10 results on '"Craig Tendler"'
Search Results
2. Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease
- Author
-
Manjula Reddy, Sheila K Pierson, Craig Tendler, Alessia Dalla Pria, Alexander Fosså, Philip Beineke, Nancy A. Shadick, Mark Bower, Sushila A. Shenoy, Christopher S. Nabel, Alexander M. Gorzewski, Michael E. Weinblatt, Sophia A. T. Parente, Jason R. Ruth, Mary Guilfoyle, Laura M. Katz, Ana B Oromendia, Daniel J. Arenas, Taku Kambayashi, David M. Lee, Ruth-Anne Langan Pai, David C. Fajgenbaum, Frits van Rhee, Yasufumi Masaki, and Jason G. Mezey
- Subjects
Oncology ,Proteomics ,medicine.medical_specialty ,Immunobiology and Immunotherapy ,medicine.medical_treatment ,Disease ,Systemic inflammation ,Siltuximab ,chemistry.chemical_compound ,Internal medicine ,medicine ,Humans ,Interleukin 6 ,biology ,business.industry ,Interleukin-6 ,Castleman Disease ,Hematology ,medicine.disease ,United States ,Cytokine ,chemistry ,Erythropoietin ,Rheumatoid arthritis ,Herpesvirus 8, Human ,biology.protein ,medicine.symptom ,business ,Janus kinase ,medicine.drug ,Signal Transduction - Abstract
Idiopathic multicentric Castleman disease (iMCD) is a poorly understood hematologic disorder involving cytokine-induced polyclonal lymphoproliferation, systemic inflammation, and potentially fatal multiorgan failure. Although the etiology of iMCD is unknown, interleukin-6 (IL-6) is an established disease driver in approximately one-third of patients. Anti–IL-6 therapy, siltuximab, is the only US Food and Drug Administration–approved treatment. Few options exist for siltuximab nonresponders, and no validated tests are available to predict likelihood of response. We procured and analyzed the largest-to-date cohort of iMCD samples, which enabled classification of iMCD into disease categories, discovery of siltuximab response biomarkers, and identification of therapeutic targets for siltuximab nonresponders. Proteomic quantification of 1178 analytes was performed on serum of 88 iMCD patients, 60 patients with clinico-pathologically overlapping diseases (human herpesvirus-8–associated MCD, N = 20; Hodgkin lymphoma, N = 20; rheumatoid arthritis, N = 20), and 42 healthy controls. Unsupervised clustering revealed iMCD patients have heterogeneous serum proteomes that did not cluster with clinico-pathologically overlapping diseases. Clustering of iMCD patients identified a novel subgroup with superior response to siltuximab, which was validated using a 7-analyte panel (apolipoprotein E, amphiregulin, serum amyloid P-component, inactivated complement C3b, immunoglobulin E, IL-6, erythropoietin) in an independent cohort. Enrichment analyses and immunohistochemistry identified Janus kinase (JAK)/signal transducer and activator of transcription 3 signaling as a candidate therapeutic target that could potentially be targeted with JAK inhibitors in siltuximab nonresponders. Our discoveries demonstrate the potential for accelerating discoveries for rare diseases through multistakeholder collaboration.
- Published
- 2021
3. Predictors of response to anti‐<scp>IL</scp>6 monoclonal antibody therapy (siltuximab) in idiopathic multicentric Castleman disease: secondary analyses of phase<scp>II</scp>clinical trial data
- Author
-
Craig Tendler, Mary Guilfoyle, Frits van Rhee, David C. Fajgenbaum, Dustin Shilling, Sepideh Nemat, Carlos Appiani, Deanna E Morra, and Sheila K Pierson
- Subjects
Adult ,Male ,medicine.medical_specialty ,Fibrinogen ,Models, Biological ,Gastroenterology ,Article ,Siltuximab ,Immunoglobulin G ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Predictive Value of Tests ,Internal medicine ,medicine ,Humans ,Monoclonal antibody therapy ,Univariate analysis ,biology ,Interleukin-6 ,business.industry ,Castleman Disease ,C-reactive protein ,Antibodies, Monoclonal ,Hematology ,Middle Aged ,Confidence interval ,Immunoglobulin A ,Clinical trial ,C-Reactive Protein ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Female ,business ,030215 immunology ,medicine.drug - Abstract
Siltuximab is the only US Food and Drug Administration-approved treatment for idiopathic multicentric Castleman disease (iMCD), a rare haematological disorder associated with substantial morbidity and mortality. Although siltuximab induces a response in a significant proportion of iMCD patients via interleukin (IL)-6 neutralization, it is not universally effective. We performed an in-depth analysis of 38 baseline laboratory parameters in iMCD patients from the phase II trial of siltuximab that met criteria for treatment response or treatment failure to develop a predictive model of response. Univariate analyses identified eight baseline laboratory parameters that were significantly different between responders and treatment failures: albumin, immunoglobulin G (IgG), immunoglobulin A, C reactive protein (CRP), fibrinogen, haemoglobin, sodium and triglycerides. Stepwise logistic regression analysis of these candidate parameters identified a top performing model that included fibrinogen, IgG, haemoglobin and CRP. Based on cross-validation of the final multivariate logistic regression model, the model accurately discriminated responders from those that failed treatment (area under the receiver operator characteristic curve 0.86 [95% confidence interval: 0.73–0.95]). All four laboratory parameters associated with response to siltuximab have biological relationships with IL6 and acute inflammation. Our model suggests that iMCD patients with laboratory evidence of an inflammatory syndrome are the best candidates for siltuximab therapy.
- Published
- 2018
4. Serum Proteomics Reveals Distinct Subtypes Associated with Treatment Response in Idiopathic Multicentric Castleman Disease
- Author
-
David C. Fajgenbaum, Philip Beineke, Jason R. Ruth, Manjula Reddy, Sheila K Pierson, Dustin Shilling, Laura M. Katz, Sofija Miljovska, Yasufumi Masaki, Jef Benbanaste, Alessia Dalla Pria, Michael E. Weinblatt, David M. Lee, Jason G. Mezey, Mark Bower, Mary Guilfoyle, Alexander Fosså, Christopher S. Nabel, Jeff Wiser, Frits van Rhee, Craig Tendler, Nancy A. Shadick, Ana B Oromendia, and Sushila A. Shenoy
- Subjects
Pathology ,medicine.medical_specialty ,Predictive marker ,biology ,business.industry ,Immunology ,Lymphoproliferative disorders ,Cell Biology ,Hematology ,medicine.disease ,Proteomics ,Biochemistry ,Siltuximab ,chemistry.chemical_compound ,chemistry ,Rheumatoid arthritis ,Proteome ,medicine ,biology.protein ,business ,Interleukin 6 ,Cytokine storm - Abstract
Human herpesvirus-8(HHV-8)-negative/idiopathic multicentric Castleman disease (iMCD) is a rare and poorly understood disorder diagnosed in ~1,000 individuals in the USA each year. It involves polyclonal lymphoproliferation, constitutional symptoms, systemic inflammation and an uncontrollable cytokine storm resulting in life-threatening multi-organ failure. Diagnosis and treatment can be difficult due to limited etiological understanding and heterogeneous presentation - clinical, laboratory, and histopathological abnormalities overlap with infectious, autoimmune and oncological diseases. iMCD symptoms and disease progression are largely believed to be driven by interleukin-6 (IL-6). However, approximately 66% of iMCD patients did not respond to anti-IL-6 therapy, siltuximab, the only FDA-approved iMCD therapy, in its phase II study (NCT01024036). Few treatment options exist for anti-IL-6 refractory patients because alternative driver cytokines and signaling pathways are not known. Herein we report the largest study to-date of iMCD serum proteomes with correlative anti-IL6 response data from 92 iMCD patients in disease flare (n=75 of which were collected as part of NCT01024036), in order to: (1) molecularly define iMCD, (2) identify predictors of response to anti-IL6 therapy, and (3) gain insights into the pathogenesis of iMCD. Proteomes of HHV8-positive MCD (n=20), Hodgkin lymphoma (n=20), rheumatoid arthritis (n=20) and healthy individuals (n=44) were also analyzed. Of the ~1,300 analytes measured using SomaLogic SOMAscan, 1,178 passed QC and were included in analyses. Each analyte was log2 transformed and capped at the 2.5th and 97.5th percentiles. Clinical and laboratory data collected at the time of sample draw were used to calculate disease activity following a modified CHAP scale: C-reactive protein, hemoglobin and albumin; missing performance status. Response to siltuximab was determined in NCT01024036. Data analysis was performed using the Medidata Rave Omics machine learning platform and R v3.4.4. Clustering of baseline proteomic data for iMCD patients identified six clusters that ranged in size from seven to 27 subjects. No associations with race, site, sex, age, or batch were found. Analytes identified among the strongest differentiators include cytokines, chemokines and inflammatory molecules. Interestingly, the largest cluster was associated with response to siltuximab (p Disclosures Guilfoyle: Janssen Pharmaceuticals, Inc.: Employment. Tendler:Janssen Pharmaceuticals, Inc.: Employment, Equity Ownership. Reddy:Janssen Pharmaceuticals, Inc.: Employment, Equity Ownership. Weinblatt:Amgen: Consultancy, Research Funding; Sanofi/Regeneron: Consultancy, Research Funding; Crescendo Bioscience: Consultancy, Research Funding; Bristol Myers Squibb: Consultancy, Research Funding; UCB Pharmaceuticals: Consultancy, Research Funding. Bower:Merck: Honoraria; ViiV: Honoraria; Gilead: Honoraria; Janssen Pharmaceuticals: Honoraria. Masaki:Ono: Research Funding; Phizer: Research Funding; Astellas: Research Funding; Eisai: Research Funding; Kyowa Hakko Kirin: Research Funding. Beineke:Medidata Solutions: Employment, Equity Ownership. Miljovska:Medidata Solutions: Employment. Katz:Medidata Solutions: Employment. Shenoy:Medidata Solutions: Consultancy. Oromendia:Medidata Solutions: Employment, Equity Ownership. Mezey:Medidata Solutions: Consultancy. Wiser:Medidata Solutions: Employment, Equity Ownership. Benbanaste:Medidata Solutions: Employment, Equity Ownership. Lee:Medidata Solutions: Employment. Fosså:Janssen Pharmaceuticals, Inc.: Honoraria. Fajgenbaum:Janssen Pharmaceuticals, Inc.: Research Funding.
- Published
- 2018
5. Randomized Phase III Trial of Pegylated Liposomal Doxorubicin Versus Vinorelbine or Mitomycin C Plus Vinblastine in Women With Taxane-Refractory Advanced Breast Cancer
- Author
-
Craig Tendler, Charles L. Vogel, Jean Marc Nabholtz, Robert G. Mennel, Lillian Mellars, Manfred Kaufmann, Anna Lluch, I. Craig Henderson, Gunther von Minckwitz, Leila Alland, Alan M. Keller, Vassilis A. Georgoulias, and Aura Erazo
- Subjects
Adult ,Bridged-Ring Compounds ,Cancer Research ,medicine.medical_specialty ,Mitomycin ,medicine.medical_treatment ,Urology ,Salvage therapy ,Breast Neoplasms ,Vinblastine ,Vinorelbine ,Breast cancer ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm Metastasis ,Aged ,Salvage Therapy ,Chemotherapy ,Taxane ,business.industry ,Mitomycin C ,Middle Aged ,medicine.disease ,Surgery ,Regimen ,Oncology ,Doxorubicin ,Drug Resistance, Neoplasm ,Liposomes ,Female ,Taxoids ,business ,medicine.drug - Abstract
Purpose To compare the efficacy of pegylated liposomal doxorubicin (PLD) with that of a common salvage regimen (comparator) in patients with taxane-refractory advanced breast cancer. Patients and Methods Following failure of a first- or second-line taxane-containing regimen for metastatic disease, 301 women were randomly assigned to receive PLD (50 mg/m2 every 28 days); or comparator-vinorelbine (30 mg/m2 weekly) or mitomycin C (10 mg/m2 day 1 and every 28 days) plus vinblastine (5 mg/m2 day 1, day 14, day 28, and day 42) every 6 to 8 weeks. Patients were stratified before random assignment based on number of previous chemotherapy regimens for metastatic disease and presence of bone metastases only. Results Progression-free survival (PFS) and overall survival (OS) were similar for PLD and comparator (PFS: hazard ratio [HR], 1.26; 95% CI, 0.98 to 1.62; P = .11; median, 2.9 months [PLD] and 2.5 months [comparator]; OS: HR, 1.05; 95% CI, 0.82 to 1.33; P = .71; median, 11.0 months [PLD] and 9.0 months [comparator]). In anthracycline-naïve patients, PFS was somewhat longer with PLD, relative to the comparator (n = 44; median PFS, 5.8 v 2.1 months; HR, 2.40; 95% CI, 1.16 to 4.95; P = .01). Most frequently reported adverse events were nausea (23% to 31%), vomiting (17% to 20%), and fatigue (9% to 20%) and were similar among treatment groups. PLD-treated patients experienced more palmar-plantar erythrodysesthesia (37%; 18% grade 3, 1 patient grade 4) and stomatitis (22%; 5% grades 3/4). Neuropathy (11%), constipation (16%), and neutropenia (14%) were more common with vinorelbine. Alopecia was low in both the PLD and vinorelbine groups (3% and 5%). Conclusion PLD has efficacy comparable to that of common salvage regimens in patients with taxane-refractory metastatic breast cancer, thereby representing a useful therapeutic option.
- Published
- 2004
6. Phase I Study of the Farnesyltransferase Inhibitor Lonafarnib with Paclitaxel in Solid Tumors
- Author
-
Dong M. Shin, Merrill S. Kies, Elizabeth Thompson, Bonnie S. Glisson, Edward S. Kim, Reginald F. Munden, Vassiliki A. Papadimitrakopoulou, W. Robert Bishop, Charles Lu, Xuemei Wang, Frank V. Fossella, Paul Kirschmeier, Peter F. Thall, Yali Zhu, Roy S. Herbst, Waun Ki Hong, Fadlo R. Khuri, Michael L. Meyers, Paul Statkevich, Craig Tendler, and Sandra Bangert
- Subjects
Adult ,Male ,Cancer Research ,Neutropenia ,Paclitaxel ,Pyridines ,Pharmacology ,chemistry.chemical_compound ,Piperidines ,Pharmacokinetics ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Farnesyltranstransferase ,Humans ,Medicine ,Lonafarnib ,Fatigue ,Aged ,Alkyl and Aryl Transferases ,Taxane ,Dose-Response Relationship, Drug ,business.industry ,Farnesyltransferase inhibitor ,Anemia ,Leukopenia ,Middle Aged ,medicine.disease ,Heart Arrest ,Regimen ,Treatment Outcome ,Oncology ,chemistry ,Tolerability ,Area Under Curve ,Female ,business - Abstract
Purpose: To establish the maximum tolerated dose of lonafarnib, a novel farnesyltransferase inhibitor, in combination with paclitaxel in patients with solid tumors and to characterize the safety, tolerability, dose-limiting toxicity, and pharmacokinetics of this combination regimen. Experimental Design: In a Phase I trial, lonafarnib was administered p.o., twice daily (b.i.d.) on continuously scheduled doses of 100 mg, 125 mg, and 150 mg in combination with i.v. paclitaxel at doses of 135 mg/m2 or 175 mg/m2 administered over 3 h on day 8 of every 21-day cycle. Plasma paclitaxel and lonafarnib concentrations were collected at selected time points from each patient. Results: Twenty-four patients were enrolled; 21 patients were evaluable. The principal grade 3/4 toxicity was diarrhea (5 of 21 patients), which was most likely due to lonafarnib. dose-limiting toxicities included grade 3 hyperbilirubinemia at dose level 3 (100 mg b.i.d. lonafarnib and 175 mg/m2 paclitaxel); grade 4 diarrhea and grade 3 peripheral neuropathy at dose level 3A (125 mg b.i.d. lonafarnib and 175 mg/m2 paclitaxel); and grade 4 neutropenia with fever and grade 4 diarrhea at level 4 (150 mg b.i.d. lonafarnib and 175 mg/m2 paclitaxel). The maximum tolerated dose established by the continual reassessment method was lonafarnib 100 mg b.i.d. and paclitaxel 175 mg/m2. Paclitaxel appeared to have no effect on the pharmacokinetics of lonafarnib. The median duration of therapy was eight cycles, including seven cycles with paclitaxel. Six of 15 previously treated patients had a durable partial response, including 3 patients who had previous taxane therapy. Notably, two of five patients with taxane-resistant metastatic non-small cell lung cancer had partial responses. Conclusions: When combined with paclitaxel, the recommended dose of lonafarnib for Phase II trials is 100 mg p.o. twice daily with 175 mg/m2 of paclitaxel i.v. every 3 weeks. Additional studies of lonafarnib in combination regimens appear warranted, particularly in patients with non-small cell lung cancer.
- Published
- 2004
7. Treating cancer with PEG Intron
- Author
-
Ronald M. Bukowski, Craig Tendler, F.R.C.P.C. David Cutler M.D., Esther Rose, Mark Laughlin, and Paul Statkevich
- Subjects
Cancer Research ,Dose-Response Relationship, Drug ,business.industry ,Area under the curve ,Interferon-alpha ,Cancer ,Alpha interferon ,Antineoplastic Agents ,Interferon alpha-2 ,Pharmacology ,medicine.disease ,Drug Administration Schedule ,Recombinant Proteins ,Polyethylene Glycols ,Oncology ,Pharmacokinetics ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Pharmacodynamics ,Immunology ,PEG ratio ,medicine ,Humans ,Dosing ,business ,Chronic myelogenous leukemia - Abstract
BACKGROUND PEG Intron (pegylated interferon-alpha-2b [IFN-α-2b]; Schering-Plough, Kenilworth, NJ) has demonstrated delayed clearance and increased area under the curve compared with native IFN-α-2b. Studies in patients with chronic hepatitis C infection and malignancies have demonstrated both biologic and clinical activity of PEG Intron and have provided empiric data to compare the pharmacokinetics (PK) and pharmacodynamics of PEG Intron and IFN-α-2b. METHODS The authors conducted a review of the available data comparing the PK and pharmacodynamic effects of PEG Intron and IFN-α-2b. Safety and efficacy data from Phase I/II studies of PEG Intron in patients with chronic myelogenous leukemia (CML) and solid tumors were also reviewed. RESULTS Data from patients with chronic hepatitis C infection suggest that exposure to IFN at a PEG Intron dose of 0.25 μg/kg per week is similar to that observed after administration of IFN-α-2b at a dose of 3 million International Units, three times per week. PEG Intron at doses up to 6 μg/kg per week was well tolerated and demonstrated clinical activity in patients with CML and solid tumors, including metastatic melanoma and renal cell carcinoma. CONCLUSIONS Dose intensification can be achieved safely in patients with CML and solid tumors using PEG Intron, which could improve efficacy. These results provide useful dosing guidelines to clinicians investigating the antitumor activity of PEG Intron in patients with malignancies. More data are needed to determine the optimal dose in various oncologic indications. However, these results provide a sound rationale for further investigation of PEG Intron. Cancer 2002;95:389–96. © 2002 American Cancer Society. DOI 10.1002/cncr.10663
- Published
- 2002
8. Recombinant interferon-? therapy in patients with follicular lymphoma
- Author
-
Howard Ozer M.D., Peter H. Wiernik, Craig Tendler, and Francis Giles
- Subjects
Oncology ,Chemotherapy ,medicine.medical_specialty ,Cancer Research ,business.industry ,medicine.medical_treatment ,Follicular lymphoma ,Induction chemotherapy ,Combination chemotherapy ,medicine.disease ,Minimal residual disease ,Non-Hodgkin's lymphoma ,Maintenance therapy ,hemic and lymphatic diseases ,Internal medicine ,Immunology ,medicine ,business ,Interferon alfa ,medicine.drug - Abstract
BACKGROUND Advanced stage, follicular, non-Hodgkin's lymphoma (NHL) has no cure and no single standard of care. Remissions induced by standard chemotherapy regimens generally are not durable and, with the exception of selected patients with limited early stage disease, most patients with follicular NHL eventually die of their disease. Recombinant interferon-α (rIFN-α) has demonstrated activity against follicular NHL in clinical trials. METHODS A comprehensive survey of current therapeutic options for follicular NHL patients was conducted with emphasis on the role of rIFN-α used in conjunction with chemotherapy regimens. RESULTS Phase III studies have demonstrated that rIFN-α delays disease progression and may improve overall survival when administered either with chemotherapy or as maintenance therapy after induction treatment for follicular lymphoma. Adverse effects from combination or maintenance regimens are not significantly different from those from chemotherapy alone. CONCLUSIONS Recombinant IFN-α is safe and effective when given in conjunction with standard chemotherapeutic regimens in selected patients with follicular NHL, and may especially benefit patients with minimal residual disease after induction chemotherapy. Cancer 1998;82:1821-30. © 1998 American Cancer Society.
- Published
- 1998
9. Medication dosages during pediatric emergencies: a simple and comprehensive guide
- Author
-
CRAIG TENDLER, SUSAN GROSSMAN, and JUDITH TENENBAUM
- Subjects
Adolescent ,Pharmaceutical Preparations ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Body Weight ,Infant, Newborn ,Humans ,Infant ,Emergencies ,Child - Abstract
Drug dosing during life-threatening pediatric emergencies is a source of stress for most physicians and nurses. This can be attributed to the lack of standardized drug doses for most pediatric medications, thus requiring time-consuming calculations with small margins of error. Anxiety may be further heightened by the infrequent occurrence of pediatric emergencies, resulting in a staffs limited experience with such crises. In an effort to reduce the potential for error and anxiety during administration of these pediatric critical care drugs, a majority of the major medical centers are currently using medication tables. Many prototypes have been published in the literature, but most require calculations and are incomplete in their content.
- Published
- 1989
10. Fusospirochetal ulcerative gingivitis in children
- Author
-
Edward J. Bottone and Craig Tendler
- Subjects
Male ,medicine.medical_specialty ,business.industry ,Spirochaetales Infections ,Dermatology ,Gingivitis, Necrotizing Ulcerative ,Stomatitis, Herpetic ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,medicine ,Fusobacterium Infections ,Ulcerative gingivitis ,Humans ,business - Published
- 1987
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.