37 results on '"Hurwitz ME"'
Search Results
2. Cost trends of metastatic renal cell carcinoma therapy: the impact of oral anticancer agents and immunotherapy.
- Author
-
Forman R, Long JB, Westvold SJ, Agnish K, Mcmanus HD, Leapman MS, Hurwitz ME, Spees LP, Wheeler SB, Gross CP, and Dinan MA
- Subjects
- Humans, Male, Female, Retrospective Studies, United States, Aged, Administration, Oral, Aged, 80 and over, Medication Adherence statistics & numerical data, Fee-for-Service Plans, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell economics, Carcinoma, Renal Cell therapy, Kidney Neoplasms drug therapy, Kidney Neoplasms pathology, Kidney Neoplasms economics, Medicare economics, Immunotherapy economics, Antineoplastic Agents economics, Antineoplastic Agents therapeutic use, Antineoplastic Agents administration & dosage, Health Expenditures statistics & numerical data
- Abstract
Background: Immunotherapy (IO) and oral anticancer agents (OAA) have improved outcomes for metastatic renal cell carcinoma (mRCC), but there is a need to understand real-world costs from the perspective of payers and patients., Methods: We used retrospective fee-for-service Medicare 100% claims data to study patients diagnosed with mRCC in 2015-2019. We identified initial treatment type and costs (the year after diagnosis) and analyzed differences in monthly and 12-month costs over time and between OAA, IO, and combination groups and the association between Out-Of-Pocket (OOP) costs and adherence., Results: We identified 15 407 patients with mRCC (61% male; 85% non-Hispanic White). A total of 6196 received OAA, IO, or combination OAA/IO as initial treatment. OAA use decreased (from 31% to 11%) with a simultaneous rise in patients receiving IO (3% to 26%) or combination IO/OAA therapy (1% to 11%). Medicare payments for all patients with mRCC increased by 41%, from $60 320 (95% confidence interval = 58 260 to 62 380) in 2015 to $85 130 (95% confidence interval = 82 630 to 87 630) in 2019. Payments increased in patients who received OAA, IO, or combination OAA/IO but were stable in those with other/no treatment. Initial higher OOP responsibility ($200-$1000) was associated with 13% decrease in percent days covered in patients receiving OAA in the first 90 days of treatment, compared with those whose OOP responsibility was less than $200., Conclusion: From 2015 to 2019, costs for Medicare patients with mRCC rose substantially due to more patients receiving IO or IO/OAA combined therapy and increases in costs among those receiving those therapies. Increased OOP costs was associated with decreased adherence., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
3. CD70-Targeted Allogeneic CAR T-Cell Therapy for Advanced Clear Cell Renal Cell Carcinoma.
- Author
-
Pal SK, Tran B, Haanen JBAG, Hurwitz ME, Sacher A, Tannir NM, Budde LE, Harrison SJ, Klobuch S, Patel SS, Meza L, Dequeant ML, Ma A, He QA, Williams LM, Keegan A, Gurary EB, Dar H, Karnik S, Guo C, Heath H, Yuen RR, Morrow PK, Agarwal N, and Srour SA
- Subjects
- Humans, Animals, Mice, Female, Male, Middle Aged, Receptors, Chimeric Antigen immunology, Aged, Xenograft Model Antitumor Assays, Cell Line, Tumor, Adult, Carcinoma, Renal Cell therapy, Carcinoma, Renal Cell immunology, Kidney Neoplasms therapy, Kidney Neoplasms immunology, Immunotherapy, Adoptive methods, CD27 Ligand
- Abstract
Therapeutic approaches for clear cell renal cell carcinoma (ccRCC) remain limited; however, chimeric antigen receptor (CAR) T-cell therapies may offer novel treatment options. CTX130, an allogeneic CD70-targeting CAR T-cell product, was developed for the treatment of advanced or refractory ccRCC. We report that CTX130 showed favorable preclinical proliferation and cytotoxicity profiles and completely regressed RCC xenograft tumors. We also report results from 16 patients with relapsed/refractory ccRCC who received CTX130 in a phase I, multicenter, first-in-human clinical trial. No patients encountered dose-limiting toxicity, and disease control was achieved in 81.3% of patients. One patient remains in a durable complete response at 3 years. Finally, we report on a next-generation CAR T construct, CTX131, in which synergistic potency edits to CTX130 confer improved expansion and efficacy in preclinical studies. These data represent a proof of concept for the treatment of ccRCC and other CD70+ malignancies with CD70- targeted allogeneic CAR T cells. Significance: Although the role of CAR T cells is well established in hematologic malignancies, the clinical experience in solid tumors has been disappointing. This clinical trial demonstrates the first complete response in a patient with RCC, reinforcing the potential benefit of CAR T cells in the treatment of solid tumors., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
4. Providing 0.1 Full-Time Equivalent (FTE) Support to Fellowship Core Faculty Improves Faculty Involvement in Fellowship Education and Recruitment.
- Author
-
Butt A, Christian J, Kress A, Lu BY, Hurwitz ME, Goldberg SB, Podoltsev NA, Gilkes L, and Lee AI
- Subjects
- Humans, Surveys and Questionnaires, Salaries and Fringe Benefits, Job Satisfaction, Medical Oncology education, Education, Medical, Graduate, Mentors, Hematology education, Personnel Selection, Female, Male, Fellowships and Scholarships, Faculty, Medical
- Abstract
In 2022, the American Council for Graduate Medical Education (ACGME) recommended that core faculty (CF) in medical subspecialty fellowships receive at least 0.1 full-time equivalent (FTE) salary support, with plans to enforce compliance in July 2023. After early feedback raised concerns about potential unintended consequences, ACGME deferred enforcement to July 2024. Hence, there is an urgent need to understand the ramifications of providing FTE support for CF. In 2020, the Yale hematology and medical oncology (HO) fellowship program began providing 0.1 FTE support to all CF. Perceptions regarding this were assessed via surveys distributed to all CF in 2021 and 2022 and to all HO fellows in 2021. The vast majority (83.3%) of CF survey respondents reported improved job satisfaction and an increased sense of involvement in the fellowship program as a result of the new 0.1 FTE-supported CF program. Most CF increased attendance at fellowship conferences, devoted more time to mentorship, and increased participation in recruitment. In free text comments, CF respondents described that providing 0.1 FTE support made them "feel rewarded," gave them "a sense of commitment" to the fellowship, and helped "offset clinical requirements." HO fellows reported "a positive impact" of the new program with faculty being "more present at lectures." The median number of times faculty were available to interview fellowship applicants rose markedly after introduction of the program. The FTE-supported CF program was viewed enthusiastically by fellows and faculty, resulting in increased CF involvement in fellowship education and recruitment., (© 2024. The Author(s) under exclusive licence to American Association for Cancer Education.)
- Published
- 2024
- Full Text
- View/download PDF
5. Area Vulnerability and Disparities in Therapy for Patients With Metastatic Renal Cell Carcinoma.
- Author
-
Rahman SN, Long JB, Westvold SJ, Leapman MS, Spees LP, Hurwitz ME, McManus HD, Gross CP, Wheeler SB, and Dinan MA
- Subjects
- Humans, Male, Female, Aged, United States, Retrospective Studies, Aged, 80 and over, Social Vulnerability, Vulnerable Populations statistics & numerical data, Socioeconomic Factors, Carcinoma, Renal Cell therapy, Carcinoma, Renal Cell ethnology, Medicare statistics & numerical data, Kidney Neoplasms therapy, Kidney Neoplasms ethnology, Healthcare Disparities statistics & numerical data, Healthcare Disparities ethnology
- Abstract
Importance: Area-level measures of sociodemographic disadvantage may be associated with racial and ethnic disparities with respect to receipt of treatment for metastatic renal cell carcinoma (mRCC) but have not been investigated previously, to our knowledge., Objective: To assess the association between area-level measures of social vulnerability and racial and ethnic disparities in the treatment of US Medicare beneficiaries with mRCC from 2015 through 2019., Design, Setting, and Participants: This retrospective cohort study included Medicare beneficiaries older than 65 years who were diagnosed with mRCC from January 2015 through December 2019 and were enrolled in fee-for-service Medicare Parts A, B, and D from 1 year before through 1 year after presumed diagnosis or until death. Data were analyzed from November 22, 2022, through January 26, 2024., Exposures: Five different county-level measures of disadvantage and 4 zip code-level measures of vulnerability or deprivation and segregation were used to dichotomize whether an individual resided in the most vulnerable quartile according to each metric. Patient-level factors included age, race and ethnicity, sex, diagnosis year, comorbidities, frailty, Medicare and Medicaid dual enrollment eligibility, and Medicare Part D low-income subsidy (LIS)., Main Outcomes and Measures: The main outcomes were receipt and type of systemic therapy (oral anticancer agent or immunotherapy from 2 months before to 1 year after diagnosis of mRCC) as a function of patient and area-level characteristics. Multivariable regression analyses were used to adjust for patient factors, and odds ratios (ORs) from logistic regression and relative risk ratios (RRRs) from multinomial logistic regression are reported., Results: The sample included 15 407 patients (mean [SD] age, 75.6 [6.8] years), of whom 9360 (60.8%) were men; 6931 (45.0%), older than 75 years; 93 (0.6%), American Indian or Alaska Native; 257 (1.7%), Asian or Pacific Islander; 757 (4.9%), Hispanic; 1017 (6.6%), non-Hispanic Black; 12 966 (84.2%), non-Hispanic White; 121 (0.8%), other; and 196 (1.3%), unknown. Overall, 8317 patients (54.0%) received some type of systemic therapy. After adjusting for individual factors, no county or zip code-level measures of social vulnerability, deprivation, or segregation were associated with disparities in treatment. In contrast, patient-level factors, including female sex (OR, 0.78; 95% CI, 0.73-0.84) and LIS (OR, 0.48; 95% CI, 0.36-0.65), were associated with lack of treatment, with particularly limited access to immunotherapy for patients with LIS (RRR, 0.25; 95% CI, 0.14-0.43). Associations between individual-level factors and treatment in multivariable analysis were not mediated by the addition of area-level metrics. Disparities by race and ethnicity were consistently and only observed within the most vulnerable areas, as indicated by the top quartile of each vulnerability deprivation index., Conclusions and Relevance: In this cohort study of older Medicare patients diagnosed with mRCC, individual-level demographics, including race and ethnicity, sex, and income, were associated with receipt of systemic therapy, whereas area-level measures were not. However, individual-level racial and ethnic disparities were largely limited to socially vulnerable areas, suggesting that efforts to improve racial and ethnic disparities may be most effective when targeted to socially vulnerable areas.
- Published
- 2024
- Full Text
- View/download PDF
6. Personalizing approaches to the management of metastatic hormone sensitive prostate cancer: role of advanced imaging, genetics and therapeutics.
- Author
-
Lokeshwar SD, Choksi AU, Haltstuch D, Rahman SN, Press BH, Syed J, Hurwitz ME, Kim IY, and Leapman MS
- Subjects
- Male, Humans, Docetaxel, Androgen Antagonists therapeutic use, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hormones therapeutic use, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms genetics, Prostatic Neoplasms therapy
- Abstract
Purpose: To summarize contemporary and emerging strategies for the diagnosis and management of metastatic hormone sensitive prostate cancer (mHSPC), focusing on diagnostic testing and therapeutics., Methods: Literature review using PUBMED-Medline databases as well as clinicaltrials.gov to include reported or ongoing clinical trials on treatment for mHSPC. We prioritized the findings from phase III randomized clinical trials, systematic reviews, meta-analyses and clinical practice guidelines., Results: There have been significant changes to the diagnosis and staging evaluation of mHSPC with the integration of increasingly accurate positron emission tomography (PET) imaging tracers that exceed the performance of conventional computerized tomography (CT) and bone scan. Germline multigene testing is recommended for the evaluation of patients newly diagnosed with mHSPC given the prevalence of actionable alterations that may create candidacy for specific therapies. Although androgen deprivation therapy (ADT) remains the backbone of treatment for mHSPC, approaches to first-line treatment include the integration of multiple agents including androgen receptor synthesis inhibitors (ARSI; abiraterone) Androgen Receptor antagonists (enzalutamide, darolutamide, apalautamide), and docetaxel chemotherapy. The combination of ADT, ARSI, and docetaxel chemotherapy has recently been evaluated in a randomized trial and was associated with significantly improved overall survival including in patients with a high burden of disease. The role of local treatment to the prostate with radiation has been evaluated in randomized trials with additional studies underway evaluating the role of cytoreductive radical prostatectomy., Conclusion: The staging and initial management of patients with mHSPC has undergone significant advances in the last decade with advancements in the diagnosis, treatment and sequencing of therapies., (© 2023. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
7. Disparities in immune and targeted therapy utilization for older US patients with metastatic renal cell carcinoma.
- Author
-
Chow RD, Long JB, Hassan S, Wheeler SB, Spees LP, Leapman MS, Hurwitz ME, McManus HD, Gross CP, and Dinan MA
- Subjects
- Humans, Male, Female, Aged, United States epidemiology, Medicare, Ethnicity, White, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy
- Abstract
Disparities in metastatic renal cell carcinoma (mRCC) outcomes persist in the era of oral anticancer agents (OAAs) and immunotherapies (IOs). We examined variation in the utilization of mRCC systemic therapies among US Medicare beneficiaries from 2015 to 2019. Logistic regression models evaluated the association between therapy receipt and demographic covariates including patient race, ethnicity, and sex. In total, 15 407 patients met study criteria. After multivariable adjustment, non-Hispanic Black race and ethnicity was associated with reduced IO (adjusted relative risk ratio [aRRR] = 0.76, 95% confidence interval [CI] = 0.61 to 0.95; P = .015) and OAA receipt (aRRR = 0.76, 95% CI = 0.64 to 0.90; P = .002) compared with non-Hispanic White race and ethnicity. Female sex was associated with reduced IO (aRRR = 0.73, 95% CI = 0.66 to 0.81; P < .001) and OAA receipt (aRRR = 0.74, 95% CI = 0.68 to 0.81; P < .001) compared with male sex. Thus, disparities by race, ethnicity, and sex were observed in mRCC systemic therapy utilization for Medicare beneficiaries from 2015 to 2019., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
8. Bempegaldesleukin plus Nivolumab in First-line Metastatic Urothelial Carcinoma: Results from PIVOT-02.
- Author
-
Siefker-Radtke AO, Cho DC, Diab A, Sznol M, Bilen MA, Balar AV, Grignani G, Puente E, Tang L, Chien D, Hoch U, Choudhury A, Yu D, Currie SL, Tagliaferri MA, Zalevsky J, Hurwitz ME, and Tannir NM
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Humans, Interleukin-2 therapeutic use, Nivolumab adverse effects, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell secondary, Prodrugs therapeutic use, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms etiology
- Abstract
Background: Despite recent changes in the treatment landscape, there remains an unmet need for effective, tolerable, chemotherapy-free treatments for patients with advanced/metastatic urothelial carcinoma (mUC), especially cisplatin-ineligible patients., Objective: To evaluate the immunostimulatory interleukin-2 cytokine prodrug bempegaldesleukin (BEMPEG) plus nivolumab in patients with advanced/mUC from the phase 2 multicenter PIVOT-02 study., Design, Setting, and Participants: This open-label, multicohort phase 1/2 study enrolled patients with previously untreated locally advanced/surgically unresectable or mUC (N = 41)., Intervention: Patients received BEMPEG 0.006 mg/kg plus nivolumab 360 mg intravenously every 3 wk., Outcome Measurements and Statistical Analysis: The primary objectives were safety and the objective response rate (ORR) in patients with measurable disease at baseline and at least one postbaseline tumor response assessment (response-evaluable). Secondary objectives were overall survival (OS) and progression-free survival (PFS). Exploratory biomarker analyses via univariate logistic regression were performed to test the association between potential biomarkers (CD8
+ tumor-infiltrating lymphocytes, tumor mutational burden, and IFN-γ gene expression profile) and response., Results and Limitations: The ORR was 35% (13/37 evaluable patients) and the complete response rate was 19% (7/37 patients); the median duration of response was not reached. Median PFS was 4.1 mo (95% confidence interval [CI] 2.1-8.7) and median OS was 23.7 mo (95% CI 15.8-not reached). Overall, 40/41 patients (98%) experienced at least one treatment-related adverse event (TRAE); grade 3/4 TRAEs occurred in 11 patients (27%), most commonly pyrexia (4.9%; 2 patients). Exploratory biomarker analyses showed no association between biomarkers and response. Limitations include the small sample size and single-arm design., Conclusions: BEMPEG plus nivolumab was well tolerated and showed antitumor activity as first-line treatment in patients with locally advanced/mUC., Patient Summary: We investigated an immune-stimulating prodrug called bempegaldesleukin plus the antibody nivolumab as the first therapy for patients with advanced or metastatic cancer of the urinary tract. This combination had manageable treatment-related side effects and was effective in a subset of patients. This trial is registered at ClinicalTrials.gov as NCT02983045., (Copyright © 2022 European Association of Urology. Published by Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
9. Translation of the 27-gene immuno-oncology test (IO score) to predict outcomes in immune checkpoint inhibitor treated metastatic urothelial cancer patients.
- Author
-
Seitz RS, Hurwitz ME, Nielsen TJ, Bailey DB, Varga MG, Ring BZ, Metts CF, Schweitzer BL, McGregor K, and Ross DT
- Subjects
- B7-H1 Antigen genetics, Biomarkers, Tumor genetics, Clinical Trials as Topic, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms pathology
- Abstract
Background: The IO Score is a 27-gene immuno-oncology (IO) classifier that has previously predicted benefit to immune checkpoint inhibitor (ICI) therapy in triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC). It generates both a continuous score and a binary result using a defined threshold that is conserved between breast and lung. Herein, we aimed to evaluate the IO Score's binary threshold in ICI-naïve TCGA bladder cancer patients (TCGA-BLCA) and assess its clinical utility in metastatic urothelial cancer (mUC) using the IMvigor210 clinical trial treated with the ICI, atezolizumab., Methods: We identified a list of tumor immune microenvironment (TIME) related genes expressed across the TCGA breast, lung squamous and lung adenocarcinoma cohorts (TCGA-BRCA, TCGA-LUSQ, and TCGA-LUAD, 939 genes total) and then examined the expression of these 939 genes in TCGA-BLCA, to identify patients as having high inflammatory gene expression. Using this as a test of classification, we assessed the previously established threshold of IO Score. We then evaluated the IO Score with this threshold in the IMvigor210 cohort for its association with overall survival (OS)., Results: In TCGA-BLCA, IO Score positive patients had a strong concordance with high inflammatory gene expression (p < 0.0001). Given this concordance, we applied the IO Score to the ICI treated IMvigor210 patients. IO Score positive patients (40%) had a significant Cox proportional hazard ratio (HR) of 0.59 (95% CI 0.45-0.78 p < 0.001) for OS and improved median OS (15.6 versus 7.5 months) compared to IO Score negative patients. The IO Score remained significant in bivariate models combined with all other clinical factors and biomarkers, including PD-L1 protein expression and tumor mutational burden., Conclusion: The IMvigor210 results demonstrate the potential for the IO Score as a clinically useful biomarker in mUC. As this is the third tumor type assessed using the same algorithm and threshold, the IO Score may be a promising candidate as a tissue agnostic marker of ICI clinical benefit. The concordance between IO Score and inflammatory gene expression suggests that the classifier is capturing common features of the TIME across cancer types., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Bempegaldesleukin plus nivolumab in first-line renal cell carcinoma: results from the PIVOT-02 study.
- Author
-
Tannir NM, Cho DC, Diab A, Sznol M, Bilen MA, Balar AV, Grignani G, Puente E, Tang L, Chien D, Hoch U, Choudhury A, Yu D, Currie SL, Tagliaferri MA, Zalevsky J, Siefker-Radtke AO, and Hurwitz ME
- Subjects
- Female, Humans, Interleukin-2 therapeutic use, Male, Nivolumab therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell pathology, Kidney Neoplasms drug therapy, Kidney Neoplasms pathology
- Abstract
Background: Immune checkpoint inhibitor-based combinations have expanded the treatment options for patients with renal cell carcinoma (RCC); however, tolerability remains challenging. The aim of this study was to evaluate the safety and efficacy of the immunostimulatory interleukin-2 cytokine prodrug bempegaldesleukin (BEMPEG) plus nivolumab (NIVO) as first-line therapy in patients with advanced clear-cell RCC., Methods: This was an open-label multicohort, multicenter, single-arm phase 1/2 study; here, we report results from the phase 1/2 first-line RCC cohort (N=49). Patients received BEMPEG 0.006 mg/kg plus NIVO 360 mg intravenously every 3 weeks. The primary objectives were safety and objective response rate (ORR; patients with measurable disease at baseline and at least one postbaseline tumor response assessment). Secondary objectives included overall survival (OS) and progression-free survival (PFS). Exploratory biomarker analyses: association between baseline biomarkers and ORR., Results: At a median follow-up of 32.7 months, the ORR was 34.7% (17/49 patients); 3/49 patients (6.1%) had a complete response. Of the 17 patients with response, 14 remained in response for >6 months, and 6 remained in response for >24 months. Median PFS was 7.7 months (95% CI 3.8 to 13.9), and median OS was not reached (95% CI 37.3 to not reached). Ninety-eight per cent (48/49) of patients experienced ≥1 treatment-related adverse event (TRAE) and 38.8% (19/49) had grade 3/4 TRAEs, most commonly syncope (8.2%; 4/49) and increased lipase (6.1%; 3/49). No association between exploratory biomarkers and ORR was observed. Limitations include the small sample size and single-arm design., Conclusions: BEMPEG plus NIVO showed preliminary antitumor activity as first-line therapy in patients with advanced clear-cell RCC and was well tolerated. These findings warrant further investigation., Competing Interests: Competing interests: AOS-R received research funding from BioClin Therapeutics, Bristol Myers Squibb, Janssen, Merck Sharp & Dohme, Michael and Sherry Sutton Fund for Urothelial Cancer, Nektar Therapeutics, US National Institutes of Health, and Takeda. She has also served as an advisor/consultant to AstraZeneca, Bavarian Nordic, BioClin Therapeutics, Bristol Myers Squibb, EMD Serono, Genentech, Inovio Pharmaceuticals, Janssen, Merck, US National Comprehensive Cancer Network, Nektar Therapeutics, and Seattle Genetics. DCC has received consulting fees from Pfizer, Nectar, Torque, and Puretech. AD has been an advisory board member for Nektar Therapeutics. MS has served as a consultant/advisor for Genentech-Roche, Bristol Myers Squibb, AstraZeneca/MedImmune, Pfizer, Novartis, Kyowa-Kirin, Amgen, Merus, Seattle Genetics, Immune Design, Prometheus, Anaeropharma, Astellas-Agensys, Immunova, Nektar Therapeutics, Neostem, Pierre-Fabre, Eli Lilly, Symphogen, Lion Biotechnologies, Amphivena, and Adaptive Biotechnologies. MAB has acted as a paid consultant for, and/or as a member of the advisory boards of, Exelixis, Bayer, BMS, Eisai, Pfizer, AstraZeneca, Janssen, Genomic Health, Nektar Therapeutics, and Sanofi and has received grants to his institution from Xencor, Bayer, Bristol Myers Squibb, Genentech/Roche, Seattle Genetics, Incyte, Nektar Therapeutics, AstraZeneca, Tricon Pharmaceuticals, Peleton Therapeutics, and Pfizer for work performed as outside of the current study. AVB has received institutional research funding from AstraZeneca/MedImmune, F Hoffmann–La Roche/Genentech, Merck, and Seattle Genetics; and honoraria from AstraZeneca/MedImmune, F Hoffmann–La Roche/Genentech and Merck. He has also served as an advisor/consultant to AstraZeneca/MedImmune, Cerulean Pharma, F Hoffmann–La Roche/Genentech, Incyte, Merck, Nektar Therapeutics, Pfizer/EMD Serono, and Seattle Genetics/Astellas. GG has received grants and personal fees from PharmaMar, grants from Novartis, and personal fees from Lilly, Pfizer, Bayer, and Eisai, outside the submitted work. EP, LT, DC, UH, AC, and DY are employees of, and have ownership interest (eg, stock) in, Nektar Therapeutics. SLC is a former employee of, and has ownership interest (eg, stock) in, Nektar Therapeutics. MAT is the chief medical officer at Nektar Therapeutics and has ownership interest (eg, stock) in the company. JZ is the chief research and development officer at Nektar Therapeutics and has ownership interest (eg, stock) in the company. MEH has had a consulting or advisory Role with Nektar Therapeutics, Janssen, Crispr Therapeutics, Bristol Myers Squibb/Celgene, and Exelixis; and has received research funding from Apexigen, Astellas Pharma, AstraZeneca/MedImmune, Bayer, Bristol Myers Squibb, Corvus Pharmaceuticals, Lilly, Endocyte, Genentech, Genmab, Innocrin Pharma, Iovance Biotherapeutics, Merck, Nektar Therapeutics, Novartis, Pfizer, Progenics, Sanofi/Aventis, Seattle Genetics, Torque, Unum Therapeutics, and Achilles Therapeutics. NMT has received grant support, consulting fees, and honoraria from Bristol Myers Squibb; grant support from Epizyme and Mirati Therapeutics; grant support, consulting fees, and honoraria from, and served on an advisory board for, Exelixis and Novartis; received consulting fees and honoraria from Argos Therapeutics and Pfizer; and received consulting fees and honoraria from, and served on an advisory board for, Eisai, Nektar Therapeutics, and Oncorena., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
11. Bempegaldesleukin Plus Nivolumab in First-Line Metastatic Melanoma.
- Author
-
Diab A, Tykodi SS, Daniels GA, Maio M, Curti BD, Lewis KD, Jang S, Kalinka E, Puzanov I, Spira AI, Cho DC, Guan S, Puente E, Nguyen T, Hoch U, Currie SL, Lin W, Tagliaferri MA, Zalevsky J, Sznol M, and Hurwitz ME
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Europe, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Interleukin-2 adverse effects, Interleukin-2 therapeutic use, Male, Melanoma mortality, Melanoma secondary, Middle Aged, Neoplasm Staging, Nivolumab adverse effects, Polyethylene Glycols adverse effects, Progression-Free Survival, Skin Neoplasms mortality, Skin Neoplasms pathology, Time Factors, United States, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Immune Checkpoint Inhibitors therapeutic use, Interleukin-2 analogs & derivatives, Melanoma drug therapy, Nivolumab therapeutic use, Polyethylene Glycols therapeutic use, Skin Neoplasms drug therapy
- Abstract
Purpose: Therapies that produce deep and durable responses in patients with metastatic melanoma are needed. This phase II cohort from the international, single-arm PIVOT-02 study evaluated the CD122-preferential interleukin-2 pathway agonist bempegaldesleukin (BEMPEG) plus nivolumab (NIVO) in first-line metastatic melanoma., Methods: A total of 41 previously untreated patients with stage III/IV melanoma received BEMPEG 0.006 mg/kg plus NIVO 360 mg once every 3 weeks for ≤ 2 years; 38 were efficacy-evaluable (≥ 1 postbaseline scan). Primary end points were safety and objective response rate (blinded independent central review); other end points included progression-free survival, overall survival (OS), and exploratory biomarkers., Results: At 29.0 months' median follow-up, the objective response rate was 52.6% (20 of 38 patients), and the complete response rate was 34.2% (13 of 38 patients). Median change in size of target lesions from baseline was -78.5% (response-evaluable population); 47.4% (18 of 38 patients) experienced complete clearance of target lesions. Median progression-free survival was 30.9 months (95% CI, 5.3 to not estimable). Median OS was not reached; the 24-month OS rate was 77.0% (95% CI, 60.4 to 87.3). Grade 3 and 4 treatment-related and immune-mediated adverse events occurred in 17.1% (7 of 41) and 4.9% (2 of 41) of patients, respectively. Increased polyfunctional responses in CD8+ and CD4+ T cells were seen in blood after treatment, driven by cytokines with effector functions. Early on-treatment blood biomarkers (CD8+ polyfunctional strength difference and eosinophils) correlated with treatment response., Conclusion: BEMPEG in combination with NIVO was tolerated, with relatively low rates of grade 3 and 4 treatment-related and immune-mediated adverse events. The combination had encouraging antitumor activity in first-line metastatic melanoma, including an extended median progression-free survival. Exploratory analyses associated noninvasive, on-treatment biomarkers with response, before radiologic evidence was observed., Competing Interests: Adi DiabHonoraria: Array BioPharmaConsulting or Advisory Role: Nektar, CureVac, Celgene, IderaResearch Funding: Nektar, Idera, Celgene, Pfizer, Merck, ApexigenTravel, Accommodations, Expenses: Nektar Scott S. TykodiConsulting or Advisory Role: Merck, Intellisphere LLC, Natera, Bristol Myers Squibb, ExelixisResearch Funding: Genentech, Bristol Myers Squibb, Merck Sharp & Dohme, Calithera Biosciences, Pfizer, Jounce Therapeutics, Nektar, Exelixis, Clinigen GroupPatents, Royalties, Other Intellectual Property: Patent pending Gregory A. DanielsHonoraria: Sanofi/RegeneronConsulting or Advisory Role: Sanofi/RegeneronSpeakers' Bureau: Regeneron, Array BioPharma, Sanofi/RegeneronResearch Funding: Bristol Myers Squibb, Amgen, Viralytics, Nektar, Merck Michele MaioStock and Other Ownership Interests: Theravance, Epigen TherapeuticsHonoraria: Bristol Myers Squibb, AstraZeneca, Roche, MSD, Merck, Amgen, Pierre Fabre, Alfasigma, Sanofi, LillyConsulting or Advisory Role: Bristol Myers Squibb, Roche, AstraZeneca, MSD, Merck, Pierre Fabre, AlfasigmaPatents, Royalties, Other Intellectual Property: DNA hypomethylating agents for cancer therapyTravel, Accommodations, Expenses: Bristol Myers Squibb, AstraZeneca, Roche, MSD, Merck, Amgen, Pierre Fabre, Alfasigma Brendan D. CurtiHonoraria: Clinigen Group, NektarConsulting or Advisory Role: MerckResearch Funding: Bristol Myers Squibb, Galectin Therapeutics, Clinigen GroupPatents, Royalties, Other Intellectual Property: Biomarkers for OX40 responseTravel, Accommodations, Expenses: Agonox Karl D. LewisHonoraria: Array BioPharma, Iovance BiotherapeuticsConsulting or Advisory Role: Array BioPharma, Merck, Roche, Regeneron, Sanofi, Iovance BiotherapeuticsResearch Funding: Roche/Genentech, Merck, Array BioPharma, Incyte, Nektar, Iovance Biotherapeutics, Bristol Myers Squibb, Kartos Therapeutics, OncoSec, Regeneron, Alkermes, Neon Therapeutics, Ultimovacs, Senhwa Biosciences, Replimune, AmgenTravel, Accommodations, Expenses: Merck, Roche/Genentech, Regeneron, Neon Therapeutics, AlkermesUncompensated Relationships: Roche/Genentech, Regeneron Sekwon JangConsulting or Advisory Role: Bristol Myers Squibb, EMD Serono, Novartis, Sanofi, Sun Biopharma, Genentech Ewa KalinkaHonoraria: Bristol Myers Squibb, MSD, AstraZeneca, Regeneron, Nektar, RocheConsulting or Advisory Role: Bristol Myers SquibbSpeakers' Bureau: Bristol Myers Squibb, RocheResearch Funding: Bristol Myers Squibb, Merck Sharp & Dohme, Nektar, AstraZeneca, RocheTravel, Accommodations, Expenses: Roche Igor PuzanovStock and Other Ownership Interests: CelldexConsulting or Advisory Role: Amgen, Iovance Biotherapeutics, Merck, Roche, Nouscom, Seneca Therapeutics Alexander I. SpiraStock and Other Ownership Interests: LillyHonoraria: CytomX Therapeutics, AstraZeneca/MedImmune, Merck, Takeda, Amgen, Janssen Oncology, Novartis, Bristol Myers Squibb, BayerConsulting or Advisory Role: Array BioPharma, Incyte, Amgen, Novartis, AstraZeneca/MedImmune, Mirati Therapeutics, Gritstone Oncology, Jazz Pharmaceuticals, Merck, Bristol Myers SquibbResearch Funding: Roche, AstraZeneca, Boehringer Ingelheim, Astellas Pharma, MedImmune, Novartis, Newlink Genetics, Incyte, AbbVie, Ignyta, LAM Therapeutics, Trovagene, Takeda, Macrogenics, CytomX Therapeutics, Astex Pharmaceuticals, Bristol Myers Squibb, Loxo, Arch Therapeutics, Gritstone Oncology, Plexxikon, Amgen, Daiichi Sankyo, ADC Therapeutics, Janssen Oncology, Mirati Therapeutics, Rubius Therapeutics Daniel C. ChoConsulting or Advisory Role: Nektar, Pfizer, Werewolf TherapeuticsExpert Testimony: Genentech, Abbott/AbbVie Shanhong GuanEmployment: Nektar TherapeuticsStock and Other Ownership Interests: Nektar Therapeutics Erika PuenteEmployment: NektarStock and Other Ownership Interests: Nektar Tuan NguyenEmployment: Nektar, Theravance TherapeuticsStock and Other Ownership Interests: Nektar, Theravance Ute HochOther Relationship: Nektar Sue L. CurrieEmployment: NektarStock and Other Ownership Interests: Nektar Wei LinEmployment: Erasca Inc, NektarLeadership: Erasca IncStock and Other Ownership Interests: Nektar, Erasca IncTravel, Accommodations, Expenses: Nektar, Erasca Inc Mary A. TagliaferriEmployment: NektarLeadership: Nektar, ENZO BiochemStock and Other Ownership Interests: NektarPatents, Royalties, Other Intellectual Property: US 10576121Travel, Accommodations, Expenses: Nektar Jonathan ZalevskyEmployment: NektarLeadership: NektarStock and Other Ownership Interests: NektarTravel, Accommodations, Expenses: Nektar Mario SznolStock and Other Ownership Interests: Amphivena, Intensity Therapeutics, Adaptive Biotechnologies, Actym Therapeutics, Torque, Nextcure, EvolveImmune Therapeutics, Johnson & Johnson/Janssen, GlaxoSmithKlineConsulting or Advisory Role: Bristol Myers Squibb, AstraZeneca/MedImmune, Nektar, Lilly, Adaptimmune, Seattle Genetics, Pierre Fabre, Molecular Partners, AbbVie, Pieris Pharmaceuticals, Innate Pharma, Immunocore, Genocea Biosciences, Anaeropharma, Zelluna, Boston Pharmaceuticals, Alligator Bioscience, Servier, Dragonfly Therapeutics, Verastem, Boehringer Ingelheim, Agenus, Numab, BioNTech AG, Genentech/Roche, Gilead Sciences, Jazz Pharmaceuticals, Targovax, Sapience Therapeutics, Pfizer, Tessa Therapeutics, OncoSec, Trillium Therapeutics, StCube, Simcha, ITeos TherapeuticsOther Relationship: Haymarket Media, Physicians' Education Resource, DAVAOncology, CEC Oncology Michael E. HurwitzEmployment: Pfizer, Gamida Cell, ArvinasConsulting or Advisory Role: Nektar, Janssen, Crispr Therapeutics, Bristol Myers Squibb/Celgene, ExelixisResearch Funding: Apexigen, Astellas Pharma, AstraZeneca/MedImmune, Bayer, Bristol Myers Squibb, Corvus Pharmaceuticals, Lilly, Endocyte, Genentech, Genmab, Innocrin Pharma, Iovance Biotherapeutics, Merck, Nektar, Novartis, Pfizer, Progenics, Sanofi/Aventis, Seattle Genetics, Torque, Unum Therapeutics, Achilles TherapeuticsNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
12. Current Understanding and Management of Intraductal Carcinoma of the Prostate.
- Author
-
Considine B, Adeniran A, and Hurwitz ME
- Subjects
- Carcinoma, Ductal pathology, Carcinoma, Ductal therapy, Humans, Male, Neoplasm Grading, Prostatic Neoplasms pathology, Prostatic Neoplasms therapy, Signal Transduction genetics, Carcinoma, Ductal genetics, DNA Mismatch Repair genetics, Genetic Predisposition to Disease genetics, Microsatellite Instability, Mutation, Prostatic Neoplasms genetics
- Abstract
Purpose of Review: This review will discuss current understanding and management approaches of Intraductal carcinoma of the prostate (IDC-P). IDC-P is a histological finding characterized by neoplastic cells that expand but do not invade prostate ducts., Recent Findings: The presence of IDC-P on a prostate biopsy is almost always associated with an invasive disease component and is independently associated with worse clinical outcomes in both early and late disease. These tumors are enriched for mutations in homologous DNA recombination repair (HRR) leading to high genomic instability. Multiparametric MRI with targeted biopsy may aid in diagnosis. Given the poor clinical outcomes associated with this histologic entity, its presence in biopsies should warrant consideration of aggressive management., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
13. Bempegaldesleukin (NKTR-214) plus Nivolumab in Patients with Advanced Solid Tumors: Phase I Dose-Escalation Study of Safety, Efficacy, and Immune Activation (PIVOT-02).
- Author
-
Diab A, Tannir NM, Bentebibel SE, Hwu P, Papadimitrakopoulou V, Haymaker C, Kluger HM, Gettinger SN, Sznol M, Tykodi SS, Curti BD, Tagliaferri MA, Zalevsky J, Hannah AL, Hoch U, Aung S, Fanton C, Rizwan A, Iacucci E, Liao Y, Bernatchez C, Hurwitz ME, and Cho DC
- Subjects
- Adult, Aged, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell immunology, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Immune Checkpoint Inhibitors adverse effects, Immunotherapy, Interleukin-2 administration & dosage, Interleukin-2 adverse effects, Kidney Neoplasms genetics, Kidney Neoplasms immunology, Lung Neoplasms genetics, Lung Neoplasms immunology, Lymphocyte Count, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Male, Melanoma genetics, Melanoma immunology, Middle Aged, Nivolumab adverse effects, Polyethylene Glycols adverse effects, Programmed Cell Death 1 Receptor antagonists & inhibitors, Treatment Outcome, Young Adult, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Renal Cell drug therapy, Immune Checkpoint Inhibitors administration & dosage, Interleukin-2 analogs & derivatives, Kidney Neoplasms drug therapy, Lung Neoplasms drug therapy, Melanoma drug therapy, Nivolumab administration & dosage, Polyethylene Glycols administration & dosage
- Abstract
This single-arm, phase I dose-escalation trial (NCT02983045) evaluated bempegaldesleukin (NKTR-214/BEMPEG), a CD122-preferential IL2 pathway agonist, plus nivolumab in 38 patients with selected immunotherapy-naïve advanced solid tumors (melanoma, renal cell carcinoma, and non-small cell lung cancer). Three dose-limiting toxicities were reported in 2 of 17 patients during dose escalation [hypotension ( n = 1), hyperglycemia ( n = 1), metabolic acidosis ( n = 1)]. The most common treatment-related adverse events (TRAE) were flu-like symptoms (86.8%), rash (78.9%), fatigue (73.7%), and pruritus (52.6%). Eight patients (21.1%) experienced grade 3/4 TRAEs; there were no treatment-related deaths. Total objective response rate across tumor types and dose cohorts was 59.5% (22/37), with 7 complete responses (18.9%). Cellular and gene expression analysis of longitudinal tumor biopsies revealed increased infiltration, activation, and cytotoxicity of CD8
+ T cells, without regulatory T-cell enhancement. At the recommended phase II dose, BEMPEG 0.006 mg/kg plus nivolumab 360 mg every 3 weeks, the combination was well tolerated and demonstrated encouraging clinical activity irrespective of baseline PD-L1 status. SIGNIFICANCE: These data show that BEMPEG can be successfully combined with a checkpoint inhibitor as dual immunotherapy for a range of advanced solid tumors. Efficacy was observed regardless of baseline PD-L1 status and baseline levels of tumor-infiltrating lymphocytes, suggesting therapeutic potential for patients with poor prognostic risk factors for response to PD-1/PD-L1 blockade. See related commentary by Rouanne et al., p. 1097 . This article is highlighted in the In This Issue feature, p. 1079 ., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
14. Bempegaldesleukin selectively depletes intratumoral Tregs and potentiates T cell-mediated cancer therapy.
- Author
-
Sharma M, Khong H, Fa'ak F, Bentebibel SE, Janssen LME, Chesson BC, Creasy CA, Forget MA, Kahn LMS, Pazdrak B, Karki B, Hailemichael Y, Singh M, Vianden C, Vennam S, Bharadwaj U, Tweardy DJ, Haymaker C, Bernatchez C, Huang S, Rajapakshe K, Coarfa C, Hurwitz ME, Sznol M, Hwu P, Hoch U, Addepalli M, Charych DH, Zalevsky J, Diab A, and Overwijk WW
- Subjects
- Animals, Antibodies, Monoclonal, Humanized administration & dosage, CD8-Positive T-Lymphocytes immunology, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell immunology, Cohort Studies, Drug Therapy, Combination, Female, Humans, Interferon-gamma genetics, Interferon-gamma immunology, Interleukin-2 administration & dosage, Interleukin-2 agonists, Interleukin-2 immunology, Ipilimumab administration & dosage, Lymphocyte Activation drug effects, Melanoma genetics, Melanoma immunology, Mice, Mice, Inbred C57BL, Receptors, Interleukin-2 genetics, Receptors, Interleukin-2 immunology, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha immunology, Carcinoma, Renal Cell drug therapy, Interleukin-2 analogs & derivatives, Melanoma drug therapy, Polyethylene Glycols administration & dosage, Prodrugs administration & dosage, T-Lymphocytes, Regulatory immunology
- Abstract
High dose interleukin-2 (IL-2) is active against metastatic melanoma and renal cell carcinoma, but treatment-associated toxicity and expansion of suppressive regulatory T cells (Tregs) limit its use in patients with cancer. Bempegaldesleukin (NKTR-214) is an engineered IL-2 cytokine prodrug that provides sustained activation of the IL-2 pathway with a bias to the IL-2 receptor CD122 (IL-2Rβ). Here we assess the therapeutic impact and mechanism of action of NKTR-214 in combination with anti-PD-1 and anti-CTLA-4 checkpoint blockade therapy or peptide-based vaccination in mice. NKTR-214 shows superior anti-tumor activity over native IL-2 and systemically expands anti-tumor CD8
+ T cells while inducing Treg depletion in tumor tissue but not in the periphery. Similar trends of intratumoral Treg dynamics are observed in a small cohort of patients treated with NKTR-214. Mechanistically, intratumoral Treg depletion is mediated by CD8+ Teff-associated cytokines IFN-γ and TNF-α. These findings demonstrate that NKTR-214 synergizes with T cell-mediated anti-cancer therapies.- Published
- 2020
- Full Text
- View/download PDF
15. Key Factors in Clinical Protocols for Adoptive Cell Therapy in Melanoma.
- Author
-
Considine B and Hurwitz ME
- Subjects
- Humans, Lymphocyte Depletion, Lymphocytes, Tumor-Infiltrating immunology, Clinical Protocols, Immunotherapy, Adoptive methods, Melanoma immunology, Melanoma therapy
- Abstract
Adoptive cell therapy (ACT) with autologous tumor infiltrating lymphocytes (TIL) has been studied for patients with advanced metastatic cancers (primarily melanoma) for decades and has changed significantly during that period. Treatment with TIL includes ex vivo cell activation and expansion followed by re-infusion of these cells into the patient. After cell infusion, patients receive Interleukin-2 (IL-2). Objective response rates up to 52% have been seen in patients with metastatic melanoma. Efforts to improve TIL therapy include better selection and expansion of tumor-reactive lymphocytes, optimization of IL-2 or other T cell activating cytokine dosing, and, potentially, genetic manipulation of the immune cell product. Here we describe methods involved in the collection, expansion, and treatment with TIL for patients with metastatic melanoma.
- Published
- 2020
- Full Text
- View/download PDF
16. A versatile flow-based assay for immunocyte-mediated cytotoxicity.
- Author
-
Rabinovich PM, Zhang J, Kerr SR, Cheng BH, Komarovskaya M, Bersenev A, Hurwitz ME, Krause DS, Weissman SM, and Katz SG
- Subjects
- Animals, Cell Line, Tumor, Cell Nucleus immunology, Cell Nucleus pathology, High-Throughput Screening Assays, Humans, Immunotherapy, Adoptive, Male, Melanoma immunology, Melanoma pathology, Mice, Inbred C57BL, Predictive Value of Tests, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology, Reproducibility of Results, Skin Neoplasms immunology, Skin Neoplasms pathology, Time Factors, Workflow, Cytotoxicity Tests, Immunologic methods, Cytotoxicity, Immunologic, Flow Cytometry, Killer Cells, Natural immunology, Lymphocytes, Tumor-Infiltrating immunology, T-Lymphocytes immunology
- Abstract
Cell-mediated cytotoxicity is a critical function of the immune system in mounting defense against pathogens and cancers. Current methods that allow direct evaluation of cell-mediated cytotoxicity suffer from a wide-range of drawbacks. Here, we present a novel strategy to measure cytotoxicity that is direct, sensitive, rapid, and highly adaptable. Moreover, it allows accurate measurement of viability of both target and effector cells. Target cells are fluorescently labeled with a non-toxic, cell-permeable dye that covalently binds to cell proteins, including nuclear proteins. The labeled target cells are incubated with effector cells to begin killing. Following the killing reaction, the cell mixture is incubated with another dye that specifically stains proteins of dead cells, including nuclear proteins. In the final step, cell nuclei are released by Triton X-100, and analyzed by flow cytometry. This results in four nuclear staining patterns that separate target and effector nuclei as well as nuclei of live and dead cells. Analyzing nuclei, instead of cells, greatly reduces flow cytometry errors caused by the presence of target-effector cell aggregates. Target killing time can often be reduced to 2 h and the assay can be done in a high throughput format. We have successfully validated this assay in a variety of cytotoxicity scenarios including those mediated by NK-92 cells, Chimeric Antigen Receptor (CAR)-T cells, and Tumor Infiltrating Lymphocytes (TIL). Therefore, this technique is broadly applicable, highly sensitive and easily administered, making it a powerful tool to assess immunotherapy-based, cell-mediated cytotoxicity., (Copyright © 2019. Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
17. A First-in-Human Study and Biomarker Analysis of NKTR-214, a Novel IL2Rβγ-Biased Cytokine, in Patients with Advanced or Metastatic Solid Tumors.
- Author
-
Bentebibel SE, Hurwitz ME, Bernatchez C, Haymaker C, Hudgens CW, Kluger HM, Tetzlaff MT, Tagliaferri MA, Zalevsky J, Hoch U, Fanton C, Aung S, Hwu P, Curti BD, Tannir NM, Sznol M, and Diab A
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biomarkers, CD8-Positive T-Lymphocytes, Cell Line, Tumor, Humans, Interleukin-2 administration & dosage, Interleukin-2 adverse effects, Interleukin-2 pharmacokinetics, Interleukin-2 therapeutic use, Neoplasms etiology, Neoplasms pathology, Polyethylene Glycols administration & dosage, Polyethylene Glycols adverse effects, Polyethylene Glycols pharmacokinetics, Treatment Outcome, Tumor Microenvironment drug effects, Interleukin-2 analogs & derivatives, Interleukin-2 metabolism, Interleukin-2 Receptor alpha Subunit metabolism, Neoplasms drug therapy, Neoplasms metabolism, Polyethylene Glycols therapeutic use, Signal Transduction drug effects
- Abstract
NKTR-214 (bempegaldesleukin) is a novel IL2 pathway agonist, designed to provide sustained signaling through heterodimeric IL2 receptor βγ to drive increased proliferation and activation of CD8
+ T and natural killer cells without unwanted expansion of T regulatory cells (Treg) in the tumor microenvironment. In this first-in-human multicenter phase I study, NKTR-214 administered as an outpatient regimen was well tolerated and showed clinical activity including tumor shrinkage and durable disease stabilization in heavily pretreated patients. Immune activation and increased numbers of immune cells were observed in the periphery across all doses and cycles with no loss of NKTR-214 activity with repeated administration. On-treatment tumor biopsies demonstrated that NKTR-214 promoted immune cell increase with limited increase of Tregs. Transcriptional analysis of tumor biopsies showed that NKTR-214 engaged the IL2 receptor pathway and significantly increased genes associated with an effector phenotype. Based on safety and pharmacodynamic markers, the recommended phase II dose was determined to be 0.006 mg/kg every three weeks. SIGNIFICANCE: We believe that IL2- and IL2 pathway-targeted agents such as NKTR-214 are key components to an optimal immunotherapy treatment algorithm. Based on its biological activity and tolerability, NKTR-214 is being studied with approved immuno-oncology agents including checkpoint inhibitors. See related commentary by Sullivan, p. 694 . This article is highlighted in the In This Issue feature, p. 681 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
18. Current Status and Future Directions of Immunotherapy in Renal Cell Carcinoma.
- Author
-
Considine B and Hurwitz ME
- Subjects
- Carcinoma, Renal Cell immunology, Humans, Immunotherapy methods, Kidney Neoplasms immunology, Antineoplastic Agents therapeutic use, Carcinoma, Renal Cell drug therapy, Immunologic Factors therapeutic use, Immunotherapy trends, Kidney Neoplasms drug therapy
- Abstract
Purpose of Review: Renal cell carcinoma (RCC) was recognized as an immunologically sensitive cancer over 30 years ago. The first therapies to affect the course of RCC were cytokines (interferon alfa-2B and interleukin-2). Subsequently, drugs that inhibit HIF (hypoxia-inducible factor)/VEGF (vascular endothelial growth factor) signaling demonstrated overall survival advantages (tyrosine kinase inhibitors and mTor inhibitors)., Recent Findings: In the last 3 years, the immune checkpoint inhibitors (ICIs) have become the standard of care treatments in the first and second lines for RCC. Emerging data show that combinations of ICI, HIF signaling inhibitors, and cytokines are potentially powerful regimens. How to combine and sequence these types of therapies and how to integrate new approaches into the management of RCC are now the key questions for the field. Treatment of RCC is likely to change dramatically in the next few years.
- Published
- 2019
- Full Text
- View/download PDF
19. Multicenter Phase 2 Trial of Gemcitabine, Carboplatin, and Sorafenib in Patients With Metastatic or Unresectable Transitional-Cell Carcinoma.
- Author
-
Hurwitz ME, Markowski P, Yao X, Deshpande H, Patel J, Mortazavi A, Donadio A, Stein MN, Kelly WK, Petrylak DP, and Mehnert JM
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carboplatin administration & dosage, Carboplatin adverse effects, Carcinoma, Transitional Cell mortality, Carcinoma, Transitional Cell pathology, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine analogs & derivatives, Disease Progression, Drug Administration Schedule, Female, Humans, Male, Middle Aged, Progression-Free Survival, Sorafenib administration & dosage, Sorafenib adverse effects, Survival Analysis, Urologic Neoplasms mortality, Urologic Neoplasms pathology, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Carcinoma, Transitional Cell drug therapy, Urologic Neoplasms drug therapy
- Abstract
Background: Sorafenib, an oral tyrosine kinase inhibitor, may enhance the antitumor activity of platinum-based chemotherapy in transitional-cell carcinoma. This study investigated the safety and clinical outcome of adding sorafenib to gemcitabine and carboplatin for patients with advanced transitional-cell carcinoma., Patients and Methods: Subjects with metastatic or unresectable chemotherapy-naive TCC with Eastern Cooperative Oncology Group performance status 0 or 1 received gemcitabine (1000 mg/m
2 on days 1 and 8) and carboplatin (area under the curve of 5 on day 1) with sorafenib (400 mg 2 times a day on days 2-19 every 21 days) for 6 cycles. Subjects with stable disease or partial or complete response continued to receive sorafenib until disease progression. The primary end point was progression-free survival (PFS) at 5 months with a secondary end point of response (partial or complete)., Results: Seventeen subjects were enrolled. The median number of cycles of gemcitabine and carboplatin with sorafenib provided was 4.4. A total of 15, 5, and 8 subjects required reductions of gemcitabine, carboplatin, and sorafenib, respectively. Thirteen subjects (76%) required multiple dose reductions. Eleven subjects (65%) were free of progression at 5 months. The overall response rate was 54% (95% confidence interval [CI], 0.28-077), with 4 patients experiencing complete response (24%; 95% CI, 0.07-0.50) and 5 partial response (29%; 95% CI, 0.10-0.56); 7 subjects (41%) had stable disease. Median PFS was 9.5 months (95% CI, 0.43-1.26), and median overall survival was 25.2 months (95% CI, 0.96-5.65). One-year PFS was 31%, and 1-year overall survival was 72%. Eleven subjects (65%) discontinued treatment because of toxicity. There were no toxic deaths., Conclusion: Gemcitabine and carboplatin with sorafenib showed clinical activity in advanced TCC, with some prolonged progression-free intervals. However, gemcitabine and carboplatin with sorafenib was associated with significant toxicity, causing discontinuation of therapy in most patients., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
20. Triclosan Is an Aminoglycoside Adjuvant for Eradication of Pseudomonas aeruginosa Biofilms.
- Author
-
Maiden MM, Hunt AMA, Zachos MP, Gibson JA, Hurwitz ME, Mulks MH, and Waters CM
- Subjects
- Biofilms growth & development, Cystic Fibrosis drug therapy, Drug Synergism, Drug Therapy, Combination, High-Throughput Screening Assays, Humans, Microbial Sensitivity Tests, Pseudomonas aeruginosa isolation & purification, Adjuvants, Pharmaceutic pharmacology, Anti-Bacterial Agents pharmacology, Biofilms drug effects, Fatty Acid Synthesis Inhibitors pharmacology, Pseudomonas aeruginosa drug effects, Tobramycin pharmacology, Triclosan pharmacology
- Abstract
One of the most important clinical obstacles in cystic fibrosis (CF) treatment is antibiotic treatment failure due to biofilms produced by Pseudomonas aeruginosa The ability of this pathogen to survive eradication by tobramycin and pathoadapt into a hyperbiofilm state leading to chronic infections is key to its success. Retrospective studies have demonstrated that preventing this pathoadaptation by improving eradication is essential to extend the lives of CF patients. To identify adjuvants that enhance tobramycin eradication of P. aeruginosa , we performed a high-throughput screen of 6,080 compounds from four drug-repurposing libraries. We identified that the Food and Drug Administration (FDA)-approved compound triclosan, in combination with tobramycin, resulted in a 100-fold reduction of viable cells within biofilms at 6 h, but neither compound alone had significant antimicrobial activity against biofilms. This synergistic treatment significantly accelerated the killing of biofilms compared to that with tobramycin treatment alone, and the combination was effective against 6/7 CF clinical isolates compared to tobramycin treatment alone, including a tobramycin-resistant strain. Further, triclosan and tobramycin killed persister cells, causing a 100-fold reduction by 8 h and complete eradication by 24 h. Triclosan also enhances tobramycin killing of multiple Burkholderia cenocepacia and Staphylococcus aureus clinical isolates grown as biofilms. Additionally, triclosan showed synergy with other aminoglycosides, such as gentamicin or streptomycin. Triclosan is a well-tolerated aminoglycoside adjuvant shown to be safe for human use that could improve the treatment of biofilm-based infections., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
21. Cancer immunotherapy: new applications in urologic oncology.
- Author
-
Hurwitz ME, Sokhn J, and Petrylak DP
- Subjects
- Humans, Male, Urology trends, Cancer Vaccines therapeutic use, Immunotherapy methods, Kidney Neoplasms therapy, Urologic Neoplasms therapy
- Abstract
Purpose of Review: Over the last 10 years, harnessing of the immune system to attack tumors has been one of the major breakthroughs in cancer, primarily through the use of immune checkpoint inhibitors (ICIs). This review will summarize current immune treatments in urologic malignancies and ongoing trials with novel combinations and in different disease settings., Recent Findings: Patients with urologic malignancies such as kidney and bladder cancer have benefited significantly from these advances with the approval of ICIs in both of these diseases. In addition, older immune therapies have also been used in these malignancies. For example, kidney cancers, which are traditionally unresponsive to chemotherapy, can respond to immune activation by cytokines. Prostate cancers, too, have immune therapies, such as sipuleucel-T, a dendritic cell vaccine. Combining ICIs with these older treatments as well as with molecularly targeted therapies and chemotherapies are currently underway., Summary: The ICIs have changed the way urologic malignancies are treated and newer combinations are likely to alter the therapeutic landscape in these diseases dramatically in the coming years.
- Published
- 2016
- Full Text
- View/download PDF
22. Clonal screens to find modifiers of partially penetrant phenotypes in C. elegans.
- Author
-
Hurwitz ME
- Subjects
- Animals, Caenorhabditis elegans embryology, Genetic Association Studies methods, Mutagenesis, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Genetic Testing methods, Penetrance, Phenotype
- Abstract
Unbiased genetic screens are an excellent way to discover novel genes involved in specific biological processes in vivo. Modifier screens, whether to suppress or enhance a phenotype, are a powerful way to find proteins that modulate biological processes responsible for specific phenotypes. However, modification of phenotypes that are only partially penetrant, which is often the case, are often extremely difficult to screen this way in a traditional F2 or non-clonal genetic screen. Here we describe an F3 or clonal screen in the nematode Caenorhabditis elegans to search for genes that modify partially penetrant phenotypes. Specifically we describe a screen to search for modifiers of genes that cause defects in migration of a specific developmentally regulated cell, the distal tip cell.
- Published
- 2014
- Full Text
- View/download PDF
23. SLI-1 Cbl inhibits the engulfment of apoptotic cells in C. elegans through a ligase-independent function.
- Author
-
Anderson C, Zhou S, Sawin E, Horvitz HR, and Hurwitz ME
- Subjects
- Actin Cytoskeleton genetics, Actin Cytoskeleton metabolism, Animals, Cytoskeletal Proteins genetics, Cytoskeletal Proteins metabolism, Gonads growth & development, Membrane Proteins genetics, Membrane Proteins metabolism, Phagocytosis, Signal Transduction, rac GTP-Binding Proteins genetics, rac GTP-Binding Proteins metabolism, Apoptosis, Caenorhabditis elegans cytology, Caenorhabditis elegans growth & development, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Cell Movement, Proto-Oncogene Proteins c-cbl genetics, Proto-Oncogene Proteins c-cbl metabolism
- Abstract
The engulfment of apoptotic cells is required for normal metazoan development and tissue remodeling. In Caenorhabditis elegans, two parallel and partially redundant conserved pathways act in cell-corpse engulfment. One pathway, which includes the small GTPase CED-10 Rac and the cytoskeletal regulator ABI-1, acts to rearrange the cytoskeleton of the engulfing cell. The CED-10 Rac pathway is also required for proper migration of the distal tip cells (DTCs) during the development of the C. elegans gonad. The second pathway includes the receptor tyrosine kinase CED-1 and might recruit membranes to extend the surface of the engulfing cell. Cbl, the mammalian homolog of the C. elegans E3 ubiquitin ligase and adaptor protein SLI-1, interacts with Rac and Abi2 and modulates the actin cytoskeleton, suggesting it might act in engulfment. Our genetic studies indicate that SLI-1 inhibits apoptotic cell engulfment and DTC migration independently of the CED-10 Rac and CED-1 pathways. We found that the RING finger domain of SLI-1 is not essential to rescue the effects of SLI-1 deletion on cell migration, suggesting that its role in this process is ubiquitin ligase-independent. We propose that SLI-1 opposes the engulfment of apoptotic cells via a previously unidentified pathway., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2012
- Full Text
- View/download PDF
24. Abl kinase inhibits the engulfment of apoptotic [corrected] cells in Caenorhabditis elegans.
- Author
-
Hurwitz ME, Vanderzalm PJ, Bloom L, Goldman J, Garriga G, and Horvitz HR
- Subjects
- Animals, Caenorhabditis elegans growth & development, Caenorhabditis elegans physiology, Cell Movement physiology, Cytoskeleton physiology, Genes, abl, Metabolic Networks and Pathways, Morphogenesis physiology, Proto-Oncogene Proteins c-crk metabolism, rac GTP-Binding Proteins metabolism, Apoptosis physiology, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Cytoskeletal Proteins metabolism, Cytoskeleton metabolism, Proto-Oncogene Proteins c-abl metabolism
- Abstract
The engulfment of apoptotic cells is required for normal metazoan development and tissue remodeling. In Caenorhabditis elegans, two parallel and partially redundant conserved pathways act in cell-corpse engulfment. One pathway includes the adaptor protein CED-2 CrkII and the small GTPase CED-10 Rac, and acts to rearrange the cytoskeleton of the engulfing cell. The other pathway includes the receptor tyrosine kinase CED-1 and might recruit membranes to extend the surface of the engulfing cell. Although many components required for engulfment have been identified, little is known about inhibition of engulfment. The tyrosine kinase Abl regulates the actin cytoskeleton in mammals and Drosophila in multiple ways. For example, Abl inhibits cell migration via phosphorylation of CrkII. We tested whether ABL-1, the C. elegans ortholog of Abl, inhibits the CED-2 CrkII-dependent engulfment of apoptotic cells. Our genetic studies indicate that ABL-1 inhibits apoptotic cell engulfment, but not through CED-2 CrkII, and instead acts in parallel to the two known engulfment pathways. The CED-10 Rac pathway is also required for proper migration of the distal tip cells (DTCs) during the development of the C. elegans gonad. The loss of ABL-1 function partially restores normal DTC migration in the CED-10 Rac pathway mutants. We found that ABI-1 the C. elegans homolog of mammalian Abi (Abl interactor) proteins, is required for engulfment of apoptotic cells and proper DTC migration. Like Abl, Abi proteins are cytoskeletal regulators. ABI-1 acts in parallel to the two known engulfment pathways, likely downstream of ABL-1. ABL-1 and ABI-1 interact physically in vitro. We propose that ABL-1 opposes the engulfment of apoptotic cells by inhibiting ABI-1 via a pathway that is distinct from the two known engulfment pathways., Competing Interests: Competing interests. The authors have declared that no competing interests exist.
- Published
- 2009
- Full Text
- View/download PDF
25. C. elegans CARMIL negatively regulates UNC-73/Trio function during neuronal development.
- Author
-
Vanderzalm PJ, Pandey A, Hurwitz ME, Bloom L, Horvitz HR, and Garriga G
- Subjects
- Amino Acid Sequence, Animals, Animals, Genetically Modified, Base Sequence, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins chemistry, Caenorhabditis elegans Proteins genetics, Cell Movement, Cloning, Molecular, DNA Primers genetics, Down-Regulation, Gene Expression Regulation, Developmental, Genes, Helminth, Models, Neurological, Molecular Sequence Data, Mutation, Nerve Tissue Proteins genetics, Neurogenesis genetics, Neurogenesis physiology, Neurons cytology, Receptors, Immunologic genetics, Receptors, Immunologic metabolism, Sequence Homology, Amino Acid, Signal Transduction, rac GTP-Binding Proteins metabolism, Roundabout Proteins, Caenorhabditis elegans growth & development, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Nerve Tissue Proteins metabolism, Neurons metabolism
- Abstract
Whereas many molecules that promote cell and axonal growth cone migrations have been identified, few are known to inhibit these processes. In genetic screens designed to identify molecules that negatively regulate such migrations, we identified CRML-1, the C. elegans homolog of CARMIL. Although mammalian CARMIL acts to promote the migration of glioblastoma cells, we found that CRML-1 acts as a negative regulator of neuronal cell and axon growth cone migrations. Genetic evidence indicates that CRML-1 regulates these migrations by inhibiting the Rac GEF activity of UNC-73, a homolog of the Rac and Rho GEF Trio. The antagonistic effects of CRML-1 and UNC-73 can control the direction of growth cone migration by regulating the levels of the SAX-3 (a Robo homolog) guidance receptor. Consistent with the hypothesis that CRML-1 negatively regulates UNC-73 activity, these two proteins form a complex in vivo. Based on these observations, we propose a role for CRML-1 as a novel regulator of cell and axon migrations that acts through inhibition of Rac signaling.
- Published
- 2009
- Full Text
- View/download PDF
26. Hypersensitivity to inhaled TOBI following reaction to gentamicin.
- Author
-
Spigarelli MG, Hurwitz ME, and Nasr SZ
- Subjects
- Administration, Inhalation, Anti-Bacterial Agents therapeutic use, Child, Cystic Fibrosis microbiology, Desensitization, Immunologic, Gentamicins therapeutic use, Humans, Male, Pseudomonas Infections prevention & control, Skin Tests, Tobramycin administration & dosage, Anti-Bacterial Agents adverse effects, Cystic Fibrosis drug therapy, Drug Eruptions etiology, Gentamicins adverse effects, Tobramycin adverse effects
- Abstract
Cystic fibrosis (CF) is the most common autosomal-recessive disease in Caucasians. Colonization with Pseudomonas aeruginosa (P. aeruginosa) of the CF airways causes deterioration of pulmonary status. TOBI (Tobramycin solution for inhalation) is an inhaled antibiotic that can improve the pulmonary disease. We report on a 9-year old boy with CF who developed a rash following a course of IV gentamicin. The rash resolved after its discontinuation. However, the rash returned all over his body, with the start of inhalation of TOBI therapy. We desensitized the patient using escalating doses of inhaled TOBI. He tolerated the procedure well, and continues to be on TOBI 9 months after desensitization on a once-a-day regimen., (Copyright 2002 Wiley-Liss, Inc.)
- Published
- 2002
- Full Text
- View/download PDF
27. Influence of sociodemographics on the health-related quality of life of pediatric patients with asthma and their caregivers.
- Author
-
Erickson SR, Munzenberger PJ, Plante MJ, Kirking DM, Hurwitz ME, and Vanuya RZ
- Subjects
- Adolescent, Adult, Child, Female, Humans, Income, Male, Regression Analysis, Asthma physiopathology, Caregivers, Health Status, Quality of Life, Socioeconomic Factors
- Abstract
The relationship between socioeconomic variables and the health-related quality of life (HQL) of children with asthma and their caregivers was examined. The Pediatric Asthma Quality of Life Questionnaire (PAQLQ) and Pediatric Asthma Caregivers Quality of Life Questionnaire (PACQLQ) were administered to 99 pediatric asthmatic patients and caregivers in two specialty clinics. Sociodemographic data was obtained from medical records and additional questions. The relationship between sociodemographic variables and HQL was determined using multiple linear regression. The mean patient age was 12.6+/-2.1 years, more were male and from a minority race. The mean age of caregivers was 41.2+/-8.5 years; most were female and were fom a minority race. Patients tended to rate their asthma severity as mild to moderate, while caregivers tended to rate patients in the moderate to severe category. Based on prescribed medications, most patients had mild to moderate asthma. Household income was consistently associated with patient-perceived HQL. Less consistent associations were seen with other variables. Household income and the caregiver's perception of asthma severity were associated with all caregiver HQL domains. It was concluded that household income was most consistently associated with the HQL of asthmatic pediatric patients and their caregivers.
- Published
- 2002
- Full Text
- View/download PDF
28. Use of computerized tomography and chest x-rays in evaluating efficacy of aerosolized recombinant human DNase in cystic fibrosis patients younger than age 5 years: a preliminary study.
- Author
-
Nasr SZ, Kuhns LR, Brown RW, Hurwitz ME, Sanders GM, and Strouse PJ
- Subjects
- Administration, Inhalation, Child, Preschool, Double-Blind Method, Female, Humans, Infant, Male, Cystic Fibrosis diagnostic imaging, Cystic Fibrosis drug therapy, Deoxyribonuclease I administration & dosage, Deoxyribonuclease I therapeutic use, Expectorants administration & dosage, Expectorants therapeutic use, Lung diagnostic imaging, Radiography, Thoracic, Recombinant Proteins administration & dosage, Recombinant Proteins therapeutic use, Tomography, X-Ray Computed
- Abstract
The aim of this study was to evaluate the ability of high-resolution computerized tomography (HRCT) of the chest and chest x-rays (CXR) to determine efficacy of inhaled recombinant human DNase (rhDNase) in cystic fibrosis (CF) patients younger than 5 years of age. A randomized, double-blind, placebo-controlled pilot study of 12 patients with CF younger than 5 years of age, attending the University of Michigan Cystic Fibrosis Center (Ann Arbor, MI) was conducted. The changes in the HRCT and CXR score from baseline to day 100 of therapy were assessed using a previously validated scoring system. The mean changes of HRCT scores between the rhDNase and placebo groups were found to be significant at the 95% level, with mean change +/- SE mean of - 1.00 +/- 0.53 and 0.58 +/- 0.24 for rhDNase and placebo groups, respectively (P = 0.02). The difference in CXR score was not significant between the two groups. An analysis was performed to relate HRCT subscores to CXR score; only thickening of the intra-interlobular septae was significantly correlated with the total CXR score (r = - 0.7, P < 0.01). There was improvement in the parents' assessments of the patients' well-being, with improvement in physical activity, decreased cough, sleep quality, and appetite in those subjects receiving rhDNase. We conclude that the administration of rhDNase was associated with improvement in the HRCT scan in CF patients younger than 5 years of age. Findings indicate that HRCT of the chest is useful and sensitive in studying responses to therapy in patients with CF lung disease. To our knowledge, this is the first report of the use of HRCT to assess the effectiveness of a therapeutic modality in so young a CF patient population., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
29. Treatment of allergic rhinitis with antihistamines and decongestants and their effects on the lower airway.
- Author
-
Hurwitz ME
- Subjects
- Asthma diagnosis, Child, Child, Preschool, Controlled Clinical Trials as Topic, Female, Humans, Male, Prognosis, Respiratory System drug effects, Rhinitis, Allergic, Seasonal diagnosis, Treatment Outcome, Asthma drug therapy, Histamine H1 Antagonists therapeutic use, Nasal Decongestants therapeutic use, Rhinitis, Allergic, Seasonal drug therapy
- Published
- 2000
- Full Text
- View/download PDF
30. Treatment of anorexia and weight loss with megestrol acetate in patients with cystic fibrosis.
- Author
-
Nasr SZ, Hurwitz ME, Brown RW, Elghoroury M, and Rosen D
- Subjects
- Adolescent, Anorexia etiology, Child, Female, Humans, Male, Anorexia drug therapy, Appetite Stimulants therapeutic use, Cystic Fibrosis complications, Megestrol Acetate therapeutic use, Weight Loss
- Abstract
Four patients with severe cystic fibrosis lung disease, anorexia and weight loss, received Megestrol Acetate (MA), as an appetite stimulant. The initial dose was 400-800 mg daily and was continued for 6-15 months. Appetite was improved, with significant weight gain in all patients and an increase in their weight for age percentile from <5% at the start of the study to approximately the 25(th) percentile after 6 months of use and improvement in quality of life. One patient discontinued MA after 6 months, and subsequently appetite and weight were depressed., (Copyright 1999 Wiley-Liss, Inc.)
- Published
- 1999
- Full Text
- View/download PDF
31. Two yeast nuclear pore complex proteins involved in mRNA export form a cytoplasmically oriented subcomplex.
- Author
-
Hurwitz ME, Strambio-de-Castillia C, and Blobel G
- Subjects
- Fungal Proteins metabolism, Gene Expression Regulation, Fungal genetics, Membrane Proteins metabolism, Microscopy, Immunoelectron, Nuclear Envelope ultrastructure, Nuclear Proteins deficiency, Phenotype, RNA Helicases, Repressor Proteins genetics, Nuclear Envelope metabolism, Nuclear Pore Complex Proteins, Nuclear Proteins metabolism, RNA, Messenger metabolism, Saccharomyces cerevisiae Proteins
- Abstract
We sublocalized the yeast nucleoporin Nup82 to the cytoplasmic side of the nuclear pore complex (NPC) by immunoelectron microscopy. Moreover, by in vitro binding assays we showed that Nup82 interacts with the C-terminal region of Nup159, a yeast nucleoporin that previously was also localized to the cytoplasmic side of the NPC. Hence, the two nucleoporins, Nup82 and Nup159, form a cytoplasmically oriented subcomplex that is likely to be part of the fibers emanating from the cytoplasmic ring of the NPC. Overexpression of Rss1/Gle1, a putative nucleoporin and/or mRNA transport factor, was shown previously to partially rescue depletion of Nup159. We show here that overexpression of Rss1/Gle1 also partially rescued depletion of Nup82. Depletion of either Nup82, Nup159, or Rss1/Gle1 was shown previously to inhibit mRNA export. As was reported previously for depletion of Nup159 or of Rss1/Gle1, we show here that depletion of Nup82 has no detectable effect on classical nuclear localization sequence-mediated nuclear import. In summary, the nucleoporins Nup159 and Nup82 form a cytoplasmically oriented subcomplex of the NPC that is likely associated with Rss1/Gle1; this complex is essential for RNA export, but not for classical nuclear localization sequence-mediated nuclear protein import.
- Published
- 1998
- Full Text
- View/download PDF
32. Physician-patient partnership in managing chronic illness.
- Author
-
Clark NM, Nothwehr F, Gong M, Evans D, Maiman LA, Hurwitz ME, Roloff D, and Mellins RB
- Subjects
- Clinical Competence, Communication, Education, Medical, Continuing, Health Behavior, Health Status, Humans, Patient Compliance, Patient Education as Topic, Patient Participation, Practice Patterns, Physicians', Chronic Disease, Physician-Patient Relations
- Published
- 1995
- Full Text
- View/download PDF
33. NUP82 is an essential yeast nucleoporin required for poly(A)+ RNA export.
- Author
-
Hurwitz ME and Blobel G
- Subjects
- Amino Acid Sequence, Base Sequence, Cloning, Molecular, Molecular Sequence Data, Nuclear Proteins chemistry, Nuclear Proteins genetics, Sequence Analysis, Temperature, Nuclear Pore Complex Proteins, Nuclear Proteins isolation & purification, Nuclear Proteins metabolism, RNA, Messenger metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins
- Abstract
We have isolated and characterized the gene encoding a novel essential nucleoporin of 82 kD, termed NUP82. Indirect immunofluorescence of cells containing an epitope tagged copy of the NUP82 localized it to the nuclear pore complex (NPC). Primary structure analysis indicates that the COOH-terminal 195 amino acids contain a putative coiled-coil domain. Deletion of the COOH-terminal 87 amino acids of this domain causes slower cell growth; deletion of the COOH-terminal 108 amino acids results in slower growth at 30 degrees C and lethality at 37 degrees C. Cells in which the last 108 amino acids of NUP82 have been deleted, when shifted to 37 degrees C, do not display any gross morphological defects in their nuclear pore complexes or nuclear envelopes. They do, however, accumulate poly(A)+ RNA in their nuclei at 37 degrees C. We propose that NUP82 acts as a linker to tether nucleoporins directly involved in nuclear transport to pore scaffolding via its coiled-coil domain.
- Published
- 1995
- Full Text
- View/download PDF
34. Heparin-precipitable cryoprecipitate in a child with cold urticaria.
- Author
-
Hurwitz ME, Salberg DJ, and Mathews KP
- Subjects
- Chemical Precipitation, Child, Female, Heparin, Humans, Urticaria blood, Cold Temperature adverse effects, Cryoglobulins analysis, Urticaria etiology
- Published
- 1980
- Full Text
- View/download PDF
35. Clinical scoring does not accurately assess hypoxemia in pediatric asthma patients.
- Author
-
Hurwitz ME, Burney RE, Howatt WF, Crowley D, and Mackenzie JR
- Subjects
- Acute Disease, Adolescent, Child, Child, Preschool, Female, Humans, Male, Partial Pressure, Prospective Studies, Asthma complications, Hypoxia diagnosis, Oxygen blood
- Abstract
Management of acute asthma in the pediatric population is based almost entirely on clinical evidence of severity. Although pulmonary function testing has been advocated to improve evaluation, it is difficult in the pediatric patient and not routinely practiced. A clinical scoring system has been devised to help standardize evaluation, but has not been validated by comparison of the results of clinical scoring with those of arterial blood oxygen levels as determined by blood gas analysis. This study was undertaken to compare clinical scoring of pediatric asthma patients with the results of arterial blood gas analysis. Thirty-eight children between the ages of 2 and 13 having 42 episodes of acute asthma were evaluated. The average age was 5.4 years. The average clinical score was 2.62; arterial blood for analysis was obtained in 37 (88%), with an average PaO2 of 81.7 mm Hg. None of the children had CO2 retention. There was no correlation between the clinical score of the children on presentation and the severity of hypoxia (correlation coefficient = -0.149). Comparison of age and arterial oxygen tension revealed a trend toward worsening hypoxemia with diminishing age from 6 to 2 years, which was not identified by clinical scoring. We conclude that clinical scoring is inaccurate for the assessment of hypoxemia in the pediatric age group. Arterial blood gas determination should be used to assess the severity of hypoxemia in the emergency treatment of pediatric asthma patients.
- Published
- 1984
- Full Text
- View/download PDF
36. Avulsed vertebral rim apophysis in a child.
- Author
-
Gooding CA and Hurwitz ME
- Subjects
- Adolescent, Diagnosis, Differential, Exostoses surgery, Humans, Intervertebral Disc Displacement surgery, Lumbar Vertebrae diagnostic imaging, Male, Radiography, Exostoses diagnostic imaging, Intervertebral Disc Displacement diagnostic imaging, Lumbar Vertebrae injuries
- Published
- 1974
- Full Text
- View/download PDF
37. Improvement in clinical asthma score and PaCO2 in children with severe asthma treated with continuously nebulized terbutaline.
- Author
-
Moler FW, Hurwitz ME, and Custer JR
- Subjects
- Administration, Inhalation instrumentation, Administration, Inhalation methods, Adolescent, Asthma blood, Asthma physiopathology, Child, Child, Preschool, Drug Administration Schedule, Female, Heart Rate drug effects, Humans, Infant, Infusions, Intravenous, Isoproterenol therapeutic use, Male, Partial Pressure, Asthma drug therapy, Carbon Dioxide blood, Nebulizers and Vaporizers, Terbutaline administration & dosage
- Abstract
We analyzed continuous nebulized terbutaline (CNT) therapy in 19 patients with 27 admissions for severe asthma and impending respiratory failure who failed to respond to our standard asthma protocol of methylprednisolone, aminophylline, and intermittently nebulized terbutaline. Terbutaline was administered by continuous face mask nebulization at a dose equaling the most frequent previous intermittent dose per hour (4 mg per hour). No patient with frank respiratory failure (i.e., PaCO2 greater than or equal to 60 torr, exhaustion, or coma) was studied. All patients improved, and therapy was stopped in a mean of 15.4 hours (range 3 to 37 hours). The average heart rate did not increase over baseline measurements through 24 hours of CNT. The mean clinical asthma score improved significantly during 8 hours, falling from 6.9 to 3.2 (p greater than 0.001). In 14 patients whose PaCO2 was greater than or equal to 39 torr (range 39 to 58 torr) and clinical asthma score was 6 or greater, PaCO2 decreased a mean of 11.7 torr during a mean of 8.1 hours. In six patients whose PaCO2 was 45 torr or greater at the start of CNT (mean 49, range 45 to 58 torr) and in whom we would have previously treated with intravenous isoproterenol, PaCO2 decreased a mean of 15 torr in an average of 8.7 hours. This preliminary study suggests that CNT is an effective therapy for severe asthma in children.
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.