65 results on '"B Mookerjee"'
Search Results
2. 414TiP An open-label phase 2a study of combination dabrafenib (D) and trametinib (T) in Asian patients (pts) with advanced BRAF V600–mutant acral lentiginous melanoma (ALM) or cutaneous melanoma (CM)
- Author
-
S.J. Shin, Arunee Dechaphunkul, D. Wu, Jun Guo, B. Mookerjee, X. Zhang, S. Thongprasert, S.-T. Cheng, Tae Min Kim, C-C. Lin, J. Maneechavakajorn, Y. Fan, X. Song, S. Bettinger, B.S. Wong, W.-C. Chang, and P. Zhang
- Subjects
Trametinib ,medicine.medical_specialty ,business.industry ,Mutant ,Dabrafenib ,Hematology ,medicine.disease ,Acral lentiginous melanoma ,Dermatology ,Oncology ,Cutaneous melanoma ,medicine ,Open label ,business ,medicine.drug - Published
- 2016
3. P2.01-24 CANOPY-2: Phase 3 Study of Canakinumab Plus Docetaxel as Second/Third Line Therapy in Locally Advanced/Metastatic NSCLC
- Author
-
L. Paz-Ares, Y. Goto, D. Lim, V. Papadimitrakopoulou, B. Mookerjee, I. Malet, Z. Zewen, and M. Reck
- Subjects
Pulmonary and Respiratory Medicine ,Oncology - Published
- 2019
4. Autotransplantation for advanced lymphoma and Hodgkin's disease followed by post-transplant rituxan/GM-CSF or radiotherapy and consolidation chemotherapy
- Author
-
Timothy Chen, Guido Tricot, Clarence Sarkodee-Adoo, Richard J.H. Smith, Kathleen Ruehle, Stanley R. Frankel, Michele Cottler-Fox, B Mookerjee, L Kight, A. Badros, A Kennedy, Naoko Takebe, A. Fassas, S Chambers, M Jacobs, Meyer R. Heyman, R Hudes, M MacFadden, Aaron P. Rapoport, Barry Meisenberg, Robert G. Fenton, and G Phillips
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,animal structures ,medicine.medical_treatment ,lymphoma ,Transplantation, Autologous ,Article ,Disease-Free Survival ,Antibodies, Monoclonal, Murine-Derived ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,consolidation chemotherapy ,Humans ,rituxan ,radiotherapy ,Aged ,Transplantation ,Chemotherapy ,autotransplantation ,business.industry ,Lymphoma, Non-Hodgkin ,Hematopoietic Stem Cell Transplantation ,Antibodies, Monoclonal ,Granulocyte-Macrophage Colony-Stimulating Factor ,Consolidation Chemotherapy ,Hematology ,Immunotherapy ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Hodgkin Disease ,Autotransplantation ,Lymphoma ,Surgery ,Radiation therapy ,Granulocyte macrophage colony-stimulating factor ,Rituximab ,Female ,Hodgkin's disease ,business ,medicine.drug - Abstract
Disease relapse occurs in 50% or more of patients who are autografted for relapsed or refractory lymphoma (NHL) or Hodgkin's disease (HD). The administration of non-cross-resistant therapies during the post-transplant phase could possibly control residual disease and delay or prevent its progression. To test this approach, 55 patients with relapsed/refractory or high-risk NHL or relapsed/refractory HD were enrolled in the following protocol: stem cell mobilization: cyclophosphamide (4.5 g/m2) + etoposide (2.0 g/m2) followed by GM-CSF or G-CSF; high-dose therapy: gemcitabine (1.0 g/m2) on day −5, BCNU (300 mg/m2) + gemcitabine (1.0 g/m2) on day −2, melphalan (140 mg/m2) on day −1, blood stem cell infusion on day 0; post-transplant immunotherapy (B cell NHL): rituxan (375 mg/m2) weekly for 4 weeks + GM-CSF (250 μg thrice weekly) (weeks 4–8); post-transplant involved-field radiotherapy (HD): 30–40 Gy to pre-transplant areas of disease (weeks 4–8); post-transplant consolidation chemotherapy (all patients): dexamethasone (40 mg daily)/cyclophosphamide (300 mg/m2/day)/etoposide (30 mg/m2/day)/cisplatin (15 mg/m2/day) by continuous intravenous infusion for 4 days + gemcitabine (1.0 g/m2, day 3) (months 3 + 9) alternating with dexamethasone/paclitaxel (135 mg/m2)/cisplatin (75 mg/m2) (months 6 + 12). Of the 33 patients with B cell lymphoma, 14 had primary refractory disease (42%), 12 had relapsed disease (36%) and seven had high-risk disease in first CR (21%). For the entire group, the 2-year Kaplan–Meier event-free survival (EFS) and overall survival (OS) were 30% and 35%, respectively, while six of 33 patients (18%) died before day 100 from transplant-related complications. The rituxan/GM-CSF phase was well-tolerated by the 26 patients who were treated and led to radiographic responses in seven patients; an eighth patient with a blastic variant of mantle-cell lymphoma had clearance of marrow involvement after rituxan/GM-CSF. Of the 22 patients with relapsed/refractory HD (21 patients) or high-risk T cell lymphoblastic lymphoma (one patient), the 2-year Kaplan–Meier EFS and OS were 70% and 85%, respectively, while two of 22 patients (9%) died before day 100 from transplant-related complications. Eight patients received involved field radiation and seven had radiographic responses within the treatment fields. A total of 72 courses of post-transplant consolidation chemotherapy were administered to 26 of the 55 total patients. Transient grade 3–4 myelosuppression was common and one patient died from neutropenic sepsis, but no patients required an infusion of backup stem cells. After adjustment for known prognostic factors, the EFS for the cohort of HD patients was significantly better than the EFS for an historical cohort of HD patients autografted after BEAC (BCNU/etoposide/cytarabine/cyclophosphamide) without consolidation chemotherapy (P = 0.015). In conclusion, post-transplant consolidation therapy is feasible and well-tolerated for patients autografted for aggressive NHL and HD and may be associated with improved progression-free survival particularly for patients with HD. Bone Marrow Transplantation (2002) 29, 303–312. doi:10.1038/sj.bmt.1703363
- Published
- 2002
5. Spontaneous delta- to beta-globin switching in K562 human leukemia cells
- Author
-
B Mookerjee, George F. Atweh, and Murat O. Arcasoy
- Subjects
Regulation of gene expression ,Messenger RNA ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Phenotype ,Molecular biology ,chemistry.chemical_compound ,chemistry ,Cell culture ,hemic and lymphatic diseases ,Globin ,Beta (finance) ,Gene ,Hemin ,K562 cells - Abstract
Previous analysis of the hemoglobin phenotype of the K562 human erythroleukemia cell line showed regulated expression of the epsilon-, zeta-, gamma-, alpha-, and delta-globin genes. Expression of the beta- globin genes has not been previously detected in this cell line. In this report, we describe the isolation of a variant of the K562 cell line that actively expresses beta-globin messenger RNA (mRNA) and polypeptide and shows greatly reduced expression of the delta-globin genes. This phenotype developed spontaneously in culture while two other K562 isolates grown under the same culture conditions have not undergone the same delta- to beta-globin switch. Analysis of this unique K562 variant shows that a construct containing a beta-globin promoter is quite active upon transient transfection into these cells. This finding suggests that the activation of the endogenous beta-globin genes results from changes in the trans-acting environment of these cells. The regulation of the beta-globin genes in this variant is characterized by a paradoxical decrease in the level of beta-globin mRNA after exposure to hemin. Other globin genes of this variant are appropriately regulated and show increased expression after hemin induction. Further study of this variant may shed light on mechanisms of gene regulation that are involved in hemoglobin switching.
- Published
- 1992
6. Biological significance and molecular mechanisms of p53-induced apoptosis
- Author
-
A, Bedi and B, Mookerjee
- Abstract
The recognition that the p53 tumour suppressor gene is frequently inactivated in human cancers has galvanized an intense pursuit of the fundamental mechanisms by which the encoded protein halts malignant transformation and tumour progression. It is now evident that p53 is a multifunctional transcription factor that is intimately involved in the cellular response to stressful stimuli such as DNA damage and hypoxia. In addition to its role in the surveillance mechanisms that arrest cell cycle progression, p53 can also trigger apoptosis in response to DNA damage or oncogenic aberrations that induce aberrant cell cycle progression. Since p53 is a critical component for DNA damage-induced apoptosis, the frequent occurrence of p53 mutations in human neoplasia provides a genetic basis for their poor response to genotoxic anticancer agents. Two recent studies offer key insights into the molecular mechanisms employed by p53 to induce cell death. One model indicates that p53 induces redox-related genes that generate reactive oxygen species and promote the oxidative degradation of mitochondrial components. The other demonstrates p53-mediated induction of DR5, a death receptor of the tumour necrosis factor receptor family, that induces death by caspase-mediated proteolysis. These insights provide an exhilarating array of possible therapeutic interventions against p53-deficient human cancers that may pay enormous dividends in the not-too-distant future.
- Published
- 2003
7. Regulation of phosphoprotein p18 in leukemic cells. Cell cycle regulated phosphorylation by p34cdc2 kinase
- Author
-
X N, Luo, B, Mookerjee, A, Ferrari, S, Mistry, and G F, Atweh
- Subjects
Base Sequence ,Recombinant Fusion Proteins ,Cell Cycle ,Molecular Sequence Data ,Phosphoproteins ,CDC2 Protein Kinase ,Microtubule Proteins ,Tumor Cells, Cultured ,Humans ,Stathmin ,Leukemia, Erythroblastic, Acute ,Phosphorylation ,DNA Primers ,Glutathione Transferase ,Signal Transduction - Abstract
p18 is a phosphoprotein that is expressed at very high levels in leukemic cells, at moderately high levels in proliferating normal lymphocytes, and at low levels in quiescent lymphocytes. Induction of terminal differentiation of leukemic cells in culture results in a decrease in cellular proliferation. These phenotypic changes are associated with rapid phosphorylation of p18, followed by a more gradual decrease in the level of its mRNA expression. More than 12 different phosphorylation products of p18 have been identified in different cells by high resolution two-dimensional polyacrylamide gel electrophoresis. Previous studies have suggested that p18 may be a substrate for protein kinase C in some cellular processes and protein kinase A in others. In this report, we show that the phosphorylation of p18 increases as cells progress toward the G2-M phases of the cell cycle in proliferating leukemic cells. We have examined the hypothesis that the putative role of p18 in cellular proliferation may be mediated by its involvement in the p34cdc2 kinase signal transduction pathway. We have produced recombinant p18 in bacterial cells and shown that it can be phosphorylated in vitro by purified p34cdc2 kinase with a stoichiometry of 0.86 mol of PO4/mol of substrate. We have used site-directed mutagenesis to demonstrate that the site of p34cdc2 phosphorylation is the serine at position 38. This same site has previously been shown to be phosphorylated in vivo in bovine brain along with another serine at position 25. The observation that p18 gets phosphorylated in the G2-M phases of the cell cycle and the demonstration that p18 is phosphorylated efficiently by p34cdc2 kinase in vitro at a residue that is also phosphorylated in vivo support the hypothesis that p18 may be a physiologic substrate for p34cdc2 kinase in vivo. We have also examined the effect of inhibiting the expression of p18 on cell cycle progression. These experiments demonstrated that antisense inhibition of the expression of p18 in K562 erythroleukemia cells is associated with a decrease in cellular proliferation and accumulation of cells in the G2-M phases of the cycle. The implications of these findings to the proposed role of p18 in the regulation of cellular proliferation are discussed.
- Published
- 1994
8. Allopurinol in prevention of reperfusion injury of hypoxically stored rat hearts
- Author
-
J, Bergsland, L, LoBalsamo, P, Lajos, M J, Feldman, and B, Mookerjee
- Subjects
Perfusion ,Superoxide Dismutase ,Allopurinol ,Myocardium ,Blood Circulation ,Hemodynamics ,Animals ,Coronary Disease ,Rats, Inbred Strains ,Organ Preservation ,Catalase ,Hypoxia ,Rats - Abstract
We have studied the effect of the xanthine-oxidase inhibitor allopurinol on the performance of hypoxically stored rat hearts to evaluate the drugs' potential value in heart transplantation. Hearts were stored for 2 hours at 15 degrees C after potassium chloride cardioplegia. Hemodynamics were measured before hypoxia and after reperfusion. Hearts were treated with allopurinol in the cardioplegia, at the time of reperfusion, or with superoxide dismutase (SOD) and catalase (CAT) and compared with control hearts that received no treatment. Hemodynamic performance after hypoxia was significantly improved in the SOD/CAT group compared with the control group. Allopurinol-treated hearts were also significantly improved, especially when the drug was given in the reperfusion phase. Our results demonstrate the importance of free radicals during reperfusion of hypoxically stored hearts. Allopurinol and SOD/CAT may be of value in reducing such damage after heart transplantation.
- Published
- 1987
9. Post-anoxic hemodynamic performance. The effect of allopurinol and superoxide dismutase/catalase
- Author
-
J, Bergsland, L, LoBalsamo, P, Lajos, and B, Mookerjee
- Subjects
Male ,Perfusion ,Ischemia ,Superoxide Dismutase ,Allopurinol ,Hemodynamics ,Animals ,Heart Transplantation ,Organ Preservation ,Catalase ,Rats - Published
- 1987
10. Hemoperfusion in digitalis intoxication: a comparative study of coated versus uncoated charcoal
- Author
-
M, Tobin, F, Cerra, J, Steinbach, and B, Mookerjee
- Subjects
Hemoperfusion ,Digoxin ,Dogs ,Acrylates ,Charcoal ,Animals ,Humans ,Cellulose ,Gels - Published
- 1977
11. In Vitro Testing for Cellular Immunity to Denatured DNA in Systemic Lupus Erythematosus
- Author
-
B. Mookerjee, Joanne Marsh, Waltraud Riedel, and John S. Percy
- Subjects
Cellular immunity ,chemistry.chemical_compound ,chemistry ,business.industry ,Immunology ,Medicine ,business ,DNA ,In vitro - Published
- 1972
12. HEMOPERFUSION IN DIGITALIS INTOXICATION
- Author
-
M Tobin, J Steinbach, F Cerra, and B Mookerjee
- Subjects
business.industry ,medicine.medical_treatment ,Biomedical Engineering ,Biophysics ,Bioengineering ,General Medicine ,DIGITALIS INTOXICATION ,Pharmacology ,Hemoperfusion ,Biomaterials ,visual_art ,visual_art.visual_art_medium ,medicine ,Digoxin intoxication ,Charcoal ,business - Published
- 1977
13. Hyperchloremic Acidosis in Early Diagnosis of Renal Allograft Rejection
- Author
-
John B. Dossetor, M H Gault, and B Mookerjee
- Subjects
Adult ,Male ,medicine.medical_specialty ,Urology ,Kidney Function Tests ,urologic and male genital diseases ,Phosphates ,Renal tubular acidosis ,Chlorides ,Transplantation Immunology ,Cadaver ,Hyperchloremic acidosis ,Internal Medicine ,Humans ,Transplantation, Homologous ,Medicine ,In patient ,Intensive care medicine ,Transplantation ,business.industry ,Acidosis, Renal Tubular ,General Medicine ,Hydrogen-Ion Concentration ,medicine.disease ,Kidney Transplantation ,Tissue Donors ,Kidney Tubules ,Creatinine ,Renal allograft ,Female ,business ,Glomerular Filtration Rate - Abstract
Hyperchloremic renal tubular acidosis has been observed to occur in a surprisingly high percentage of episodes of rejection in patients who have had cadaver renal allografts. Impairment of...
- Published
- 1969
14. Canakinumab in combination with docetaxel compared with docetaxel alone for the treatment of advanced non-small cell lung cancer following platinum-based doublet chemotherapy and immunotherapy (CANOPY-2): A multicenter, randomized, double-blind, phase 3 trial.
- Author
-
Paz-Ares L, Goto Y, Wan-Teck Lim D, Halmos B, Chul Cho B, Cobo M, Luis González Larriba J, Zhou C, Demedts I, Atmaca A, Baka S, Mookerjee B, Portella S, Zhu Z, Wu J, Demanse D, Dharan B, and Reck M
- Subjects
- Adult, Humans, Docetaxel therapeutic use, Immunotherapy, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Antibodies, Monoclonal, Humanized
- Abstract
Objectives: Canakinumab, an interleukin-1 beta inhibitor, previously showed reduced lung cancer incidence and mortality (CANTOS). Here, we compare the efficacy/safety of canakinumab versus placebo in patients with advanced non-small cell lung cancer (NSCLC) who had progressed after platinum-based doublet chemotherapy (PDC) and immunotherapy., Materials and Methods: CANOPY-2, a randomized, double-blind, phase 3 trial, enrolled adult patients with stage IIIB/IV NSCLC, without EGFR or ALK alterations, who had received one prior PDC regimen and one prior programmed death-1/programmed death-ligand 1 inhibitor and experienced subsequent disease progression. Patients were randomized to canakinumab plus docetaxel or placebo plus docetaxel., Results: A total of 237 patients were randomly allocated: 120 (51 %) to canakinumab and 117 (49 %) to placebo, stratified by histology and prior lines of therapy. Three patients in the placebo arm did not receive study treatment. The trial did not meet its primary endpoint of overall survival: median 10.6 months (95 % confidence interval [CI], 8.2-12.4) for the canakinumab arm and 11.3 months (95 % CI, 8.5-13.8) for the placebo arm (hazard ratio, 1.06 [95 % CI, 0.76-1.48]; one-sided P-value = 0.633). AEs (any grade) were reported in 95 % of patients in the canakinumab group and in 98 % of patients in the placebo group. Grade 3-4 AEs were experienced by 62 % and 64 % of patients in the canakinumab and placebo groups, respectively, and grade 5 AEs were experienced by 8 % and 5 %. Prespecified, post-hoc subgroup analyses showed that patients with undetected circulating tumor DNA (ctDNA) and/or lower levels (< 10 mg/L) of C-reactive protein (CRP) achieved longer progression-free and overall survival than those with detected ctDNA or higher (≥ 10 mg/L) CRP levels. There was no association with treatment arm., Conclusion: Adding canakinumab to docetaxel did not provide additional benefit for patients with advanced NSCLC who had progressed after PDC and immunotherapy., Clinical Registration: NCT03626545., Competing Interests: Declaration of competing interest Luis Paz-Ares reports grants from AstraZeneca, Bristol Myers Squibb, Merck, and Pfizer; consulting fees from Amgen, AstraZeneca, Bayer, Bristol Myers Squibb, GlaxoSmithKline, Ipsen, Janssen, Lilly, Merck, Mirati, Novartis, Pfizer, PharmaMar, Roche, Sanofi, Servier, and Takeda; speaker honoraria from AstraZeneca, Janssen, Merck, and Mirati; leadership roles in Altum Sequencing and Genomica, all outside the submitted work. Yasushi Goto reports clinical trial grants from AZK and Pfizer; institutional grants from AbbVie, Bristol Myers Squibb, Daiichi Sankyo, Eli Lilly, Kyorin, Novartis, Ono Pharmaceutical, and Pfizer; speaker honoraria from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Chugai, Daiichi Sankyo, Eli Lilly, Guardant Health Inc., Illumina, Merck, Novartis, Ono Pharmaceutical, Pfizer, and Taiho; leadership roles in Cancer Net Japan and JAMT, all outside the submitted work. Darren Lim reports grants from Bristol Myers Squibb; speaker honoraria from Boehringer Ingelheim, Merck, and Roche; and support for attending meetings from AstraZeneca and Taiho Pharmaceuticals, all outside the submitted work. Balazs Halmos reports research funding and honoraria for steering committee work from Novartis during the conduct of the study; reports grants from AbbVie, Advaxis, AstraZeneca, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi Sankyo, GlaxoSmithKline, Janssen, Merck, and Pfizer; consulting fees from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, Genentech, Janssen, Merck, Pfizer, Takeda, and Veracyte; and participation in advisory board meetings for Apollomics and TPT, all outside the submitted work. Byoung Chul Cho reports research funding from AbbVie, AstraZeneca, Bayer, Blueprint Medicines, Champions Oncology, Dizal Pharma, Dong-A ST, Eli Lilly, GI Innovation, Interpark Bio Convergence Corp., Janssen, MedPacto, Merck, MOGAN Institute, Novartis, Ono Pharmaceutical, and Yuhan; royalties from Champions Oncology; consulting fees from AstraZeneca, Blueprint Medicines, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, Janssen, MedPacto, Merck, Novartis, Ono Pharmaceutical, Pfizer, Roche, Takeda, and Yuhan; participation in advisory board meetings for Bridgebio Therapeutics, Cyrus Therapeutics, Guardant Health, Joseah BIO, and KANAPH Therapeutics Inc.; stock ownership of Bridgebio Therapeutics, Cyrus Therapeutics, Gencurix Inc., Interpark Bio Convergence Corp., KANAPH Therapeutics Inc., and TheraCanVac Inc.; is a board of director for Gencurix Inc. and Interpark Bio Convergence Corp.; and is a founder of DAAN Biotherapeutics, all outside the submitted work. José Luis González Larriba reports consulting fees from Boehringer Ingelheim, Bristol Myers Squibb, Janssen, and Merck; speaker honoraria from Astellas Pharma, AstraZeneca, Bristol Myers Squibb, Janssen, Merck, Novartis, Pfizer, and Roche; and support for attending meetings from Bristol Myers Squibb, Janssen, Merck, Pfizer, Roche, and Takeda, all outside the submitted work. Caicun Zhou reports speaker honoraria from Amoy Diagnostics, Boehringer Ingelheim, C-stone, Eli Lilly, LUYE Pharma, Merck, Qilu, Roche, and Sanofi; and speaker and advisor fees from Hengrui, Innovent Biologics and TopAlliance Biosciences Inc., all outside the submitted work. Ingel Demedts reports consultancy and speaker honoraria from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Merck, and Roche, outside the submitted work. Jincheng Wu, David Demanse, and Bharani Dharan are Novartis employees. Bijoyesh Mookerjee, Socorro Portella, and Zewen Zhu were Novartis employees at the time the study was conducted. Martin Reck reports consulting fees, speaker honoraria and support for attending meetings from Amgen, AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Eli Lilly, Merck, Mirati Therapeutics, Novartis, Pfizer, Sanofi, and Roche; and participation in advisory board meetings for Sanofi, all outside of submitted work. Manuel Cobo, Akin Atmaca, and Sofia Baka declare no competing interests., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Long-term outcomes in patients with BRAF V600-mutant metastatic melanoma receiving dabrafenib monotherapy: Analysis from phase 2 and 3 clinical trials.
- Author
-
Hauschild A, Ascierto PA, Schadendorf D, Grob JJ, Ribas A, Kiecker F, Dutriaux C, Demidov LV, Lebbé C, Rutkowski P, Blank CU, Gutzmer R, Millward M, Kefford R, Haas T, D'Amelio A Jr, Gasal E, Mookerjee B, and Chapman PB
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Dacarbazine adverse effects, Disease Progression, Female, Follow-Up Studies, Humans, Imidazoles adverse effects, Male, Melanoma genetics, Melanoma mortality, Melanoma secondary, Middle Aged, Oximes adverse effects, Patient Selection, Progression-Free Survival, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Time Factors, Young Adult, Antineoplastic Agents administration & dosage, Dacarbazine administration & dosage, Imidazoles administration & dosage, Melanoma drug therapy, Oximes administration & dosage, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Skin Neoplasms drug therapy
- Abstract
Background: Previous analyses of BREAK-2 and BREAK-3 showed that durable outcomes lasting ≥3 years are achievable with dabrafenib in some patients with BRAF V600-mutant metastatic melanoma (MM); however, additional follow-up is needed to fully characterise the long-term impact of dabrafenib in these patients., Methods: BREAK-2 was a single-arm phase 2 study evaluating dabrafenib in treatment-naive or previously treated BRAF V600E/K-mutant MM. BREAK-3, a randomised (3:1) phase 3 study, assessed dabrafenib versus dacarbazine in previously untreated unresectable or metastatic BRAF V600E-mutant melanoma. Five-year analyses were performed., Results: All BREAK-2 patients (N = 92 [V600E, n = 76; V600K, n = 16]) discontinued treatment by the data cutoff. Median follow-up was 13.0 months. In BRAF V600E patients, 5-year progression-free survival (PFS) and overall survival (OS) were 11% and 20%, respectively. Subsequent immunotherapy was received by 22% of patients. In BREAK-3, median follow-up was 17.0 and 12.0 months in the dabrafenib (n = 187) and dacarbazine (n = 63) arms, respectively. Thirty-seven patients (59%) receiving dacarbazine crossed over to dabrafenib following disease progression as per protocol. Five-year PFS was 12% in the dabrafenib arm; all dacarbazine-arm patients progressed or were censored by 5 years. Dabrafenib improved PFS versus dacarbazine, regardless of baseline lactate dehydrogenase levels. Five-year OS rates were 24% and 22% in the dabrafenib and dacarbazine arms, respectively. Subsequent therapy in each arm included anti-CTLA-4 (dabrafenib [24%] and dacarbazine [24%]) and/or anti-PD-1 (8% and 2%) treatment. No new safety signals were observed., Conclusions and Relevance: These data, representing extended follow-up for dabrafenib monotherapy, demonstrate that durable benefit lasting ≥5 years is achievable in a subset of patients., Trial Registration: ClinicalTrials.gov (BREAK-2, NCT01153763; BREAK-3, NCT01227889)., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: long-term survival and safety analysis of a phase 3 study.
- Author
-
Long GV, Flaherty KT, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Chiarion-Sileni V, Lebbe C, Mandalá M, Millward M, Arance A, Bondarenko I, Haanen JBAG, Hansson J, Utikal J, Ferraresi V, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, Davies MA, Lane SR, Legos JJ, Mookerjee B, and Grob JJ
- Published
- 2019
- Full Text
- View/download PDF
17. Publisher Correction: Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma.
- Author
-
Ribas A, Lawrence D, Atkinson V, Agarwal S, Miller WH Jr, Carlino MS, Fisher R, Long GV, Hodi FS, Tsoi J, Grasso CS, Mookerjee B, Zhao Q, Ghori R, Moreno BH, Ibrahim N, and Hamid O
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
18. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma.
- Author
-
Ascierto PA, Ferrucci PF, Fisher R, Del Vecchio M, Atkinson V, Schmidt H, Schachter J, Queirolo P, Long GV, Di Giacomo AM, Svane IM, Lotem M, Bar-Sela G, Couture F, Mookerjee B, Ghori R, Ibrahim N, Moreno BH, and Ribas A
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Double-Blind Method, Female, Humans, Imidazoles administration & dosage, Kaplan-Meier Estimate, MAP Kinase Kinase Kinases antagonists & inhibitors, Male, Melanoma pathology, Middle Aged, Mutation, Oximes administration & dosage, Programmed Cell Death 1 Receptor antagonists & inhibitors, Progression-Free Survival, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Pyridones administration & dosage, Pyrimidinones administration & dosage, Skin Neoplasms pathology, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melanoma drug therapy, Melanoma genetics, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms drug therapy, Skin Neoplasms genetics
- Abstract
Blocking programmed death 1 (PD-1) may enhance the durability of anti-tumor responses that are induced by the combined inhibition of BRAF and MEK
1 . Here we performed a randomized phase 2 trial ( NCT02130466 ), in which patients with treatment-naive BRAFV600E/K -mutant, advanced melanoma received the BRAF inhibitor dabrafenib and the MEK inhibitor trametinib together with the PD-1-blocking antibody pembrolizumab (triplet; n = 60) or placebo (doublet; n = 60). The primary end point of progression-free survival was numerically improved in the triplet group-16.0 months-compared with 10.3 months in the doublet group (hazard ratio, 0.66; P = 0.043); however, the trial did not reach the planned benefit for a statistically significant improvement. Median duration of response was 18.7 months (95% confidence interval, 10.1-22.1) and 12.5 months (95% confidence interval, 6.0-14.1); 59.8 and 27.8% of responses were estimated to have lasted for more than 18 months for triplet and doublet treatment, respectively. Grade 3-5 treatment-related adverse events occurred in 58.3 and 26.7% of patients treated with triplet and doublet therapies, respectively, which were most commonly fever, increased transaminase levels and rash. One patient who received triplet therapy died of pneumonitis. In summary, triplet therapy with dabrafenib, trametinib and pembrolizumab conferred numerically longer progression-free survival and duration of response with a higher rate of grade 3/4 adverse events compared with the doublet therapy of dabrafenib, trametinib and placebo.- Published
- 2019
- Full Text
- View/download PDF
19. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma.
- Author
-
Ribas A, Lawrence D, Atkinson V, Agarwal S, Miller WH Jr, Carlino MS, Fisher R, Long GV, Hodi FS, Tsoi J, Grasso CS, Mookerjee B, Zhao Q, Ghori R, Moreno BH, Ibrahim N, and Hamid O
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Female, Humans, Imidazoles administration & dosage, Immunotherapy, Male, Melanoma secondary, Middle Aged, Mutation, Oximes administration & dosage, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Mas, Pyridones administration & dosage, Pyrimidinones administration & dosage, Skin Neoplasms therapy, Young Adult, MAP Kinase Kinase Kinases antagonists & inhibitors, Melanoma drug therapy, Melanoma genetics, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms drug therapy, Skin Neoplasms genetics
- Abstract
Oncogene-targeted therapy with B-Raf proto-oncogene (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors induces a high initial response rate in patients with BRAF
V600 -mutated melanoma, with a median duration of response of approximately 1 year1-3 . Immunotherapy with antibodies to programmed death 1 (PD-1) produces lower response rates but with long response duration. Preclinical models suggest that combining BRAF and MEK inhibitors with PD-1 blockade therapy improves antitumor activity4-6 , which may provide additional treatment options for patients unlikely to have long-lasting responses to either mode of therapy alone. We enrolled 15 patients with BRAFV600 -mutated metastatic melanoma in a first-in-human clinical trial of dabrafenib, trametinib and pembrolizumab ( NCT02130466 ). Eleven patients (73%) experienced grade 3/4 treatment-related adverse events, the most common being elevation of liver function tests and pyrexia, most of which resolved with drug interruption or discontinuation of either the anti-PD-1 antibody or the targeted therapy combination. Eleven patients (73%; 95% confidence interval = 45-92%) had an objective response, and six (40%; 95% confidence interval = 16-68%) continued with a response at a median follow-up of 27 months (range = 10.3-38.4+ months) for all patients. This study suggests that this triple-combined therapy may benefit a subset of patients with BRAFV600 -mutated metastatic melanoma by increasing the frequency of long-lasting antitumor responses.- Published
- 2019
- Full Text
- View/download PDF
20. Patient-reported outcomes in patients with resected, high-risk melanoma with BRAF V600E or BRAF V600K mutations treated with adjuvant dabrafenib plus trametinib (COMBI-AD): a randomised, placebo-controlled, phase 3 trial.
- Author
-
Schadendorf D, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, Robert C, Mortier L, Lesimple T, Plummer R, Schachter J, Dasgupta K, Manson S, Koruth R, Mookerjee B, Kefford R, Dummer R, Kirkwood JM, and Long GV
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Biomarkers, Tumor genetics, Chemotherapy, Adjuvant, Disease Progression, Humans, Imidazoles adverse effects, Melanoma genetics, Melanoma mortality, Melanoma secondary, Neoplasm Recurrence, Local, Neoplasm Staging, Oximes adverse effects, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins B-raf genetics, Pyridones adverse effects, Pyrimidinones adverse effects, Quality of Life, Risk Assessment, Risk Factors, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Time Factors, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Biomarkers, Tumor antagonists & inhibitors, Dermatologic Surgical Procedures adverse effects, Dermatologic Surgical Procedures mortality, Imidazoles administration & dosage, Melanoma therapy, Mutation, Oximes administration & dosage, Patient Reported Outcome Measures, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Pyridones administration & dosage, Pyrimidinones administration & dosage, Skin Neoplasms therapy
- Abstract
Background: In the phase 3 COMBI-AD study, patients with resected, stage III melanoma with BRAF
V600E or BRAFV600K mutations received adjuvant dabrafenib plus trametinib or placebo. The primary analysis showed that dabrafenib plus trametinib significantly improved relapse-free survival at 3 years. These results led to US Food and Drug Administration approval of dabrafenib plus trametinib as adjuvant treatment for patients with resected stage III melanoma with BRAFV600E or BRAFV600K mutations. Here, we report the patient-reported outcomes from COMBI-AD., Methods: COMBI-AD was a randomised, double-blind, placebo-controlled, phase 3 study done at 169 sites in 25 countries. Study participants were aged 18 years or older and had complete resection of stage IIIA (lymph node metastases >1 mm), IIIB, or IIIC cutaneous melanoma as per American Joint Committee on Cancer 7th edition criteria, with BRAFV600E or BRAFV600K mutations, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) via an interactive voice response system, stratified by mutation type and disease stage, to receive oral dabrafenib (150 mg twice daily) plus oral trametinib (2 mg once daily) or matching placebos for 12 months. Patients, physicians, and the investigators who analysed the data were masked to treatment allocation. The primary endpoint was relapse-free survival, reported elsewhere. Health-related quality of life, reported here, was a prespecified exploratory endpoint, and was assessed with the European Quality of Life 5-Dimensions 3-Levels (EQ-5D-3L) questionnaire in the intention-to-treat population. We used a mixed-model repeated-measures analysis to assess differences in health-related quality of life between groups. This study is registered with ClinicalTrials.gov, number NCT01682083. The trial is ongoing, but is no longer recruiting participants., Findings: Between Jan 31, 2013, and Dec 11, 2014, 870 patients were enrolled and randomly assigned to receive dabrafenib plus trametinib (n=438) or matching placebos (n=432). Data were collected until the data cutoff for analyses of the primary endpoint (June 30, 2017). The median follow-up was 34 months (IQR 28-39) in the dabrafenib plus trametinib group and 33 months (20·5-39) in the placebo group. During the 12-month treatment phase, there were no significant or clinically meaningful changes from baseline between groups in EQ-5D-3L visual analogue scale (EQ-VAS) or utility scores. During treatment, there were no clinically meaningful differences in VAS scores or utility scores in the dabrafenib plus trametinib group between patients who did and did not experience the most common adverse events. During long-term follow-up (range 15-48 months), VAS and utility scores were similar between groups and did not differ from baseline scores. At recurrence, there were significant decreases in VAS scores in both the dabrafenib plus trametinib group (mean change -6·02, SD 20·57; p=0·0032) and the placebo group (-6·84, 20·86; p<0·0001); the mean change in utility score also differed significantly at recurrence for both groups (dabrafenib plus trametinib -0·0626, 0·1911, p<0·0001; placebo -0·0748, 0·2182, p<0·0001)., Interpretation: These findings show that dabrafenib plus trametinib did not affect patient-reported outcome scores during or after adjuvant treatment, and suggest that preventing or delaying relapse with adjuvant therapy could be beneficial in this setting., Funding: Novartis., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
21. Five-year outcomes from a phase 3 METRIC study in patients with BRAF V600 E/K-mutant advanced or metastatic melanoma.
- Author
-
Robert C, Flaherty K, Nathan P, Hersey P, Garbe C, Milhem M, Demidov L, Mohr P, Hassel JC, Rutkowski P, Dummer R, Utikal J, Kiecker F, Larkin J, D'Amelio A Jr, Mookerjee B, and Schadendorf D
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms genetics, Brain Neoplasms secondary, Dacarbazine administration & dosage, Female, Follow-Up Studies, Humans, Male, Melanoma genetics, Melanoma pathology, Melanoma secondary, Middle Aged, Paclitaxel administration & dosage, Prognosis, Pyridones administration & dosage, Pyrimidinones administration & dosage, Skin Neoplasms genetics, Skin Neoplasms pathology, Skin Neoplasms secondary, Survival Rate, Time Factors, Young Adult, Melanoma, Cutaneous Malignant, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Melanoma drug therapy, Mutation, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms drug therapy
- Abstract
Background: Primary findings from the METRIC (TMT212A2301) study demonstrated that trametinib improved progression-free survival (PFS) and overall survival (OS) compared with chemotherapy in patients with unresectable or metastatic cutaneous melanoma with a BRAF V600 E/K mutation. However, clinical data characterising the long-term use of these therapies in combination with BRAF inhibitors or as monotherapies are limited., Methods: In this open-label, phase 3 study, 322 patients with BRAF V600 E/K-mutant metastatic melanoma were randomised in a 2:1 ratio to receive trametinib (2 mg orally, once daily; n = 214) or chemotherapy (dacarbazine [1000 mg/m
2 ] or paclitaxel [175 mg/m2 ] intravenously, every 3 weeks; n = 108). Patients who progressed on chemotherapy were allowed to cross over and receive trametinib. Five-year results of efficacy and safety analyses are reported., Results: The median PFS was 4.9 months in the trametinib arm versus 1.5 months in the chemotherapy arm (hazard ratio, 0.54; 95% confidence interval, 0.41-0.73). Landmark OS rates for trametinib versus chemotherapy arms at 1 year, 2 years and 5 years were 60.9% versus 49.6%, 32.0% versus 29.4% and 13.3% versus 17.0%, respectively. Most patients (n = 70 [65%]) from the chemotherapy arm crossed over to the trametinib arm early in their treatment. No unexpected adverse events were reported., Conclusions: This 5-year follow-up of patients with BRAF V600 E/K-mutant metastatic melanoma on a targeted therapy demonstrates that long-term use of trametinib is possible with no new or unexpected adverse events. Some patients experienced long-term survival benefit with trametinib monotherapy (METRIC ClinicalTrials.gov number, NCT01245062.)., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
22. Evaluation of the effect of dabrafenib and metabolites on QTc interval in patients with BRAF V600-mutant tumours.
- Author
-
Nebot N, Arkenau HT, Infante JR, Chandler JC, Weickhardt A, Lickliter JD, Sarantopoulos J, Gordon MS, Mak G, St-Pierre A, Tang L, Mookerjee B, Carson SW, Hayes S, and Grossmann KF
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Area Under Curve, Computer Simulation, Dose-Response Relationship, Drug, Electrocardiography, Ambulatory methods, Female, Humans, Imidazoles adverse effects, Imidazoles pharmacokinetics, Male, Middle Aged, Models, Biological, Mutation, Neoplasms genetics, Neoplasms pathology, Oximes adverse effects, Oximes pharmacokinetics, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Antineoplastic Agents administration & dosage, Imidazoles administration & dosage, Neoplasms drug therapy, Oximes administration & dosage, Proto-Oncogene Proteins B-raf genetics
- Abstract
Aims: The effect of repeat oral supratherapeutic dosing of the BRAF inhibitor dabrafenib on QTc interval was assessed in patients with BRAF V600-mutant tumours., Methods: Part 1 of this phase 1, multicentre, 2-part study (BRF113773/NCT01738451) assessed safety/tolerability of dabrafenib 225 or 300 mg twice daily (BID) to inform part 2 dosing. Patients in part 2 received dabrafenib-matched placebo on day -1, single-dose dabrafenib 300 mg on day 1, 300 mg BID on days 2 to 7, and 300 mg on day 8 (morning), followed by 24-h Holter electrocardiographic monitoring and pharmacokinetics sample collection each dose day. Pharmacokinetics/pharmacodynamics analysis assessed combined dabrafenib and metabolite effects on QTc interval., Results: Part 1 (n = 12) determined supratherapeutic dosing, 300 mg BID, for part 2. Thirty-one patients completed part 2. Mean maximum ΔΔQTcF occurred on day 8, 10 h postdose (2.86 msec; 90% CI, -1.36 to 7.07). Categorical analysis showed no placebo and dabrafenib outliers (increase >60 msec; QTcF >500 msec). Day 1 dabrafenib 300 mg C
max and AUC(0-∞) were ≈ 2-fold higher than with single-dose 150 mg. Day 8 AUC(0-τ) with 300 mg BID was ≈ 2.7-fold higher than with 150 mg BID. Dabrafenib metabolites showed similar trends. Pharmacokinetics/pharmacodynamics modelling/simulation showed that median QTc increase was <5 msec (upper 90% CI, <10 msec). No unexpected toxicities occurred with supratherapeutic dosing., Conclusion: Repeat oral supratherapeutic dabrafenib 300 mg BID dosing had no clinically relevant effect on QTc interval, with no new safety signals seen., (© 2017 The British Pharmacological Society.)- Published
- 2018
- Full Text
- View/download PDF
23. Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAF V600E -Mutant Colorectal Cancer.
- Author
-
Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, Muro K, Gordon MS, Tabernero J, Yaeger R, O'Dwyer PJ, Humblet Y, De Vos F, Jung AS, Brase JC, Jaeger S, Bettinger S, Mookerjee B, Rangwala F, and Van Cutsem E
- Subjects
- Colorectal Neoplasms enzymology, Colorectal Neoplasms metabolism, Drug Resistance, Neoplasm, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Female, Humans, Imidazoles therapeutic use, Male, Mitogen-Activated Protein Kinase Kinases drug effects, Mitogen-Activated Protein Kinase Kinases metabolism, Oximes therapeutic use, Panitumumab therapeutic use, Proto-Oncogene Proteins B-raf metabolism, Pyridones therapeutic use, Pyrimidinones therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, MAP Kinase Signaling System, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors
- Abstract
Although BRAF inhibitor monotherapy yields response rates >50% in BRAF
V600 -mutant melanoma, only approximately 5% of patients with BRAFV600E colorectal cancer respond. Preclinical studies suggest that the lack of efficacy in BRAFV600E colorectal cancer is due to adaptive feedback reactivation of MAPK signaling, often mediated by EGFR. This clinical trial evaluated BRAF and EGFR inhibition with dabrafenib (D) + panitumumab (P) ± MEK inhibition with trametinib (T) to achieve greater MAPK suppression and improved efficacy in 142 patients with BRAFV600E colorectal cancer. Confirmed response rates for D+P, D+T+P, and T+P were 10%, 21%, and 0%, respectively. Pharmacodynamic analysis of paired pretreatment and on-treatment biopsies found that efficacy of D+T+P correlated with increased MAPK suppression. Serial cell-free DNA analysis revealed additional correlates of response and emergence of KRAS and NRAS mutations on disease progression. Thus, targeting adaptive feedback pathways in BRAFV600E colorectal cancer can improve efficacy, but MAPK reactivation remains an important primary and acquired resistance mechanism. Significance: This trial demonstrates that combined BRAF + EGFR + MEK inhibition is tolerable, with promising activity in patients with BRAFV600E colorectal cancer. Our findings highlight the MAPK pathway as a critical target in BRAFV600E colorectal cancer and the need to optimize strategies inhibiting this pathway to overcome both primary and acquired resistance. Cancer Discov; 8(4); 428-43. ©2018 AACR. See related commentary by Janku, p. 389 See related article by Hazar-Rethinam et al., p. 417 This article is highlighted in the In This Issue feature, p. 371 ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
24. Long-Term Outcomes in Patients With BRAF V600-Mutant Metastatic Melanoma Who Received Dabrafenib Combined With Trametinib.
- Author
-
Long GV, Eroglu Z, Infante J, Patel S, Daud A, Johnson DB, Gonzalez R, Kefford R, Hamid O, Schuchter L, Cebon J, Sharfman W, McWilliams R, Sznol M, Redhu S, Gasal E, Mookerjee B, Weber J, and Flaherty KT
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Imidazoles administration & dosage, Imidazoles adverse effects, Male, Melanoma genetics, Melanoma mortality, Middle Aged, Oximes administration & dosage, Oximes adverse effects, Pyridones administration & dosage, Pyridones adverse effects, Pyrimidinones administration & dosage, Pyrimidinones adverse effects, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melanoma drug therapy, Mutation, Proto-Oncogene Proteins B-raf genetics
- Abstract
Purpose To report 5-year landmark analysis efficacy and safety outcomes in patients with BRAF V600-mutant metastatic melanoma (MM) who received BRAF inhibitor dabrafenib (D) and MEK inhibitor trametinib (T) combination therapy versus D monotherapy in the randomized phase II BRF113220 study part C. Patients and Methods BRAF inhibitor-naive patients with BRAF V600-mutant MM were randomly assigned 1:1:1 to receive D 150 mg twice a day, D 150 mg twice a day plus T 1 mg once daily, or D 150 mg twice a day plus T 2 mg once daily (D + T 150/2). Patients who received D monotherapy could cross over to D + T 150/2 postprogression. Efficacy and safety were analyzed 4 and 5 years after initiation in patients with ≥ 5 years of follow-up. Results As of October 13, 2016, 18 patients who received D + T 150/2 remained in the study (13 [24%] of 54 enrolled at this dose plus five [11%] of 45 initially administered D who crossed over to D + T). With D + T 150/2, overall survival (OS; 4 years, 30%; 5 years, 28%) and progression-free survival (4 and 5 years, both 13%) appeared to stabilize with extended follow-up. Increased OS was observed in patients who received D + T with baseline normal lactate dehydrogenase (5 years, 45%) and normal lactate dehydrogenase with fewer than three organ sites with metastasis (5 years, 51%). With extended follow-up, one additional patient who received D + T 150/2 improved from a partial to a complete response. No new safety signals were observed. Conclusion This 5-year analysis represents the longest follow-up to date with BRAF + MEK inhibitor combination therapy in BRAF V600-mutant MM. Consistent with trends observed in landmark analyses with shorter follow-up, this therapy elicits durable plateaus of long-term OS and progression-free survival that last ≥ 5 years in some patients with MM.
- Published
- 2018
- Full Text
- View/download PDF
25. Dabrafenib and Trametinib Treatment in Patients With Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer.
- Author
-
Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski C, Cabanillas ME, Urbanowitz G, Mookerjee B, Wang D, Rangwala F, and Keam B
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Disease-Free Survival, Fatigue chemically induced, Female, Fever chemically induced, Humans, Imidazoles administration & dosage, Imidazoles adverse effects, Male, Middle Aged, Nausea chemically induced, Neoplasm Metastasis, Oximes administration & dosage, Oximes adverse effects, Pyridones administration & dosage, Pyridones adverse effects, Pyrimidinones administration & dosage, Pyrimidinones adverse effects, Thyroid Carcinoma, Anaplastic genetics, Thyroid Carcinoma, Anaplastic pathology, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Mutation, Proto-Oncogene Proteins B-raf genetics, Thyroid Carcinoma, Anaplastic drug therapy
- Abstract
Purpose We report the efficacy and safety of dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) combination therapy in BRAF V600E-mutated anaplastic thyroid cancer, a rare, aggressive, and highly lethal malignancy with poor patient outcomes and no systemic therapies with clinical benefit. Methods In this phase II, open-label trial, patients with predefined BRAF V600E-mutated malignancies received dabrafenib 150 mg twice daily and trametinib 2 mg once daily until unacceptable toxicity, disease progression, or death. The primary end point was investigator-assessed overall response rate. Secondary end points included duration of response, progression-free survival, overall survival, and safety. Results Sixteen patients with BRAF V600E-mutated anaplastic thyroid cancer were evaluable (median follow-up, 47 weeks; range, 4 to 120 weeks). All patients had received prior radiation treatment and/or surgery, and six had received prior systemic therapy. The confirmed overall response rate was 69% (11 of 16; 95% CI, 41% to 89%), with seven ongoing responses. Median duration of response, progression-free survival, and overall survival were not reached as a result of a lack of events, with 12-month estimates of 90%, 79%, and 80%, respectively. The safety population was composed of 100 patients who were enrolled with seven rare tumor histologies. Common adverse events were fatigue (38%), pyrexia (37%), and nausea (35%). No new safety signals were detected. Conclusion Dabrafenib plus trametinib is the first regimen demonstrated to have robust clinical activity in BRAF V600E-mutated anaplastic thyroid cancer and was well tolerated. These findings represent a meaningful therapeutic advance for this orphan disease.
- Published
- 2018
- Full Text
- View/download PDF
26. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma.
- Author
-
Long GV, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, Robert C, Mortier L, Schachter J, Schadendorf D, Lesimple T, Plummer R, Ji R, Zhang P, Mookerjee B, Legos J, Kefford R, Dummer R, and Kirkwood JM
- Subjects
- Adjuvants, Immunologic adverse effects, Adolescent, Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Double-Blind Method, Female, Humans, Imidazoles adverse effects, Male, Melanoma genetics, Melanoma mortality, Melanoma surgery, Middle Aged, Mutation, Neoplasm Recurrence, Local, Neoplasm Staging, Oximes adverse effects, Proto-Oncogene Proteins B-raf genetics, Pyridones adverse effects, Pyrimidinones adverse effects, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms surgery, Survival Analysis, Young Adult, Melanoma, Cutaneous Malignant, Adjuvants, Immunologic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Imidazoles therapeutic use, Melanoma drug therapy, Oximes therapeutic use, Pyridones therapeutic use, Pyrimidinones therapeutic use, Skin Neoplasms drug therapy
- Abstract
Background: Combination therapy with the BRAF inhibitor dabrafenib plus the MEK inhibitor trametinib improved survival in patients with advanced melanoma with BRAF V600 mutations. We sought to determine whether adjuvant dabrafenib plus trametinib would improve outcomes in patients with resected, stage III melanoma with BRAF V600 mutations., Methods: In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned 870 patients with completely resected, stage III melanoma with BRAF V600E or V600K mutations to receive oral dabrafenib at a dose of 150 mg twice daily plus trametinib at a dose of 2 mg once daily (combination therapy, 438 patients) or two matched placebo tablets (432 patients) for 12 months. The primary end point was relapse-free survival. Secondary end points included overall survival, distant metastasis-free survival, freedom from relapse, and safety., Results: At a median follow-up of 2.8 years, the estimated 3-year rate of relapse-free survival was 58% in the combination-therapy group and 39% in the placebo group (hazard ratio for relapse or death, 0.47; 95% confidence interval [CI], 0.39 to 0.58; P<0.001). The 3-year overall survival rate was 86% in the combination-therapy group and 77% in the placebo group (hazard ratio for death, 0.57; 95% CI, 0.42 to 0.79; P=0.0006), but this level of improvement did not cross the prespecified interim analysis boundary of P=0.000019. Rates of distant metastasis-free survival and freedom from relapse were also higher in the combination-therapy group than in the placebo group. The safety profile of dabrafenib plus trametinib was consistent with that observed with the combination in patients with metastatic melanoma., Conclusions: Adjuvant use of combination therapy with dabrafenib plus trametinib resulted in a significantly lower risk of recurrence in patients with stage III melanoma with BRAF V600E or V600K mutations than the adjuvant use of placebo and was not associated with new toxic effects. (Funded by GlaxoSmithKline and Novartis; COMBI-AD ClinicalTrials.gov, NCT01682083 ; EudraCT number, 2012-001266-15 .).
- Published
- 2017
- Full Text
- View/download PDF
27. Dabrafenib plus trametinib in patients with previously untreated BRAF V600E -mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial.
- Author
-
Planchard D, Smit EF, Groen HJM, Mazieres J, Besse B, Helland Å, Giannone V, D'Amelio AM Jr, Zhang P, Mookerjee B, and Johnson BE
- Subjects
- Administration, Oral, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Confidence Intervals, Disease-Free Survival, Dose-Response Relationship, Drug, Drug Administration Schedule, Gene Expression Regulation, Neoplastic, Humans, Imidazoles adverse effects, Imidazoles therapeutic use, Lung Neoplasms genetics, Lung Neoplasms mortality, Lung Neoplasms pathology, Maximum Tolerated Dose, Middle Aged, Mutation, Neoplasm Invasiveness pathology, Neoplasm Metastasis, Neoplasm Staging, Oximes adverse effects, Oximes therapeutic use, Prognosis, Prospective Studies, Pyridones adverse effects, Pyrimidinones adverse effects, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Proto-Oncogene Proteins B-raf genetics, Pyridones therapeutic use, Pyrimidinones therapeutic use
- Abstract
Background: BRAF
V600E mutation occurs in 1-2% of lung adenocarcinomas and acts as an oncogenic driver. Dabrafenib, alone or combined with trametinib, has shown substantial antitumour activity in patients with previously treated BRAFV600E -mutant metastatic non-small-cell lung cancer (NSCLC). We aimed to assess the activity and safety of dabrafenib plus trametinib treatment in previously untreated patients with BRAFV600E -mutant metastatic NSCLC., Methods: In this phase 2, sequentially enrolled, multicohort, multicentre, non-randomised, open-label study, adults (≥18 years of age) with previously untreated metastatic BRAFV600E -mutant NSCLC were enrolled into cohort C from 19 centres in eight countries within North America, Europe, and Asia. Patients received oral dabrafenib 150 mg twice per day plus oral trametinib 2 mg once per day until disease progression, unacceptable adverse events, consent withdrawal, or death. The primary endpoint was investigator-assessed overall response, defined as the percentage of patients who achieved a confirmed complete response or partial response per Response Evaluation Criteria In Solid Tumors version 1.1. The primary and safety analyses were by intention to treat in the protocol-defined population (previously untreated patients). The study is ongoing, but no longer recruiting patients. This trial is registered with ClinicalTrials.gov, number NCT01336634., Findings: Between April 16, 2014, and Dec 28, 2015, 36 patients were enrolled and treated with first-line dabrafenib plus trametinib. Median follow-up was 15·9 months (IQR 7·8-22·0) at the data cutoff (April 28, 2017). The proportion of patients with investigator-assessed confirmed overall response was 23 (64%, 95% CI 46-79), with two (6%) patients achieving a complete response and 21 (58%) a partial response. All patients had one or more adverse event of any grade, and 25 (69%) had one or more grade 3 or 4 event. The most common (occurring in more than two patients) grade 3 or 4 adverse events were pyrexia (four [11%]), alanine aminotransferase increase (four [11%]), hypertension (four [11%]), and vomiting (three [8%]). Serious adverse events occurring in more than two patients included alanine aminotransferase increase (five [14%]), pyrexia (four [11%]), aspartate aminotransferase increase (three [8%]), and ejection fraction decrease (three [8%]). One fatal serious adverse event deemed unrelated to study treatment was reported (cardiorespiratory arrest)., Interpretation: Dabrafenib plus trametinib represents a new therapy with clinically meaningful antitumour activity and a manageable safety profile in patients with previously untreated BRAFV600E -mutant NSCLC., Funding: Novartis., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
28. Three-year pooled analysis of factors associated with clinical outcomes across dabrafenib and trametinib combination therapy phase 3 randomised trials.
- Author
-
Schadendorf D, Long GV, Stroiakovski D, Karaszewska B, Hauschild A, Levchenko E, Chiarion-Sileni V, Schachter J, Garbe C, Dutriaux C, Gogas H, Mandalà M, Haanen JBAG, Lebbé C, Mackiewicz A, Rutkowski P, Grob JJ, Nathan P, Ribas A, Davies MA, Zhang Y, Kaper M, Mookerjee B, Legos JJ, Flaherty KT, and Robert C
- Subjects
- Adult, Aged, Female, Humans, Imidazoles administration & dosage, L-Lactate Dehydrogenase metabolism, Male, Melanoma metabolism, Melanoma pathology, Middle Aged, Neoplasm Metastasis pathology, Oximes administration & dosage, Predictive Value of Tests, Prognosis, Pyridones administration & dosage, Pyrimidinones administration & dosage, Risk Factors, Survival Analysis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melanoma drug therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
Aim: Understanding predictors of long-term benefit with currently available melanoma therapies is the key for optimising individualised treatments. A prior pooled analysis of dabrafenib plus trametinib (D + T)-randomised trials (median follow-up, 20.0 months) identified baseline lactate dehydrogenase (LDH) and number of organ sites with metastasis as predictive factors for progression-free (PFS) and overall (OS) survival. However, longer-term follow-up analyses are needed to confirm which patients treated with D + T can achieve maximum benefit., Methods: Three-year landmark data were retrospectively pooled for D + T patients in phase 3 trials (COMBI-d [NCT01584648]; COMBI-v [NCT01597908]). Univariate and multivariate analyses assessed prognostic values of predefined baseline factors; regression tree analysis determined hierarchy and interactions between variables., Results: Long-term pooled outcomes were consistent with individual trial results (N = 563; 3-year PFS, 23%; 3-year OS, 44%). Baseline LDH level and number of organ sites remained strongly associated with and/or predictive of PFS and OS. In addition, baseline sum of lesion diameters (SLD) was identified as a predictor for progression. In the most favourable prognostic group (normal LDH, SLD <66 mm, <3 organ sites; n = 183/563 [33%]), 3-year PFS was 42%. Baseline number of organ sites was also predictive of outcomes in patients with PFS ≥ 6 months., Conclusion: Using the largest phase 3 data set available for BRAF/MEK inhibitor combination therapy in melanoma, these results demonstrate that durable responses lasting ≥3 years are possible in subsets of patients with BRAF-mutant melanoma receiving D + T. Although the best predictive model evolved with longer follow-up, factors predicting clinical outcomes with the combination remained consistent with previous analyses., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. Dabrafenib plus trametinib in patients with BRAF V600 -mutant melanoma brain metastases (COMBI-MB): a multicentre, multicohort, open-label, phase 2 trial.
- Author
-
Davies MA, Saiag P, Robert C, Grob JJ, Flaherty KT, Arance A, Chiarion-Sileni V, Thomas L, Lesimple T, Mortier L, Moschos SJ, Hogg D, Márquez-Rodas I, Del Vecchio M, Lebbé C, Meyer N, Zhang Y, Huang Y, Mookerjee B, and Long GV
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Brain Neoplasms diagnostic imaging, Brain Neoplasms genetics, Brain Neoplasms secondary, Female, Fever chemically induced, Headache chemically induced, Humans, Imidazoles administration & dosage, Imidazoles adverse effects, Magnetic Resonance Imaging, Male, Melanoma diagnostic imaging, Melanoma genetics, Melanoma secondary, Middle Aged, Mutation, Oximes administration & dosage, Oximes adverse effects, Prospective Studies, Pyridones administration & dosage, Pyridones adverse effects, Pyrimidinones administration & dosage, Pyrimidinones adverse effects, Stroke Volume drug effects, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Melanoma drug therapy, Proto-Oncogene Proteins B-raf genetics
- Abstract
Background: Dabrafenib plus trametinib improves clinical outcomes in BRAF
V600 -mutant metastatic melanoma without brain metastases; however, the activity of dabrafenib plus trametinib has not been studied in active melanoma brain metastases. Here, we report results from the phase 2 COMBI-MB trial. Our aim was to build on the current body of evidence of targeted therapy in melanoma brain metastases through an evaluation of dabrafenib plus trametinib in patients with BRAFV600 -mutant melanoma brain metastases., Methods: This ongoing, multicentre, multicohort, open-label, phase 2 study evaluated oral dabrafenib (150 mg twice per day) plus oral trametinib (2 mg once per day) in four patient cohorts with melanoma brain metastases enrolled from 32 hospitals and institutions in Europe, North America, and Australia: (A) BRAFV600E -positive, asymptomatic melanoma brain metastases, with no previous local brain therapy, and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1; (B) BRAFV600E -positive, asymptomatic melanoma brain metastases, with previous local brain therapy, and an ECOG performance status of 0 or 1; (C) BRAFV600D/K/R -positive, asymptomatic melanoma brain metastases, with or without previous local brain therapy, and an ECOG performance status of 0 or 1; and (D) BRAFV600D/E/K/R -positive, symptomatic melanoma brain metastases, with or without previous local brain therapy, and an ECOG performance status of 0, 1, or 2. The primary endpoint was investigator-assessed intracranial response in cohort A in the all-treated-patients population. Secondary endpoints included intracranial response in cohorts B, C, and D. This study is registered with ClinicalTrials.gov, number NCT02039947., Findings: Between Feb 28, 2014, and Aug 5, 2016, 125 patients were enrolled in the study: 76 patients in cohort A; 16 patients in cohort B; 16 patients in cohort C; and 17 patients in cohort D. At the data cutoff (Nov 28, 2016) after a median follow-up of 8·5 months (IQR 5·5-14·0), 44 (58%; 95% CI 46-69) of 76 patients in cohort A achieved an intracranial response. Intracranial response by investigator assessment was also achieved in nine (56%; 95% CI 30-80) of 16 patients in cohort B, seven (44%; 20-70) of 16 patients in cohort C, and ten (59%; 33-82) of 17 patients in cohort D. The most common serious adverse events related to study treatment were pyrexia for dabrafenib (eight [6%] of 125 patients) and decreased ejection fraction (five [4%]) for trametinib. The most common grade 3 or worse adverse events, regardless of study drug relationship, were pyrexia (four [3%] of 125) and headache (three [2%])., Interpretation: Dabrafenib plus trametinib was active with a manageable safety profile in this melanoma population that was consistent with previous dabrafenib plus trametinib studies in patients with BRAFV600 -mutant melanoma without brain metastases, but the median duration of response was relatively short. These results provide evidence of clinical benefit with dabrafenib plus trametinib and support the need for additional research to further improve outcomes in patients with melanoma brain metastases., Funding: Novartis., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
30. Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: long-term survival and safety analysis of a phase 3 study.
- Author
-
Long GV, Flaherty KT, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Chiarion-Sileni V, Lebbe C, Mandalà M, Millward M, Arance A, Bondarenko I, Haanen JBAG, Hansson J, Utikal J, Ferraresi V, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, Davies MA, Lane SR, Legos JJ, Mookerjee B, and Grob JJ
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Disease Progression, Disease-Free Survival, Double-Blind Method, Drug Administration Schedule, Humans, Imidazoles adverse effects, Imidazoles pharmacokinetics, Kaplan-Meier Estimate, Melanoma genetics, Melanoma mortality, Melanoma secondary, Oximes adverse effects, Oximes pharmacokinetics, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Pyridones adverse effects, Pyridones pharmacokinetics, Pyrimidinones adverse effects, Pyrimidinones pharmacokinetics, Risk Factors, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Time Factors, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Imidazoles administration & dosage, Melanoma drug therapy, Mutation, Oximes administration & dosage, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins B-raf genetics, Pyridones administration & dosage, Pyrimidinones administration & dosage, Skin Neoplasms drug therapy
- Abstract
Background: Previous analysis of COMBI-d (NCT01584648) demonstrated improved progression-free survival (PFS) and overall survival (OS) with combination dabrafenib and trametinib versus dabrafenib monotherapy in BRAF V600E/K-mutant metastatic melanoma. This study was continued to assess 3-year landmark efficacy and safety after ≥36-month follow-up for all living patients., Patients and Methods: This double-blind, phase 3 study enrolled previously untreated patients with BRAF V600E/K-mutant unresectable stage IIIC or stage IV melanoma. Patients were randomized to receive dabrafenib (150 mg twice daily) plus trametinib (2 mg once daily) or dabrafenib plus placebo. The primary endpoint was PFS; secondary endpoints were OS, overall response, duration of response, safety, and pharmacokinetics., Results: Between 4 May and 30 November 2012, a total of 423 of 947 screened patients were randomly assigned to receive dabrafenib plus trametinib (n = 211) or dabrafenib monotherapy (n = 212). At data cut-off (15 February 2016), outcomes remained superior with the combination: 3-year PFS was 22% with dabrafenib plus trametinib versus 12% with monotherapy, and 3-year OS was 44% versus 32%, respectively. Twenty-five patients receiving monotherapy crossed over to combination therapy, with continued follow-up under the monotherapy arm (per intent-to-treat principle). Of combination-arm patients alive at 3 years, 58% remained on dabrafenib plus trametinib. Three-year OS with the combination reached 62% in the most favourable subgroup (normal lactate dehydrogenase and <3 organ sites with metastasis) versus only 25% in the unfavourable subgroup (elevated lactate dehydrogenase). The dabrafenib plus trametinib safety profile was consistent with previous clinical trial observations, and no new safety signals were detected with long-term use., Conclusions: These data demonstrate that durable (≥3 years) survival is achievable with dabrafenib plus trametinib in patients with BRAF V600-mutant metastatic melanoma and support long-term first-line use of the combination in this setting., (© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.)
- Published
- 2017
- Full Text
- View/download PDF
31. A Phase 1/1b Study Evaluating Trametinib Plus Docetaxel or Pemetrexed in Patients With Advanced Non-Small Cell Lung Cancer.
- Author
-
Gandara DR, Leighl N, Delord JP, Barlesi F, Bennouna J, Zalcman G, Infante JR, Reckamp KL, Kelly K, Shepherd FA, Mazieres J, Janku F, Gardner OS, Mookerjee B, Wu Y, Cox DS, Schramek D, Peddareddigari V, Liu Y, D'Amelio AM Jr, and Blumenschein G Jr
- Subjects
- Adenocarcinoma secondary, Adult, Aged, Aged, 80 and over, Carcinoma, Large Cell secondary, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell secondary, Docetaxel, Female, Follow-Up Studies, Humans, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Pemetrexed administration & dosage, Prognosis, Pyridones administration & dosage, Pyrimidinones administration & dosage, Survival Rate, Taxoids administration & dosage, Adenocarcinoma drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Large Cell drug therapy, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Squamous Cell drug therapy, Lung Neoplasms drug therapy
- Abstract
Objectives: This two-part study evaluated trametinib, a MEK1/2 inhibitor, in combination with anticancer agents. Inhibition of MEK, a downstream effector of KRAS, demonstrated preclinical synergy with chemotherapy in KRAS-mutant NSCLC cell lines. Part 1 of this study identified recommended phase 2 doses of trametinib combinations. Part 2, reported herein, evaluated the safety, tolerability, pharmacokinetics, and efficacy of trametinib combinations in patients with NSCLC with and without KRAS mutations., Methods: Phase 1b evaluated trametinib plus docetaxel with growth factor support (trametinib, 2.0 mg once daily, and docetaxel, 75 mg/m
2 every 3 weeks) or pemetrexed (trametinib, 1.5 mg once daily, and pemetrexed, 500 mg/m2 every 3 weeks). Eligibility criteria for the expansion cohorts included metastatic NSCLC with measurable disease, known KRAS mutation status, Eastern Cooperative Oncology Group performance status of 1 or lower, and no more than two prior regimens., Results: The primary end point of overall response rate (ORR) was met for both combinations. A confirmed partial response (PR) was observed in 10 of the 47 patients with NSCLC who received trametinib plus docetaxel (21%). The ORR was 18% (four PRs in 22 patients) in those with KRAS wild-type NSCLC versus 24% (six PRs in 25 patients) in those with KRAS-mutant NSCLC. Of the 42 patients with NSCLC treated with trametinib plus pemetrexed, six (14%) had a PR; the ORR was 17% (four of 23) in patients with KRAS-mutated NSCLC versus 11% (two of 19) in KRAS wild-type NSCLC. Adverse events-most commonly diarrhea, nausea, and fatigue-were manageable., Conclusions: Trametinib-plus-chemotherapy combinations were tolerable. Clinical activity exceeding the ORRs previously reported with docetaxel or pemetrexed alone in KRAS-mutated NSCLC and meeting prespecified criteria was observed., (Copyright © 2016 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
32. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial.
- Author
-
Planchard D, Besse B, Groen HJM, Souquet PJ, Quoix E, Baik CS, Barlesi F, Kim TM, Mazieres J, Novello S, Rigas JR, Upalawanna A, D'Amelio AM Jr, Zhang P, Mookerjee B, and Johnson BE
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma secondary, Adult, Aged, Biomarkers, Tumor genetics, Carcinoma, Large Cell drug therapy, Carcinoma, Large Cell genetics, Carcinoma, Large Cell secondary, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung secondary, Female, Follow-Up Studies, Humans, Imidazoles administration & dosage, Lung Neoplasms genetics, Lung Neoplasms pathology, Lymphatic Metastasis, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Oximes administration & dosage, Prognosis, Pyridones administration & dosage, Pyrimidinones administration & dosage, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Mutation genetics, Neoplasm Recurrence, Local drug therapy, Proto-Oncogene Proteins B-raf genetics
- Abstract
Background: BRAF mutations act as an oncogenic driver via the mitogen-activated protein kinase (MAPK) pathway in non-small cell lung cancer (NSCLC). BRAF inhibition has shown antitumour activity in patients with BRAF(V600E)-mutant NSCLC. Dual MAPK pathway inhibition with BRAF and MEK inhibitors in BRAF(V600E)-mutant NSCLC might improve efficacy over BRAF inhibitor monotherapy based on observations in BRAF(V600)-mutant melanoma. We aimed to assess the antitumour activity and safety of dabrafenib plus trametinib in patients with BRAF(V600E)-mutant NSCLC., Methods: In this phase 2, multicentre, non-randomised, open-label study, we enrolled adult patients (aged ≥18 years) with pretreated metastatic stage IV BRAF(V600E)-mutant NSCLC who had documented tumour progression after at least one previous platinum-based chemotherapy and had had no more than three previous systemic anticancer therapies. Patients with previous BRAF or MEK inhibitor treatment were ineligible. Patients with brain metastases were allowed to enrol only if the lesions were asymptomatic, untreated (or stable more than 3 weeks after local therapy if treated), and measured less than 1 cm. Enrolled patients received oral dabrafenib (150 mg twice daily) plus oral trametinib (2 mg once daily) in continuous 21-day cycles until disease progression, unacceptable adverse events, withdrawal of consent, or death. The primary endpoint was investigator-assessed overall response, which was assessed by intention to treat in the protocol-defined population (patients who received second-line or later treatment); safety was also assessed in this population and was assessed at least once every 3 weeks, with adverse events, laboratory values, and vital signs graded according to the Common Terminology Criteria for Adverse Events version 4.0. The study is ongoing but no longer recruiting patients. This trial is registered with ClinicalTrials.gov, number NCT01336634., Findings: Between Dec 20, 2013, and Jan 14, 2015, 59 patients from 30 centres in nine countries across North America, Europe, and Asia met eligibility criteria. Two patients who had previously been untreated due to protocol deviation were excluded; thus, 57 eligible patients were enrolled. 36 patients (63·2% [95% CI 49·3-75·6]) achieved an investigator-assessed overall response. Serious adverse events were reported in 32 (56%) of 57 patients and included pyrexia in nine (16%), anaemia in three (5%), confusional state in two (4%), decreased appetite in two (4%), haemoptysis in two (4%), hypercalcaemia in two (4%), nausea in two (4%), and cutaneous squamous cell carcinoma in two (4%). The most common grade 3-4 adverse events were neutropenia in five patients (9%), hyponatraemia in four (7%), and anaemia in three (5%). Four patients died during the study from fatal adverse events judged to be unrelated to treatment (one retroperitoneal haemorrhage, one subarachnoid haemorrhage, one respiratory distress, and one from disease progression that was more severe than typical progression, as assessed by the investigator)., Interpretation: Dabrafenib plus trametinib could represent a new targeted therapy with robust antitumour activity and a manageable safety profile in patients with BRAF(V600E)-mutant NSCLC., Funding: GlaxoSmithKline., Competing Interests: Declaration of interests DP acts as an advisor for AstraZeneca, Boehringer, Bristol-Myers Squibb, Clovis, Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Roche, and Sanofi and has received research funding from Novartis unrelated to the current trial. BB’s institution received a grant from Novartis for this study. HJMG’s institution has received payments from Eli Lilly, Merck Sharp & Dohme, and Roche. P-JS has been involved in clinical trials for GlaxoSmithKline and Novartis. EQ has received personal fees from AbbVie, Bristol-Myers Squibb, Clovis, Lilly, Pfizer, and Roche, has received grants from Amgen, Boehringer, Bristol-Myers Squibb, Lilly, Merck Sharp & Dohme, Mundipharma, and Roche, and has received non-financial support from Boehringer. CSB’s institution received a grant for this study from Novartis and CSB has received personal fees from Novartis. FB has received personal fees from Novartis. SN has received personal fees from Boehringer, Bristol-Myers Squibb, Eli Lilly, Merck Sharp & Dohme, and Roche. AU, PZ, AD’A, and BM are currently employees of Novartis. AD’A and BM were employees of GlaxoSmithKline during a portion of the study. AD’A and BM own stock in GlaxoSmithKline and Novartis and BM owns stock in Incyte and AstraZeneca. BEJ has received personal fees from Amgen, AstraZeneca, Boehringer, Chugai Pharmaceuticals, Clovis, Genentech, KEW Group, Merck, and Novartis, and shares of post-market revenue for an EGFR genotyping patent. The other authors declare no competing interests., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial.
- Author
-
Planchard D, Kim TM, Mazieres J, Quoix E, Riely G, Barlesi F, Souquet PJ, Smit EF, Groen HJ, Kelly RJ, Cho BC, Socinski MA, Pandite L, Nase C, Ma B, D'Amelio A Jr, Mookerjee B, Curtis CM Jr, and Johnson BE
- Subjects
- Administration, Oral, Adolescent, Adult, Aged, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Disease-Free Survival, Dose-Response Relationship, Drug, Female, Humans, Imidazoles adverse effects, Male, Middle Aged, Mutation, Neoplasm Staging, Oximes adverse effects, Protein Kinase Inhibitors adverse effects, Carcinoma, Non-Small-Cell Lung drug therapy, Imidazoles administration & dosage, Oximes administration & dosage, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins B-raf genetics
- Abstract
Background: Activating BRAF(V600E) (Val600Glu) mutations are found in about 1-2% of lung adenocarcinomas, which might provide an opportunity for targeted treatment in these patients. Dabrafenib is an oral selective inhibitor of BRAF kinase. We did a trial to assess the clinical activity of dabrafenib in patients with advanced non-small-cell lung cancer (NSCLC) positive for the BRAF(V600E) mutation., Methods: In this phase 2, multicentre, non-randomised, open-label study, we enrolled previously treated and untreated patients with stage IV metastatic BRAF(V600E)-positive NSCLC. Patients received oral dabrafenib 150 mg twice daily. The primary endpoint was investigator-assessed overall response, which was assessed in patients who had received at least one dose of dabrafenib; safety was also assessed in this population. The study is ongoing but not enrolling patients in this cohort. This trial is registered with ClinicalTrials.gov, number NCT01336634., Findings: Between Aug 3, 2011, and Feb 25, 2014, 84 patients were enrolled, six of whom had not previously received systemic treatment for NSCLC. 26 of the 78 previously treated patients achieved an investigator-assessed overall response (33% [95% CI 23-45]). Four of the six previously untreated patients had an objective response. One patient died from an intracranial haemorrhage that was judged by the investigator to be due to the study drug. Serious adverse events were reported in 35 (42%) of 84 patients. The most frequent grade 3 or worse adverse events were cutaneous squamous-cell carcinoma in ten (12%), asthenia in four (5%), and basal-cell carcinoma in four (5%)., Interpretation: Dabrafenib showed clinical activity in BRAF(V600E)-positive NSCLC. Our findings suggest that dabrafenib could represent a treatment option for a population of patients with limited therapeutic options., Funding: GlaxoSmithKline., Competing Interests: Declaration of interests DP reports advisory role for Novartis, Pfizer, Roche, Boehinger, Lilly, Bristol-Myers Squibb, AstraZeneca, MSD and Pierre Fabre. GR reports grants from GlaxoSmithKline and Novartis; consulting with Novartis; and employer receives research funding from Pfizer, Chugai/Roche and Millennium for support of research led by him. FB reports personal fees from GlaxoSmithKline and Novartis. P-J Souquet reports clinical trial for GlaxoSmithKline. HJMG reports payments to his institution from Roche, MSD, Pfizer and GlaxoSmithKline. LP at the time of the study, was an employee of GlaxoSmithKline. CN reports employment at GlaxoSmithKline at the time the study was conducted, former employment at Novartis and current employment at GlaxoSmithKline. B. Ma reports employment at GlaxoSmithKline at the time the study was conducted. AD, at the time of the study, was an employee of GlaxoSmithKline, is now an employee of Novartis. B Mookerjee, at the time of the study, was an employee of GlaxoSmithKline, is now an employee of Novartis; and owns stock in GlaxoSmithKline and Novartis. CMC was an employee of Novartis and GlaxoSmithKline. BEJ reports personal fees from AstraZeneca, Clovis Oncology, Novartis, Merck, Genentech; honoraria from Chugai Pharmaceuticals; received shares of post-market revenue for EGFR Genotyping patent. TMK, JM, EQ, EFS, RJK, BCC, and MAS report nothing to disclose., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Pyrexia in dabrafenib-treated melanoma patients is not associated with common genetic variation or HLA polymorphisms.
- Author
-
Kulkarni D, Song K, Briley L, King K, Dabrowski C, Mookerjee B, Legos J, and Spraggs C
- Subjects
- Case-Control Studies, Fever genetics, Genetic Association Studies, Humans, Melanoma genetics, Polymorphism, Single Nucleotide, Proto-Oncogene Proteins B-raf genetics, Antineoplastic Agents adverse effects, Fever chemically induced, HLA Antigens genetics, Imidazoles adverse effects, Melanoma drug therapy, Oximes adverse effects, Proto-Oncogene Proteins B-raf antagonists & inhibitors
- Abstract
Aim: Pyrexia is a common adverse event (AE) on dabrafenib treatment (monotherapy or combination with trametinib). Since germline SNPs and HLA alleles are implicated in drug-induced AEs, this study investigated their association with pyrexia., Patients & Methods: 1006 melanoma subjects from five dabrafenib-trametinib clinical studies underwent genotyping for genome-wide SNPs, which enabled imputation of 150 HLA alleles. SNP/HLA allele frequencies were compared between pyrexia cases (n = 218) and controls (n = 361) out of the 1006 subjects by meta-analysis., Results: This analysis had adequate power to detect association of common SNPs or HLA alleles with moderate to large effects on pyrexia (odds ratio >6), but no significant association was found., Conclusion: The study suggests that common genetic variation or HLA polymorphisms do not contribute substantially to dabrafenib-induced pyrexia.
- Published
- 2016
- Full Text
- View/download PDF
35. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial.
- Author
-
Grob JJ, Amonkar MM, Karaszewska B, Schachter J, Dummer R, Mackiewicz A, Stroyakovskiy D, Drucis K, Grange F, Chiarion-Sileni V, Rutkowski P, Lichinitser M, Levchenko E, Wolter P, Hauschild A, Long GV, Nathan P, Ribas A, Flaherty K, Sun P, Legos JJ, McDowell DO, Mookerjee B, Schadendorf D, and Robert C
- Subjects
- Administration, Oral, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, DNA Mutational Analysis, Disease Progression, Disease-Free Survival, Drug Administration Schedule, Genetic Predisposition to Disease, Humans, Imidazoles administration & dosage, Imidazoles adverse effects, Indoles administration & dosage, Indoles adverse effects, Intention to Treat Analysis, MAP Kinase Kinase Kinases antagonists & inhibitors, MAP Kinase Kinase Kinases metabolism, Melanoma genetics, Melanoma mortality, Melanoma secondary, Oximes administration & dosage, Oximes adverse effects, Phenotype, Prospective Studies, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Pyridones administration & dosage, Pyridones adverse effects, Pyrimidinones administration & dosage, Pyrimidinones adverse effects, Risk Factors, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Sulfonamides administration & dosage, Sulfonamides adverse effects, Surveys and Questionnaires, Time Factors, Treatment Outcome, Vemurafenib, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Imidazoles therapeutic use, Indoles therapeutic use, Melanoma drug therapy, Mutation, Oximes therapeutic use, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Pyridones therapeutic use, Pyrimidinones therapeutic use, Quality of Life, Skin Neoplasms drug therapy, Sulfonamides therapeutic use
- Abstract
Background: In the COMBI-v trial, patients with previously untreated BRAF Val600Glu or Val600Lys mutant unresectable or metastatic melanoma who were treated with the combination of dabrafenib and trametinib had significantly longer overall and progression-free survival than those treated with vemurafenib alone. Here, we present the effects of treatments on health-related quality of life (HRQoL), an exploratory endpoint in the COMBI-v study., Methods: COMBI-v was an open-label, randomised phase 3 study in which 704 patients with metastatic melanoma with a BRAF Val600 mutation were randomly assigned (1:1) by an interactive voice response system to receive either a combination of dabrafenib (150 mg twice-daily) and trametinib (2 mg once-daily) or vemurafenib monotherapy (960 mg twice-daily) orally as first-line therapy. The primary endpoint was overall survival. In this pre-specified exploratory analysis, we prospectively assessed HRQoL in the intention-to-treat population with the European Organisation for Research and Treatment of Cancer quality of life (EORTC QLQ-C30), EuroQoL-5D (EQ-5D), and Melanoma Subscale of the Functional Assessment of Cancer Therapy-Melanoma (FACT-M), completed at baseline, during study treatment, at disease progression, and after progression. We used a mixed-model, repeated measures ANCOVA to assess differences in mean scores between groups with baseline score as covariate; all p-values are descriptive. The COMBI-v trial is registered with ClinicalTrials.gov, number NCT01597908, and is ongoing for the primary endpoint, but is not recruiting patients., Findings: From June 4, 2012, to Oct 7, 2013, 1645 patients at 193 centres worldwide were screened for eligibility, and 704 patients were randomly assigned to dabrafenib plus trametinib (n=352) or vemurafenib (n=352). Questionnaire completion rates for both groups were high (>95% at baseline, >80% at follow-up assessments, and >70% at disease progression) with similar HRQoL and symptom scores reported at baseline in both treatment groups for all questionnaires. Differences in mean scores between treatment groups were significant and clinically meaningful in favour of the combination compared with vemurafenib monotherapy for most domains across all three questionnaires during study treatment and at disease progression, including EORTC QLQ-C30 global health (7·92, 7·62, 6·86, 7·47, 5·16, 7·56, and 7·57 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; p<0·001 for all assessments except p=0·005 at week 40), EORTC QLQ-C30 pain (-13·20, -8·05, -8·82, -12·69, -12·46, -11·41, and -10·57 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; all p<0·001), EQ-5D thermometer scores (7·96, 8·05, 6·83, 11·53, 7·41, 9·08, and 10·51 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; p<0·001 for all assessments except p=0·006 at week 32), and FACT-M Melanoma Subscale score (3·62, 2·93, 2·45, 3·39, 2·85, 3·00, and 3·68 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; all p<0·001)., Interpretation: From the patient's perspective, which integrates not only survival advantage but also disease-associated and adverse-event-associated symptoms, treatment with the combination of a BRAF inhibitor plus a MEK inhibitor (dabrafenib plus trametinib) adds a clear benefit over monotherapy with the BRAF inhibitor vemurafenib and supports the combination therapy as standard of care in this population., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial.
- Author
-
Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion-Sileni V, Lebbe C, Mandalà M, Millward M, Arance A, Bondarenko I, Haanen JB, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Swann S, Legos JJ, Jin F, Mookerjee B, and Flaherty K
- Subjects
- Adult, Aged, Aged, 80 and over, Disease-Free Survival, Double-Blind Method, Drug Therapy, Combination, Female, Humans, Male, Middle Aged, Proto-Oncogene Proteins B-raf genetics, Treatment Outcome, Young Adult, Imidazoles administration & dosage, Melanoma drug therapy, Oximes administration & dosage, Pyridones administration & dosage, Pyrimidinones administration & dosage
- Abstract
Background: Previously, a study of ours showed that the combination of dabrafenib and trametinib improves progression-free survival compared with dabrafenib and placebo in patients with BRAF Val600Lys/Glu mutation-positive metastatic melanoma. The study was continued to assess the secondary endpoint of overall survival, which we report in this Article., Methods: We did this double-blind phase 3 study at 113 sites in 14 countries. We enrolled previously untreated patients with BRAF Val600Glu or Val600Lys mutation-positive unresectable stage IIIC or stage IV melanoma. Participants were computer-randomised (1:1) to receive a combination of dabrafenib (150 mg orally twice daily) and trametinib (2 mg orally once daily), or dabrafenib and placebo. The primary endpoint was progression-free survival and overall survival was a secondary endpoint. This study is registered with ClinicalTrials.gov, number NCT01584648., Findings: Between May 4, 2012, and Nov 30, 2012, we screened 947 patients for eligibility, of whom 423 were randomly assigned to receive dabrafenib and trametinib (n=211) or dabrafenib only (n=212). The final data cutoff was Jan 12, 2015, at which time 222 patients had died. Median overall survival was 25·1 months (95% CI 19·2-not reached) in the dabrafenib and trametinib group versus 18·7 months (15·2-23·7) in the dabrafenib only group (hazard ratio [HR] 0·71, 95% CI 0·55-0·92; p=0·0107). Overall survival was 74% at 1 year and 51% at 2 years in the dabrafenib and trametinib group versus 68% and 42%, respectively, in the dabrafenib only group. Based on 301 events, median progression-free survival was 11·0 months (95% CI 8·0-13·9) in the dabrafenib and trametinib group and 8·8 months (5·9-9·3) in the dabrafenib only group (HR 0·67, 95% CI 0·53-0·84; p=0·0004; unadjusted for multiple testing). Treatment-related adverse events occurred in 181 (87%) of 209 patients in the dabrafenib and trametinib group and 189 (90%) of 211 patients in the dabrafenib only group; the most common was pyrexia (108 patients, 52%) in the dabrafenib and trametinib group, and hyperkeratosis (70 patients, 33%) in the dabrafenib only group. Grade 3 or 4 adverse events occurred in 67 (32%) patients in the dabrafenib and trametinib group and 66 (31%) patients in the dabrafenib only group., Interpretation: The improvement in overall survival establishes the combination of dabrafenib and trametinib as the standard targeted treatment for BRAF Val600 mutation-positive melanoma. Studies assessing dabrafenib and trametinib in combination with immunotherapies are ongoing., Funding: GlaxoSmithKline., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
37. A phase 2 study of ruxolitinib, an oral JAK1 and JAK2 Inhibitor, in patients with advanced polycythemia vera who are refractory or intolerant to hydroxyurea.
- Author
-
Verstovsek S, Passamonti F, Rambaldi A, Barosi G, Rosen PJ, Rumi E, Gattoni E, Pieri L, Guglielmelli P, Elena C, He S, Contel N, Mookerjee B, Sandor V, Cazzola M, Kantarjian HM, Barbui T, and Vannucchi AM
- Subjects
- Adult, Aged, Aged, 80 and over, Anemia blood, Anemia chemically induced, Anemia pathology, Contraindications, Drug-Related Side Effects and Adverse Reactions classification, Drug-Related Side Effects and Adverse Reactions pathology, Female, Granulocytes pathology, Hematocrit, Humans, Hydroxyurea adverse effects, Janus Kinase 1 antagonists & inhibitors, Janus Kinase 2 antagonists & inhibitors, Male, Middle Aged, Nitriles, Polycythemia Vera blood, Polycythemia Vera pathology, Pyrimidines, Thrombocytopenia blood, Thrombocytopenia chemically induced, Thrombocytopenia pathology, Young Adult, Janus Kinase 1 genetics, Janus Kinase 2 genetics, Polycythemia Vera drug therapy, Pyrazoles administration & dosage
- Abstract
Background: Polycythemia vera (PV) is a myeloproliferative neoplasm associated with somatic gain-of-function mutations of Janus kinase-2 (JAK2). Therapeutic options are limited in patients with advanced disease. Ruxolitinib, an oral JAK1/JAK2 inhibitor, is active in preclinical models of PV. The long-term efficacy and safety of ruxolitinib in patients with advanced PV who are refractory or intolerant to hydroxyurea were studied in a phase 2 trial., Methods: Response was assessed using modified European LeukemiaNet criteria, which included a reduction in hematocrit to <45% without phlebotomy, resolution of palpable splenomegaly, normalization of white blood cell and platelet counts, and reduction in PV-associated symptoms., Results: Thirty-four patients received ruxolitinib for a median of 152 weeks (range, 31 weeks-177 weeks) or 35.0 months (range, 7.1 months-40.7 months). Hematocrit <45% without phlebotomy was achieved in 97% of patients by week 24.Only 1 patient required a phlebotomy after week 4. Among patients with palpable splenomegaly at baseline, 44% and 63%, respectively, achieved nonpalpable spleen measurements at weeks 24 and 144. Clinically meaningful improvements in pruritus, night sweats, and bone pain were observed within 4 weeks of the initiation of therapy and maintained with continued treatment. Ruxolitinib treatment also reduced elevated levels of inflammatory cytokines and granulocyte activation. Thrombocytopenia and anemia were the most common adverse events.Thrombocytopenia of grade 3 or anemia of grade 3 (according to National Cancer Institute Common Terminology Criteria for Adverse Events,version 3.0) occurred in 3 patients each (9%) (1 patient had both) and were managed with dose modification., Conclusions: Ruxolitinib was generally well tolerated and provided rapid and durable clinical benefits in patients with advanced PV who were refractory or intolerant to hydroxyurea.
- Published
- 2014
- Full Text
- View/download PDF
38. Cediranib in combination with fulvestrant in hormone-sensitive metastatic breast cancer: a randomized Phase II study.
- Author
-
Hyams DM, Chan A, de Oliveira C, Snyder R, Vinholes J, Audeh MW, Alencar VM, Lombard J, Mookerjee B, Xu J, Brown K, and Klein P
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Breast Neoplasms blood, Breast Neoplasms pathology, Estradiol administration & dosage, Estradiol adverse effects, Estradiol analogs & derivatives, Estradiol pharmacokinetics, Estrogen Antagonists administration & dosage, Estrogen Antagonists adverse effects, Estrogen Antagonists pharmacokinetics, Female, Fibroblast Growth Factor 2 blood, Fulvestrant, Humans, Middle Aged, Neoplasm Metastasis, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Quinazolines administration & dosage, Quinazolines adverse effects, Quinazolines pharmacokinetics, Vascular Endothelial Growth Factor A blood, Vascular Endothelial Growth Factor Receptor-2 blood, Young Adult, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms drug therapy
- Abstract
Hormone receptor-positive breast cancer is treated with estrogen inhibitors. Fulvestrant (FASLODEX™), an estrogen receptor (ER) antagonist with no known agonist effects, competitively binds, blocks and degrades the ER. Vascular endothelial growth factor (VEGF) may mediate resistance to ER antagonists. Cediranib is a highly potent VEGF signaling inhibitor with activity against all three VEGF receptors. This randomized Phase II study evaluated cediranib plus fulvestrant. Postmenopausal women with hormone-sensitive metastatic breast cancer were eligible. The primary endpoint was progression-free survival (PFS). Secondary endpoints included objective response rate (ORR), duration of response, clinical benefit rate (CBR), safety/tolerability and pharmacokinetics (PK). Patients received cediranib 45 mg/day (n=31) or placebo (n=31) both plus fulvestrant. Demographic/baseline characteristics were well balanced. Patients treated with cediranib had a numerical advantage in PFS (hazard ratio=0.867, P=0.669; median 223 vs. 112 days, respectively) and ORR (22 vs. 8 %, respectively) vs. placebo, although not statistically significant. CBR was 42 % in both arms. The most common adverse events (AEs) in the cediranib arm were diarrhea (68 %), fatigue (61 %) and hypertension (55 %). The incidence of grade ≥ 3 AEs (68 % vs. 32 %), serious AEs (48 % vs. 13 %), discontinuation AEs (39 % vs. 10 %), and cediranib dose reductions/interruptions (74 % vs. 32 %) were higher in the cediranib arm. There was no evidence of a clinically relevant effect of cediranib on fulvestrant PK. Cediranib plus fulvestrant may demonstrate clinical activity in this population, but cediranib 45 mg was not sufficiently well tolerated. Investigation of lower doses of cediranib plus hormonal/chemotherapy could be considered.
- Published
- 2013
- Full Text
- View/download PDF
39. Cediranib with mFOLFOX6 versus bevacizumab with mFOLFOX6 as first-line treatment for patients with advanced colorectal cancer: a double-blind, randomized phase III study (HORIZON III).
- Author
-
Schmoll HJ, Cunningham D, Sobrero A, Karapetis CS, Rougier P, Koski SL, Kocakova I, Bondarenko I, Bodoky G, Mainwaring P, Salazar R, Barker P, Mookerjee B, Robertson J, and Van Cutsem E
- Subjects
- Adult, Aged, Bevacizumab, Biopsy, Needle, Colorectal Neoplasms mortality, Confidence Intervals, Disease-Free Survival, Dose-Response Relationship, Drug, Double-Blind Method, Drug Administration Schedule, Female, Fluorouracil therapeutic use, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Leucovorin therapeutic use, Male, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Staging, Organoplatinum Compounds therapeutic use, Patient Selection, Proportional Hazards Models, Risk Assessment, Survival Analysis, Treatment Outcome, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Quinazolines therapeutic use
- Abstract
Purpose: To compare the efficacy of cediranib (a vascular endothelial growth factor receptor tyrosine kinase inhibitor [VEGFR TKI]) with that of bevacizumab (anti-VEGF-A monoclonal antibody) in combination with chemotherapy as first-line treatment for advanced metastatic colorectal cancer (mCRC)., Patients and Methods: HORIZON III [Cediranib Plus FOLFOX6 Versus Bevacizumab Plus FOLFOX6 in Patients With Untreated Metastatic Colorectal Cancer] had an adaptive phase II/III design. Patients randomly assigned 1:1:1 received mFOLFOX6 [oxaliplatin 85 mg/m(2) and leucovorin 400 mg/m(2) intravenously followed by fluorouracil 400 mg/m(2) intravenously on day 1 and then continuous infusion of 2,400 mg/m(2) over the next 46 hours every 2 weeks] with cediranib (20 or 30 mg per day) or bevacizumab (5 mg/kg every 14 days). An independent end-of-phase II analysis concluded that mFOLFOX6/cediranib 20 mg met predefined criteria for continuation; subsequent patients received mFOLFOX6/cediranib 20 mg or mFOLFOX6/bevacizumab (randomly assigned 1:1). The primary objective was to compare progression-free survival (PFS)., Results: In all, 1,422 patients received mFOLFOX6/cediranib 20 mg (n = 709) or mFOLFOX6/bevacizumab (n = 713). Primary analysis revealed no significant difference between arms for PFS (hazard ratio [HR], 1.10; 95% CI, 0.97 to 1.25; P = .119), overall survival (OS; HR, 0.95; 95% CI, 0.82 to 1.10; P = .541), or overall response rate (46.3% v 47.3%). Median PFS and OS were 9.9 and 22.8 months for mFOLFOX6/cediranib and 10.3 and 21.3 months for mFOLFOX6/bevacizumab. The PFS upper 95% CI was outside the predefined noninferiority limit (HR < 1.2). Common adverse events with more than 5% incidence in the cediranib arm included diarrhea, neutropenia, and hypertension. Cediranib-treated patients completed fewer chemotherapy cycles than bevacizumab-treated patients (median 10 v 12 cycles). Patient-reported outcomes (PROs) were significantly less favorable in cediranib-treated versus bevacizumab-treated patients (P < .001)., Conclusion: Cediranib activity, in terms of PFS and OS, was comparable to that of bevacizumab when added to mFOLFOX6; however, the predefined boundary for PFS noninferiority was not met. The cediranib safety profile was consistent with previous studies but led to less favorable PROs compared with bevacizumab. Investigation of oral TKIs in CRC continues.
- Published
- 2012
- Full Text
- View/download PDF
40. Cediranib monotherapy in patients with advanced renal cell carcinoma: results of a randomised phase II study.
- Author
-
Mulders P, Hawkins R, Nathan P, de Jong I, Osanto S, Porfiri E, Protheroe A, van Herpen CM, Mookerjee B, Pike L, Jürgensmeier JM, and Gore ME
- Subjects
- Aged, Algorithms, Antineoplastic Agents adverse effects, Antineoplastic Agents blood, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Carcinoma, Renal Cell blood, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell pathology, Double-Blind Method, Female, Humans, Kidney Neoplasms blood, Kidney Neoplasms metabolism, Kidney Neoplasms pathology, Male, Middle Aged, Placebos, Quinazolines adverse effects, Quinazolines blood, Quinazolines pharmacokinetics, Treatment Outcome, Tumor Burden drug effects, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy, Quinazolines therapeutic use
- Abstract
Background: Cediranib is a highly potent vascular endothelial growth factor (VEGF) signalling inhibitor with activity against VEGF receptors 1, 2 and 3. This Phase II, randomised, double-blind, parallel-group study compared the efficacy of cediranib with placebo in patients with metastatic or recurrent clear cell renal cell carcinoma who had not previously received a VEGF signalling inhibitor., Methods: Patients were randomised (3:1) to cediranib 45 mg/day or placebo. The primary objective was comparison of change from baseline in tumour size after 12 weeks of therapy. Secondary objectives included response rate and duration, progression-free survival (PFS) and safety and tolerability. Patients in the placebo group could cross over to open-label cediranib at 12 weeks or earlier if their disease had progressed. This study has been completed and is registered with ClinicalTrials.gov, number NCT00423332., Findings: Patients (n=71) were randomised to receive cediranib (n=53) or placebo (n=18). The primary study outcome revealed that, after 12weeks of therapy, there was a significant difference in mean percentage change from baseline in tumour size between the cediranib (-20%) and placebo (+20%) arms (p<0.0001). Eighteen patients (34%) on cediranib achieved a partial response and 25 (47%) experienced stable disease. Cediranib treatment prolonged PFS significantly compared with placebo (hazard ratio (HR)=0.45, 90%confidence interval: 0.26-0.76, p=0.017; median PFS 12.1 versus 2.8 months). The most common adverse events in patients receiving cediranib were diarrhoea (74%), hypertension (64%), fatigue (58%) and dysphonia (58%)., Interpretation: Cediranib monotherapy demonstrated significant evidence of antitumour activity in patients with advanced renal cell carcinoma. The adverse event profile was consistent with previous studies of cediranib 45 mg., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. A 2-step approach to myeloablative haploidentical stem cell transplantation: a phase 1/2 trial performed with optimized T-cell dosing.
- Author
-
Grosso D, Carabasi M, Filicko-O'Hara J, Kasner M, Wagner JL, Colombe B, Cornett Farley P, O'Hara W, Flomenberg P, Werner-Wasik M, Brunner J, Mookerjee B, Hyslop T, Weiss M, and Flomenberg N
- Subjects
- Adult, Aged, Calibration, Cell Count, Female, Graft vs Host Disease epidemiology, Graft vs Host Disease etiology, Haplotypes, Hematologic Neoplasms immunology, Hematologic Neoplasms mortality, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cell Transplantation standards, Humans, Male, Middle Aged, Myeloablative Agonists adverse effects, Transplantation Conditioning adverse effects, Transplantation, Homologous, Young Adult, Hematologic Neoplasms therapy, Hematopoietic Stem Cell Transplantation methods, Myeloablative Agonists therapeutic use, T-Lymphocytes cytology, Transplantation Conditioning methods
- Abstract
Studies of haploidentical hematopoietic stem cell transplantation (HSCT) have identified threshold doses of T cells below which severe GVHD is usually absent. However, little is known regarding optimal T-cell dosing as it relates to engraftment, immune reconstitution, and relapse. To begin to address this question, we developed a 2-step myeloablative approach to haploidentical HSCT in which 27 patients conditioned with total body irradiation (TBI) were given a fixed dose of donor T cells (HSCT step 1), followed by cyclophosphamide (CY) for T-cell tolerization. A CD34-selected HSC product (HSCT step 2) was infused after CY. A dose of 2 × 10(8)/kg of T cells resulted in consistent engraftment, immune reconstitution, and acceptable rates of GVHD. Cumulative incidences of grade III-IV GVHD, nonrelapse mortality (NRM), and relapse-related mortality were 7.4%, 22.2%, and 29.6%, respectively. With a follow-up of 28-56 months, the 3-year probability of overall survival for the whole cohort is 48% and 75% in patients without disease at HSCT. In the context of CY tolerization, a high, fixed dose of haploidentical T cells was associated with encouraging outcomes, especially in good-risk patients, and can serve as the basis for further exploration and optimization of this 2-step approach. This study is registered at www.clinicaltrials.gov as NCT00429143.
- Published
- 2011
- Full Text
- View/download PDF
42. Antiviral responses following L-leucyl-L-leucine methyl esther (LLME)-treated lymphocyte infusions: graft-versus-infection without graft-versus-host disease.
- Author
-
Filicko-O'Hara J, Grosso D, Flomenberg PR, Friedman TM, Brunner J, Drobyski W, Ferber A, Kakhniashvili I, Keever-Taylor C, Mookerjee B, Talano JA, Wagner JI, Korngold R, and Flomenberg N
- Subjects
- Adult, Aged, Cohort Studies, Flow Cytometry, Graft vs Host Disease prevention & control, Humans, Male, Middle Aged, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocytes immunology, T-Lymphocytes transplantation, Young Adult, Dipeptides therapeutic use, Graft vs Host Disease immunology, Hematopoietic Stem Cell Transplantation methods, Lymphocyte Transfusion methods, T-Lymphocyte Subsets drug effects, T-Lymphocyte Subsets transplantation
- Abstract
Although allogeneic hematopoietic progenitor cell transplant (HPCT) is curative therapy for many disorders, it is associated with significant morbidity and mortality, which can be related to graft-versus-host disease (GVHD) and the immunosuppressive measures required for its prevention and/or treatment. Whether the immunosuppression is pharmacologic or secondary to graft manipulation, the graft recipient is left at increased risk of the threatening opportunistic infection. Refractory viral diseases in the immunocompromised host have been treated by infusion of virus-specific lymphotyces and by unmanipulated donor lymphocyte infusion (DLI) therapy. L-leucyl-L-leucine methyl ester (LLME) is a compound that induces programmed cell death of natural killer (NK) cells, monocytes, granulocytes, most CD8(+) T cells, and a small fraction of CD4(+) T cells. We have undertaken a study of the use of LLME-treated DLI following T cell-depleted allogeneic HPCT, specifically to aid with immune reconstitution. In this ongoing clinical trial, we have demonstrated the rapid emergence of virus-specific responses following LLME DLI with minimal associated GVHD. This paper examines the pace of immune recovery and the rapid development of antiviral responses in 6 patients who developed viral infections during the time period immediately preceding or coincident with the administration of the LLME DLI.
- Published
- 2009
- Full Text
- View/download PDF
43. Adenovirus DNA polymerase is recognized by human CD8+ T cells.
- Author
-
Joshi A, Tang J, Kuzma M, Wagner J, Mookerjee B, Filicko J, Carabasi M, Flomenberg N, and Flomenberg P
- Subjects
- Adult, Cytotoxicity Tests, Immunologic, DNA-Binding Proteins immunology, Humans, Interferon-gamma metabolism, Lymphocyte Activation, T-Lymphocyte Subsets immunology, Adenoviridae immunology, CD8-Positive T-Lymphocytes immunology, DNA-Directed DNA Polymerase immunology, Viral Nonstructural Proteins immunology
- Abstract
Donor lymphocytes have potential as a treatment for adenovirus (Ad) disease in haematopoietic stem cell transplant (SCT) recipients, but better understanding of Ad-specific T-cell responses is required. Most healthy adults exhibit memory T-cell responses to hexon, a capsid protein synthesized late after infection. However, since the Ad E3-19k downregulates major histocompatibility complex (MHC) class I molecules, cytotoxic T cells (CTLs) targeted to early viral proteins may be more effective in eliminating Ad-infected cells in vivo. Here we show that Ad-specific CTLs recognize the early region 2 proteins DNA polymerase (Pol) and DNA-binding protein (DBP). Firstly, memory Ad-specific CD8(+) T cells were amplified from healthy donors by in vitro stimulation with Ad-infected dendritic cells and found to exhibit MHC-restricted cytotoxicity to targets expressing Pol and DBP. Secondly, gamma interferon responses to HLA A2-binding motif peptides from Pol and DBP were directly detected in peripheral blood mononuclear cells (PBMCs) from a recently infected normal donor. Peptide-specific CTLs generated to Pol and DBP epitopes were confirmed to exhibit HLA A2-restricted killing of targets expressing Pol or DBP. Lastly, Pol-epitope-specific T cells were detected at similar or higher frequencies than hexon and DBP in three of three SCT recipients recovering from invasive Ad disease. Pol epitopes were well conserved among different Ad serotypes. Therefore, Pol is a promising target for immunotherapy of Ad disease.
- Published
- 2009
- Full Text
- View/download PDF
44. Purification of melanoma reactive T cell by using a monocyte-based solid phase T-cell selection system for adoptive therapy.
- Author
-
Li J, Mookerjee B, and Wagner J
- Subjects
- Antigens, Neoplasm immunology, Cell Communication immunology, Cell Culture Techniques, Cell Line, Tumor, Cytotoxicity Tests, Immunologic, Dendritic Cells cytology, Dendritic Cells immunology, Epitopes, T-Lymphocyte immunology, HLA-A Antigens immunology, HLA-A2 Antigen, Humans, Interferon-gamma metabolism, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear immunology, MART-1 Antigen, Monocytes cytology, Neoplasm Proteins immunology, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic metabolism, Cell Separation methods, Immunotherapy, Adoptive methods, Melanoma immunology, Monocytes immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
The generation of melanoma-reactive T cells with the characteristics necessary for in vivo effectiveness remains a considerable obstacle to the application of adoptive cell therapy. Recent clinical success with adoptive cell therapy for melanoma is motivating additional investigation to improve the technology of generating such tumor reactive lymphocytes. Here we describe a novel solid phase T-cell selection system, in which monocytes are immobilized on solid support for antigen-specific T-cell purification. We hypothesized and proved that antigen-specific T cells recognize their cognate antigens and bind to them faster than nonantigen-specific T cells and are concentrated on the surface after removing the nonadherent cells by washing. Moreover, activated antigen-specific T cells proliferated more rapidly than nonspecific T cells, further increasing the frequency and purity of antigen-specific T cells. Optimal selection times for Melan-A-specific T cells are studied. Our data demonstrated that T-cell selection can usually increase the frequency of tumor antigen-specific T cells by >10-fold, whereas T-cell expansion after the selection boost the frequency of tumor antigen-specific T cells by another approximately 10-fold. More importantly, these T cells are generated under more physiologic conditions. This new T-cell selection system is superior to traditional repeated stimulation methods in generating tumor antigen-specific T cells for adoptive cell immunotherapy. This inexpensive and simple T-cell selection system can produce large quantity of highly purified Melan-A-specific T cells within 2 weeks after T-cell activation.
- Published
- 2008
- Full Text
- View/download PDF
45. In vitro methods for generating highly purified EBV associated tumor antigen-specific T cells by using solid phase T cell selection system for immunotherapy.
- Author
-
Li J, Mookerjee B, Wagner J, and Flomenberg N
- Subjects
- Humans, In Vitro Techniques, Lymphocyte Activation, Time, Viral Matrix Proteins immunology, Antigens, Neoplasm immunology, Cell Separation methods, Herpesvirus 4, Human immunology, Immunotherapy methods, T-Lymphocytes, Cytotoxic immunology
- Abstract
Adoptive cell transfer immunotherapy has been utilized to treat EBV related human malignancies including post-transplant lymphoproliferative diseases, Hodgkin's lymphoma and nasopharyngeal carcinoma. However, there are limited options available for tumor antigen-specific T cell purification. Here we describe a novel solid phase T cell selection system, in which monocytes or EBV transformed B-lymphocytes are immobilized on solid support for antigen-specific T cell purification. We hypothesize and prove that antigen-specific T cells recognize their cognate antigens and bind to them faster than non-antigen specific T cells. Therefore antigen-specific T cells can be concentrated on the surface after removing the non-adherent cells by washing. The optimal selection time for both EBV-specific T cells and LMP2-specific T cells is studied. Our data demonstrate that the frequency of antigen-specific T cells can be increased by >20-fold after selection. Moreover, activated antigen-specific T cells proliferate more rapidly than non-specific T cells, further increasing the frequency and purity of antigen-specific T cells. This new T cell selection system is superior to traditional repeated stimulation methods in generating tumor antigen-specific T cells. We are able to generate large quantities of highly purified T cells of subdominant antigens LMP2 within 2 weeks after T cell activation for adoptive cell transfer immunotherapy with this simple, rapid and inexpensive T cell selection system.
- Published
- 2007
- Full Text
- View/download PDF
46. T cell repertoire complexity is conserved after LLME treatment of donor lymphocyte infusions.
- Author
-
Friedman TM, Filicko-O'Hara J, Mookerjee B, Wagner JL, Grosso DA, Flomenberg N, and Korngold R
- Subjects
- Adult, Female, Graft vs Host Disease prevention & control, Humans, Male, Middle Aged, T-Lymphocyte Subsets cytology, Transplantation Chimera, Dipeptides therapeutic use, Hematopoietic Stem Cell Transplantation methods, Immunosuppressive Agents therapeutic use, Lymphocyte Transfusion methods, T-Lymphocyte Subsets transplantation
- Abstract
Slow reconstitution of the T cell repertoire after allogeneic blood or bone marrow stem cell transplantation is a major risk factor for patient mortality. The delivery of immunocompetent T cells as delayed donor lymphocyte infusions (DLIs) is a potential way of counteracting this problem. The development of graft-versus-host disease (GVHD) is a potential complication of this procedure, however. We previously found that in P-->F1 haploidentical murine models, the ex-vivo treatment of donor lymphocytes with L-leucyl-L-leucine methyl ester (LLME) can prevent the onset of GVHD after DLI, likely by inducing cell death in most of the perforin-positive CD8(+) T cells and in a fraction of CD4(+) T cells. Our previous preclinical studies have formed the basis of an ongoing phase I clinical trial in which patients received LLME-treated DLI from their original donor in an attempt to accelerate T cell reconstitution. To understand how this treatment strategy might affect the complexity of the DLI T cell repertoire, we used T cell receptor Vbeta spectratype analysis to evaluate the DLI product pre-LLME and post-LLME treatment. The results indicated that the LLME-treated DLI product exhibited CDR3-size distribution complexities similar to those of its untreated donor sample. In addition, comparisons of the CD4(+) and CD8(+) T cell repertoire from the donor before LLME treatment with that of the recipient post-DLI demonstrated equal complexity for most of the resolvable Vbeta families. Finally, the in vitro proliferative capacity of LLME-treated DLI product in response to allo-stimulation in a one-way mixed lymphocyte reaction was comparable to that of the untreated product.
- Published
- 2007
- Full Text
- View/download PDF
47. Treatment of adenovirus disease in stem cell transplant recipients with cidofovir.
- Author
-
Neofytos D, Ojha A, Mookerjee B, Wagner J, Filicko J, Ferber A, Dessain S, Grosso D, Brunner J, Flomenberg N, and Flomenberg P
- Subjects
- Adenovirus Infections, Human etiology, Adult, Aged, CD4 Lymphocyte Count, Cidofovir, Colitis drug therapy, Colitis virology, Cytosine therapeutic use, Female, Hepatitis, Viral, Human drug therapy, Humans, Immunoglobulins, Intravenous therapeutic use, Immunologic Factors therapeutic use, Male, Nephritis drug therapy, Nephritis virology, Transplantation, Homologous, Viral Load, Adenovirus Infections, Human drug therapy, Antiviral Agents therapeutic use, Cytosine analogs & derivatives, Hematopoietic Stem Cell Transplantation adverse effects, Organophosphonates therapeutic use
- Abstract
Invasive adenovirus (AdV) disease is fatal in >50% of allogeneic hematopoietic stem cell transplant (SCT) recipients. Treatment with cidofovir may improve outcomes based on in vitro susceptibility data and case reports. Six consecutive cases of invasive AdV disease treated with cidofovir were reviewed among 84 allogeneic adult SCT recipients (incidence, 7.1%). Cidofovir was administered intravenously at 5 mg/kg per dose (1-7 doses). All patients received intravenous immune globulin. Blood AdV DNA levels (viral loads, VLs) were monitored with a real-time quantitative polymerase chain reaction assay. Published reports of cidofovir treatment of AdV disease in SCT recipients were critically reviewed. The primary manifestations of AdV disease were hepatitis (n = 3), colitis (n = 2), and nephritis (n = 1). All patients had detectable AdV VLs, with peak values from 5 x 10(5) to 2 x 10(8) copies/mL. All patients received CD34+ selected grafts (n = 3) and/or had graft-versus-host disease (n = 4) and had CD4 counts <100 cells/mm3. Only 1 of 5 patients (20%) who received >or=2 doses of cidofovir died with active AdV disease. Four patients exhibited improvement within days of treatment with cidofovir as documented by clinical criteria and declines in AdV VLs (without a change in immunosuppression). In contrast, 1 patient treated late after onset of AdV disease died after 1 dose of cidofovir. In our review of 70 published cases treated with >or=2 doses of cidofovir, 13 (19%) died from AdV disease. In conclusion, early treatment of AdV disease with cidofovir inhibits viral replication in vivo and reduces mortality in allogeneic SCT recipients compared with historical data.
- Published
- 2007
- Full Text
- View/download PDF
48. Cytoprotection in acute myelogenous leukemia (AML) therapy.
- Author
-
Grosso D, Filicko J, Garcia-Manero G, Beardell F, Brunner J, Cohn J, Ferbér A, Martinez J, Mookerjee B, Rose L, Tice D, Wagner JL, Capizzi R, and Flomenberg N
- Subjects
- Adult, Aged, Amifostine adverse effects, Bone Marrow drug effects, Cytoprotection, Female, Humans, Idarubicin adverse effects, Male, Middle Aged, Survival Analysis, Amifostine administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Idarubicin administration & dosage, Leukemia, Myeloid, Acute drug therapy
- Abstract
Planning therapy for acute myelogenous leukemia (AML) is difficult because of the heterogeneous nature of the disease and varying patient age at presentation. Cytogenetics and patient age at the time of diagnosis are two major factors determining treatment outcome in AML. Patients with poor-risk cytogenetics have much lower complete remission rates than other groups. In addition, AML in patients greater than 55 to 60 years of age often exhibits a resistant phenotype, more akin to secondary AML or AML arising from myelodysplastic syndromes. This group is also characterized by lower complete remission rates, and often requires the delivery of intensive therapy to a patient population that is the least likely to tolerate it. At the Jefferson Health System (Philadelphia, PA), we wished to develop a regimen that was maximally intensive to treat stubborn disease, but gentle enough to be given to all patients regardless of age. Toward this end, 33 patients received a maximal dose of the cytoprotective agent, amifostine, before each infusion of idarubicin in the "7 + 3" regimen, escalating the dose of idarubicin in a phase I fashion to a maximum dose of 24 mg/m2 . The data indicate that the addition of amifostine to "7 + 3" AML induction therapy enables a substantial escalation of the idarubicin dose through the 21-mg/m2 dose level, without a concomitant increase in side effects, thus providing a regimen that is both intensive and applicable to patients of all ages. Currently, phase II studies are ongoing on a national basis to evaluate the efficacy of this regimen.
- Published
- 2004
- Full Text
- View/download PDF
49. Activity of single-agent melphalan 220-300 mg/m2 with amifostine cytoprotection and autologous hematopoietic stem cell support in non-Hodgkin and Hodgkin lymphoma.
- Author
-
Phillips GL, Meisenberg BR, Reece DE, Adams VR, Badros AZ, Brunner JL, Fenton RG, Filicko J, Grosso DL, Hale GA, Howard DS, Johnson VP, Kniska A, Marshall KW, Mookerjee B, Nath R, Rapoport AP, Sarkodee-Adoo C, Takebe N, Vesole DH, Wagner JL, and Flomenberg N
- Subjects
- Adult, Combined Modality Therapy, Female, Humans, Male, Middle Aged, Transplantation Conditioning methods, Transplantation, Autologous, Amifostine administration & dosage, Antineoplastic Agents, Alkylating administration & dosage, Hematopoietic Stem Cell Transplantation, Hodgkin Disease therapy, Lymphoma, Non-Hodgkin therapy, Melphalan administration & dosage, Radiation-Protective Agents administration & dosage
- Abstract
High-dose chemotherapy using melphalan (HDMEL) is an important component of many conditioning regimens that are given before autologous hematopoietic stem cell transplantation (AHSCT). In contrast to the situation in myeloma, and to a lesser degree acute leukemia, only a very limited published experience exists with the use of HDMEL conditioning as a single agent in doses requiring AHSCT for lymphoma, both Hodgkin lymphoma (HL) and especially non-Hodgkin lymphoma (NHL). Thus, we report results of treating 26 lymphoma patients (22 with NHL and four with HL) with HDMEL 220-300 mg/m(2) plus amifostine (AF) cytoprotection and AHSCT as part of a phase I-II trial. Median age was 51 years (range 24-62 years); NHL histology was varied, but was aggressive (including transformed from indolent) in 19 patients, indolent in two patients and mantle cell in one. All 26 patients had been extensively treated; 11 were refractory to the immediate prior therapy on protocol entry and two had undergone prior AHSCT. All were deemed ineligible for other, 'first-line' AHSCT regimens. Of these 26 patients, 22 survived to initial tumor evaluation on D +100. At this time, 13 were in complete remission, including four patients who were in second CR before HDMEL+AF+AHSCT. Responses occurred at all HDMEL doses. Currently, seven patients are alive, including five without progression, with a median follow-up in these latter patients of D +1163 (range D +824 to D +1630); one of these patients had a nonmyeloablative allograft as consolidation on D +106. Conversely, 14 patients relapsed or progressed, including five who had previously achieved CR with the AHSCT procedure. Two patients, both with HL, remain alive after progression; one is in CR following salvage radiotherapy. Six patients died due to nonrelapse causes, including two NHL patients who died while in CR. We conclude that HDMEL+AF+AHSCT has significant single-agent activity in relapsed or refractory NHL and HL. This experience may be used as a starting point for subsequent dose escalation of HDMEL (probably with AF) in established combination regimens.
- Published
- 2004
- Full Text
- View/download PDF
50. Autotransplantation for advanced lymphoma and Hodgkin's disease followed by post-transplant rituxan/GM-CSF or radiotherapy and consolidation chemotherapy.
- Author
-
Rapoport AP, Meisenberg B, Sarkodee-Adoo C, Fassas A, Frankel SR, Mookerjee B, Takebe N, Fenton R, Heyman M, Badros A, Kennedy A, Jacobs M, Hudes R, Ruehle K, Smith R, Kight L, Chambers S, MacFadden M, Cottler-Fox M, Chen T, Phillips G, and Tricot G
- Subjects
- Adult, Aged, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Murine-Derived, Antineoplastic Combined Chemotherapy Protocols, Combined Modality Therapy, Disease-Free Survival, Female, Granulocyte-Macrophage Colony-Stimulating Factor administration & dosage, Hodgkin Disease drug therapy, Hodgkin Disease pathology, Hodgkin Disease radiotherapy, Humans, Immunotherapy, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Non-Hodgkin pathology, Lymphoma, Non-Hodgkin radiotherapy, Male, Middle Aged, Rituximab, Transplantation, Autologous, Hematopoietic Stem Cell Transplantation, Hodgkin Disease therapy, Lymphoma, Non-Hodgkin therapy
- Abstract
Disease relapse occurs in 50% or more of patients who are autografted for relapsed or refractory lymphoma (NHL) or Hodgkin's disease (HD). The administration of non-cross-resistant therapies during the post-transplant phase could possibly control residual disease and delay or prevent its progression. To test this approach, 55 patients with relapsed/refractory or high-risk NHL or relapsed/refractory HD were enrolled in the following protocol: stem cell mobilization: cyclophosphamide (4.5 g/m(2)) + etoposide (2.0 g/m(2)) followed by GM-CSF or G-CSF; high-dose therapy: gemcitabine (1.0 g/m(2)) on day -5, BCNU (300 mg/m(2)) + gemcitabine (1.0 g/m(2)) on day -2, melphalan (140 mg/m(2)) on day -1, blood stem cell infusion on day 0; post-transplant immunotherapy (B cell NHL): rituxan (375 mg/m(2)) weekly for 4 weeks + GM-CSF (250 microg thrice weekly) (weeks 4-8); post-transplant involved-field radiotherapy (HD): 30-40 Gy to pre-transplant areas of disease (weeks 4-8); post-transplant consolidation chemotherapy (all patients): dexamethasone (40 mg daily)/cyclophosphamide (300 mg/m(2)/day)/etoposide (30 mg/m(2)/day)/cisplatin (15 mg/m(2)/day) by continuous intravenous infusion for 4 days + gemcitabine (1.0 g/m(2), day 3) (months 3 + 9) alternating with dexamethasone/paclitaxel (135 mg/m(2))/cisplatin (75 mg/m(2)) (months 6 + 12). Of the 33 patients with B cell lymphoma, 14 had primary refractory disease (42%), 12 had relapsed disease (36%) and seven had high-risk disease in first CR (21%). For the entire group, the 2-year Kaplan-Meier event-free survival (EFS) and overall survival (OS) were 30% and 35%, respectively, while six of 33 patients (18%) died before day 100 from transplant-related complications. The rituxan/GM-CSF phase was well-tolerated by the 26 patients who were treated and led to radiographic responses in seven patients; an eighth patient with a blastic variant of mantle-cell lymphoma had clearance of marrow involvement after rituxan/GM-CSF. Of the 22 patients with relapsed/refractory HD (21 patients) or high-risk T cell lymphoblastic lymphoma (one patient), the 2-year Kaplan-Meier EFS and OS were 70% and 85%, respectively, while two of 22 patients (9%) died before day 100 from transplant-related complications. Eight patients received involved field radiation and seven had radiographic responses within the treatment fields. A total of 72 courses of post-transplant consolidation chemotherapy were administered to 26 of the 55 total patients. Transient grade 3-4 myelosuppression was common and one patient died from neutropenic sepsis, but no patients required an infusion of backup stem cells. After adjustment for known prognostic factors, the EFS for the cohort of HD patients was significantly better than the EFS for an historical cohort of HD patients autografted after BEAC (BCNU/etoposide/cytarabine/cyclophosphamide) without consolidation chemotherapy (P = 0.015). In conclusion, post-transplant consolidation therapy is feasible and well-tolerated for patients autografted for aggressive NHL and HD and may be associated with improved progression-free survival particularly for patients with HD.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.