19 results on '"Kurzrock R"'
Search Results
2. The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians.
- Author
-
Subbiah, V, Solit, DB, Chan, TA, and Kurzrock, R
- Subjects
Oncology & Carcinogenesis ,Oncology and Carcinogenesis - Published
- 2020
3. Targeted therapy for advanced salivary gland carcinoma based on molecular profiling: results from MyPathway, a phase IIa multiple basket study
- Author
-
Kurzrock, R, Bowles, DW, Kang, H, Meric-Bernstam, F, Hainsworth, J, Spigel, DR, Bose, R, Burris, H, Sweeney, CJ, Beattie, MS, Blotner, S, Schulze, K, Cuchelkar, V, and Swanton, C
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Clinical Research ,Digestive Diseases ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Good Health and Well Being ,Antineoplastic Combined Chemotherapy Protocols ,Breast Neoplasms ,Carcinoma ,Humans ,Molecular Targeted Therapy ,Receptor ,ErbB-2 ,Salivary Gland Neoplasms ,Salivary Glands ,Trastuzumab ,advanced salivary gland carcinoma ,targeted therapy ,HER2-positive ,pertuzumab ,trastuzumab ,molecular profiling ,Receptor ,erbB-2 ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
BackgroundSystemic therapy options for salivary cancers are limited. MyPathway (NCT02091141), a phase IIa study, evaluates targeted therapies in non-indicated tumor types with actionable molecular alterations. Here, we present the efficacy and safety results for a subgroup of MyPathway patients with advanced salivary gland cancer (SGC) matched to targeted therapies based on tumor molecular characteristics.Patients and methodsMyPathway is an ongoing, multiple basket, open-label, non-randomized, multi-center study. Patients with advanced SGC received pertuzumab + trastuzumab (HER2 alteration), vismodegib (PTCH-1/SMO mutation), vemurafenib (BRAF V600 mutation), or atezolizumab [high tumor mutational burden (TMB)]. The primary endpoint is the objective response rate (ORR).ResultsAs of January 15, 2018, 19 patients with SGC were enrolled and treated in MyPathway (15 with HER2 amplification and/or overexpression and one each with a HER2 mutation without amplification or overexpression, PTCH-1 mutation, BRAF mutation, and high TMB). In the 15 patients with HER2 amplification/overexpression (with or without mutations) who were treated with pertuzumab + trastuzumab, 9 had an objective response (1 complete response, 8 partial responses) for an ORR of 60% (9.2 months median response duration). The clinical benefit rate (defined by patients with objective responses or stable disease >4 months) was 67% (10/15), median progression-free survival (PFS) was 8.6 months, and median overall survival was 20.4 months. Stable disease was observed in the patient with a HER2 mutation (pertuzumab + trastuzumab, n = 1/1, PFS 11.0 months), and partial responses in patients with the PTCH-1 mutation (vismodegib, n = 1/1, PFS 14.3 months), BRAF mutation (vemurafenib, n = 1/1, PFS 18.5 months), and high TMB (atezolizumab, n = 1/1, PFS 5.5+ months). No unexpected toxicity occurred.ConclusionsOverall, 12 of 19 patients (63%) with advanced SGC, treated with chemotherapy-free regimens matched to specific molecular alterations, experienced an objective response. Data from MyPathway suggest that matched targeted therapy for SGC has promising efficacy, supporting molecular profiling in treatment determination.
- Published
- 2020
4. Targeted therapy for advanced salivary gland carcinoma based on molecular profiling: results from MyPathway, a phase IIa multiple basket study.
- Author
-
Kurzrock, R, Bowles, DW, Kang, H, Meric-Bernstam, F, Hainsworth, J, Spigel, DR, Bose, R, Burris, H, Sweeney, CJ, Beattie, MS, Blotner, S, Schulze, K, Cuchelkar, V, and Swanton, C
- Subjects
Salivary Glands ,Humans ,Carcinoma ,Breast Neoplasms ,Salivary Gland Neoplasms ,Receptor ,erbB-2 ,Antineoplastic Combined Chemotherapy Protocols ,Molecular Targeted Therapy ,Trastuzumab ,HER2-positive ,advanced salivary gland carcinoma ,molecular profiling ,pertuzumab ,targeted therapy ,trastuzumab ,Oncology & Carcinogenesis ,Oncology and Carcinogenesis - Abstract
BackgroundSystemic therapy options for salivary cancers are limited. MyPathway (NCT02091141), a phase IIa study, evaluates targeted therapies in non-indicated tumor types with actionable molecular alterations. Here, we present the efficacy and safety results for a subgroup of MyPathway patients with advanced salivary gland cancer (SGC) matched to targeted therapies based on tumor molecular characteristics.Patients and methodsMyPathway is an ongoing, multiple basket, open-label, non-randomized, multi-center study. Patients with advanced SGC received pertuzumab + trastuzumab (HER2 alteration), vismodegib (PTCH-1/SMO mutation), vemurafenib (BRAF V600 mutation), or atezolizumab [high tumor mutational burden (TMB)]. The primary endpoint is the objective response rate (ORR).ResultsAs of January 15, 2018, 19 patients with SGC were enrolled and treated in MyPathway (15 with HER2 amplification and/or overexpression and one each with a HER2 mutation without amplification or overexpression, PTCH-1 mutation, BRAF mutation, and high TMB). In the 15 patients with HER2 amplification/overexpression (with or without mutations) who were treated with pertuzumab + trastuzumab, 9 had an objective response (1 complete response, 8 partial responses) for an ORR of 60% (9.2 months median response duration). The clinical benefit rate (defined by patients with objective responses or stable disease >4 months) was 67% (10/15), median progression-free survival (PFS) was 8.6 months, and median overall survival was 20.4 months. Stable disease was observed in the patient with a HER2 mutation (pertuzumab + trastuzumab, n = 1/1, PFS 11.0 months), and partial responses in patients with the PTCH-1 mutation (vismodegib, n = 1/1, PFS 14.3 months), BRAF mutation (vemurafenib, n = 1/1, PFS 18.5 months), and high TMB (atezolizumab, n = 1/1, PFS 5.5+ months). No unexpected toxicity occurred.ConclusionsOverall, 12 of 19 patients (63%) with advanced SGC, treated with chemotherapy-free regimens matched to specific molecular alterations, experienced an objective response. Data from MyPathway suggest that matched targeted therapy for SGC has promising efficacy, supporting molecular profiling in treatment determination.
- Published
- 2020
5. Level of evidence used in recommendations by the National Comprehensive Cancer Network (NCCN) guidelines beyond Food and Drug Administration approvals
- Author
-
Kurzrock, R, Gurski, LA, Carlson, RW, Ettinger, DS, Horwitz, SM, Kumar, SK, Million, L, von Mehren, M, and Benson, AB
- Subjects
Cancer ,Prevention ,Clinical Research ,Comparative Effectiveness Research ,Clinical Trials and Supportive Activities ,6.1 Pharmaceuticals ,5.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Development of treatments and therapeutic interventions ,Antineoplastic Agents ,Drug Approval ,Evidence-Based Medicine ,Humans ,Neoplasms ,Off-Label Use ,Patient Care Management ,Practice Guidelines as Topic ,Prognosis ,Randomized Controlled Trials as Topic ,United States ,United States Food and Drug Administration ,oncology ,guidelines ,off-label drug use ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
BackgroundA previous analysis of 113 National Comprehensive Cancer Network® (NCCN®) recommendations reported that NCCN frequently recommends beyond Food and Drug Administration (FDA)-approved indications (44 off-label recommendations) and claimed that the evidence for these recommendations was weak.MethodsIn order to determine the strength of the evidence, we carried out an in-depth re-analysis of the 44 off-label recommendations listed in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®).ResultsOf the 44 off-label recommendations, 14 were later approved by the FDA and/or are supported by randomized controlled trial (RCT) data. In addition, 13 recommendations were either very minor extrapolations from the FDA label (n = 8) or were actually on-label (n = 5). Of the 17 remaining extrapolations, 8 were for mechanism-based agents applied in rare cancers or subsets with few available treatment options (median response rate = 43%), 7 were based on non-RCT data showing significant efficacy (>50% response rates), and 2 were later removed from the NCCN Guidelines because newer therapies with better activity and/or safety became available.ConclusionOff-label drug use is a frequent component of care for patients with cancer in the United States. Our findings indicate that when the NCCN recommends beyond the FDA-approved indications, the strength of the evidence supporting such recommendations is robust, with a significant subset of these drugs later becoming FDA approved or supported by RCT. Recommendations without RCT data are often for mechanism-based drugs with high response rates in rare cancers or subsets without effective therapies.
- Published
- 2019
6. Level of evidence used in recommendations by the National Comprehensive Cancer Network (NCCN) guidelines beyond Food and Drug Administration approvals.
- Author
-
Kurzrock, R, Gurski, LA, Carlson, RW, Ettinger, DS, Horwitz, SM, Kumar, SK, Million, L, von Mehren, M, and Benson, AB
- Subjects
Humans ,Neoplasms ,Antineoplastic Agents ,Prognosis ,Drug Approval ,Evidence-Based Medicine ,United States Food and Drug Administration ,Patient Care Management ,United States ,Randomized Controlled Trials as Topic ,Practice Guidelines as Topic ,Off-Label Use ,guidelines ,off-label drug use ,oncology ,Prevention ,Cancer ,Clinical Trials and Supportive Activities ,Comparative Effectiveness Research ,Clinical Research ,5.1 Pharmaceuticals ,6.1 Pharmaceuticals ,Oncology & Carcinogenesis ,Oncology and Carcinogenesis - Abstract
BackgroundA previous analysis of 113 National Comprehensive Cancer Network® (NCCN®) recommendations reported that NCCN frequently recommends beyond Food and Drug Administration (FDA)-approved indications (44 off-label recommendations) and claimed that the evidence for these recommendations was weak.MethodsIn order to determine the strength of the evidence, we carried out an in-depth re-analysis of the 44 off-label recommendations listed in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®).ResultsOf the 44 off-label recommendations, 14 were later approved by the FDA and/or are supported by randomized controlled trial (RCT) data. In addition, 13 recommendations were either very minor extrapolations from the FDA label (n = 8) or were actually on-label (n = 5). Of the 17 remaining extrapolations, 8 were for mechanism-based agents applied in rare cancers or subsets with few available treatment options (median response rate = 43%), 7 were based on non-RCT data showing significant efficacy (>50% response rates), and 2 were later removed from the NCCN Guidelines because newer therapies with better activity and/or safety became available.ConclusionOff-label drug use is a frequent component of care for patients with cancer in the United States. Our findings indicate that when the NCCN recommends beyond the FDA-approved indications, the strength of the evidence supporting such recommendations is robust, with a significant subset of these drugs later becoming FDA approved or supported by RCT. Recommendations without RCT data are often for mechanism-based drugs with high response rates in rare cancers or subsets without effective therapies.
- Published
- 2019
7. Preoperative Circulating Tumor DNA in Patients with Peritoneal Carcinomatosis Is an Independent Predictor of Progression-Free Survival
- Author
-
Baumgartner, J, Banks, K, Lanman, R, Tran, L, Kelly, KJ, Lowy, A, and Kurzrock, R
- Subjects
Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Published
- 2018
8. Retreatment with anti-EGFR based therapies in metastatic colorectal cancer: impact of intervening time interval and prior anti-EGFR response
- Author
-
Liu, X, George, GC, Tsimberidou, AM, Naing, A, Wheler, JJ, Kopetz, S, Fu, S, Piha-Paul, SA, Eng, C, Falchook, GS, Janku, F, Garrett, C, Karp, D, Kurzrock, R, Zinner, R, Raghav, K, Subbiah, V, Hess, K, Meric-Bernstam, F, Hong, DS, and Overman, MJ
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Digestive Diseases ,Clinical Research ,Colo-Rectal Cancer ,Adult ,Aged ,Antibodies ,Monoclonal ,Antibodies ,Monoclonal ,Humanized ,Cetuximab ,Colorectal Neoplasms ,Disease-Free Survival ,ErbB Receptors ,Erlotinib Hydrochloride ,Female ,Humans ,Male ,Middle Aged ,Neoplasm Metastasis ,Panitumumab ,Proto-Oncogene Proteins p21(ras) ,Retreatment ,Anti-EGFR treatment ,KRAS-wt CRC ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis ,Epidemiology - Abstract
BackgroundThis retrospective study aims to investigate the activity of retreatment with anti-EGFR-based therapies in order to explore the concept of clonal evolution by evaluating the impact of prior activity and intervening time interval.MethodsEighty-nine KRAS exon 2-wild-type metastatic colorectal patients were retreated on phase I/II clinical trials containing anti-EGFR therapies after progressing on prior cetuximab or panitumumab. Response on prior anti-EGFR therapy was defined retrospectively per physician-records as response or stable disease ≥6 months. Multivariable statistical methods included a multiple logistic regression model for response, and Cox proportional hazards model for progression-free survival.ResultsRetreatment anti-EGFR agents were cetuximab (n = 76) or cetuximab plus erlotinib (n = 13). The median interval time between prior and retreatment regimens was 4.57 months (range: 0.46-58.7). Patients who responded to the prior cetuximab or panitumumab were more likely to obtain clinical benefit to the retreatment compared to the non-responders in both univariate (p = 0.007) and multivariate analyses (OR: 3.38, 95 % CI: 1.27, 9.31, p = 0.019). The clinical benefit rate on retreatment also showed a marginally significant association with interval time between the two anti-EGFR based therapies (p = 0.053). Median progression-free survival on retreatment was increased in prior responders (4.9 months, 95 % CI: 3.6, 6.2) compared to prior non-responders (2.5 months, 95 % CI, 1.58, 3.42) in univariate (p = 0.064) and multivariate analysis (HR: 0.70, 95 % CI: 0.43-1.15, p = 0.156).ConclusionOur data lends support to the concept of clonal evolution, though the clinical impact appears less robust than previously reported. Further work to determine which patients benefit from retreatment post progression is needed.
- Published
- 2015
9. Retreatment with anti-EGFR based therapies in metastatic colorectal cancer: impact of intervening time interval and prior anti-EGFR response.
- Author
-
Liu, X, George, GC, Tsimberidou, AM, Naing, A, Wheler, JJ, Kopetz, S, Fu, S, Piha-Paul, SA, Eng, C, Falchook, GS, Janku, F, Garrett, C, Karp, D, Kurzrock, R, Zinner, R, Raghav, K, Subbiah, V, Hess, K, Meric-Bernstam, F, Hong, DS, and Overman, MJ
- Subjects
Humans ,Colorectal Neoplasms ,Neoplasm Metastasis ,Antibodies ,Monoclonal ,Disease-Free Survival ,Retreatment ,Adult ,Aged ,Middle Aged ,Female ,Male ,Proto-Oncogene Proteins p21(ras) ,Antibodies ,Monoclonal ,Humanized ,ErbB Receptors ,Cetuximab ,Erlotinib Hydrochloride ,Panitumumab ,Anti-EGFR treatment ,KRAS-wt CRC ,Receptor ,Epidermal Growth Factor ,Antibodies ,Monoclonal ,Humanized ,Oncology and Carcinogenesis ,Public Health and Health Services ,Oncology & Carcinogenesis - Abstract
BackgroundThis retrospective study aims to investigate the activity of retreatment with anti-EGFR-based therapies in order to explore the concept of clonal evolution by evaluating the impact of prior activity and intervening time interval.MethodsEighty-nine KRAS exon 2-wild-type metastatic colorectal patients were retreated on phase I/II clinical trials containing anti-EGFR therapies after progressing on prior cetuximab or panitumumab. Response on prior anti-EGFR therapy was defined retrospectively per physician-records as response or stable disease ≥6 months. Multivariable statistical methods included a multiple logistic regression model for response, and Cox proportional hazards model for progression-free survival.ResultsRetreatment anti-EGFR agents were cetuximab (n = 76) or cetuximab plus erlotinib (n = 13). The median interval time between prior and retreatment regimens was 4.57 months (range: 0.46-58.7). Patients who responded to the prior cetuximab or panitumumab were more likely to obtain clinical benefit to the retreatment compared to the non-responders in both univariate (p = 0.007) and multivariate analyses (OR: 3.38, 95 % CI: 1.27, 9.31, p = 0.019). The clinical benefit rate on retreatment also showed a marginally significant association with interval time between the two anti-EGFR based therapies (p = 0.053). Median progression-free survival on retreatment was increased in prior responders (4.9 months, 95 % CI: 3.6, 6.2) compared to prior non-responders (2.5 months, 95 % CI, 1.58, 3.42) in univariate (p = 0.064) and multivariate analysis (HR: 0.70, 95 % CI: 0.43-1.15, p = 0.156).ConclusionOur data lends support to the concept of clonal evolution, though the clinical impact appears less robust than previously reported. Further work to determine which patients benefit from retreatment post progression is needed.
- Published
- 2015
10. Fibroblast growth factor family aberrations in cancers: clinical and molecular characteristics
- Author
-
Parish, A, Schwaederle, M, Daniels, G, Piccioni, D, Fanta, P, Schwab, R, Shimabukuro, K, Parker, BA, Helsten, T, and Kurzrock, R
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Aged ,Female ,Fibroblast Growth Factors ,Humans ,Male ,Middle Aged ,Neoplasms ,Receptors ,Fibroblast Growth Factor ,Signal Transduction ,cancer ,FGF ,FGFR ,sequencing ,profiling ,cancer ,FGF ,FGFR ,sequencing ,profiling ,Developmental Biology ,Biochemistry and cell biology - Abstract
Fibroblast growth factor ligands and receptors (FGF and FGFR) play critical roles in tumorigenesis, and several drugs have been developed to target them. We report the biologic correlates of FGF/FGFR abnormalities in diverse malignancies. The medical records of patients with cancers that underwent targeted next generation sequencing (182 or 236 cancer-related genes) were reviewed. The following FGF/FGFR genes were tested: FGF3, 4, 6, 7, 10, 12, 14, 19, 23 and FGFR1, 2, 3, and 4. Of 391 patients, 56 (14.3%) had aberrant FGF (N = 38, all amplifications) and/or FGFR (N = 22 including 5 mutations and one FGFR3-TACC3 fusion). FGF/FGFR aberrations were most frequent in breast cancers (26/81, 32.1%, p = 0.0003). In multivariate analysis, FGF/FGFR abnormalities were independently associated with CCND1/2, RICTOR, ZNF703, RPTOR, AKT2, and CDK8 alterations (all P < 0.02), as well as with an increased median number of alterations (P < 0.0001). FGF3, FGF4, FGF19 and CCND1 were co-amplified in 22 of 391 patients (5.6%, P < 0.0001), most likely because they co-localize on the same chromosomal region (11q13). There was no significant difference in time to metastasis or overall survival when comparing patients harboring FGF/FGFR alterations versus those not. Overall, FGF/FGFR was one of the most frequently aberrant pathways in our population comprising patients with diverse malignancies. These aberrations frequently co-exist with anomalies in a variety of other genes, suggesting that tailored combination therapy may be necessary in these patients.
- Published
- 2015
11. Fibroblast growth factor family aberrations in cancers: clinical and molecular characteristics.
- Author
-
Parish, A, Schwaederle, M, Daniels, G, Piccioni, D, Fanta, P, Schwab, R, Shimabukuro, K, Parker, BA, Helsten, T, and Kurzrock, R
- Subjects
Humans ,Neoplasms ,Fibroblast Growth Factors ,Receptors ,Fibroblast Growth Factor ,Signal Transduction ,Aged ,Middle Aged ,Female ,Male ,cancer ,FGF ,FGFR ,sequencing ,profiling ,cancer ,FGF ,FGFR ,sequencing ,profiling ,Receptors ,Fibroblast Growth Factor ,Cancer ,4.1 Discovery and preclinical testing of markers and technologies ,2.1 Biological and endogenous factors ,Developmental Biology ,Biochemistry and Cell Biology - Abstract
Fibroblast growth factor ligands and receptors (FGF and FGFR) play critical roles in tumorigenesis, and several drugs have been developed to target them. We report the biologic correlates of FGF/FGFR abnormalities in diverse malignancies. The medical records of patients with cancers that underwent targeted next generation sequencing (182 or 236 cancer-related genes) were reviewed. The following FGF/FGFR genes were tested: FGF3, 4, 6, 7, 10, 12, 14, 19, 23 and FGFR1, 2, 3, and 4. Of 391 patients, 56 (14.3%) had aberrant FGF (N = 38, all amplifications) and/or FGFR (N = 22 including 5 mutations and one FGFR3-TACC3 fusion). FGF/FGFR aberrations were most frequent in breast cancers (26/81, 32.1%, p = 0.0003). In multivariate analysis, FGF/FGFR abnormalities were independently associated with CCND1/2, RICTOR, ZNF703, RPTOR, AKT2, and CDK8 alterations (all P < 0.02), as well as with an increased median number of alterations (P < 0.0001). FGF3, FGF4, FGF19 and CCND1 were co-amplified in 22 of 391 patients (5.6%, P < 0.0001), most likely because they co-localize on the same chromosomal region (11q13). There was no significant difference in time to metastasis or overall survival when comparing patients harboring FGF/FGFR alterations versus those not. Overall, FGF/FGFR was one of the most frequently aberrant pathways in our population comprising patients with diverse malignancies. These aberrations frequently co-exist with anomalies in a variety of other genes, suggesting that tailored combination therapy may be necessary in these patients.
- Published
- 2015
12. Insulin growth factor receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with metastatic adrenocortical carcinoma
- Author
-
Naing, A, LoRusso, P, Fu, S, Hong, D, Chen, HX, Doyle, LA, Phan, AT, Habra, MA, and Kurzrock, R
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Biotechnology ,Cancer ,Adrenocortical Carcinoma ,Adult ,Aged ,Antibodies ,Monoclonal ,Antibodies ,Monoclonal ,Humanized ,Antineoplastic Combined Chemotherapy Protocols ,Disease-Free Survival ,Female ,Humans ,Male ,Middle Aged ,Sirolimus ,Young Adult ,phase I clinical trials ,IGF-1R pathway ,mTOR pathway ,adrenocortical carcinoma ,cixutumumab ,Oncology and Carcinogenesis ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundAdrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy without an available effective systemic chemotherapy. Insulin growth factor 2 (IGF-2) overexpression leading to the activation of the IGF-1 receptor (IGF-1R)/mammalian target of rapamycin (mTOR) pathway is well described in ACC. Cixutumumab, a fully human IgG1 monoclonal antibody directed at IGF-1R was combined with temsirolimus on the basis of preclinical data.MethodsPatients received cixutumumab, 3-6 mg kg(-1) intravenously (IV) weekly, and temsirolimus, 25-37.5 mg IV weekly (4-week cycles), with restaging after 8 weeks.ResultsTwenty-six patients were enrolled (13 (50%) men); median age, 47 years; median number of prior therapies, 4. Five patients previously received an IGF-1R inhibitor and one, temsirolimus. The most frequent toxicities, at least possibly drug related, were grade 1-2 thrombocytopenia (38%), mucositis (58%), hypercholesterolaemia (31%), hypertriglyceridemia (35%), and hyperglycaemia (31%). In all, 11 of 26 patients (42%) achieved stable disease (SD) >6 months (duration range=6-21 months) with 3 of the 11 having received a prior IGF-1R inhibitor.ConclusionCixutumumab combined with temsirolimus was well tolerated and >40% of patients achieved prolonged SD.
- Published
- 2013
13. Insulin growth factor receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with metastatic adrenocortical carcinoma.
- Author
-
Naing, A, Lorusso, P, Fu, S, Hong, D, Chen, HX, Doyle, LA, Phan, AT, Habra, MA, and Kurzrock, R
- Subjects
Humans ,Adrenocortical Carcinoma ,Sirolimus ,Antineoplastic Combined Chemotherapy Protocols ,Antibodies ,Monoclonal ,Disease-Free Survival ,Adult ,Aged ,Middle Aged ,Female ,Male ,Young Adult ,Antibodies ,Monoclonal ,Humanized ,phase I clinical trials ,IGF-1R pathway ,mTOR pathway ,adrenocortical carcinoma ,cixutumumab ,Antibodies ,Monoclonal ,Humanized ,Oncology and Carcinogenesis ,Public Health and Health Services ,Oncology & Carcinogenesis - Abstract
BackgroundAdrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy without an available effective systemic chemotherapy. Insulin growth factor 2 (IGF-2) overexpression leading to the activation of the IGF-1 receptor (IGF-1R)/mammalian target of rapamycin (mTOR) pathway is well described in ACC. Cixutumumab, a fully human IgG1 monoclonal antibody directed at IGF-1R was combined with temsirolimus on the basis of preclinical data.MethodsPatients received cixutumumab, 3-6 mg kg(-1) intravenously (IV) weekly, and temsirolimus, 25-37.5 mg IV weekly (4-week cycles), with restaging after 8 weeks.ResultsTwenty-six patients were enrolled (13 (50%) men); median age, 47 years; median number of prior therapies, 4. Five patients previously received an IGF-1R inhibitor and one, temsirolimus. The most frequent toxicities, at least possibly drug related, were grade 1-2 thrombocytopenia (38%), mucositis (58%), hypercholesterolaemia (31%), hypertriglyceridemia (35%), and hyperglycaemia (31%). In all, 11 of 26 patients (42%) achieved stable disease (SD) >6 months (duration range=6-21 months) with 3 of the 11 having received a prior IGF-1R inhibitor.ConclusionCixutumumab combined with temsirolimus was well tolerated and >40% of patients achieved prolonged SD.
- Published
- 2013
14. Cell cycle-related shifts in subcellular localization of BCR: association with mitotic chromosomes and with heterochromatin.
- Author
-
Wetzler, M, Talpaz, M, Yee, G, Stass, S A, Van Etten, R A, Andreeff, M, Goodacre, A M, Kleine, H D, Mahadevia, R K, and Kurzrock, R
- Subjects
Cell Compartmentation ,Cell Cycle: physiology ,Cell Nucleus: chemistry ,ultrastructure ,Chromosomes: chemistry ,Flow Cytometry ,Fluorescent Antibody Technique ,Heterochromatin: chemistry ,Humans ,Interphase: physiology ,Leukemia ,Mitosis: physiology ,Oncogene Proteins: isolation & purification ,Protein-Tyrosine Kinases ,Proto-Oncogene Proteins ,Proto-Oncogene Proteins c-bcr ,Tumor Cells ,Cultured - Abstract
The disruption of the BCR gene and its juxtaposition to and consequent activation of the ABL gene has been implicated as the critical molecular defect in Philadelphia chromosome-positive leukemias. The normal BCR protein is a multifunctional molecule with domains that suggest its participation in phosphokinase and GTP-binding pathways. Taken together with its localization to the cytoplasm of uncycled cells, it is therefore presumed to be involved in cytoplasmic signaling. By performing a double aphidicolin block for cell cycle synchronization, we currently demonstrate that the subcellular localization of BCR shifts from being largely cytoplasmic in interphase cells to being predominantly perichromosomal in mitosis. Furthermore, with the use of immunogold labeling and electron microscopy, association of BCR with DNA, in particular heterochromatin, can be demonstrated even in quiescent cells. Results were similar in cell lines of lymphoid or myeloid origin. These observations suggest a role for BCR in the phosphokinase interactions linked to condensed chromatin, a network previously implicated in cell cycle regulation.
- Published
- 1995
15. Subcellular localization of Bcr, Abl, and Bcr-Abl proteins in normal and leukemic cells and correlation of expression with myeloid differentiation.
- Author
-
Wetzler, M, Talpaz, M, Van Etten, R A, Hirsh-Ginsberg, C, Beran, M, and Kurzrock, R
- Subjects
Animals ,Base Sequence ,Blast Crisis ,Blotting ,Southern ,Cell Differentiation ,DNA Primers ,DNA ,Complementary ,Fluorescent Antibody Technique ,Fusion Proteins ,bcr-abl: analysis ,biosynthesis ,Humans ,Immunohistochemistry: methods ,Leukemia: metabolism ,pathology ,Leukemia ,Promyelocytic ,Acute ,Mice ,Molecular Sequence Data ,Protein-Tyrosine Kinases ,Proto-Oncogene Proteins: analysis ,biosynthesis ,Proto-Oncogene Proteins c-abl: analysis ,biosynthesis ,Proto-Oncogene Proteins c-bcr ,Proto-Oncogenes ,Transfection ,Tumor Cells ,Cultured - Abstract
We used specific antisera and immunohistochemical methods to investigate the subcellular localization and expression of Bcr, Abl, and Bcr-Abl proteins in leukemic cell lines and in fresh human leukemic and normal samples at various stages of myeloid differentiation. Earlier studies of the subcellular localization of transfected murine type IV c-Abl protein in fibroblasts have shown that this molecule resides largely in the nucleus, whereas transforming deletion variants are localized exclusively in the cytoplasm. Here, we demonstrate that the murine type IV c-Abl protein is also found in the nucleus when overexpressed in a mouse hematopoietic cell line. However, in both normal and leukemic human hematopoietic cells, c-Abl is discerned predominantly in the cytoplasm, with nuclear staining present, albeit at a lower level. In contrast, normal endogenous Bcr protein, as well as the aberrant p210BCR-ABL and p190BCR-ABL proteins consistently localize to the cytoplasm in both cell lines and fresh cells. The results with p210BCR-ABL were confirmed in a unique Ph1-positive chronic myelogenous leukemia (CML) cell line, KBM5, which lacks the normal chromosome 9 and hence the normal c-Abl product. Because the p210BCR-ABL protein appears cytoplasmic in both chronic phase and blast crisis CML cells, as does the p190BCR-ABL in Ph1-positive acute leukemia, a change in subcellular location of Bcr-Abl proteins between cytoplasm and nucleus cannot explain the different spectrum of leukemias associated with p210 and p190, nor the transition from the chronic to the acute leukemia phenotype seen in CML. Further analysis of fresh CML and normal hematopoietic bone marrow cells reveals that p210BCR-ABL, as well as the normal Bcr and Abl proteins, are expressed primarily in the early stages of myeloid maturation, and that levels of expression are reduced significantly as the cells mature to polymorphonuclear leukocytes. Similarly, a decrease in Bcr and Abl levels occurs in HL-60 cells induced by DMSO to undergo granulocytic differentiation. The action of p210BCR-ABL and its normal counterparts may, therefore, take place during the earlier stages of myeloid development.
- Published
- 1993
16. Lymphotoxin is an autocrine growth factor for Epstein-Barr virus-infected B cell lines.
- Author
-
Estrov, Z, Kurzrock, R, Pocsik, E, Pathak, S, Kantarjian, HM, Zipf, TF, Harris, D, Talpaz, M, and Aggarwal, BB
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Clinical Research ,Pediatric Cancer ,Rare Diseases ,Pediatric ,Hematology ,Biotechnology ,Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Adult ,Aged ,Aged ,80 and over ,Antibodies ,Antibodies ,Monoclonal ,B-Lymphocytes ,Blotting ,Southern ,Cell Division ,Cells ,Cultured ,DNA ,Viral ,Dose-Response Relationship ,Drug ,Growth Substances ,Herpesviridae Infections ,Herpesvirus 4 ,Human ,Humans ,Immunoglobulin Heavy Chains ,Immunophenotyping ,Karyotyping ,Leukemia ,Myeloid ,Acute ,Lymphotoxin-alpha ,Myelodysplastic Syndromes ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Receptors ,Cell Surface ,Receptors ,Tumor Necrosis Factor ,Tumor Necrosis Factor-alpha ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Because human lymphotoxin (LT) was originally isolated from a lymphoblastoid cell line, we investigated the role of this molecule in three newly established Epstein-Barr virus (EBV)-infected human B cell lines. These lines were derived from acute lymphoblastic leukemia (Z-6), myelodysplastic syndrome (Z-43), and acute myelogenous leukemia (Z-55) patients who had a prior EBV infection. Each lymphoblastoid cell line had a karyotype that was different from that of the original parent leukemic cells, and all expressed B cell, but not T cell or myeloid surface markers. In all three lines, rearranged immunoglobulin heavy chain joining region (JH) bands were found, and the presence of EBV DNA was confirmed by Southern blotting. Z-6, Z-43, and Z-55 cell lines constitutively produced 192, 48, and 78 U/ml LT, respectively, as assessed by a cytotoxicity assay and antibody neutralization. Levels of tumor necrosis factor (TNF) were undetectable. Scatchard analysis revealed that all the cell lines expressed high-affinity TNF/LT receptors with receptor densities of 4197, 1258, and 1209 sites/cell on Z-6, Z-43, and Z-55, respectively. Furthermore, labeled TNF binding could be reversed by both unlabeled TNF, as well as by LT. Studies with p60 and p80 receptor-specific antibodies revealed that the three lines expressed primarily the p80 form of the TNF receptor. When studied in a clonogenic assay, exogenous LT stimulated proliferation of all three cell lines in a dose-dependent fashion at concentrations ranging from 25 to 500 U/ml. Similar results were obtained with [3H]TdR incorporation. Monoclonal anti-LT neutralizing antibodies at concentrations of 25-500 U/ml inhibited cellular multiplication in a dose-dependent manner. It is interesting that in spite of a common receptor, TNF (1,000 U/ml) had no direct effect on Z-55 cell growth, whereas it partially reversed the stimulatory effect of exogenous LT. In addition, TNF inhibited Z-6 and Z-43 cell proliferation, and its suppressive effect was reversed by exogenous LT. Both p80 and p60 forms of soluble TNF receptors suppressed the lymphoblastoid cell line proliferation and their inhibitory effect was partially reversed by LT. Our data suggest that (a) LT is an autocrine growth factor for EBV-transformed lymphoblastoid B cell lines; and (b) anti-LT antibodies, soluble TNF/LT receptors, and TNF itself can suppress the growth of lymphoblastoid cells, probably by modulating or competing with LT.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1993
17. Lymphotoxin is an autocrine growth factor for Epstein-Barr virus-infected B cell lines.
- Author
-
Estrov, Z, Kurzrock, R, Pocsik, E, Pathak, S, Kantarjian, HM, Zipf, TF, Harris, D, Talpaz, M, and Aggarwal, BB
- Subjects
B-Lymphocytes ,Cells ,Cultured ,Humans ,Herpesvirus 4 ,Human ,Herpesviridae Infections ,Myelodysplastic Syndromes ,Growth Substances ,Tumor Necrosis Factor-alpha ,Receptors ,Cell Surface ,Receptors ,Tumor Necrosis Factor ,DNA ,Viral ,Antibodies ,Antibodies ,Monoclonal ,Blotting ,Southern ,Karyotyping ,Immunophenotyping ,Cell Division ,Dose-Response Relationship ,Drug ,Adult ,Aged ,Aged ,80 and over ,Immunoglobulin Heavy Chains ,Lymphotoxin-alpha ,Leukemia ,Myeloid ,Acute ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,and over ,Monoclonal ,Blotting ,Southern ,Cells ,Cultured ,DNA ,Viral ,Dose-Response Relationship ,Drug ,Herpesvirus 4 ,Human ,Leukemia ,Myeloid ,Acute ,Receptors ,Cell Surface ,Tumor Necrosis Factor ,Medical and Health Sciences ,Immunology - Abstract
Because human lymphotoxin (LT) was originally isolated from a lymphoblastoid cell line, we investigated the role of this molecule in three newly established Epstein-Barr virus (EBV)-infected human B cell lines. These lines were derived from acute lymphoblastic leukemia (Z-6), myelodysplastic syndrome (Z-43), and acute myelogenous leukemia (Z-55) patients who had a prior EBV infection. Each lymphoblastoid cell line had a karyotype that was different from that of the original parent leukemic cells, and all expressed B cell, but not T cell or myeloid surface markers. In all three lines, rearranged immunoglobulin heavy chain joining region (JH) bands were found, and the presence of EBV DNA was confirmed by Southern blotting. Z-6, Z-43, and Z-55 cell lines constitutively produced 192, 48, and 78 U/ml LT, respectively, as assessed by a cytotoxicity assay and antibody neutralization. Levels of tumor necrosis factor (TNF) were undetectable. Scatchard analysis revealed that all the cell lines expressed high-affinity TNF/LT receptors with receptor densities of 4197, 1258, and 1209 sites/cell on Z-6, Z-43, and Z-55, respectively. Furthermore, labeled TNF binding could be reversed by both unlabeled TNF, as well as by LT. Studies with p60 and p80 receptor-specific antibodies revealed that the three lines expressed primarily the p80 form of the TNF receptor. When studied in a clonogenic assay, exogenous LT stimulated proliferation of all three cell lines in a dose-dependent fashion at concentrations ranging from 25 to 500 U/ml. Similar results were obtained with [3H]TdR incorporation. Monoclonal anti-LT neutralizing antibodies at concentrations of 25-500 U/ml inhibited cellular multiplication in a dose-dependent manner. It is interesting that in spite of a common receptor, TNF (1,000 U/ml) had no direct effect on Z-55 cell growth, whereas it partially reversed the stimulatory effect of exogenous LT. In addition, TNF inhibited Z-6 and Z-43 cell proliferation, and its suppressive effect was reversed by exogenous LT. Both p80 and p60 forms of soluble TNF receptors suppressed the lymphoblastoid cell line proliferation and their inhibitory effect was partially reversed by LT. Our data suggest that (a) LT is an autocrine growth factor for EBV-transformed lymphoblastoid B cell lines; and (b) anti-LT antibodies, soluble TNF/LT receptors, and TNF itself can suppress the growth of lymphoblastoid cells, probably by modulating or competing with LT.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1993
18. Recombinant gamma interferon induces hypertriglyceridemia and inhibits post-heparin lipase activity in cancer patients.
- Author
-
Kurzrock, R, Rohde, MF, Quesada, JR, Gianturco, SH, Bradley, WA, Sherwin, SA, and Gutterman, JU
- Subjects
Clinical Research ,Cancer ,Adult ,Aged ,Female ,Glycoproteins ,Humans ,Interferon-gamma ,Lipoprotein Lipase ,Male ,Middle Aged ,Neoplasms ,Recombinant Proteins ,Triglycerides ,Tumor Necrosis Factor-alpha ,Medical and Health Sciences ,Immunology - Abstract
Animals suffering from malignancy or chronic infection develop characteristic metabolic abnormalities, including a well-defined hypertriglyceridemic state. These abnormalities have been attributed to release of one or more mediators from activated macrophages. We report that cancer patients receiving RIFN-gamma, a potent macrophage activator, at doses of greater than or equal to 0.25 mg/m2/d i.m. show marked increases in triglyceride but not in cholesterol levels (pretreatment triglyceride level of 180 +/- 190 mg/dl [mean +/- SD] vs. a day-14 level of 370 +/- 242 mg/dl, n = 23, p less than 0.001 by the paired t test). This hypertriglyceridemia was characterized by an increase in very low-density lipoproteins and a decrease in plasma post-heparin lipase activity, consistent with defective triglyceride clearance (mean pretreatment lipase level of 2.1 mumol/ml/h vs. a day-14 level of 1.2 mumol/ml/h, n = 6, p = 0.02 by the paired t test). rIFN-gamma did not directly inhibit lipoprotein lipase enzymatic activity in vitro. Other possible mechanisms of action, such as suppression of lipase by an rIFN-gamma-induced mediator released from activated macrophages, or a direct effect of interferon on lipase biosynthesis, require further investigation. Our observations provide evidence that factors produced by the immune system can regulate lipid metabolism in man.
- Published
- 1986
19. Recombinant gamma interferon induces hypertriglyceridemia and inhibits post-heparin lipase activity in cancer patients.
- Author
-
Kurzrock, R, Rohde, MF, Quesada, JR, Gianturco, SH, Bradley, WA, Sherwin, SA, and Gutterman, JU
- Subjects
Humans ,Neoplasms ,Lipoprotein Lipase ,Glycoproteins ,Triglycerides ,Tumor Necrosis Factor-alpha ,Recombinant Proteins ,Adult ,Aged ,Middle Aged ,Female ,Male ,Interferon-gamma ,Medical and Health Sciences ,Immunology - Abstract
Animals suffering from malignancy or chronic infection develop characteristic metabolic abnormalities, including a well-defined hypertriglyceridemic state. These abnormalities have been attributed to release of one or more mediators from activated macrophages. We report that cancer patients receiving RIFN-gamma, a potent macrophage activator, at doses of greater than or equal to 0.25 mg/m2/d i.m. show marked increases in triglyceride but not in cholesterol levels (pretreatment triglyceride level of 180 +/- 190 mg/dl [mean +/- SD] vs. a day-14 level of 370 +/- 242 mg/dl, n = 23, p less than 0.001 by the paired t test). This hypertriglyceridemia was characterized by an increase in very low-density lipoproteins and a decrease in plasma post-heparin lipase activity, consistent with defective triglyceride clearance (mean pretreatment lipase level of 2.1 mumol/ml/h vs. a day-14 level of 1.2 mumol/ml/h, n = 6, p = 0.02 by the paired t test). rIFN-gamma did not directly inhibit lipoprotein lipase enzymatic activity in vitro. Other possible mechanisms of action, such as suppression of lipase by an rIFN-gamma-induced mediator released from activated macrophages, or a direct effect of interferon on lipase biosynthesis, require further investigation. Our observations provide evidence that factors produced by the immune system can regulate lipid metabolism in man.
- Published
- 1986
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.