Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Food Security and Safety Volume 2: African Perspectives ; 2:265-282, 2022.
Article in English | Scopus | ID: covidwho-20240804

ABSTRACT

Africa ranks second in the number of undernourished people globally and has the highest prevalence of food insecurity, twice the world's average. The continent could not meet the past Millennium Development Goals and targets for 2015, and the current projection shows that Africa is not on track to achieve Sustainable Development Goal 2 of Zero Hunger by 2030. Prospects for achieving these goals are dismal because of inherent primary drivers of food insecurity in each African region. This chapter identifies the primary drivers of food and nutrition insecurity in Africa and suggests strategies to attenuate its effect. Climate shocks (drought and flood) and insecurity are the primary agents driving food insecurity in Western, Central, and Southern Africa. Migratory pests (desert locusts) are a big challenge in Eastern and Southern Africa that have destroyed thousands of farmlands, while dependence on food subsidies, climate change, and political instability are the primary drivers of food insecurity in North Africa. In summary, the prevalence of food insecurity in Africa differs owing to the influence of food insecurity drivers in each region. Consequently, the COVID-19 widespread is expected to exacerbate Africa's current food insecurity. Sustainable strategies such as investing in the agricultural system through sustainable policies;reducing food prices;preventing localized desert locust outbreaks from attaining plague proportions and counterinsurgency;managing climate;and investing in food assistance in severe, catastrophic food insecurity that best fits each region would play a key role in mitigating food and nutrition insecurity in Africa. © The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland AG 2023.

2.
Transplantation and Cellular Therapy ; 29(2 Supplement):S379-S380, 2023.
Article in English | EMBASE | ID: covidwho-2317836

ABSTRACT

Background: The ZUMA-1 safety management Cohort 6 (N=40), which evaluated whether prophylactic corticosteroids and earlier corticosteroids and/or tocilizumab could improve safety outcomes, demonstrated an improved safety profile (no Grade >=3 cytokine release syndrome [CRS];15% Grade >=3 neurologic events [NEs]) vs pivotal Cohorts 1+2, without compromising response rate or durability (95% ORR, 80% CR rate, and 53% ongoing response rate with >=1 y of follow-up;Oluwole, et al. ASH 2021. 2832). Here, 2-y updated outcomes are reported. Method(s): Eligible pts with R/R LBCL underwent leukapheresis (followed by optional bridging therapy) and conditioning chemotherapy, then a single axi-cel infusion. Pts received corticosteroid prophylaxis (once-daily oral dexamethasone 10 mg on Days 0 [before axi-cel], 1, and 2) and earlier corticosteroids and/or tocilizumab for CRS and NE management vs Cohorts 1+2 (Oluwole, et al. Br J Haematol. 2021). The primary endpoints were incidence and severity of CRS and NEs. Secondary endpoints included ORR (investigator-assessed), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and chimeric antigen receptor (CAR) T-cell levels in blood. Result(s): As of December 16, 2021, the median follow-up time for the 40 treated pts was 26.9 mo. Since the 1-y analysis, no new CRS events were reported (no pts had Grade >=3 CRS to date). The incidence of Grade >=3 NEs increased from 15% to 18%between the 1-y and 2-y analyses. Two new NEs occurred in 2 pts: 1 pt had Grade 2 dementia (onset on Day 685 and ongoing at time of data cutoff;not related to axi-cel) and 1 had Grade 5 axi-cel-related leukoencephalopathy. Since the 1-y analysis, 6 new infections were reported (Grades 1, 2, and 5 COVID-19 [n=1 each], Grade 3 Pneumocystis jirovecii pneumonia [n=1], Grade 3 unknown infectious episode with inflammatory syndrome [n=1], and Grade 2 herpes zoster [n=1]). In total, 8 deaths occurred since the 1-y analysis (progressive disease [n=5], leukoencephalopathy [n=1], and COVID-19 [n=2]). The ORR was 95% (80% CR), which was unchanged from the 1-y analysis. Median DOR and PFS were since reached (25.9 mo [95% CI, 7.8-not estimable] and 26.8 mo [95% CI, 8.7-not estimable], respectively). Median OS was still not reached. Kaplan- Meier estimates of the 2-y DOR, PFS, and OS rates were 53%, 53%, and 62%, respectively. Of 18 pts (45%) in ongoing response at data cutoff, all achieved CR as the best response. By Month 24, 14/20 pts with evaluable samples (70%) had detectable CAR T cells (vs 23/36 pts [64%] in Cohorts 1+2). Conclusion(s): With 2 y of follow-up, the ZUMA-1 Cohort 6 toxicity management strategy continued to demonstrate an improved long-term safety profile of axi-cel in pts with R/R LBCL. Further, responses remained high, durable, and similar to those observed in Cohorts 1+2 (Locke, et al. Lancet Oncol. 2019).Copyright © 2023 American Society for Transplantation and Cellular Therapy

3.
West Afr J Med ; 40(4): 435-437, 2023 Apr 28.
Article in English | MEDLINE | ID: covidwho-2301220

ABSTRACT

BACKGROUND: Endocrine diseases are ubiquitous. In our environment, diabetes mellitus (DM), obesity and thyroid disorders represent the most common examples. Diabetes mellitus is a global health problem with a myriad of complications. We sought to evaluate outcome in terms of fatality in those with common endocrine diseases who were infected with COVID-19. AIMS AND OBJECTIVES: To determine outcome in terms of mortality in patients with common endocrine diseases who contracted COVID-19. MATERIALS AND METHODS: We conducted an observational, descriptive, cross-sectional study with 120 participants drawn from the endocrinology/DM clinic at the Lagos University Teaching Hospital and Serenity Hospital, Surulere (a private medical clinic). Data collected included age, gender, type of endocrine disease, comorbid diseases, and COVID-19 status. Through charts from the medical records department, outcome of participants in terms of mortality was determined. RESULTS: Data of 120 subjects were analyzed. There were 61males and 59 females, yielding a male:female ratio of 1:1. Mean age was 58 years and the mode was 46 years. Over half (88) of the patients had diabetes mellitus, 22 had obesity, and 17 had thyroid disorders. The case fatality rate of patients with endocrine diseases who had COVID-19 was 11%, with about 85% of these deaths occurring in the elderly (those aged above 60 years). Ninety-two percent of the patients who died had type 2 DM. Approximately 80% of patients who were infected with COVID-19 had at least one co-morbid disease. CONCLUSION: Older age, type 2 diabetes mellitus, and the presence of at least one comorbidity were associated with increased mortality in patients with endocrine diseases who were infected with COVID-19 in our study.


CONTEXTE: Les maladies endocriniennes sont omniprésentes. Dans notre environnement, le diabète sucré, l'obésité et les troubles thyroïdiens en sont les exemples les plus courants. Le diabète est un problème de santé mondial qui s'accompagne d'une myriade de complications. Nous avons cherché à évaluer l'issue en termes de mortalité chez les personnes atteintes de maladies endocriniennes courantes qui ont été infectées par COVID-19. BUTS ET OBJECTIFS: Déterminer l'issue en termes de mortalité chez les patients atteints de maladies endocriniennes courantes qui ont contracté COVID 19. MATÉRIEL ET MÉTHODOLOGIES: Nous avons mené une étude observationnelle, descriptive et transversale auprès de 120 participants provenant de la clinique d'endocrinologie/DM de l'hôpital universitaire de Lagos et de l'hôpital Serenity, Surulere (clinique médicale privée). Les données recueillies comprenaient l'âge, le sexe, le type de maladie endocrinienne, les maladies concomitantes et le statut COVID-19. Les résultats des participants en termes de mortalité ont été déterminés à partir des dossiers médicaux. RÉSULTATS: Les données de 120 sujets ont été analysées. Il y avait 61 hommes et 59 femmes, avec un ratio homme/femme de 1:1. L'âge moyen était de 58 ans, le mode de 46 ans. Plus de la moitié [88] des patients souffraient de diabète sucré. 22 patients souffraient d'obésité et 17 de troubles thyroïdiens. Le taux de létalité des patients souffrant de maladiesendocriniennes et atteints de COVID-19 était de 11 %, 85 % de ces décès survenant chez des personnes âgées, c'est-à-dire de plus de 60 ans. 92 % des patients décédés souffraient de diabète de type 2. Environ 80 % des patients infectés par COVID-19 présentaient au moins une maladie concomitante. CONCLUSION: L'âge avancé, le diabète de type 2, la présence d'au moins une comorbidité sont associés à une mortalité accrue chez les patients atteints de maladies endocriniennes et infectés par COVID-19 dans notre étude. Mots-clés: Maladies endocriniennes, COVID-19, comorbidités, syndrome métabolique.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Aged , Humans , Male , Female , Middle Aged , Diabetes Mellitus, Type 2/epidemiology , Cross-Sectional Studies , Nigeria/epidemiology , COVID-19/epidemiology , Obesity/epidemiology
4.
Paediatrics and Child Health (Canada) ; 27(Supplement 3):e3, 2022.
Article in English | EMBASE | ID: covidwho-2190135

ABSTRACT

BACKGROUND: The COVID-19 pandemic has had marked effects on mental health, including in pediatric populations. Pediatric patients have faced mental health concerns at increased rates including anxiety and depression. Furthermore, patients with eating disorders represent a vulnerable group who have been negatively impacted as well, as a result of lack of support, loss of in-person follow-up and increased relapse. In our centre, and nationally, clinicians have noted a trend towards increased eating disorder referrals and increased hospitalizations during the pandemic. OBJECTIVE(S): The objective of this study was to determine the incidence, severity and triggers for eating disorders in the adolescent population during the COVID-19 pandemic and how it compares to the year prior. As well, the subset of patients who were hospitalized for medical stabilization were further analyzed to determine severity of illness. DESIGN/METHODS: A retrospective chart review compared the first year of the COVID-19 pandemic (March 2020-March 2021), to the previous 12 months. Inclusion criteria included referrals to an eating disorder clinic and inpatient admissions to pediatrics or mental health services during the specified time frame. Data collected included age of onset, triggers, comorbid mental health conditions, and weight measures. Among hospitalized patients, orthostatic vital changes, need for NG feeds, length of medical stabilization and length of mental health hospitalization were included. RESULT(S): Overall, 76 patients were included in the study. 44 (57.9%) were referred after COVID, which was significantly increased from the prior year (p=0.05). On average, patients presented at a younger age (14.2 +/- 2.3 vs. 14.9 +/- 1.9;p=0.08). Pre-COVID, approximately 44% of referrals were from family physicians and 19% from pediatrics. During COVID, approximately 39% were from family doctors and 25% from pediatricians. There was an increase in the number of patients requiring hospitalization for treatment (16 vs. 3), with 50% of the post-COVID admissions being direct from the ED Clinic on initial assessment. The reason for hospitalization was unstable vitals/ bradycardia in 68.7% of admissions;self-harm comprised the majority of the other admissions. CONCLUSION(S): Our results support national and international reports that eating disorder incidence has increased during COVID-19. Patients described loss of routine, anxiety, and isolation as triggers related to the pandemic. Disruptions to daily life including school, sports, recreation, and relationships had profound effects on the mental health of children. The effect of social media on body image has also contributed. It is important for clinicians to screen for mental health conditions, including eating disorders at all available opportunities. Furthermore, this study demonstrates the need for increased services at our centre. Limitations for this study include that it is a single-centre study with a relatively small patient population. As well, it does not capture patients who may have been referred only to psychiatry.

5.
Blood ; 138(SUPPL 1):3826, 2021.
Article in English | EMBASE | ID: covidwho-1770242

ABSTRACT

Introduction: Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 Chimeric Antigen Receptor (CAR) T-cell therapy that induces durable responses in patients with relapsed or refractory large B-cell lymphoma. At a median of 27.1 months follow-up on the ZUMA-1 trial, median overall survival (OS) was 25.8 months with 39% progression free survival (PFS) at 2 years post-infusion (Locke, Lancet Onc 2019). We previously reported outcomes of axi-cel patients treated with standard of care therapy at a median follow up of 12.9 months, including 42% who did not meet eligibility criteria for ZUMA-1 based on co-morbidities (Nastoupil, JCO 2020). Here we report results from this cohort at a median follow up of 32.4 months, as well as late outcomes of interest including cytopenias, infections and secondary malignancies. Methods and Results: The US Lymphoma CAR-T Consortium comprised of 17 US academic centers who contributed data independent of the manufacturer. Two hundred and ninety-eight patients underwent leukapheresis with intent to manufacture standard of care axi-cel as of September 30, 2018. In infused patients (n=275), OS and PFS were calculated from date of infusion. After median follow-up of 32.4 months (95% CI 31.1 - 34.3), median OS was not reached (95% CI 25.6 - not evaluable) (Figure 1A) with 1-, 2- and 3-year OS of 68.5% (95% CI 62.6-73.7), 56.4% (95% CI 50.1-62.2) and 52.2% (95% CI 45.7-58.2%), respectively. Median PFS was 9 months (95% CI 5.9-19.6) (Figure 1B);1-, 2- and 3-year PFS was 47.4% (95% CI 41.4-53.2), 41.6% (95% CI 35.6-47.5) and 37.3% (95% CI 31.3-43.2), respectively. Twenty-seven PFS events occurred at or after 1 year post infusion;19 events were progressive lymphoma, with the latest relapse observed 28 months after axi-cel infusion. Eight patients died while in remission from their lymphoma: 4 from secondary malignancy, 3 from infection, and 1 from unknown causes. Results of multivariable modeling were similar to our prior analysis: factors associated with both a shorter PFS and shorter OS included male sex, elevated pre-lymphodepletion LDH, and poor ECOG status. Complete blood count and B- and T-cell recovery data were collected at 1 and 2-years post-infusion, excluding patients who had relapsed or been treated for secondary malignancy at time of collection (Table 1). Rates of neutropenia (absolute neutrophil count ≤1000) at 1- and 2- years were 9.2% (10/109) and 11.2% (9/80) and rates of CD4 count ≤200/ul were 62% (23/37) and 27% (7/26). Recovery of B cells was seen in 54% (15/28) and 57% (13/23) at 1-and 2-years post infusion. Infections were reported in 31.2% (34/109) patients between 6- and 12-months post infusion, and 17% (18/109) were severe, requiring either hospitalization and/or IV antibiotics. Twenty-one patients (24%, 21/89) had an infection between 1- and 2- years, 11% of which were severe. Twenty percent (10/49) of patients between 2- and 3-years had an infection and 4 (8%) were severe. Neutropenia, low CD4 counts, and IgG levels were not associated with infection, though patients with infection between 6-12 months were more likely to have received IVIG (p<0.001). No patient in this cohort died of COVID-19. Twenty-two of 275 (8%) patients were diagnosed with subsequent malignancy after axi-cel treatment: 14/275 (5%) patients were diagnosed with myeloid malignancies (MDS (n=12), AML (n=1), CMML (n=1));other malignancies included squamous cell carcinoma of skin (n=3);sarcoma (n=1);endometrial (n=1);lung (n=1);mesothelioma (n=1) and AITL (n=1). Patients with myeloid malignancy had a median age of 62 at axi-cel apheresis (IQR 56-67), 64% were male and median lines of prior therapy was 4 (IQR 3-6), including 36% with a prior autologous stem cell transplant. Eleven patients were in remission from lymphoma at myeloid malignancy diagnosis, while 3 were diagnosed after progression and interval therapy. Conclusion: This multi-center retrospective study showed similar long-term results to the ZUMA-1 trial, despite including patients who did not meet ZUMA-1 eligibility criteria ba ed on comorbidities. Sixteen percent of PFS events were seen after 1 year, largely due to disease progression. Late infection was common but was not explained by persistent neutropenia or low CD4 counts. Subsequent malignancy, including MDS, occurred in 8% of patients and require further study to better identify patients at risk. (Figure Presented).

6.
Blood ; 138:739, 2021.
Article in English | EMBASE | ID: covidwho-1582364

ABSTRACT

Background: High-risk LBCL is associated with poor prognosis after first-line anti-CD20 mAb-containing regimens, highlighting the need for novel treatments. Axi-cel, an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, is approved for treatment of relapsed/refractory (R/R) LBCL after ≥2 lines of systemic therapy. Here we report the primary analysis of ZUMA-12, a Phase 2, multicenter, single-arm study of axi-cel as part of first-line therapy in patients with high-risk LBCL. Methods: Eligible adults had high-risk LBCL, defined by histology (double- or triple-hit status [MYC and BCL2 and/or BCL6 translocations] per investigator) or an IPI score ≥3, plus a positive interim PET per Lugano Classification (Deauville score [DS] 4/5) after 2 cycles of an anti-CD20 mAb and anthracycline-containing regimen. Patients underwent leukapheresis and received conditioning chemotherapy (cyclophosphamide and fludarabine) followed by a single axi-cel infusion at 2×10 6 CAR T cells/kg. Non-chemotherapy bridging could be administered before conditioning per investigator discretion. The primary endpoint was investigator-assessed complete response (CR) rate per Lugano. Secondary endpoints included objective response rate (ORR;CR + partial response), duration of response (DOR), event-free survival (EFS), progression-free survival (PFS), overall survival (OS), incidence of adverse events (AEs), and levels of CAR T cells in blood and cytokines in serum. The primary analysis occurred after all treated patients had ≥6 months of follow-up. Results: As of May 17, 2021, 42 patients were enrolled and 40 were treated with axi-cel. Median age was 61 years (range, 23-86);68% of patients were male, 63% had ECOG 1, 95% had stage III/IV disease, 48% had DS4, 53% had DS5, 25% had double- or triple-hit status per central assessment, and 78% had IPI score ≥3. A total of 37 patients had centrally confirmed double- or triple-hit histology or an IPI score ≥3 and were evaluable for response, with 15.9 months of median follow-up (range, 6.0-26.7). The CR rate was 78% (n=29;95% CI, 62-90);89% of patients had an objective response, and median time to initial response was 1 month. Among all 40 treated patients, 90% had an objective response (80% CR rate). At data cutoff, 73% of response-evaluable patients had ongoing responses. Medians for DOR, EFS, and PFS were not reached;12-month estimates were 81%, 73%, and 75%, respectively. The estimated OS at 12 months was 91%. All 40 treated patients had AEs of any grade;85% of patients had Grade ≥3 AEs, most commonly cytopenias (68%). Grade ≥3 cytokine release syndrome (CRS) and neurologic events (NEs) occurred in 3 patients (8%) and 9 patients (23%), respectively. Median times to onset of CRS and NEs were 4 days (range, 1-10) and 9 days (range, 2-44), with median durations of 6 days and 7 days, respectively. All CRS and most NEs (28/29) of any grade resolved by data cutoff (1 ongoing Grade 1 tremor);39/40 CRS events resolved by 14 days post-infusion and 19/29 NEs resolved by 21 days post-infusion. Tocilizumab was administered to 63% and 3% of patients for management of CRS or NEs, respectively;corticosteroids were administered to 35% and 33% of patients for CRS and NE management. One Grade 5 event of COVID-19 occurred (Day 350). Median peak CAR T-cell level in all treated patients was 36 cells/µL (range, 7-560), and median expansion by AUC 0-28 was 495 cells/µL × days (range, 74-4288). CAR T-cell levels peaked at a median of 8 days post-infusion (range, 8-37). Higher frequency of CCR7+CD45RA+ T cells in axi-cel product, previously associated with greater expansion of CAR T cells (Locke et al. Blood Adv. 2020), was observed in ZUMA-12, compared with the ZUMA-1 study in R/R LBCL (Neelapu et al. New Engl J Med. 2017). Conclusion: In the primary analysis of ZUMA-12, axi-cel demonstrated a high rate of rapid and complete responses in patients with high-risk LBCL, a population with high unmet need. With 15.9 months of median follow-up, responses were durable as medians for DOR, EFS, nd PFS were not yet reached and over 70% of patients remained in response at data cutoff. No new safety signals were reported with axi-cel in an earlier line. Overall, axi-cel may benefit patients exposed to fewer prior therapies, and further trials in first-line high-risk LBCL are warranted to assess axi-cel in this setting. [Formula presented] Disclosures: Neelapu: Kite, a Gilead Company, Merck, Bristol Myers Squibb, Novartis, Celgene, Pfizer, Allogene, Kuur, Incyte, Precision BioSciences, Legend, Adicet Bio, Calibr, and Unum Therapeutics: Other: personal fees;Kite, a Gilead Company, Bristol Myers Squibb, Merck, Poseida, Cellectis, Celgene, Karus Therapeutics, Unum Therapeutics (Cogent Biosciences), Allogene, Precision BioSciences, Acerta and Adicet Bio: Research Funding;Takeda Pharmaceuticals and related to cell therapy: Patents & Royalties;Kite, a Gilead Company, Merck, Bristol Myers Squibb, Novartis, Celgene, Pfizer, Allogene Therapeutics, Cell Medica/Kuur, Incyte, Precision Biosciences, Legend Biotech, Adicet Bio, Calibr, Unum Therapeutics and Bluebird Bio: Honoraria. Dickinson: Janssen: Consultancy, Honoraria;Takeda: Research Funding;Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau;Amgen: Honoraria;Celgene: Research Funding;Bristol-Myers Squibb: Consultancy, Honoraria;MSD: Consultancy, Honoraria, Research Funding, Speakers Bureau;Roche: Consultancy, Honoraria, Other: travel, accommodation, expenses, Research Funding, Speakers Bureau;Gilead Sciences: Consultancy, Honoraria, Speakers Bureau. Munoz: Kite, a Gilead Company, Kyowa, Bayer, Pharmacyclics/Janssen, Seagen, Acrotech/Aurobindo, Beigene, Verastem, AstraZeneca, Celgene/BMS, Genentech/Roche.: Speakers Bureau;Bayer, Gilead/Kite Pharma, Celgene, Merck, Portola, Incyte, Genentech, Pharmacyclics, Seattle Genetics, Janssen, and Millennium: Research Funding;Pharmacyclics/Abbvie, Bayer, Kite, a Gilead Company, Pfizer, Janssen, Juno/Celgene, Bristol Myers Squibb, Kyowa Kirin, Alexion, Fosun Kite, Innovent, Seagen, BeiGene, Debiopharm, Epizyme, Karyopharm, ADC Therapeutics, Servier, and Genmab: Consultancy, Other: advisory role;Alexion, AstraZeneca Rare Disease: Other: Study investigator;Targeted Oncology, OncView, Kyowa Kirin, Physicians' Education Resource, and Seagen: Honoraria. Thieblemont: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Gilead Sciences: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees;Kyte: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Bristol Myers Squibb/Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees;Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses, Research Funding;Incyte: Honoraria, Membership on an entity's Board of Directors or advisory committees;Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Cellectis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses;Hospira: Research Funding;Bayer: Honoraria;Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses. Oluwole: Pfizer: Consultancy;Curio Science: Consultancy;Janssen: Consultancy;Kite, a Gilead Company: Consultancy, Research Funding. Herrera: Takeda: Consultancy;Genentech: Consultancy, Research Funding;Merck: Consultancy, Research Funding;Seagen: Consultancy, Research Fundi g;AstraZeneca: Consultancy, Research Funding;Kite, a Gilead Company: Research Funding;Gilead Sciences: Research Funding;Tubulis: Consultancy;ADC Therapeutics: Consultancy, Research Funding;Bristol Myers Squibb: Consultancy, Research Funding;Karyopharm: Consultancy. Ujjani: Loxo: Research Funding;AstraZeneca: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding;Epizyme: Consultancy, Membership on an entity's Board of Directors or advisory committees;Janssen: Consultancy;TG Therapeutics: Honoraria;Gilead: Honoraria;ACDT: Honoraria;Kite, a Gilead Company: Honoraria;Adaptive Biotechnologies: Research Funding;Atara Bio: Consultancy;AbbVie: Consultancy, Research Funding;Pharmacyclics: Consultancy, Research Funding. Lin: Sorrento: Consultancy;Legend: Consultancy;Novartis: Consultancy;Bluebird Bio: Consultancy, Research Funding;Gamida Cell: Consultancy;Janssen: Consultancy, Research Funding;Celgene: Consultancy, Research Funding;Juno: Consultancy;Vineti: Consultancy;Takeda: Research Funding;Merck: Research Funding;Kite, a Gilead Company: Consultancy, Research Funding. Riedell: Bayer: Honoraria;Karyopharm Therapeutics: Consultancy, Honoraria;Morphosys: Research Funding;Celgene/Bristol-Myers Squibb Company: Consultancy, Honoraria, Research Funding;Verastem Oncology: Honoraria;Kite, a Gilead Company: Honoraria, Research Funding, Speakers Bureau;Novartis: Consultancy, Honoraria, Research Funding;Takeda: Consultancy;BeiGene: Consultancy;Calibr: Research Funding;Xencor: Research Funding;Tessa Therapeutics: Research Funding. Kekre: Gilead: Consultancy, Honoraria;Novartis: Consultancy, Honoraria;Celgene: Consultancy, Honoraria. Lui: Gilead Sciences: Other: stock or other ownership;Kite, a Gilead Company: Current Employment, Other: travel support. Milletti: Kite, aGilead company: Current Employment;Gilead Sciences: Other: stock or other ownership. Dong: Kite, a Gilead Company: Current Employment;Gilead Sciences: Other: stock or other ownership;GliaCure/Tufts: Consultancy, Other: advisory role, Patents & Royalties. Xu: Kite, A Gilead Company: Current Employment;Gilead Sciences: Other: stock or other ownership. Chavez: MorphoSys, Bayer, Karyopharm, Kite, a Gilead Company, Novartis, Janssen, AbbVie, TeneoBio, and Pfizer: Consultancy;ADC Therapeutics: Consultancy, Research Funding;Merk: Research Funding;AstraZeneca: Research Funding;MorphoSys, AstraZeneca, BeiGene, Genentech, Kite, a Gilead Company, and Epizyme: Speakers Bureau;BMS: Speakers Bureau.

SELECTION OF CITATIONS
SEARCH DETAIL