furthermore, dimensionality increased for the DoC group as compared to healthy controls, and could distinguish different diagnostic groups of DoC patients.

16.
Front Neurol ; 9: 861, 2018.
Article in English | MEDLINE | ID: mdl-30405513

ABSTRACT

Behavioral assessments could not suffice to provide accurate diagnostic information in individuals with disorders of consciousness (DoC). Multimodal neuroimaging markers have been developed to support clinical assessments of these patients. Here we present findings obtained by hybrid fludeoxyglucose (FDG-)PET/MR imaging in three severely brain-injured patients, one in an unresponsive wakefulness syndrome (UWS), one in a minimally conscious state (MCS), and one patient emerged from MCS (EMCS). Repeated behavioral assessment by means of Coma Recovery Scale-Revised and neurophysiological evaluation were performed in the two weeks before and after neuroimaging acquisition, to ascertain that clinical diagnosis was stable. The three patients underwent one imaging session, during which two resting-state fMRI (rs-fMRI) blocks were run with a temporal gap of about 30 min. rs-fMRI data were analyzed with a graph theory approach applied to nine independent networks. We also analyzed the benefits of concatenating the two acquisitions for each patient or to select for each network the graph strength map with a higher ratio of fitness. Finally, as for clinical assessment, we considered the best functional connectivity pattern for each network and correlated graph strength maps to FDG uptake. Functional connectivity analysis showed several differences between the two rs-fMRI acquisitions, affecting in a different way each network and with a different variability for the three patients, as assessed by ratio of fitness. Moreover, combined PET/fMRI analysis demonstrated a higher functional/metabolic correlation for patients in EMCS and MCS compared to UWS. In conclusion, we observed for the first time, through a test-retest approach, a variability in the appearance and temporal/spatial patterns of resting-state networks in severely brain-injured patients, proposing a new method to select the most informative connectivity pattern.

17.
J Neurol Sci ; 375: 464-470, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28320187

ABSTRACT

OBJECTIVE: To evaluate effects of 5 sessions of transcranial direct current stimulation (tDCS) over the left dorsolateral prefrontal cortex in patients with prolonged disorders of consciousness (DOC). METHODS: Seven patients in vegetative state (VS) and 6 in minimally conscious state (MCS), at ≥3months after brain injury, were randomized into two groups: group 1 received one week of active tDCS and 1week of sham stimulation, separated by 1 resting week; group 2 received active and sham stimulation in reverse order. We performed clinical and EEG evaluations before and after the first stimulation session, two hours after the last weekly stimulation, twice during the resting week, and during a 3-month follow-up. RESULTS: We observed small changes of patients' conditions after the first tDCS session and immediately after the 5 active stimulations. Substantial clinical and EEG changes were observed in 5/13 patients (3 in MCS and 2 in VS) starting after entire (active and sham) stimulation protocol and further progressing during the next months. No baseline features distinguished patients who improved from patients who did not improve. CONCLUSIONS: Repeated tDCS did not exert remarkable short-term clinical and EEG effects in patients with prolonged DOC. Further studies should ascertain whether tDCS might promote clinical recovery in the long-term period.


Subject(s)
Consciousness Disorders/therapy , Transcranial Direct Current Stimulation/methods , Adult , Aged , Aged, 80 and over , Brain Injuries/complications , Consciousness Disorders/etiology , Cross-Over Studies , Double-Blind Method , Electroencephalography , Female , Humans , Male , Middle Aged , Severity of Illness Index , Treatment Outcome
18.
J Immunol ; 189(12): 5622-31, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23144496

ABSTRACT

Adoptive T cell therapy uses the specificity of the adaptive immune system to target cancer and virally infected cells. Yet the mechanism and means by which to enhance T cell function are incompletely described, especially in the skin. In this study, we use a murine model of immunotherapy to optimize cell-mediated immunity in the skin. We show that in vitro-derived central but not effector memory-like T cells bring about rapid regression of skin-expressing cognate Ag as a transgene in keratinocytes. Local inflammation induced by the TLR7 receptor agonist imiquimod subtly yet reproducibly decreases time to skin graft rejection elicited by central but not effector memory T cells in an immunodeficient mouse model. Local CCL4, a chemokine liberated by TLR7 agonism, similarly enhances central memory T cell function. In this model, IL-2 facilitates the development in vivo of effector function from central memory but not effector memory T cells. In a model of T cell tolerogenesis, we further show that adoptively transferred central but not effector memory T cells can give rise to successful cutaneous immunity, which is dependent on a local inflammatory cue in the target tissue at the time of adoptive T cell transfer. Thus, adoptive T cell therapy efficacy can be enhanced if CD8(+) T cells with a central memory T cell phenotype are transferred, and IL-2 is present with contemporaneous local inflammation.


Subject(s)
Immunologic Memory/immunology , Skin/immunology , Skin/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Adoptive Transfer/methods , Animals , Cell Communication/immunology , Cells, Cultured , Epidermal Cells , Epidermis/immunology , Epidermis/transplantation , Immune Tolerance , Immunity, Cellular , Inflammation/immunology , Inflammation/surgery , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Skin/cytology , Skin Transplantation/immunology , Skin Transplantation/methods , Skin Transplantation/pathology , T-Lymphocyte Subsets/transplantation
19.
Dev Dyn ; 238(12): 3175-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19877269

ABSTRACT

Pitrm1 is a zinc metalloendopeptidase that has been implicated in Alzheimer's disease and mitochondrial peptide degradation, but to date no major role in embryonic development has been documented. In a screen for genes regulated by hedgehog signaling in the mouse limb, we showed that expression of Pitrm1 is upregulated in response to loss of the Gli3 transcription factor. Here we confirm spatial changes in Pitrm1 expression in the Gli3 mutant mouse limb and examine Pitrm1 expression in Shh null and Ptch1 conditional deletion mouse mutants. In wild-type mice, Pitrm1 is expressed in a number of developing tissues known to be patterned by Sonic hedgehog, including the limbs, face, cortex, hippocampus, cerebellum, tectum, sub-mandibular gland, lung, genital tubercle, hair follicles, and the enamel knot of the teeth. Additionally, Pitrm1 is expressed in Pax3-expressing myoblast progenitors in the limb, the dermomyotome, and developing muscles of the face and torso.


Subject(s)
Extremities/embryology , Hedgehog Proteins/physiology , Metalloendopeptidases/genetics , Muscle, Skeletal/embryology , Stem Cells/metabolism , Animals , Embryo, Mammalian , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Metalloendopeptidases/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Signal Transduction/genetics , Tissue Distribution
20.
Mech Dev ; 122(11): 1218-33, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16169709

ABSTRACT

From early in limb development the transcription factor Gli3 acts to define boundaries of gene expression along the anterior-posterior (AP) axis, establishing asymmetric patterns required to provide positional information. As limb development proceeds, posterior mesenchyme expression of Sonic hedgehog (Shh) regulates Gli3 transcription and post-translational processing to specify digit number and identity. The molecular cascades dependent on Gli3 at later stages of limb development, which link early patterning events with final digit morphogenesis, remain poorly characterised. By analysing the transcriptional consequences of loss of Gli3 in the anterior margin of the E11.5 and E12.5 limb bud in the polydactylous mouse mutant extra-toes (Gli3(Xt/Xt)), we have identified a number of known and novel transcripts dependent on Gli3 in the limb. In particular, we demonstrated that the genes encoding the paired box transcription factor Pax9, the Notch ligand Jagged1 and the cell surface receptor Cdo are dependent on Gli3 for correct expression in the anterior limb mesenchyme. Analysis of expression in compound Shh;Gli3 mutant mouse embryos and in both in vitro and in vivo Shh signaling assays, further defined the importance of Shh regulated processing of Gli3 in controlling gene expression. In particular Pax9 regulation by Shh and Gli3 was shown to be context dependent, with major differences between the limb and somite revealed by Shh bead implantation experiments in the chick. Jagged1 was shown to be induced by Shh in the chick limb and in a C3H10T1/2 cell based signaling assay, with Shh;Gli3 mutant analysis indicating that expression is dependent on Gli3 derepression. Our data have also revealed that perturbation of early patterning events within the Gli3(Xt/Xt) limb culminates in a specific delay of anterior chondrogenesis which is subsequently realised as extra digits.


Subject(s)
Calcium-Binding Proteins/physiology , Extremities/embryology , Intercellular Signaling Peptides and Proteins/physiology , Kruppel-Like Transcription Factors/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Paired Box Transcription Factors/metabolism , Animals , Biomarkers , Body Patterning/physiology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Line , Chick Embryo , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , PAX9 Transcription Factor , Paired Box Transcription Factors/antagonists & inhibitors , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/physiology , Serrate-Jagged Proteins , Zinc Finger Protein Gli3
SELECTION OF CITATIONS
SEARCH DETAIL
...