116 results on '"Berlin JD"'
Search Results
2. Abstract PD3-06: Safety and efficacy of anti-Trop-2 antibody drug conjugate, sacituzumab govitecan (IMMU-132), in heavily pretreated patients with TNBC
- Author
-
Bardia, A, primary, Diamond, JR, additional, Mayer, IA, additional, Starodub, AN, additional, Moroose, RL, additional, Isakoff, SJ, additional, Ocean, AJ, additional, Guarino, MJ, additional, Berlin, JD, additional, Messersmith, WA, additional, Thomas, SS, additional, O'Shaughnessy, JA, additional, Kalinsky, K, additional, Maurer, M, additional, Chang, JC, additional, Forero, A, additional, Traina, T, additional, Gucalp, A, additional, Wilhelm, F, additional, Wegener, WA, additional, Maliakal, P, additional, Sharkey, RM, additional, Goldenberg, DM, additional, and Vahdat, LT, additional
- Published
- 2016
- Full Text
- View/download PDF
3. Phase I study of oral irinotecan as a single-agent and given sequentially with capecitabine.
- Author
-
Goff LW, Benson AB 3rd, Lorusso PM, Tan AR, Berlin JD, Denis LJ, Benner RJ, Yin D, and Rothenberg ML
- Published
- 2012
4. Adjuvant therapy for pancreatic cancer. To treat or not to treat?
- Author
-
Keedy VL and Berlin JD
- Abstract
Despite attempted curative resection of localized adenocarcinoma of the pancreas, most patients experience a recurrence and die of their disease. The Gastrointestinal Tumor Study Group, European Organisation for Research and Treatment of Cancer, and European Study Group for Pancreatic Cancer trials have suggested the benefit of adjuvant therapy. However, the relatively few randomized trials available have not established a definite standard of care due to study limitations. Although these trials, and the recently published Chariti Onkologie (CONKO)-001 trial, have shown a definite advantage of adjuvant chemotherapy, the most effective chemotherapy and the role of radiation therapy remain unclear. This review will discuss the data available from reported trials of adjuvant and neoadjuvant therapy in pancreatic cancer, address the issues leading to the ongoing controversies, and consider future directions for clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2007
5. Omission of 5-Fluorouracil Bolus From Multidrug Regimens for Advanced Gastrointestinal Cancers: A Multicenter Cohort Study.
- Author
-
Peng C, Saffo S, Oberstein PE, Shusterman M, Thomas C, Becker DJ, Berlin JD, Leichman LP, Boursi B, Nagar AB, and Yu S
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Leucovorin therapeutic use, Leucovorin administration & dosage, Adult, Kaplan-Meier Estimate, Treatment Outcome, Cohort Studies, Retrospective Studies, Neoplasm Staging, Fluorouracil administration & dosage, Fluorouracil therapeutic use, Fluorouracil adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Gastrointestinal Neoplasms drug therapy, Gastrointestinal Neoplasms mortality, Gastrointestinal Neoplasms pathology
- Abstract
Background: 5-Fluorouracil (5-FU) is a major component of gastrointestinal cancer treatments. In multidrug regimens such as FOLFOX, FOLFIRI, and FOLFIRINOX, 5-FU is commonly administered as a bolus followed by an infusion. However, the pharmacologic rationale for incorporating the 5-FU bolus in these regimens is unclear, and there are other effective regimens for gastrointestinal cancers that do not include the bolus. The purpose of this study was to determine whether omission of the 5-FU bolus was associated with a difference in survival and toxicity., Methods: A real-world database from Flatiron Health was queried for patients with advanced colorectal, gastroesophageal, and pancreatic cancers who received first-line FOLFOX, FOLFIRI, and FOLFIRINOX regimens. Cox proportional hazards and Kaplan-Meier analyses were performed to compare survival outcomes between patients who received the 5-FU bolus and those who did not. Inverse probability of treatment weighted (IPTW) analysis was performed to adjust for treatment selection bias., Results: This study included 11,765 patients with advanced colorectal (n=8,670), gastroesophageal (n=1,481), and pancreatic (n=1,614) cancers. Among all first-line 5-FU multidrug regimens, 10,148 (86.3%) patients received a 5-FU bolus and 1,617 (13.7%) did not. After IPTW analysis, we found that omitting the bolus was not associated with a decrease in overall survival (hazard ratio, 0.99; 95% CI, 0.91-1.07; P=.74). However, omitting the bolus was associated with reductions in neutropenia (10.7% vs 22.7%; P<.01), thrombocytopenia (11.2% vs 16.1%; P<.01), and use of granulocyte colony-stimulating factors after treatment (19.6% vs 29.1%; P<.01)., Conclusions: After adjusting for baseline clinical factors, we found that omission of the 5-FU bolus from FOLFOX, FOLFIRI, and FOLFIRINOX regimens was not associated with decreased survival, but resulted in decreased toxicity and possible health care savings.
- Published
- 2024
- Full Text
- View/download PDF
6. A phase I study to evaluate the safety, pharmacokinetics, and pharmacodynamics of PF-06939999 (PRMT5 inhibitor) in patients with selected advanced or metastatic tumors with high incidence of splicing factor gene mutations.
- Author
-
Rodon J, Rodriguez E, Maitland ML, Tsai FY, Socinski MA, Berlin JD, Thomas JS, Al Baghdadi T, Wang IM, Guo C, Golmakani M, Clark LN, Gazdoiu M, Li M, and Tolcher AW
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Mutation, Maximum Tolerated Dose, RNA Splicing Factors, Dose-Response Relationship, Drug, Neoplasms drug therapy, Neoplasms genetics, Protein-Arginine N-Methyltransferases genetics
- Abstract
Background: Protein arginine methyltransferase 5 (PRMT5) methylates multiple substrates dysregulated in cancer, including spliceosome machinery components. PF-06939999 is a selective small-molecule PRMT5 inhibitor., Patients and Methods: This phase I dose-escalation and -expansion trial (NCT03854227) enrolled patients with selected solid tumors. PF-06939999 was administered orally once or twice a day (q.d./b.i.d.) in 28-day cycles. The objectives were to evaluate PF-06939999 safety and tolerability to identify maximum tolerated dose (MTD) and recommended part 2 dose (RP2D), and assess pharmacokinetics (PK), pharmacodynamics [changes in plasma symmetric dimethylarginine (SDMA) levels], and antitumor activities., Results: In part 1 dose escalation, 28 patients received PF-06939999 (0.5 mg q.d. to 6 mg b.i.d.). Four of 24 (17%) patients reported dose-limiting toxicities: thrombocytopenia (n = 2, 6 mg b.i.d.), anemia (n = 1, 8 mg q.d.), and neutropenia (n = 1, 6 mg q.d.). PF-06939999 exposure increased with dose. Steady-state PK was achieved by day 15. Plasma SDMA was reduced at steady state (58%-88%). Modulation of plasma SDMA was dose dependent. No MTD was determined. In part 2 dose expansion, 26 patients received PF-06939999 6 mg q.d. (RP2D). Overall (part 1 + part 2), the most common grade ≥3 treatment-related adverse events included anemia (28%), thrombocytopenia/platelet count decreased (22%), fatigue (6%), and neutropenia (4%). Three patients (6.8%) had confirmed partial response (head and neck squamous cell carcinoma, n = 1; non-small-cell lung cancer, n = 2), and 19 (43.2%) had stable disease. No predictive biomarkers were identified., Conclusions: PF-06939999 demonstrated a tolerable safety profile and objective clinical responses in a subset of patients, suggesting that PRMT5 is an interesting cancer target with clinical validation. However, no predictive biomarker was identified. The role of PRMT5 in cancer biology is complex and requires further preclinical, mechanistic investigation to identify predictive biomarkers for patient selection., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Phase Ib/II Trial of Ribociclib in Combination with Binimetinib in Patients with NRAS-mutant Melanoma.
- Author
-
Schuler M, Zimmer L, Kim KB, Sosman JA, Ascierto PA, Postow MA, De Vos FYFL, van Herpen CML, Carlino MS, Johnson DB, Berking C, Reddy MB, Harney AS, Berlin JD, and Amaria RN
- Subjects
- Aminopyridines adverse effects, Benzimidazoles administration & dosage, GTP Phosphohydrolases genetics, Humans, Membrane Proteins genetics, Purines, Melanoma drug therapy, Melanoma genetics, Melanoma pathology
- Abstract
Purpose: Enhanced MAPK pathway signaling and cell-cycle checkpoint dysregulation are frequent in NRAS-mutant melanoma and, as such, the regimen of the MEK inhibitor binimetinib and the selective CDK4/6 inhibitor ribociclib is a rational combination., Patients and Methods: This is a phase Ib/II, open-label study of ribociclib + binimetinib in patients with NRAS-mutant melanoma (NCT01781572). Primary objectives were to estimate the MTD/recommended phase II dose (RP2D) of the combination (phase Ib) and to characterize combination antitumor activity at the RP2D (phase II). Tumor genomic characterization and pharmacokinetics/pharmacodynamics were also evaluated., Results: Ten patients (16.4%) experienced dose-limiting toxicities in cycle 1 of phase Ib. Overall response rate in the phase II cohort (n = 41) for the selected RP2D (binimetinib 45 mg twice daily + ribociclib 200 mg once daily, 21 days on/7 days off) was 19.5% [8/41; 95% confidence interval (CI), 8.8-34.9]. The response rate was 32.5% (13/40; 95% CI, 20.1-48.0) in patients with NRAS mutation with concurrent alterations of CDKN2A, CDK4, or CCND1. Median progression-free survival was 3.7 months (95% CI, 3.5-5.6) and median overall survival was 11.3 months (95% CI, 9.3-14.2) for all patients. Common treatment-related toxicities included creatine phosphokinase elevation, rash, edema, anemia, nausea, diarrhea, and fatigue. Pharmacokinetics and safety were consistent with single-agent data, supporting a lack of drug-drug interaction., Conclusions: Ribociclib + binimetinib can be safely administered and is clinically active in patients with NRAS-mutant melanoma. Co-mutations of cell-cycle genes may define a population with greater likelihood of treatment benefit. See related commentary by Moschos, p. 2977., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
8. Exploring the safety, effect on the tumor microenvironment, and efficacy of itacitinib in combination with epacadostat or parsaclisib in advanced solid tumors: a phase I study.
- Author
-
Naing A, Powderly JD, Nemunaitis JJ, Luke JJ, Mansfield AS, Messersmith WA, Sahebjam S, LoRusso PM, Garrido-Laguna I, Leopold L, Geschwindt R, Ding K, Smith M, and Berlin JD
- Subjects
- Acetonitriles, Humans, Oximes, Pyrazoles, Pyrimidines, Pyrroles, Pyrrolidines, Sulfonamides, Neoplasms drug therapy, Tumor Microenvironment
- Abstract
Background: This phase I multicenter study was designed to evaluate the safety, tolerability, efficacy, and translational effects on the tumor microenvironment of itacitinib (Janus-associated kinase 1 (JAK1) inhibitor) in combination with epacadostat (indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor) or parsaclisib (phosphatidylinositol 3-kinase δ (PI3Kδ) inhibitor)., Methods: Patients with advanced or metastatic solid tumors were enrolled and received itacitinib (100-400 mg once a day) plus epacadostat (50-300 mg two times per day; group A), or itacitinib (100-400 mg once a day) plus parsaclisib or parsaclisib monotherapy (0.3-10 mg once a day; group B)., Results: A total of 142 patients were enrolled in the study. The maximum tolerated dose was not reached for either the combination of itacitinib plus epacadostat (n=47) or itacitinib plus parsaclisib (n=90). One dose-limiting toxicity of serious, grade 3 aseptic meningitis was reported in a patient receiving itacitinib 300 mg once a day plus parsaclisib 10 mg once a day, which resolved when the study drugs were withdrawn. The most common treatment-related adverse events among patients treated with itacitinib plus epacadostat included fatigue, nausea, pyrexia, and vomiting, and for patients treated with itacitinib plus parsaclisib were fatigue, pyrexia, and diarrhea. In the itacitinib plus epacadostat group, no patient had an objective response. Among patients receiving itacitinib 100 mg once a day plus parsaclisib 0.3 mg once a day, three achieved partial response for an objective response rate (95% CI) of 7.1% (1.50 to 19.48). Treatment with itacitinib plus epacadostat demonstrated some increase in tumor CD8
+ T cell infiltration and minor changes in six plasma proteins, whereas treatment with itacitinib plus high-dose parsaclisib resulted in downregulation of 20 plasma proteins mostly involved in immune cell function, with no observed change in intratumoral CD8+ T cell infiltration., Conclusion: Adverse events with JAK1 inhibition combined with either IDO1 or PI3Kδ inhibition were manageable, but the combinations demonstrated limited clinical activity or enhancement of immune activation in the tumor microenvironment., Trial Registration Number: NCT02559492., Competing Interests: Competing interests: AN reports research funding from Amplimmune, Arcus Biosciences, ARMO BioSciences, Atterocor/Millendo, Bristol Myers Squibb, Calithera Biosciences, CytomX Therapeutics, Eli Lilly, EMD Serono, Healios Oncology Nutrition, ImmuneOncia, Incyte, Karyopharm Therapeutics, Kymab, MedImmune, Merck, NCI, NeoimmuneTech, Neon Therapeutics, Novartis, Pfizer, PsiOxus, Regeneron, Surface Oncology, and TopAlliance Biosciences; advisory board of CytomX Therapeutics, Genome & Company, Kymab, Novartis, OncoSec KEYNOTE-695, and STCube Pharmaceuticals; and travel and accommodation expense from ARMO BioSciences. JDP reports clinical trial funding from AbbVie, Alkermes, AstraZeneca, BMS, Corvus, Curis, EMD Serono, Incyte, Macrogenics, MT Group, Precision for Medicine, RAPT Therapeutics, Sequenom, StemCell, Tempest, and Top Alliance BioSciences; advisory for AstraZeneca, Curis, and Merck; consultancy for AstraZeneca, BMS, and Curis; speakers’ bureau for BMS and Merck; laboratory contract research for Merck; and is founder and owner of BioCytics and Carolina BioOncology Institute. JJN reports board membership, employment, and stock ownership with Gradalis. JJL reports scientific advisory board (no stock) for 7 Hills, Spring Bank; scientific advisory board (stock) for Actym, Alphamab Oncology, Arch Oncology, Kanaph, Mavu, Onc.AI, Pyxis, and Tempest; consultancy with compensation from AbbVie, Alnylam, Array, Bayer, Bristol Myers Squibb, Checkmate, Cstone, Eisai, EMD Serono, Inzen Therapeutics, Janssen, KSQ, Macrogenics, Merck, Mersana, Nektar, Novartis, Pfizer, Regeneron, Ribon, Rubius, Silicon, Synlogic, TRex, Werewolf, Xencor, and Xilio; research support (all to institutions for clinical trials unless noted) from AbbVie, Agios (IIT), Array (IIT), Astellas, Bristol Myers Squibb (IIT and industry), Corvus, EMD Serono, Immatics, Incyte, Kadmon, Macrogenics, Merck, Moderna, Nektar, Numab, Replimune, Rubius, Spring Bank, Synlogic, Takeda, Tizona, Trishula, and Xencor; and patents (both provisional) serial #15/612,657 (Cancer Immunotherapy) and PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof). ASM reports honoraria to institution from AbbVie, AstraZeneca, Bristol Myers Squibb, Genentech/Roche, and Janssen; grant funding from Novartis and Verily Life Sciences; travel expenses from Roche; and is non-remunerated Director of Mesothelioma Applied Research Foundation. WAM reports institutional research funding (for this study) from Incyte. SS reports research support from Bristol Myers Squibb, Brooklyn ImmunoTherapeutics, and Merck; and advisory board for Boehringer Ingelheim and Merck. PML reports advisory board membership for AbbVie, ABL Bio, Agenus, Astellas, AstraZeneca, Bayer, Black Diamond, Cybrexa, CytomX, EMD Serono, Genentech, GenMab, GlaxoSmithKline, ImmunoMet, IQVIA, Kineta, Kyowa Kirin Pharmaceutical Development, MacroGenics, Molecular Templates, Pfizer, QED Therapeutics, Salarius, Shattuck, Silverback, STCube Pharmaceuticals, Takeda, TRIGR Therapeutics, and Zentalis Pharmaceuticals; advisory board and consultant for I-Mab; consultant for SK Life Science and Sotio; data safety monitoring board for Agios, Five Prime, Halozyme, and Tyme; and imCORE Alliance with Roche-Genentech. IG-L reports institutional research funding from Bayer, GSK, Incyte, Novartis, Pfizer, and Trishula; ad-hoc advisory board for Eisai and Pfizer; and data and safety monitoring committee for Sotio. LL, RG, KD, and MS report employment and stock ownership with Incyte. JDB reports nothing to disclose., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2022
- Full Text
- View/download PDF
9. First-in-Human PET Imaging and Estimated Radiation Dosimetry of l-[5- 11 C]-Glutamine in Patients with Metastatic Colorectal Cancer.
- Author
-
Cohen AS, Grudzinski J, Smith GT, Peterson TE, Whisenant JG, Hickman TL, Ciombor KK, Cardin D, Eng C, Goff LW, Das S, Coffey RJ, Berlin JD, and Manning HC
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Positron-Emission Tomography, Tissue Distribution, Positron Emission Tomography Computed Tomography, Carbon Radioisotopes, Radiopharmaceuticals pharmacokinetics, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Glutamine metabolism, Radiometry, Neoplasm Metastasis
- Abstract
Altered metabolism is a hallmark of cancer. In addition to glucose, glutamine is an important nutrient for cellular growth and proliferation. Noninvasive imaging via PET may help facilitate precision treatment of cancer through patient selection and monitoring of treatment response. l-[5-
11 C]-glutamine (11 C-glutamine) is a PET tracer designed to study glutamine uptake and metabolism. The aim of this first-in-human study was to evaluate the radiologic safety and biodistribution of11 C-glutamine for oncologic PET imaging. Methods: Nine patients with confirmed metastatic colorectal cancer underwent PET/CT imaging. Patients received 337.97 ± 44.08 MBq of11 C-glutamine. Dynamic PET acquisitions that were centered over the abdomen or thorax were initiated simultaneously with intravenous tracer administration. After the dynamic acquisition, a whole-body PET/CT scan was acquired. Volume-of-interest analyses were performed to obtain estimates of organ-based absorbed doses of radiation. Results:11 C-glutamine was well tolerated in all patients, with no observed safety concerns. The organs with the highest radiation exposure included the bladder, pancreas, and liver. The estimated effective dose was 4.46E-03 ± 7.67E-04 mSv/MBq. Accumulation of11 C-glutamine was elevated and visualized in lung, brain, bone, and liver metastases, suggesting utility for cancer imaging. Conclusion: PET using11 C-glutamine appears safe for human use and allows noninvasive visualization of metastatic colon cancer lesions in multiple organs. Further studies are needed to elucidate its potential for other cancers and for monitoring response to treatment., (© 2022 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2022
- Full Text
- View/download PDF
10. Sacituzumab govitecan, a Trop-2-directed antibody-drug conjugate, for patients with epithelial cancer: final safety and efficacy results from the phase I/II IMMU-132-01 basket trial.
- Author
-
Bardia A, Messersmith WA, Kio EA, Berlin JD, Vahdat L, Masters GA, Moroose R, Santin AD, Kalinsky K, Picozzi V, O'Shaughnessy J, Gray JE, Komiya T, Lang JM, Chang JC, Starodub A, Goldenberg DM, Sharkey RM, Maliakal P, Hong Q, Wegener WA, Goswami T, and Ocean AJ
- Subjects
- Antibodies, Monoclonal, Humanized, Camptothecin analogs & derivatives, Female, Humans, Male, Middle Aged, Immunoconjugates, Lung Neoplasms
- Abstract
Background: Sacituzumab govitecan (SG), a trophoblast cell surface antigen-2 (Trop-2)-directed antibody-drug conjugate, has demonstrated antitumor efficacy and acceptable tolerability in a phase I/II multicenter trial (NCT01631552) in patients with advanced epithelial cancers. This report summarizes the safety data from the overall safety population (OSP) and efficacy data, including additional disease cohorts not published previously., Patients and Methods: Patients with refractory metastatic epithelial cancers received intravenous SG (8, 10, 12, or 18 mg/kg) on days 1 and 8 of 21-day cycles until disease progression or unacceptable toxicity. Endpoints for the OSP included safety and pharmacokinetic parameters with investigator-evaluated objective response rate (ORR per RECIST 1.1), duration of response, clinical benefit rate, progression-free survival, and overall survival evaluated for cohorts (n > 10 patients) of small-cell lung, colorectal, esophageal, endometrial, pancreatic ductal adenocarcinoma, and castrate-resistant prostate cancer., Results: In the OSP (n = 495, median age 61 years, 68% female; UGT1A1∗28 homozygous, n = 46; 9.3%), 41 (8.3%) permanently discontinued treatment due to adverse events (AEs). Most common treatment-related AEs were nausea (62.6%), diarrhea (56.2%), fatigue (48.3%), alopecia (40.4%), and neutropenia (57.8%). Most common treatment-related serious AEs (n = 75; 15.2%) were febrile neutropenia (4.0%) and diarrhea (2.8%). Grade ≥3 neutropenia and febrile neutropenia occurred in 42.4% and 5.3% of patients, respectively. Neutropenia (all grades) was numerically more frequent in UGT1A1∗28 homozygotes (28/46; 60.9%) than heterozygotes (69/180; 38.3%) or UGT1A1∗1 wild type (59/177; 33.3%). There was one treatment-related death due to an AE of aspiration pneumonia. Partial responses were seen in endometrial cancer (4/18, 22.2% ORR) and small-cell lung cancer (11/62, 17.7% ORR), and one castrate-resistant prostate cancer patient had a complete response (n = 1/11; 9.1% ORR)., Conclusions: SG demonstrated a toxicity profile consistent with previous published reports. Efficacy was seen in several cancer cohorts, which validates Trop-2 as a broad target in solid tumors., Competing Interests: Disclosure AB has received research support (paid to institution) from Genentech, Novartis, Pfizer, Merck, Sanofi, Radius Health, Immunomedics, Inc., Mersana, Innocrin, and Biotheranostics Inc.; has served as a consultant to Biotheranostics Inc., Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics, Inc., Spectrum Pharma, Taiho, Sanofi, Daiichi Pharma/AstraZeneca, Puma, Philipps, and Eli Lilly; and has received travel support from Biotheranostics Inc., Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics, Inc., Spectrum Pharma, Taiho, Sanofi, and Philipps. WM has received research support (paid to institution) from Immunomedics, Inc. EAK is a stockholder of Immunomedics, Inc. JDB has received research support (paid to institution) from AbbVie/Pharmacyclics, Beigene, EMD Serono, Immunomedics, Inc., Symphogen, Pfizer, Taiho, Novartis, Genentech/Roche, Bayer, Lilly/Loxo, Incyte, Boston Biomedical, Macrogenics, and I-Mab Biopharma; has served as a consultant to Ipsen, Clovis, and QED Therapeutics, EMD Serono, Seattle Genetics, Bayer, Celgene, LSK Biopharma, Eisai, AstraZeneca, Five Prime, Armo, AbbVie, Symphogen, Erytech, and Gritstone; participates in a Data Safety Monitoring Board for the Pancreatic Cancer Action Network; and receives compensation from the National Cancer Institute for study sections and work on an Investigational Drug Steering Committee. LV has received research support from Weill Cornell Medicine, serves on advisory boards for Immunomedics, Inc., Berg Pharma, and Seattle Genetics, and has served as a consultant to Osmol Therapeutics. RM has received research support from the Orlando Health UF Health Cancer Center and has received honorarium from Eli Lilly, Pfizer, Genentech, Immunomedics, Inc., Seattle Genetics, and Bristol-Myers Squibb (BMS). ADS has served as a consultant to Puma and Merck, and has received research support from Gilead, Puma, Immunomedics, Inc., Synthon, Boehringer-Ingelheim, Genentech, and Tesaro. KK has received research support from Immunomedics, Inc., Novartis, Incyte, Genentech/Roche, Eli Lilly, Pfizer, Calithera Biosciences, Acetylon, Seattle Genetics, Amgen, Zentalis Pharmaceuticals, and CytomX Therapeutics; has served as a consultant to Immunomedics, Inc., Merck, Seattle Genetics, Pfizer, Novartis, Eisai, Eli Lilly, Amgen, and AstraZeneca. His spouse is currently employed by Grail and was previously employed at Array and Pfizer. JO has served as a consultant to AbbVie, Agendia, AstraZeneca, BMS, Celgene, Eisai, Genentech, Immunomedics, Inc., Jounce Therapeutics, Lilly, Merck, Novartis, Pfizer, Puma, Roche, and Seattle Genetics. JEG has received research support from BMS, Merck, AstraZeneca, Array, Boehringer Ingelheim, and Genentech, and has served as a consultant to BMS, Merck, Inivata, EMD Serono/Merck KGaA, AstraZeneca, Celgene, and Novartis. TK has received travel support from Merck and has received honoraria from Boehringer Ingelheim. JML holds equity interest in Salus Discovery, LLC. He has served as a consultant to Janssen, Sanofi, Astellas, Pfizer, and Immunomedics, Inc. AS has served as a consultant to Sandoz and Bayer and has received speaking honoraria from BMS. DMG reports previous employment by Immunomedics, Inc. and status as company founder; serving as Chairman of the Board and Chief Scientific Officer of Immunomedics, Inc.; and was a stockholder. He is an inventor and co-inventor of patents pertaining to sacituzumab govitecan, with potential royalties from product sales. RMS was previously employed by Immunomedics, Inc., at the time this study was conducted; has served as a consultant to Immunomedics, Inc.; and was a stockholder of Immunomedics, Inc. PM reports previous employment by Immunomedics, Inc., while study was performed and being a stockholder in Immunomedics, Inc. QH was an employee and stock shareholder of Immunomedics, Inc., a subsidiary of Gilead Sciences, Inc. and has received travel support from Advaxis; spouse is an employee and stock shareholder of Merck. WW reports previous employment by Immunomedics, Inc., while study was performed; served as a consultant to Immunomedics, Inc., and was a stockholder in Immunomedics, Inc. TG was an employee and stockholder of Immunomedics, Inc. a subsidiary of Gilead Sciences, Inc. AJO has served in a consulting or advisory role for Immunomedics, Inc., Celgene, Tyme Therapeutics, Array, Merck, BMS, ProStrakan, Novartis, Pfizer, Eli Lilly, and Genentech; has served in Speaker's Bureau for Daiichi Sankyo; and has received travel, accommodations, expenses from Daiichi Sankyo. All other authors have declared no conflicts of interest., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Evaluation of determinants for age disparities in the survival improvement of colon cancer: results from a cohort of more than 486,000 patients in the United States.
- Author
-
Chen F, Wang F, Bailey CE, Murff HJ, Berlin JD, Shu XO, and Zheng W
- Abstract
Over the past two decades, elderly colon cancer patients experienced less improvement in survival than their younger counterparts, yet the contributing factors remain unknown. We aimed to evaluate factors that may contribute to the age disparity of survival improvement among patients with colon cancer. Using data from the National Cancer Database, we identified patients diagnosed with colon cancer between 2004 and 2012 with follow-up data up to 2017. The hazard ratios (HR) and 95% confidence intervals (CI) for 5-year OS associated with study variables were estimated using multivariable Cox regression. Among 486,284 patients included in this study, elderly patients (aged ≥75) had a lower adherence to National Comprehensive Cancer Network (NCCN) treatment guidelines (% of non-adherence: 45.3%) than younger patients (aged <50, 19.3%; P <0.001). After adjusting for demographics, access to care and clinical characteristics, compared with patients diagnosed between 2004 and 2006, younger and older patients diagnosed between 2010 and 2012 experienced 16% (HR: 0.84, 95% CI: 0.81-0.88) and 6% (HR: 0.94, 95% CI: 0.93-0.95) reductions in mortality ( P
for interaction =1.42×10-5 ), respectively. After an additional adjustment for guideline adherence status, no significant difference in the improvement of survival was noted ( Pfor interaction =0.17). The association patterns were similar regardless of tumor stage, race, and high comorbidity scores (all Pfor interaction >0.05). Several patient-related factors were identified in association with noncompliance to NCCN guidelines, including comorbidity status. However, over 60% of noncompliance elderly patients had a Charlson comorbidity score of 0. The observed age disparity in survival improvement among colon cancer patients was primarily explained by a slower improvement in adherence to NCCN treatment guidelines in elderly than younger patients. Many older adults were not receiving recommended therapies despite minimal comorbidities. Our findings call for measures to increase adherence to treatment guidelines among elderly patients to improve survival., Competing Interests: None., (AJCR Copyright © 2020.)- Published
- 2020
12. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials.
- Author
-
Hong DS, DuBois SG, Kummar S, Farago AF, Albert CM, Rohrberg KS, van Tilburg CM, Nagasubramanian R, Berlin JD, Federman N, Mascarenhas L, Geoerger B, Dowlati A, Pappo AS, Bielack S, Doz F, McDermott R, Patel JD, Schilder RJ, Tahara M, Pfister SM, Witt O, Ladanyi M, Rudzinski ER, Nanda S, Childs BH, Laetsch TW, Hyman DM, and Drilon A
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Male, Middle Aged, Young Adult, Neoplasms chemistry, Neoplasms drug therapy, Proteins analysis, Pyrazoles therapeutic use, Pyrimidines therapeutic use
- Abstract
Background: The selective TRK inhibitor larotrectinib was approved for paediatric and adult patients with advanced TRK fusion-positive solid tumours based on a primary analysis set of 55 patients. The aim of our analysis was to explore the efficacy and long-term safety of larotrectinib in a larger population of patients with TRK fusion-positive solid tumours., Methods: Patients were enrolled and treated in a phase 1 adult, a phase 1/2 paediatric, or a phase 2 adolescent and adult trial. Some eligibility criteria differed between these studies. For this pooled analysis, eligible patients were aged 1 month or older, with a locally advanced or metastatic non-CNS primary, TRK fusion-positive solid tumour, who had received standard therapy previously if available. This analysis set includes the 55 patients on which approval of larotrectinib was based. Larotrectinib was administered orally (capsule or liquid formulation), on a continuous 28-day schedule, to adults mostly at a dose of 100 mg twice daily, and to paediatric patients mostly at a dose of 100 mg/m
2 (maximum of 100 mg) twice daily. The primary endpoint was objective response as assessed by local investigators in an intention-to-treat analysis. Contributing trials are registered with ClinicalTrials.gov, NCT02122913 (active not recruiting), NCT02637687 (recruiting), and NCT02576431 (recruiting)., Findings: Between May 1, 2014, and Feb 19, 2019, 159 patients with TRK fusion-positive cancer were enrolled and treated with larotrectinib. Ages ranged from less than 1 month to 84 years. The proportion of patients with an objective response according to investigator assessment was 121 (79%, 95% CI 72-85) of 153 evaluable patients, with 24 (16%) having complete responses. In a safety population of 260 patients treated regardless of TRK fusion status, the most common grade 3 or 4 larotrectinib-related adverse events were increased alanine aminotransferase (eight [3%] of 260 patients), anaemia (six, 2%), and decreased neutrophil count (five [2%]). The most common larotrectinib-related serious adverse events were increased alanine aminotransferase (two [<1%] of 260 patients), increased aspartate aminotransferase (two [<1%]), and nausea (two [<1%]). No treatment-related deaths occurred., Interpretation: These data confirm that TRK fusions define a unique molecular subgroup of advanced solid tumours for which larotrectinib is highly active. Safety data indicate that long-term administration of larotrectinib is feasible., Funding: Bayer and Loxo Oncology., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
13. Phase I Study of Trifluridine/Tipiracil Plus Irinotecan and Bevacizumab in Advanced Gastrointestinal Tumors.
- Author
-
Varghese AM, Cardin DB, Hersch J, Benson AB, Hochster HS, Makris L, Hamada K, Berlin JD, and Saltz LB
- Subjects
- Adult, Aged, Aged, 80 and over, Bevacizumab administration & dosage, Drug Combinations, Female, Gastrointestinal Neoplasms pathology, Humans, Irinotecan administration & dosage, Male, Maximum Tolerated Dose, Middle Aged, Patient Safety, Progression-Free Survival, Pyrrolidines administration & dosage, Thymine administration & dosage, Tissue Distribution, Treatment Outcome, Trifluridine administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Gastrointestinal Neoplasms drug therapy
- Abstract
Purpose: This two-part phase Ib trial determined the maximum tolerated dose (MTD) of the combination of trifluridine/tipiracil (FTD/TPI) and irinotecan in patients with advanced gastrointestinal tumors, and evaluated the safety, pharmacokinetics, and antitumor activity of the FTD/TPI, irinotecan, and bevacizumab triplet combination in previously treated metastatic colorectal cancer (mCRC)., Patients and Methods: Dose escalation (3+3 design) in advanced gastrointestinal tumors was followed by expansion in mCRC. During dose escalation, patients received FTD/TPI (20-35 mg/m
2 twice daily; days 1-5 of a 14-day cycle) and irinotecan (120-180 mg/m2 ; day 1). During expansion, the MTD of FTD/TPI and irinotecan plus bevacizumab (5 mg/kg; day 1) was administered., Results: Fifty patients (26 across six dose-escalation cohorts and 24 in the expansion phase) were enrolled. Two dose-limiting toxicities (fatigue and neutropenia) were observed in the dose-escalation phase, and MTD was defined as FTD/TPI 25 mg/m2 twice daily plus irinotecan 180 mg/m2 . In the expansion phase, 83% (20/24) experienced any-cause grade ≥3 adverse events (AEs) with the triplet combination, most frequently neutropenia (42%), leukopenia (25%), and diarrhea (12%). AEs of any-cause led to dosing interruptions, modifications, and discontinuations in 29%, 17%, and 4% of patients, respectively. No treatment-related deaths occurred. Three patients (12%) experienced partial responses and 16 (67%) patients had stable disease lasting >4 months. The median progression-free survival was 7.9 months (95% confidence interval, 5.1-13.4 months)., Conclusions: Tolerability and activity observed in this phase I trial support further investigation of the FTD/TPI-irinotecan-bevacizumab combination in previously treated mCRC., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
14. Recent advances in the treatment of pancreatic cancer.
- Author
-
Roth MT, Cardin DB, and Berlin JD
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Humans, Neoadjuvant Therapy, Quality of Life, Carcinoma, Pancreatic Ductal therapy, Pancreatic Neoplasms therapy
- Abstract
Pancreatic ductal adenocarcinoma is one of the deadliest solid tumor malignancies and is projected to become a leading cause of cancer-related death in coming years. Improving quality of life and survival amongst these patients will require new ideas and novel therapies in a multidisciplinary approach. This review will cover the most recent advances in the comprehensive treatment of pancreatic cancer and place them within a historical context when necessary. Treatment of all disease stages will be discussed, but the focus is on systemic therapy as novel drugs and new treatment combinations enter the clinic. This will include more aggressive chemotherapy in earlier disease stages, approved uses for immunotherapy, and targetable mutations. In addition, negative trials of importance and controversial topics will be noted., Competing Interests: No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed., (Copyright: © 2020 Roth MT et al.)
- Published
- 2020
- Full Text
- View/download PDF
15. Correction: A phase 2 randomised study of veliparib plus FOLFIRI±bevacizumab versus placebo plus FOLFIRI±bevacizumab in metastatic colorectal cancer.
- Author
-
Gorbunova V, Beck JT, Hofheinz RD, Garcia-Alfonso P, Nechaeva M, Gracian AC, Mangel L, Fernandez EE, Deming DA, Ramanathan RK, Torres AH, Sullivan D, Luo Y, and Berlin JD
- Abstract
The original version of this article contained an error in Figure 1a. The number of patients at risk listed in the Veliparib arm of Figure 1a should have read "65" instead of "35".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
16. Advanced pancreatic cancer clinical trials: The continued underrepresentation of older patients.
- Author
-
White MN, Dotan E, Catalano PJ, Cardin DB, and Berlin JD
- Subjects
- Adenocarcinoma pathology, Age Distribution, Aged, Fatigue chemically induced, Humans, Infections chemically induced, Linear Models, Middle Aged, Pancreatic Neoplasms pathology, Progression-Free Survival, Survival Rate, Thrombocytopenia chemically induced, Adenocarcinoma drug therapy, Antineoplastic Agents therapeutic use, Clinical Trials, Phase III as Topic statistics & numerical data, Pancreatic Neoplasms drug therapy, Research Subjects statistics & numerical data
- Abstract
Objectives: Older patients make up the majority of patients with pancreatic cancer, with a median age of 71 years at diagnosis. However, older patients are underrepresented in clinical trials in pancreatic cancer. This study investigates trends in age distribution of patients enrolled in clinical trials for advanced pancreatic cancer over time, and examines outcomes and toxicity in older patient subgroups from two studies conducted by Eastern Cooperative Oncology Group and American College of Radiology Imaging Network (ECOG-ACRIN) in this disease., Materials and Methods: 16,042 patients from 38 phase III clinical trials for locally advanced or metastatic pancreatic adenocarcinoma published between 1997 and 2016 were identified and included in this analysis. Outcomes and toxicity by age were examined in two of the trials, ECOG-ACRIN trials E2297 and E6201, which included a total of 1146 patients., Results: The median age across the trials was 62.7 years; median ages for individual trials ranged from 57 years to 66 years. Weighted linear regression showed no significant change in median age over time. Combined analysis of the two ECOG-ACRIN trials demonstrated higher rates of fatigue, thrombocytopenia, and infection in those ≥75 years compared with those <75 years, but despite this showed no difference in overall survival (OS) or progression-free survival (PFS) (OS: 5.7 vs. 5.6 months and PFS: 2.8 vs 3.5 months)., Conclusions: Enrollment of older adults in phase III pancreatic cancer clinical trials has not increased over time, despite increasing number of older patients seen in clinic. Increased efforts are needed to enhance enrollment of older patients in clinical trials, and to promote trials specifically for older patients, in order to improve the evidence base for treating this patient population., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
17. Impact of Peritoneal Metastasis on Survival of Patients With Small Intestinal Neuroendocrine Tumor.
- Author
-
Wright MF, Cates J, Gonzalez RS, Das S, Berlin JD, and Shi C
- Subjects
- Adult, Aged, Aged, 80 and over, Disease-Free Survival, Female, Humans, Intestinal Neoplasms mortality, Intestine, Small pathology, Liver Neoplasms mortality, Liver Neoplasms secondary, Male, Middle Aged, Neuroendocrine Tumors mortality, Peritoneal Neoplasms mortality, Prognosis, Young Adult, Intestinal Neoplasms pathology, Neuroendocrine Tumors secondary, Peritoneal Neoplasms secondary
- Abstract
The liver and peritoneum are the 2 most common distant metastatic sites for small intestinal neuroendocrine tumors (SI-NET). In this study, we evaluated the differential impact of hepatic and/or peritoneal metastasis on prognosis of SI-NET patients. Surgical Pathology archives were searched for SI-NET resections performed between January 1, 1994 and August 31, 2017. Two hundred nineteen cases with clinical follow-up data were identified. Pathology reports and electronic medical records were reviewed. The 219 patients included 104 females and 115 males with a median age of 59 years (range, 19 to 85 y). There were 71 (33%) cases without hepatic or peritoneal metastasis, 80 (37%) with hepatic metastasis only, 14 (6%) with peritoneal metastasis only, and 53 (24%) with both hepatic and peritoneal metastasis at the time of surgery or during follow-up. The number of primary tumors, largest tumor size, lymph node metastasis, pT category, and sex were not significant independent prognostic factors in multivariate Cox proportional hazard regression. Age was the only variable other than presence of metastatic disease that was associated with worse prognosis (5% increase in risk/year of age; 95% confidence interval, 1.7%-8.2%; P=0.003). After controlling for patient age, pairwise comparisons of marginal linear predictions showed increased risk with peritoneal metastasis, with or without associated hepatic metastasis, compared to hepatic metastasis only. In conclusion, although limited by the number of patients with peritoneal metastasis only, these results support substratifying patients with metastatic SI-NET by anatomic site of metastasis.
- Published
- 2019
- Full Text
- View/download PDF
18. Phase I study combining the aurora kinase a inhibitor alisertib with mFOLFOX in gastrointestinal cancer.
- Author
-
Goff LW, Azad NS, Stein S, Whisenant JG, Koyama T, Vaishampayan U, Hochster H, Connolly R, Weise A, LoRusso PM, Salaria SN, El-Rifai W, and Berlin JD
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Azepines administration & dosage, Female, Fluorouracil administration & dosage, Gastrointestinal Neoplasms metabolism, Gastrointestinal Neoplasms pathology, Humans, Leucovorin administration & dosage, Male, Maximum Tolerated Dose, Middle Aged, Organoplatinum Compounds administration & dosage, Prognosis, Pyrimidines administration & dosage, Survival Rate, Tissue Distribution, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Aurora Kinase A antagonists & inhibitors, Gastrointestinal Neoplasms drug therapy, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Neoplastic drug effects
- Abstract
Overexpression and cellular mis-localization of aurora kinase A (AURKA) in gastrointestinal cancers results in chromosomal instability, activation of multiple oncogenic pathways, and inhibition of pro-apoptotic signaling. Inhibition of AURKA causes mitotic delays, severe chromosome congression, and activation of p53/p73 leading to cell death. Our preclinical data showed cooperative activity with the AURKA inhibitor alisertib and platinum agents in cell lines and xenografts, and suggested an optimal treatment window. Therefore, this study was designed to determine the maximum-tolerated dose (MTD) of alisertib in combination with modified FOLFOX (mFOLFOX), as this is a standard platinum-based therapy for gastrointestinal cancers. Standard 3 + 3 dose escalation was used, where the starting dose of alisertib was 10 mg twice daily (Days 1-3), with leucovorin (400 mg/m
2 ) and oxaliplatin (85 mg/m2 ) on Day 2 followed by continuous 46-h 5-FU (2400 mg/m2 ) infusion on Days 2-4 in 14-day cycles. Fourteen patients with advanced gastrointestinal cancers were enrolled and two doses explored; two patients were not evaluable for dose-limiting toxicity (DLT) and replaced. Two patients experienced DLTs at 20 mg of alisertib (Grade 3 fatigue (n = 2); Grade 3 nausea, vomiting, dehydration with hospitalization (n = 1)). MTD was 10 mg alisertib with 85 mg/m2 oxaliplatin and 2400 mg/m2 5-FU. Most frequent toxicities were nausea (57%), diarrhea, fatigue, neuropathy, and vomiting (43%), and anorexia and anemia (36%); most were Grade 1-2. One patient with colorectal cancer had a partial response of 12 evaluable patients, and four patients had stable disease. Alisertib in combination with mFOLFOX did not demonstrate unexpected side effects, but the regimen was only tolerable at the lowest dose investigated.- Published
- 2019
- Full Text
- View/download PDF
19. A phase 2 randomised study of veliparib plus FOLFIRI±bevacizumab versus placebo plus FOLFIRI±bevacizumab in metastatic colorectal cancer.
- Author
-
Gorbunova V, Beck JT, Hofheinz RD, Garcia-Alfonso P, Nechaeva M, Cubillo Gracian A, Mangel L, Elez Fernandez E, Deming DA, Ramanathan RK, Torres AH, Sullivan D, Luo Y, and Berlin JD
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Benzimidazoles adverse effects, Bevacizumab administration & dosage, Bevacizumab adverse effects, Camptothecin administration & dosage, Camptothecin adverse effects, Colorectal Neoplasms pathology, Drug-Related Side Effects and Adverse Reactions classification, Drug-Related Side Effects and Adverse Reactions pathology, Female, Fluorouracil administration & dosage, Fluorouracil adverse effects, Humans, Kaplan-Meier Estimate, Leucovorin administration & dosage, Leucovorin adverse effects, Male, Middle Aged, Neoplasm Metastasis, Progression-Free Survival, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Benzimidazoles administration & dosage, Camptothecin analogs & derivatives, Colorectal Neoplasms drug therapy
- Abstract
Background: Metastatic colorectal cancer (mCRC) has low survival rates. We assessed if addition of veliparib, concurrent to FOLFIRI, improves survival in patients with previously untreated mCRC., Methods: This study compared veliparib (200 mg BID for 7 days of each 14-day cycle) to placebo, each with FOLFIRI. Bevacizumab was allowed in both arms. The primary endpoint was progression-free survival (PFS)., Results: Patients were randomised to receive veliparib (n = 65) or placebo (n = 65) in combination with FOLFIRI. Median PFS was 12 vs 11 months (veliparib vs placebo) [HR = 0.94 (95% CI: 0.60, 1.48)]. Median OS was 25 vs 27 months [HR = 1.26 (95% CI: 0.74, 2.16)]. Response rate was 57% vs 62%. Median DOR was 11 vs 9 months [HR = 0.73 (95% CI: 0.38, 1.40)]. AEs with significantly higher frequency (p < 0.05) in the veliparib group were anaemia (39% vs 19%, p = 0.019) and neutropenia (66% vs 37%, p = 0.001) for common AEs (≥20%); neutropenia (59% vs 22%, p < 0.001) for common Grade 3/4 AEs (≥5%); none in serious AEs. Haematopoietic cytopenias were more common with veliparib (79% vs 52%, p = 0.003). Fourteen percent of patients on veliparib and 15% on placebo discontinued treatment due to AEs., Conclusion: Veliparib added to FOLFIRI ± bevacizumab demonstrated similar efficacy as FOLFIRI ± bevacizumab in frontline mCRC patients. No unexpected safety concerns occurred.
- Published
- 2019
- Full Text
- View/download PDF
20. A Phase 1b/2 Study of the Bruton Tyrosine Kinase Inhibitor Ibrutinib and the PD-L1 Inhibitor Durvalumab in Patients with Pretreated Solid Tumors.
- Author
-
Hong D, Rasco D, Veeder M, Luke JJ, Chandler J, Balmanoukian A, George TJ, Munster P, Berlin JD, Gutierrez M, Mita A, Wakelee H, Samakoglu S, Guan S, Dimery I, Graef T, and Borazanci E
- Subjects
- Adenine analogs & derivatives, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Disease-Free Survival, Female, Humans, Male, Middle Aged, Piperidines, Prospective Studies, Pyrazoles adverse effects, Pyrazoles pharmacokinetics, Pyrimidines adverse effects, Pyrimidines pharmacokinetics, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, B7-H1 Antigen antagonists & inhibitors, Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Pyrimidines therapeutic use
- Abstract
Background: Ibrutinib, a first-in-class, once-daily inhibitor of Bruton's tyrosine kinase, is approved in the United States for the treatment of various B-cell malignancies. Preclinical data suggest synergistic antitumor activity of ibrutinib with programmed death-ligand 1 (PD-L1) inhibitors in solid tumors. This study evaluated ibrutinib plus durvalumab, a PD-L1-targeting antibody, in patients with relapsed/refractory solid tumors., Methods: This open-label, multicenter, phase 1b/2 study enrolled previously treated patients with stage III/IV pancreatic adenocarcinoma, breast cancer, or non-small cell lung cancer (NSCLC). Phase 1b determined the recommended phase 2 dose (RP2D). In phase 2, patients were treated at the RP2D to evaluate the safety and antitumor activity of ibrutinib plus durvalumab., Results: The RP2D was identified as ibrutinib 560 mg p.o. daily and durvalumab 10 mg/kg i.v. every 2 weeks, with 122 patients treated at the RP2D. Median age was 61 years, and the majority of patients (94%) had stage IV disease. Overall response rates (complete or partial responses) were 2% for pancreatic cancer, 3% for breast cancer, and 0% for NSCLC. Median progression-free survival was 1.7, 1.7, and 2.0 months in the pancreatic cancer, breast cancer, and NSCLC cohorts, respectively. Median overall survival was 4.2, 4.2, and 7.9 months in the pancreatic cancer, breast cancer, and NSCLC cohorts, respectively. The safety profiles observed across tumor types were consistent with the known safety profiles for ibrutinib and durvalumab. Grade ≥3 adverse events in ≥5% of all patients were hyponatremia (10%), dyspnea (7%), maculopapular rash (7%), pneumonia (7%), anemia (6%), and diarrhea (6%)., Conclusions: The combination of ibrutinib 560 mg daily and durvalumab 10 mg/kg every 2 weeks had an acceptable safety profile. The antitumor activity of the ibrutinib-durvalumab combination was limited in our study population., (© 2019 The Author(s) Published by S. Karger AG, Basel.)
- Published
- 2019
- Full Text
- View/download PDF
21. Correction to: Phase I study combining the aurora kinase a inhibitor alisertib with mFOLFOX in gastrointestinal cancer.
- Author
-
Goff LW, Azad NS, Stein S, Whisenant JG, Koyama T, Vaishampayan U, Hochster H, Connolly R, Weise A, LoRusso PM, Salaria SN, El-Rifai W, and Berlin JD
- Abstract
The authors would like to note that the investigator affiliations have been corrected to reflect the actual affiliations of each author. The authors would also like to note an amendment to the first name of the second author. Nilo Azad was changed to reflect the full name of the author, which is Nilofer S. Azad as shown above. The original article has been corrected.
- Published
- 2018
- Full Text
- View/download PDF
22. First-in-human phase I dose escalation study of MK-8033 in patients with advanced solid tumors.
- Author
-
Keedy VL, Lenz HJ, Saltz L, Whisenant JG, Berlin JD, and Camacho LH
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Antineoplastic Agents blood, Antineoplastic Agents pharmacokinetics, Benzocycloheptenes adverse effects, Benzocycloheptenes blood, Benzocycloheptenes pharmacokinetics, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms metabolism, Progression-Free Survival, Sulfonamides adverse effects, Sulfonamides blood, Sulfonamides pharmacokinetics, Antineoplastic Agents administration & dosage, Benzocycloheptenes administration & dosage, Neoplasms drug therapy, Proto-Oncogene Proteins c-met antagonists & inhibitors, Sulfonamides administration & dosage
- Abstract
Background C-Met, which is frequently activated in multiple cancers, has been implicated in tumor formation, progression, metastasis, angiogenesis, and resistance to multiple therapies. MK-8033 is a small-molecule inhibitor of c-Met that binds preferentially to the activated conformation, and has demonstrated anti-tumor activity in preclinical models. This first-in-human trial was performed to establish the safety and maximum tolerated dose (MTD), as well as preliminary pharmacokinetics (PK) and clinical activity. Methods Forty-seven patients were enrolled in three parts. The primary objective of Parts A and B was safety, whereas Part C evaluated the effect of proton-pump inhibitors on MK-8033 absorption. Dose escalation used an accelerated continual reassessment method, and dose-limiting toxicities (DLTs) were any treatment-related, first course non-hematologic grade ≥ 3 toxicity (except alopecia or inadequately treated nausea/vomiting/diarrhea), grade 4 hematologic toxicity (except grade 3 neutropenic fever and thrombocytopenia), or toxicity where treatment is held >3 weeks. Results Forty-six patients were treated across nine dose levels, and the MTD was 750 mg twice daily. DLTs were fatigue, nausea, vomiting, transaminitis, and hypokalemia. Most frequent toxicities were fatigue (28.3%), nausea (21.7%), and alopecia (19.6%), predominately grade ≤ 2. One patient with endometriod adenocarcinoma achieved a partial response and eight had stable disease. Median progression-free survival (PFS) was 57 days. Strikingly, the PFS for the one responder was 846 days. PK results showed that proton-pump inhibitors have no effect on MK-8033 absorption. Conclusion MK-8033 was well tolerated with no significant toxicity issues, albeit with limited clinical activity. Unfortunately, the company decided to discontinue further clinical development of MK-8033.
- Published
- 2018
- Full Text
- View/download PDF
23. Consensus statement on essential patient characteristics in systemic treatment trials for metastatic colorectal cancer: Supported by the ARCAD Group.
- Author
-
Goey KKH, Sørbye H, Glimelius B, Adams RA, André T, Arnold D, Berlin JD, Bodoky G, de Gramont A, Díaz-Rubio E, Eng C, Falcone A, Grothey A, Heinemann V, Hochster HS, Kaplan RS, Kopetz S, Labianca R, Lieu CH, Meropol NJ, Price TJ, Schilsky RL, Schmoll HJ, Shacham-Shmueli E, Shi Q, Sobrero AF, Souglakos J, Van Cutsem E, Zalcberg J, van Oijen MGH, Punt CJA, and Koopman M
- Subjects
- Biomarkers, Tumor genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Consensus, DNA Mismatch Repair, Delphi Technique, Humans, Karnofsky Performance Status, Liver Neoplasms genetics, Liver Neoplasms secondary, Microsatellite Instability, Mutation, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Treatment Outcome, Clinical Trials, Phase III as Topic standards, Colorectal Neoplasms therapy, Liver Neoplasms therapy, Patient Selection
- Abstract
Background: Patient characteristics and stratification factors are key features influencing trial outcomes. However, there is substantial heterogeneity in reporting of patient characteristics and use of stratification factors in phase 3 trials investigating systemic treatment of metastatic colorectal cancer (mCRC). We aimed to develop a minimum set of essential baseline characteristics and stratification factors to include in such trials., Methods: We performed a modified, two-round Delphi survey among international experts with wide experience in the conduct and methodology of phase 3 trials of systemic treatment of mCRC., Results: Thirty mCRC experts from 15 different countries completed both consensus rounds. A total of 14 patient characteristics were included in the recommended set: age, performance status, primary tumour location, primary tumour resection, prior chemotherapy, number of metastatic sites, liver-only disease, liver involvement, surgical resection of metastases, synchronous versus metachronous metastases, (K)RAS and BRAF mutation status, microsatellite instability/mismatch repair status and number of prior treatment lines. A total of five patient characteristics were considered the most relevant stratification factors: RAS/BRAF mutation status, performance status, primary tumour sidedness and liver-only disease., Conclusions: This survey provides a minimum set of essential baseline patient characteristics and stratification factors to include in phase 3 trials of systemic treatment of mCRC. Inclusion of these patient characteristics and strata in study protocols and final study reports will improve interpretation of trial results and facilitate cross-study comparisons., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
24. A Phase II Study of Ganetespib as Second-line or Third-line Therapy for Metastatic Pancreatic Cancer.
- Author
-
Cardin DB, Thota R, Goff LW, Berlin JD, Jones CM, Ayers GD, Whisenant JG, and Chan E
- Subjects
- Adult, Aged, Carcinoma, Pancreatic Ductal secondary, Female, Follow-Up Studies, Humans, Lymphatic Metastasis, Male, Middle Aged, Pancreatic Neoplasms pathology, Prognosis, Survival Rate, Carcinoma, Pancreatic Ductal drug therapy, Drug Resistance, Neoplasm, Pancreatic Neoplasms drug therapy, Salvage Therapy, Triazoles therapeutic use
- Abstract
Objectives: Heat shock protein 90 regulates multiple signaling proteins involved in key pathways of pancreatic cancer pathogenesis. Ganetespib binds to heat shock protein 90 and interferes with its binding to client proteins thus leading to inactivation and degradation of the signaling proteins that promote cancer progression. This phase II study was designed to evaluate the efficacy of ganetespib in patients with refractory metastatic pancreatic cancer (rMPC)., Methods: Patients with rMPC received 175 mg/m ganetespib intravenously once weekly for 3 weeks in 4-week cycles. Primary endpoint was disease control rate at 8 weeks, with a goal of 70%. Secondary endpoints were progression-free survival, overall survival, and safety. Simon's 2-stage design was used to assess futility and efficacy. Ganetespib was considered inactive if ≤8 patients among the first 15 treated had disease control after 8 weeks of treatment., Results: Fourteen patients were treated on study. Grade 3 treatment-related toxicities were diarrhea, abdominal pain, fatigue, nausea, vomiting, and hyponatremia. Disease control rate at 8 weeks was 28.6%, and median progression-free survival and overall survival were 1.58 months and 4.57 months, respectively. Early stopping rules for lack of clinical efficacy led to study closure., Conclusions: Single-agent ganetespib was tolerable with only modest disease control in rMPC. This disease is resistant to chemotherapy, and given the emerging data in lung and rectal cancers, as well as in pancreatic cancer cell lines, suggesting improved activity of ganetespib in combination with cytotoxic agents, studies combining this agent with chemotherapy in rMPC are more likely to yield success.
- Published
- 2018
- Full Text
- View/download PDF
25. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018.
- Author
-
Shah MH, Goldner WS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, Chan J, Kulke MH, Benson AB, Blaszkowsky LS, Eads J, Engstrom PF, Fanta P, Giordano T, He J, Heslin MJ, Kalemkerian GP, Kandeel F, Khan SA, Kidwai WZ, Kunz PL, Kuvshinoff BW, Lieu C, Pillarisetty VG, Saltz L, Sosa JA, Strosberg JR, Sussman CA, Trikalinos NA, Uboha NA, Whisenant J, Wong T, Yao JC, Burns JL, Ogba N, and Zuccarino-Catania G
- Subjects
- Adrenal Gland Neoplasms diagnosis, Adult, Humans, Neuroendocrine Tumors diagnosis, Societies, Medical standards, United States, Adrenal Gland Neoplasms therapy, Delivery of Health Care, Integrated standards, Medical Oncology standards, Neuroendocrine Tumors therapy
- Abstract
The NCCN Guidelines for Neuroendocrine and Adrenal Tumors provide recommendations for the management of adult patients with neuroendocrine tumors (NETs), adrenal gland tumors, pheochromocytomas, and paragangliomas. Management of NETs relies heavily on the site of the primary NET. These NCCN Guidelines Insights summarize the management options and the 2018 updates to the guidelines for locoregional advanced disease, and/or distant metastasis originating from gastrointestinal tract, bronchopulmonary, and thymus primary NETs., (Copyright © 2018 by the National Comprehensive Cancer Network.)
- Published
- 2018
- Full Text
- View/download PDF
26. Current Concepts in the Treatment of Resectable Pancreatic Cancer.
- Author
-
Roth MT and Berlin JD
- Subjects
- Combined Modality Therapy, Humans, Pancreatic Neoplasms surgery, Prognosis, Neoadjuvant Therapy, Pancreatectomy methods, Pancreatic Neoplasms therapy
- Abstract
Purpose of Review: The diagnosis of pancreatic cancer carries with it a high mortality rate. Despite advances in the field, this has remained relatively unchanged over the last few decades. Current options for the treatment of resectable pancreatic ductal adenocarcinoma will be reviewed here in conjunction with the historical data that support them. We will focus on updates in treatment guidelines and ongoing clinical trials of interest., Recent Findings: For localized disease, standard of care includes resection followed by adjuvant chemotherapy ± chemoradiation. Recently, a report was published supporting the use of doublet therapy with gemcitabine and capecitabine (as opposed to gemcitabine monotherapy), which prompted a practice-changing update to major treatment guidelines. Multiple trials using neoadjuvant treatment, novel therapies, and different forms of radiation are ongoing. Although pancreatic cancer is an active area of research, outcomes remain dismal. Clinical trials will need to be more robust and innovative to drastically improve survival statistics.
- Published
- 2018
- Full Text
- View/download PDF
27. Consensus statement on mandatory measurements in pancreatic cancer trials (COMM-PACT) for systemic treatment of unresectable disease.
- Author
-
Ter Veer E, van Rijssen LB, Besselink MG, Mali RMA, Berlin JD, Boeck S, Bonnetain F, Chau I, Conroy T, Van Cutsem E, Deplanque G, Friess H, Glimelius B, Goldstein D, Herrmann R, Labianca R, Van Laethem JL, Macarulla T, van der Meer JHM, Neoptolemos JP, Okusaka T, O'Reilly EM, Pelzer U, Philip PA, van der Poel MJ, Reni M, Scheithauer W, Siveke JT, Verslype C, Busch OR, Wilmink JW, van Oijen MGH, and van Laarhoven HWM
- Subjects
- Biomarkers blood, Consensus, Delphi Technique, Health Status, Humans, Pancreatic Neoplasms blood, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Treatment Outcome, Clinical Trials, Phase III as Topic standards, Data Accuracy, Pancreatic Neoplasms therapy, Randomized Controlled Trials as Topic standards, Research Design standards
- Abstract
Variations in the reporting of potentially confounding variables in studies investigating systemic treatments for unresectable pancreatic cancer pose challenges in drawing accurate comparisons between findings. In this Review, we establish the first international consensus on mandatory baseline and prognostic characteristics in future trials for the treatment of unresectable pancreatic cancer. We did a systematic literature search to find phase 3 trials investigating first-line systemic treatment for locally advanced or metastatic pancreatic cancer to identify baseline characteristics and prognostic variables. We created a structured overview showing the reporting frequencies of baseline characteristics and the prognostic relevance of identified variables. We used a modified Delphi panel of two rounds involving an international panel of 23 leading medical oncologists in the field of pancreatic cancer to develop a consensus on the various variables identified. In total, 39 randomised controlled trials that had data on 15 863 patients were included, of which 32 baseline characteristics and 26 prognostic characteristics were identified. After two consensus rounds, 23 baseline characteristics and 12 prognostic characteristics were designated as mandatory for future pancreatic cancer trials. The COnsensus statement on Mandatory Measurements in unresectable PAncreatic Cancer Trials (COMM-PACT) identifies a mandatory set of baseline and prognostic characteristics to allow adequate comparison of outcomes between pancreatic cancer studies., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. An Intergroup Randomized Phase II Study of Bevacizumab or Cetuximab in Combination with Gemcitabine and in Combination with Chemoradiation in Patients with Resected Pancreatic Carcinoma: A Trial of the ECOG-ACRIN Cancer Research Group (E2204).
- Author
-
Berlin JD, Feng Y, Catalano P, Abbruzzese JL, Philip PA, McWilliams RR, Lowy AM, Benson AB III, and Blackstock AW
- Subjects
- Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic administration & dosage, Antineoplastic Agents, Immunological administration & dosage, Bevacizumab administration & dosage, Cetuximab administration & dosage, Chemoradiotherapy methods, Combined Modality Therapy, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Disease-Free Survival, Female, Humans, Male, Middle Aged, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms therapy
- Abstract
Objectives: Evaluate toxicity of two treatment arms, A (cetuximab) and B (bevacizumab), when combined with gemcitabine, and chemoradiation in patients with completely resected pancreatic carcinoma. Secondary objectives included overall survival (OS) and disease-free survival (DFS)., Methods: Patients with R0/R1 resection were randomized 1:1 to cetuximab or bevacizumab administered in combination with gemcitabine for two treatment cycles. Next three cycles included concurrent cetuximab/bevacizumab plus chemoradiation, followed by one cycle of cetuximab/bevacizumab. Cycles 7-12 included cetuximab/bevacizumab with gemcitabine. Cycles were 2 weeks. Frequency of specific toxicities was summarized for each treatment arm at two times during the study, after chemotherapy but prior to chemoradiation and after all therapy., Results: A total of 127 patients were randomized (A, n = 65; B, n = 62). Prior to chemoradiation, the overall rate for toxicities of interest was 10% for arm A and 2% for arm B. After all therapy, the overall rates for toxicities of interest were 30 and 25% for arms A and B, respectively. Overall median OS and DFS were 17 and 11 months, respectively, which is not a significant improvement over expected survival rates for historical controls., Conclusions: Both treatments were tolerable with manageable toxicities, and were safe enough for a phase III trial had this been indicated., (© 2017 S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
29. Phase I/II Trial of Labetuzumab Govitecan (Anti-CEACAM5/SN-38 Antibody-Drug Conjugate) in Patients With Refractory or Relapsing Metastatic Colorectal Cancer.
- Author
-
Dotan E, Cohen SJ, Starodub AN, Lieu CH, Messersmith WA, Simpson PS, Guarino MJ, Marshall JL, Goldberg RM, Hecht JR, Wegener WA, Sharkey RM, Govindan SV, Goldenberg DM, and Berlin JD
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Camptothecin administration & dosage, Camptothecin adverse effects, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Disease Progression, Disease-Free Survival, Drug Administration Schedule, Female, Half-Life, Humans, Infusions, Intravenous, Irinotecan, Male, Middle Aged, Neoplasm Metastasis, Survival Analysis, Treatment Outcome, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents administration & dosage, Camptothecin analogs & derivatives, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm
- Abstract
Purpose The objectives were to evaluate dosing schedules of labetuzumab govitecan, an antibody-drug conjugate targeting carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) for tumor delivery of 7-ethyl-10-hydroxycamptothecin (SN-38), in an expanded phase II trial of patients with relapsed or refractory metastatic colorectal cancer. Patients and Methods Eligible patients with at least one prior irinotecan-containing therapy received labetuzumab govitecan once weekly at 8 and 10 mg/kg, or two times per week at 4 and 6 mg/km on weeks 1 and 2 of 3-week repeated cycles. End points were safety, response, pharmacokinetics, and immunogenicity. Results Eighty-six patients who had undergone a median of five prior therapies (range, one to 13) were each enrolled into one of the four cohorts. On the basis of Response Evaluation Criteria in Solid Tumors 1.1, 38% of these patients had a tumor as well as plasma carcinoembryonic antigen reduction from baseline after labetuzumab govitecan treatment; one patient achieved a partial response with a sustained response spanning > 2 years, whereas 42 patients had stable disease as the best overall response. Median progression-free survival and overall survival were 3.6 and 6.9 months, respectively. The major toxicities (grade ≥ 3) among all cohorts were neutropenia (16%), leukopenia (11%), anemia (9%), and diarrhea (7%). The antibody-drug conjugate's mean half-life was 16.5 hours for the four cohorts. Anti-drug/anti-antibody antibodies were not detected. The two once-weekly dose schedules, showing comparable toxicity and efficacy, were chosen for further study. Conclusion Monotherapy with labetuzumab govitecan demonstrated a manageable safety profile and therapeutic activity in heavily pretreated patients with metastatic colorectal cancer, all with prior irinotecan therapy. Further studies of labetuzumab govitecan treatment alone or in combination with other therapies in earlier settings are indicated.
- Published
- 2017
- Full Text
- View/download PDF
30. A Phase Ib Dose-Escalation Study of the Safety, Tolerability, and Pharmacokinetics of Cobimetinib and Duligotuzumab in Patients with Previously Treated Locally Advanced or Metastatic Cancers with Mutant KRAS.
- Author
-
Lieu CH, Hidalgo M, Berlin JD, Ko AH, Cervantes A, LoRusso P, Gerber DE, Eder JP, Eckhardt SG, Kapp AV, Tsuhako A, McCall B, Pirzkall A, Uyei A, and Tabernero J
- Subjects
- Acneiform Eruptions epidemiology, Acneiform Eruptions etiology, Administration, Oral, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Asthenia epidemiology, Asthenia etiology, Azetidines pharmacology, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Dose-Response Relationship, Drug, Drug Eruptions epidemiology, Drug Eruptions etiology, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Female, Humans, Hypokalemia epidemiology, Hypokalemia etiology, Immunoglobulin G pharmacology, MAP Kinase Kinase 1 antagonists & inhibitors, MAP Kinase Kinase 1 metabolism, Male, Middle Aged, Neoplasm Staging, Piperidines pharmacology, Prospective Studies, Receptor, ErbB-3 antagonists & inhibitors, Receptor, ErbB-3 metabolism, Signal Transduction drug effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols pharmacology, Azetidines therapeutic use, Colorectal Neoplasms drug therapy, Immunoglobulin G therapeutic use, Piperidines therapeutic use, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Lessons Learned: Cobimetinib and duligotuzumab were well tolerated as single agents and in combination with other agents.The cobimetinib and duligotuzumab combination was associated with increased toxicity, most notably gastrointestinal, and limited efficacy in the patient population tested., Background: KRAS -mutant tumors possess abnormal mitogen-activated protein kinases (MAPK) pathway signaling, leading to dysregulated cell proliferation. Cobimetinib blocks MAPK signaling. The dual-action antibody duligotuzumab (MEHD7945A) inhibits ligand binding to both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 3 (HER3). Blockade of EGFR/HER3 and inhibition of mitogen-activated protein kinase (MEK) in KRAS -mutant tumors may provide additive benefit., Methods: Patients with KRAS -mutant solid tumors were eligible for this phase Ib dose-escalation study with a planned expansion phase. Duligotuzumab was given intravenously (IV) at 1,100 mg every 2 weeks (q2w), while cobimetinib was given orally in a standard 3 + 3 design to identify the recommended phase II dose (RP2D). The primary objective was to evaluate the safety and tolerability of this combination., Results: Twenty-three patients were enrolled. Dose-limiting toxicities (DLTs) included grade 4 hypokalemia and grade 3 mucosal inflammation, asthenia, and dermatitis acneiform. Seventy percent of patients experienced grade 3 or worse adverse events (AEs). Five (22%) and 12 (52%) patients missed at least 1 dose of duligotuzumab and cobimetinib, respectively, and 9 (39%) patients required a cobimetinib dose reduction. Three (13%) patients discontinued due to an AE. Best response was limited to 9 patients with stable disease and 13 patients with progressive disease., Conclusion: Given the limited tolerability and efficacy of this combination, the study did not proceed to expansion stage and closed for enrollment., (© AlphaMedPress; the data published online to support this summary is the property of the authors.)
- Published
- 2017
- Full Text
- View/download PDF
31. Phase II study of the Multikinase inhibitor of angiogenesis, Linifanib, in patients with metastatic and refractory colorectal cancer expressing mutated KRAS.
- Author
-
Chan E, Goff LW, Cardin DB, Ancell K, Smith SJ, Whisenant JG, Ye F, and Berlin JD
- Subjects
- Adult, Aged, Angiogenesis Inhibitors adverse effects, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Disease-Free Survival, Drug Resistance, Neoplasm, Female, Humans, Indazoles adverse effects, Kaplan-Meier Estimate, Lung Neoplasms diagnostic imaging, Lung Neoplasms drug therapy, Lung Neoplasms secondary, Male, Middle Aged, Mutation, Phenylurea Compounds adverse effects, Protein Kinase Inhibitors adverse effects, Tomography, X-Ray Computed, Treatment Outcome, Angiogenesis Inhibitors therapeutic use, Colorectal Neoplasms drug therapy, Indazoles therapeutic use, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Background Targeting angiogenesis in advanced colorectal cancer (CRC) has been one of the many factors prolonging survival. Bevacizumab was the first agent to demonstrate this, but even after progression on bevacizumab, continued VEGF-inhibition continues to improve survival. Combining epidermal growth factor receptor monoclonal antibodies with standard frontline therapies have also improved clinical outcomes, yet the improved benefit is not observed in patients with mutant KRAS. Thus, an unmet medical need exists to develop additional therapeutic options for patients with KRAS mutant CRC. Methods Patients received the anti-angiogenic agent linifanib at the recommended phase II dose of 17.5 mg. Primary endpoint was objective response rate (ORR), with a goal of 10%. Secondary endpoints were progression-free survival (PFS), overall survival (OS), and safety. Simon's optimal two-stage design was used to assess futility. Linifanib was considered inactive if two or fewer patients among the first 30 achieved an objective response. Results Thirty patients were enrolled on study. Grade 3 treatment-related toxicities occurring in at least two patients were fatigue, hypertension, proteinuria, diarrhea, nausea, oral pain, vomiting, thrombocytopenia, and arthralgia. Although no responses were observed, 63.5% of patients achieved stable disease. The median PFS and OS were 4.7 months and 9.5 months, respectively. Stopping rules for lack of clinical efficacy led to study closure. Conclusion Despite observing zero responses, a majority of patients had stable disease and eight patients had stable disease lasting longer than 5 months. These results suggest that linifanib has some anti-tumor activity in KRAS mutant metastatic and refractory CRC.
- Published
- 2017
- Full Text
- View/download PDF
32. A phase 1 study of anti-TGFβ receptor type-II monoclonal antibody LY3022859 in patients with advanced solid tumors.
- Author
-
Tolcher AW, Berlin JD, Cosaert J, Kauh J, Chan E, Piha-Paul SA, Amaya A, Tang S, Driscoll K, Kimbung R, Kambhampati SR, Gueorguieva I, and Hong DS
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Cohort Studies, Cytokines metabolism, Dose-Response Relationship, Drug, Female, Half-Life, Humans, Infusions, Intravenous, Male, Maximum Tolerated Dose, Middle Aged, Treatment Outcome, Young Adult, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Receptors, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
Purpose: LY3022859 is an anti-TGFβRII IgG
1 monoclonal antibody that inhibits receptor-mediated signaling activation. The primary objective of this phase I study was to determine a phase II dose in patients with advanced solid tumors. Secondary objectives were to assess safety and pharmacokinetics (PK)., Methods: LY3022859 was infused intravenously (IV) at 1.25 mg/kg over 1 h every 2 weeks (Q2W) (cohort 1A) and at flat doses of 12.5 mg (cohort 1B) and 25 mg (cohort 2) over 3 h Q2W., Results: Fourteen patients were enrolled in cohorts 1A (n = 2), 1B (n = 5), and 2 (n = 7). DLTs were experienced by both patients in cohort 1A (infusion-related reaction) and 2 patients in cohort 2 (cytokine release syndrome and infusion-related reaction). No MTD was determined. At the 25 mg dose level (cohort 2), after fifth infusion, LY3022859 had a short t1/2 (4.37-7.80 h) and rapid clearance (CLss , 0.412 L/h). Exposure increased twofold (from 28.5 to 60.2 μg·h/mL) with increase in dose from 12.5 to 25 mg. No accumulation was observed after repeat administration., Conclusions: The MTD for LY3022859 was not determined. Dose escalation beyond 25 mg was considered unsafe due to worsening symptoms (uncontrolled cytokine release) despite prophylaxis (corticosteroids and antihistamines)., Trial Registration: clinicaltrials.gov Identifier: NCT01646203.- Published
- 2017
- Full Text
- View/download PDF
33. Academic Cancer Center Phase I Program Development.
- Author
-
Frankel AE, Flaherty KT, Weiner GJ, Chen R, Azad NS, Pishvaian MJ, Thompson JA, Taylor MH, Mahadevan D, Lockhart AC, Vaishampayan UN, Berlin JD, Smith DC, Sarantopoulos J, Riese M, Saleh MN, Ahn C, and Frenkel EP
- Subjects
- Clinical Trials as Topic, Humans, Neoplasms genetics, Program Development, United States, Academic Medical Centers, Neoplasms epidemiology
- Abstract
Multiple factors critical to the effectiveness of academic phase I cancer programs were assessed among 16 academic centers in the U.S. Successful cancer centers were defined as having broad phase I and I/II clinical trial portfolios, multiple investigator-initiated studies, and correlative science. The most significant elements were institutional philanthropic support, experienced clinical research managers, robust institutional basic research, institutional administrative efforts to reduce bureaucratic regulatory delays, phase I navigators to inform patients and physicians of new studies, and a large cancer center patient base. New programs may benefit from a separate stand-alone operation, but mature phase I programs work well when many of the activities are transferred to disease-oriented teams. The metrics may be useful as a rubric for new and established academic phase I programs. The Oncologist 2017;22:369-374., (© The Authors. The Oncologist published by Wiley Periodicals, Inc. on behalf of AlphaMed Press 2017.)
- Published
- 2017
- Full Text
- View/download PDF
34. Preclinical Rationale for the Phase III Trials in Metastatic Pancreatic Cancer: Is Wishful Thinking Clouding Successful Drug Development for Pancreatic Cancer?
- Author
-
Thota R, Maitra A, and Berlin JD
- Subjects
- Animals, Drug Therapy methods, Drug Therapy trends, Humans, Neoplasm Metastasis, Pancreatic Neoplasms pathology, Randomized Controlled Trials as Topic methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clinical Trials, Phase III as Topic methods, Drug Screening Assays, Antitumor methods, Pancreatic Neoplasms drug therapy
- Abstract
Prior phase III trials in advanced pancreatic cancer have been predominantly unsuccessful. In this review, we attempt to understand how past preclinical data were translated into phase III clinical trials in metastatic pancreatic cancer as described in the article. A systematic literature review conducted through the MEDLINE, Embase, and Cochrane Central Register of Controlled Trials databases, from January 1997 to June 2015 using key words-phase III clinical trials, metastatic/advanced pancreatic adenocarcinoma or pancreatic cancer identified 30 randomized controlled trials (RCTs) that met criteria. The trials were limited to RCTs in the first-line treatment of patients with metastatic pancreatic cancer. The success rate of first-line phase III studies in advanced pancreatic cancer was only 13%. In 60% of the RCTs, no preclinical experiments were referenced in biologically cognate pancreatic models. Nine (30%) of the RCTs were designed based on preclinical evidence from in vitro cell lines alone without additional in vivo validation in xenograft models. It remains uncertain how strongly the preclinical data influence the development of clinical regimens but so far the studies developed based on more solid preclinical evidence have been successful.
- Published
- 2017
- Full Text
- View/download PDF
35. Evaluation of Pancreatic Cancer Clinical Trials and Benchmarks for Clinically Meaningful Future Trials: A Systematic Review.
- Author
-
Rahib L, Fleshman JM, Matrisian LM, and Berlin JD
- Subjects
- Clinical Trials as Topic, Disease-Free Survival, Humans, Survival Rate, Adenocarcinoma drug therapy, Antineoplastic Agents therapeutic use, Benchmarking, Clinical Trials, Phase II as Topic, Clinical Trials, Phase III as Topic, Pancreatic Neoplasms drug therapy
- Abstract
Importance: Progress in the treatment of pancreatic adenocarcinoma has been minimal; it remains the only major cancer type with a 5-year survival rate of less than 10%., Objective: To explore why a large proportion of advanced pancreatic cancer clinical trials executed over the past 25 years have had negative results and to identify benchmarks that could have predicted success., Evidence Review: Phase 3 studies of patients with advanced pancreatic cancer were identified by searching clinicaltrials.gov and the scientific literature., Findings: Thirty-two phase 3 studies in 13 675 chemotherapy-naive patients resulted in 3 agents or combinations being considered clinically meaningful. Nineteen agents or combinations (70%) were tested in phase 2 trials preceding the phase 3 trial. In cases with paired phase 2 and 3 results, meeting the primary end point of the phase 2 trial predicted the outcome of the phase 3 trial 76% of the time but proceeded despite phase 2 negative results in 10 cases. We applied criteria for a clinically meaningful result identified by the American Society of Clinical Oncology (ASCO) Cancer Research Committee to these historical cases. Overall, progression-free and 1-year survival of experimental arms was compared with time period-controlled median values of control arms to normalize for the observed increase in response to gemcitabine over time., Conclusions and Relevance: Applying the benchmark of a 50% improvement in overall survival as the primary end point to phase 2 data, or secondary end points of a 90% increase in 1-year survival or an 80% to 100% increase in progression-free survival, showed the greatest ability to predict a clinically meaningful phase 3 trial. Had these criteria been applied to these trials over the past 25 years, more than 11 571 patients enrolled in phase 3 trials that did not meet the primary end point could theoretically have been diverted to earlier-stage trials in an attempt to more rapidly advance the field.
- Published
- 2016
- Full Text
- View/download PDF
36. A Phase I Study of the Anti-Activin Receptor-Like Kinase 1 (ALK-1) Monoclonal Antibody PF-03446962 in Patients with Advanced Solid Tumors.
- Author
-
Goff LW, Cohen RB, Berlin JD, de Braud FG, Lyshchik A, Noberasco C, Bertolini F, Carpentieri M, Stampino CG, Abbattista A, Wang E, and Borghaei H
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized, Dose-Response Relationship, Drug, Female, Humans, Male, Middle Aged, Treatment Outcome, Activin Receptors, Type II immunology, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Neoplasms drug therapy
- Abstract
Purpose: Objectives of this dose-finding study were to determine the MTD and recommended phase II dose (RP2D) of the first-in-class anti-activin receptor-like kinase 1 (ALK-1) monoclonal antibody PF-03446962, and assess safety and antitumor activity in patients with advanced solid tumors., Experimental Design: This open-label, multicenter study was based on a 3+3 design. PF-03446962 was administered biweekly by intravenous infusion, at doses ranging from 0.5 to 15 mg/kg., Results: Forty-four patients received treatment with PF-03446962. Dose-limiting toxicities observed during dose escalation included grade 3 increased amylase, grade 3/4 increased lipase, and grade 3/4 thrombocytopenia. The MTD was determined to be 10 mg/kg. The RP2D was set at 7 mg/kg for patients with advanced solid tumors, based on the observed safety, pharmacokinetics, and antitumor activity. The most-frequent treatment-related, all-grade adverse events included thrombocytopenia (20.5%), fatigue (15.9%), and nausea, increased amylase, and increased lipase (each 11.4%). Treatment-related telangiectasia was noted in 7% of patients, suggesting in vivo inhibition of the ALK-1 pathway. None of the deaths was deemed to be treatment-related. Three (6.8%) patients with advanced hepatocellular carcinoma, renal cell carcinoma, or non-small cell lung cancer achieved a partial response, and 12 (27.3%) patients had stable disease, across dose levels. Contrast-enhanced ultrasound analysis of tumor vascularity showed reduction in tumor perfusion in 2 patients with stable disease following treatment with PF-03446962., Conclusions: The clinical activity demonstrated in this study points to PF-03446962 as a novel approach to antiangiogenic therapy, with manageable safety profile and single-agent, antitumor activity in patients with advanced solid tumors. Clin Cancer Res; 22(9); 2146-54. ©2015 AACR., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
37. Phase I trial of vorinostat added to chemoradiation with capecitabine in pancreatic cancer.
- Author
-
Chan E, Arlinghaus LR, Cardin DB, Goff L, Berlin JD, Parikh A, Abramson RG, Yankeelov TE, Hiebert S, Merchant N, Bhaskara S, and Chakravarthy AB
- Subjects
- Adult, Aged, Aged, 80 and over, Diffusion Magnetic Resonance Imaging, Humans, Male, Maximum Tolerated Dose, Middle Aged, Pancreatic Neoplasms diagnostic imaging, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Capecitabine administration & dosage, Chemoradiotherapy, Pancreatic Neoplasms therapy
- Abstract
Background and Purpose: This single institution phase I trial determined the maximum tolerated dose (MTD) of concurrent vorinostat and capecitabine with radiation in non-metastatic pancreatic cancer., Material and Methods: Twenty-one patients received escalating doses of vorinostat (100-400mg daily) during radiation. Capecitabine was given 1000mg q12 on the days of radiation. Radiation consisted of 30Gy in 10 fractions. Vorinostat dose escalation followed the standard 3+3 design. No dose escalation beyond 400mg vorinostat was planned. Diffusion-weighted (DW)-MRI pre- and post-treatment was used to evaluate in vivo tumor cellularity., Results: The MTD of vorinostat was 400mg. Dose limiting toxicities occurred in one patient each at dose levels 100mg, 300mg, and 400mg: 2 gastrointestinal toxicities and one thrombocytopenia. The most common adverse events were lymphopenia (76%) and nausea (14%). The apparent diffusion coefficient (ADC) increased in most tumors. Nineteen (90%) patients had stable disease, and two (10%) had progressive disease at time of surgery. Eleven patients underwent surgical exploration with four R0 resections and one R1 resection. Median overall survival was 1.1years (95% confidence interval 0.78-1.35)., Conclusions: The combination of vorinostat 400mg daily M-F and capecitabine 1000mg q12 M-F with radiation (30Gy in 10 fractions) was well tolerated with encouraging median overall survival., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
38. The Past, Present, and Future of Pancreatic Cancer Clinical Trials.
- Author
-
Matrisian LM and Berlin JD
- Subjects
- Biomarkers, Tumor genetics, Clinical Trials, Phase III as Topic, Humans, Pancreatic Neoplasms epidemiology, Pancreatic Neoplasms genetics, Survival Rate, Immunotherapy, Molecular Targeted Therapy, Pancreatic Neoplasms therapy
- Abstract
Upper gastrointestinal malignancies comprise half of the deadliest cancers as defined by those with a 5-year survival rate less than 50%. Using pancreatic adenocarcinoma (PAC) as an example, we retrospectively evaluated the success of phase III clinical trials, examined the current landscape of clinical trials, and identified emerging areas that foretell the future for this disease. Pancreatic and liver cancers are on the rise and will be the second and third leading causes of cancer deaths in 2030. A total of 35 different agents or combinations have been tested in randomized phase III clinical trials for patients with advanced PAC over the past 25 years, but only 11% have been incorporated into clinical practice. There has been a 37% increase in the number of PAC trials open in the United States between 2011 to 2012 and 2014 to 2015. Enrollment has also increased slightly, from 3.85% of the newly diagnosed cases in 2011 to 4.15% in 2014. However, the demand for patients far exceeds the number of patients available for these trials. On the horizon is the realization that stratification of patients with PAC using biomarkers that predict a high probability of a response could reallocate patients to faster, smaller trials with a greater chance of a survival benefit. The current landscape of PAC clinical trials and the launch of the Pancreatic Cancer Action Network's Know Your Tumor initiative indicate this shift is starting to occur, with particular emphasis on targeted therapies, immunotherapies, and agents that disrupt the stroma.
- Published
- 2016
- Full Text
- View/download PDF
39. Neuroendocrine tumors, version 1.2015.
- Author
-
Kulke MH, Shah MH, Benson AB 3rd, Bergsland E, Berlin JD, Blaszkowsky LS, Emerson L, Engstrom PF, Fanta P, Giordano T, Goldner WS, Halfdanarson TR, Heslin MJ, Kandeel F, Kunz PL, Kuvshinoff BW 2nd, Lieu C, Moley JF, Munene G, Pillarisetty VG, Saltz L, Sosa JA, Strosberg JR, Vauthey JN, Wolfgang C, Yao JC, Burns J, and Freedman-Cass D
- Subjects
- Disease Management, Humans, Neuroendocrine Tumors diagnosis, Neuroendocrine Tumors therapy
- Abstract
Neuroendocrine tumors (NETs) comprise a broad family of tumors that may or may not be associated with symptoms attributable to hormonal hypersecretion. The NCCN Clinical Practice Guidelines in Oncology for Neuroendocrine Tumors discuss the diagnosis and management of both sporadic and hereditary NETs. This selection from the guidelines focuses on sporadic NETs of the pancreas, gastrointestinal tract, lung, and thymus., (Copyright © 2015 by the National Comprehensive Cancer Network.)
- Published
- 2015
- Full Text
- View/download PDF
40. Thymidylate synthase genotype-directed chemotherapy for patients with gastric and gastroesophageal junction cancers.
- Author
-
Goff LW, Thakkar N, Du L, Chan E, Tan BR, Cardin DB, McLeod HL, Berlin JD, Zehnbauer B, Fournier C, Picus J, Wang-Gillam A, Lee W, and Lockhart AC
- Subjects
- Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic adverse effects, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cardia pathology, Esophageal Neoplasms drug therapy, Female, Fluorouracil adverse effects, Fluorouracil therapeutic use, Gene Frequency genetics, Genetic Variation genetics, Humans, Leucovorin adverse effects, Leucovorin therapeutic use, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local prevention & control, Organoplatinum Compounds adverse effects, Organoplatinum Compounds therapeutic use, Prospective Studies, Risk, Stomach Neoplasms drug therapy, Treatment Outcome, Enhancer Elements, Genetic genetics, Esophageal Neoplasms genetics, Esophagogastric Junction pathology, Stomach Neoplasms genetics, Thymidylate Synthase genetics
- Abstract
Background: Retrospective studies indicate associations between TSER (thymidylate synthase enhancer region) genotypes and clinical outcomes in patients receiving 5-FU based chemotherapy, but well-controlled prospective validation has been lacking., Methods: In this phase II study (NCT00515216 registered through ClinicalTrials.gov, http://clinicaltrials.gov/show/NCT00515216), patients with "good risk" TSER genotypes (at least one TSER*2 allele) were treated with FOLFOX chemotherapy to determine whether prospective patient selection can improve overall response rates (ORR) in patients with gastric and gastroesophageal junction (GEJ) cancers, compared with historical outcomes in unselected patients (estimated 43%)., Results: The ORR in genotype-selected patients was 39.1% (9 partial responses out of 23 evaluable patients, 95% CI, 22.2 to 59.2), not achieving the primary objective of improving ORR. An encouraging disease control rate (DCR, consisting of partial responses and stable diseases) of 95.7% was noted and patients with homozygous TSER*2 genotype showed better tumor response., Conclusions: In this first prospective, multi-institutional study in patients with gastric or GEJ cancers, selecting patients with at least one TSER*2 allele did not improve the ORR but led to an encouraging DCR. Further studies are needed to investigate the utility of selecting patients homozygous for the TSER*2 allele and additional genomic markers in improving clinical outcomes for patients with gastric and GEJ cancers., Trial Registration: ClinicalTrials.gov NCT00515216.
- Published
- 2014
- Full Text
- View/download PDF
41. Phase 1/2 study of KRN330, a fully human anti-A33 monoclonal antibody, plus irinotecan as second-line treatment for patients with metastatic colorectal cancer.
- Author
-
Bendell JC, Lenz HJ, Ryan T, El-Rayes BF, Marshall JL, Modiano MR, Hart LL, Kingsley CD, George TJ, Nakashima D, and Berlin JD
- Subjects
- Adolescent, Adult, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Antineoplastic Agents, Phytogenic therapeutic use, Camptothecin analogs & derivatives, Camptothecin therapeutic use, Colorectal Neoplasms pathology, Disease Progression, Disease-Free Survival, Female, Humans, Irinotecan, Male, Maximum Tolerated Dose, Middle Aged, Young Adult, Antibodies, Monoclonal therapeutic use, Colorectal Neoplasms drug therapy, Membrane Glycoproteins antagonists & inhibitors
- Abstract
KRN330 is a recombinant, fully-human monoclonal antibody directed against A33, a surface differentiation antigen that is uniformly expressed in 95 % of colorectal cancers. A previous Phase 1 study of single-agent KRN330 identified a maximum tolerated dose (MTD) of 3 mg/kg q2w and preliminary evidence of clinical activity among patients with advanced and metastatic colorectal cancer (mCRC). This Phase 1/2 trial sought to assess the safety and activity of second-line KRN330 plus irinotecan in patients with mCRC. Patients with mCRC who showed disease progression after FOLFOX/CapOx received intravenous doses of KRN330 (0.5 or 1.0 mg/kg qw or q2w) plus irinotecan (180 mg/m(2)) in a standard 3 + 3 dose escalation. The MTD of KRN330 with irinotecan in 19 patients was 0.5 mg/kg qw in the Phase 1 study with gastrointestinal effects and neutropenia being the predominant dose-limiting toxicities. In the Phase 2 study, the most frequent treatment-related Grade ≥3 toxicities in 44 patients were fatigue (15.9 %), neutropenia (13.6 %), leukopenia (6.8 %), diarrhea (4.5 %), and dehydration (4.5 %). Objective response rate (ORR) was 4.5 % and disease control rate was 45.5 % for the intent-to-treat population. Median progression-free survival was 87 days (95 % CI, 43-136 days). The prespecified ORR of KRN330 plus irinotecan was not met. Further investigation of KRN330 plus other agents may be warranted.
- Published
- 2014
- Full Text
- View/download PDF
42. Enabling a genetically informed approach to cancer medicine: a retrospective evaluation of the impact of comprehensive tumor profiling using a targeted next-generation sequencing panel.
- Author
-
Johnson DB, Dahlman KH, Knol J, Gilbert J, Puzanov I, Means-Powell J, Balko JM, Lovly CM, Murphy BA, Goff LW, Abramson VG, Crispens MA, Mayer IA, Berlin JD, Horn L, Keedy VL, Reddy NM, Arteaga CL, Sosman JA, and Pao W
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms pathology, Female, Head and Neck Neoplasms pathology, High-Throughput Nucleotide Sequencing, Humans, Male, Melanoma pathology, Middle Aged, Neoplasm Staging, Breast Neoplasms genetics, Head and Neck Neoplasms genetics, Melanoma genetics, Neoplasm Proteins genetics
- Abstract
Background: Oncogenic genetic alterations "drive" neoplastic cell proliferation. Small molecule inhibitors and antibodies are being developed that target an increasing number of these altered gene products. Next-generation sequencing (NGS) is a powerful tool to identify tumor-specific genetic changes. To determine the clinical impact of extensive genetic analysis, we reviewed our experience using a targeted NGS platform (FoundationOne) in advanced cancer patients., Patients and Methods: We retrospectively assessed demographics, NGS results, and therapies received for patients undergoing targeted NGS (exonic sequencing of 236 genes and selective intronic sequencing from 19 genes) between April 2012 and August 2013. Coprimary endpoints were the percentage of patients with targeted therapy options uncovered by mutational profiling and the percentage who received genotype-directed therapy., Results: Samples from 103 patients were tested, most frequently breast carcinoma (26%), head and neck cancers (23%), and melanoma (10%). Most patients (83%) were found to harbor potentially actionable genetic alterations, involving cell-cycle regulation (44%), phosphatidylinositol 3-kinase-AKT (31%), and mitogen-activated protein kinase (19%) pathways. With median follow-up of 4.1 months, 21% received genotype-directed treatments, most in clinical trials (61%), leading to significant benefit in several cases. The most common reasons for not receiving genotype-directed therapy were selection of standard therapy (35%) and clinical deterioration (13%)., Conclusion: Mutational profiling using a targeted NGS panel identified potentially actionable alterations in a majority of advanced cancer patients. The assay identified additional therapeutic options and facilitated clinical trial enrollment. As time progresses, NGS results will be used to guide therapy in an increasing proportion of patients., (©AlphaMed Press.)
- Published
- 2014
- Full Text
- View/download PDF
43. American Society of Clinical Oncology perspective: Raising the bar for clinical trials by defining clinically meaningful outcomes.
- Author
-
Ellis LM, Bernstein DS, Voest EE, Berlin JD, Sargent D, Cortazar P, Garrett-Mayer E, Herbst RS, Lilenbaum RC, Sima C, Venook AP, Gonen M, Schilsky RL, Meropol NJ, and Schnipper LE
- Subjects
- Clinical Trials, Phase II as Topic standards, Clinical Trials, Phase III as Topic standards, Humans, Societies, Medical organization & administration, Societies, Medical standards, Treatment Outcome, United States, Clinical Trials as Topic standards, Medical Oncology organization & administration
- Published
- 2014
- Full Text
- View/download PDF
44. Treatment of metastatic pancreatic adenocarcinoma: a review.
- Author
-
Thota R, Pauff JM, and Berlin JD
- Subjects
- Adenocarcinoma mortality, Adenocarcinoma secondary, Albumins administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clinical Trials, Phase III as Topic, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Humans, Neoplasm Metastasis, Paclitaxel administration & dosage, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Gemcitabine, Pancreatic Neoplasms, Adenocarcinoma drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Gemcitabine monotherapy has been the standard of care for patients with metastatic pancreatic cancer for several decades. Despite recent advances in various chemotherapeutic regimens and in the development of targeted therapies, metastatic pancreatic cancer remains highly resistant to chemotherapy. Previous studies of several combination regimens showed minimal or no significant change in overall survival compared with gemcitabine alone. Secreted protein acidic and rich in cysteine (SPARC) overexpression in pancreatic stromal fibroblasts is considered one of the major causes of chemotherapy resistance. The nanoparticle albumin-bound formulation of paclitaxel (nab-paclitaxel) has been found to be superior to other formulations of paclitaxel because of its favorable pharmacokinetic properties. Initial preclinical studies showed its synergistic effect with gemcitabine in pancreatic cancer, in which nab-paclitaxel is sequestered by SPARC to cause stromal depletion and increasing microvasculature, resulting in higher gemcitabine concentration within the tumor. In the recent phase III multinational Metastatic Pancreatic Adenocarcinoma Clinical Trial (MPACT), the combination of gemcitabine and nab-paclitaxel was shown to be superior to gemcitabine monotherapy, with an increase in median survival of 1.8 months. Combination therapy with gemcitabine plus erlotinib, or with gemcitabine plus nab-paclitaxel, or the multidrug regimen of leucovorin, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX) can be considered as first-line chemotherapy for patients with metastatic pancreatic cancer. In this review we will discuss details of the recently approved combination of gemcitabine and nab-paclitaxel for first-line treatment of metastatic pancreatic adenocarcinoma and compare it with other therapeutic options.
- Published
- 2014
45. Pancreas cancer on the rise: are we up to the challenge?
- Author
-
Cardin DB and Berlin JD
- Subjects
- Female, Humans, Male, Black or African American statistics & numerical data, Pancreatic Neoplasms mortality, White People statistics & numerical data
- Published
- 2013
- Full Text
- View/download PDF
46. Clinical utility of KRAS and BRAF mutations in a cohort of patients with colorectal neoplasms submitted for microsatellite instability testing.
- Author
-
Cushman-Vokoun AM, Stover DG, Zhao Z, Koehler EA, Berlin JD, and Vnencak-Jones CL
- Subjects
- Adult, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Staging, Prognosis, Proto-Oncogene Proteins p21(ras), Retrospective Studies, Survival Rate, Biomarkers, Tumor genetics, Colorectal Neoplasms genetics, Microsatellite Instability, Mutation genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf genetics, ras Proteins genetics
- Abstract
Background: Molecular analysis has become important in colorectal carcinoma (CRC) evaluation. Alterations in KRAS, BRAF, or mismatch repair (MMR) genes may determine therapeutic response or define a hereditary cancer syndrome. Correlation of DNA studies with clinical findings will further clarify the clinical utility of these markers., Patients and Methods: A retrospective study was performed on 111 paraffin-embedded tumor specimens submitted for microsatellite instability (MSI) testing based on clinical history or histologic examination, or both. DNA samples were screened for 7 KRAS mutations and the BRAF p.V600E mutation using fluorescent allele-specific polymerase-chain reaction (PCR) and capillary electrophoresis. Clinical data were collected through chart review., Results: Fifty-eight male and 53 female patients were studied. The incidence of KRAS and BRAF mutations was 49.5% and 7.2%, respectively. Dideoxy sequencing verified KRAS mutation status in 46 of 49 specimens tested. There was a trend toward significance of individual KRAS mutations on survival (P = .003). Dually positive KRAS and MSI tumors exclusively demonstrated p.G12D and p.G13D mutations (G>A transitions). BRAF-mutated tumors were predominantly right-sided and associated with a borderline worse prognosis. Forty-eight percent of tumors with MSI were present in the left colon or rectum., Conclusion: Allele-specific PCR is an accurate and convenient method to assess KRAS and BRAF mutations and may detect mutations not identified by dideoxy sequencing. KRAS mutation status, in conjunction with morphologic or clinical parameters, may be useful in determining whether a tumor should be tested for MSI. MSI testing should not be considered exclusively in right-sided lesions. BRAF analysis may not be useful in rectal adenocarcinomas and should be evaluated in larger studies., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Borderline resectable pancreatic cancer: need for standardization and methods for optimal clinical trial design.
- Author
-
Katz MH, Marsh R, Herman JM, Shi Q, Collison E, Venook AP, Kindler HL, Alberts SR, Philip P, Lowy AM, Pisters PW, Posner MC, Berlin JD, and Ahmad SA
- Subjects
- Adenocarcinoma pathology, Humans, Neoadjuvant Therapy, Pancreatectomy, Pancreatic Neoplasms pathology, Patient Selection, Adenocarcinoma therapy, Clinical Trials as Topic standards, Pancreatic Neoplasms therapy, Research Design standards
- Abstract
Background: Methodological limitations of prior studies have prevented progress in the treatment of patients with borderline resectable pancreatic adenocarcinoma. Shortcomings have included an absence of staging and treatment standards and pre-existing biases with regard to the use of neoadjuvant therapy and the role of vascular resection at pancreatectomy., Methods: In this manuscript, we review limitations of studies of borderline resectable PDAC reported to date, highlight important controversies related to this disease stage, emphasize the research infrastructure necessary for its future study, and present a recently-approved Intergroup pilot study (Alliance A021101) that will provide a foundation upon which subsequent well-designed clinical trials can be performed., Results: We identified twenty-three studies published since 2001 which report outcomes of patients with tumors labeled as borderline resectable and who were treated with neoadjuvant therapy prior to planned pancreatectomy. These studies were heterogeneous in terms of the populations studied, the metrics used to characterize therapeutic response, and the indications used to select patients for surgery. Mechanisms used to standardize these and other issues that are incorporated into Alliance A021101 are reviewed., Conclusions: Rigorous standards of clinical trial design incorporated into trials of other disease stages must be adopted in all future studies of borderline resectable pancreatic cancer. The Intergroup trial should serve as a paradigm for such investigations.
- Published
- 2013
- Full Text
- View/download PDF
48. Safety, pharmacokinetics and pharmacodynamics of the anti-A33 fully-human monoclonal antibody, KRN330, in patients with advanced colorectal cancer.
- Author
-
Infante JR, Bendell JC, Goff LW, Jones SF, Chan E, Sudo T, Burris HA, and Berlin JD
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Area Under Curve, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Diarrhea chemically induced, Dose-Response Relationship, Drug, Drug Administration Schedule, Drug Resistance, Neoplasm drug effects, Female, Humans, Immunohistochemistry, Male, Metabolic Clearance Rate, Middle Aged, Nausea chemically induced, Neoplasm Metastasis, Time Factors, Treatment Outcome, Vomiting chemically induced, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal therapeutic use, Colorectal Neoplasms drug therapy, Membrane Glycoproteins immunology
- Abstract
Purpose: The objective of this first-in-human trial included the safety, maximum tolerated dose (MTD), pharmacokinetics, immunogenicity and antitumour effects of KRN330, a novel fully-human monoclonal antibody directed against A33, a membrane bound glycoprotein uniformly expressed in 95% of colorectal cancers., Methods: Patients with advanced or metastatic colorectal cancer (CRC) refractory to standard therapy were eligible. Twenty-nine patients received weekly intravenous KRN330 (0.1-10mg/kg) for a minimum of 4 weeks in a standard 3+3 design, and nine patients received q2 week doses at 3mg/kg with pre- and post-biopsies to evaluate tumour binding and safety on this schedule., Results: The most common KRN330 related adverse events (all grades) were nausea (66%), diarrhoea (61%) and vomiting (47%). The MTD was 3mg/kg weekly, with dose-limiting grade 3 gastrointestinal toxicities at 10mg/kg and the intermediate dose level of 6 mg/kg. Pharmacokinetics of KRN330 was linear. Stable disease was reported in 12/38 patients (32%), with a median duration of 155 days. There was no evidence of human anti-human antibodies, and immunohistochemistry on biopsy samples demonstrated that KRN330 remained bound to tumour 2 weeks after dosing., Conclusions: KRN330 is safe and tolerable at the MTD of 3mg/kg once weekly in patients with advanced CRC. Dosing on alternate weeks is supported by tumour binding. The long treatment durations and lack of immunogenicity warrant further investigation of KRN330 in combination., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Systemic cytotoxic and biological therapies of colorectal liver metastases: expert consensus statement.
- Author
-
Schwarz RE, Berlin JD, Lenz HJ, Nordlinger B, Rubbia-Brandt L, and Choti MA
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chemotherapy, Adjuvant, Clinical Trials, Phase III as Topic, Colorectal Neoplasms diagnosis, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Consensus Development Conferences as Topic, Hepatectomy, Humans, Liver Neoplasms diagnosis, Liver Neoplasms mortality, Liver Neoplasms secondary, Neoplasm Metastasis, Patient Selection, Practice Guidelines as Topic, Radiotherapy, Adjuvant, Survival Analysis, Treatment Outcome, Colorectal Neoplasms therapy, Liver Neoplasms therapy
- Abstract
Systemic therapy for colorectal cancer liver metastases (CRLM) has undergone significant development in the past 15 years. Therapy regimens consisting of combinations of cytotoxic chemotherapeutic agents have demonstrated greater efficacy and contributed to a significant survival improvement. As the majority of patients who undergo resection for liver-only CRLM are at risk of disease recurrence and cancer-related death, combining resection with systemic therapy appears sensible. However, trial-based evidence is sparse to support this concept. Peri-operative FOLFOX has demonstrated a progression-free survival benefit in a single Phase III trial; the safety of chemotherapy and subsequent operations was acceptable and only a few patients showed initial progression. Chemotherapy-associated liver injury (CALI), including sinusoidal obstruction syndrome and steatohepatitis, has been observed after cytotoxic therapy, and should have implications for chemotherapy plans prior to hepatectomy. In general, pre-operative chemotherapy should not extend beyond 3 months. For patients with unresectable liver-only CRLM, a response to chemotherapy could establish resectability and should be considered an initial treatment goal. In patients with unresectable CRLM, oxaliplatin- or irinotecan-containing combinations represent the standard options, although single-agent choices may be appropriate for individual patients. The addition of bevacizumab carries the potential for a greater response and possibly for reduced CALI risks. In tumours without K-ras mutations, anti-epidermal growth factor receptor (EGFR) agents are also reasonable choices for a greater response and improved survival outcomes. It is crucial that all systemic CRLM treatment decisions include proper definitions of treatment goals and endpoints, and are derived based on appropriate multidisciplinary considerations for other potentially applicable local or regional modalities., (© 2012 International Hepato-Pancreato-Biliary Association.)
- Published
- 2013
- Full Text
- View/download PDF
50. A phase I study of cetuximab in combination with gemcitabine and radiation for locally advanced pancreatic cancer.
- Author
-
Chakravarthy AB, Tsai CJ, O'Brien N, Lockhart AC, Chan E, Parikh A, Berlin JD, and Merchant N
- Abstract
Background: Cetuximab is a monoclonal antibody against the epidermal growth factor receptor (EGFR). The primary goal of this phase I study was to determine the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs) of gemcitabine when combined with cetuximab plus radiation in patients with locally advanced pancreatic cancer., Patients and Methods: Patients with locally unresectable adenocarcinoma of the pancreas were treated with gemcitabine (200 mg/m(2)/week before dose escalation) plus cetuximab (400 mg/m(2) loading dose, 250 mg/m(2) weekly maintenance dose) concurrent with radiation (50.4 Gy)., Results: Nine patients were enrolled in the study. One withdrew due to declining performance status before receiving any therapy. Grade 4 allergic reactions to cetuximab caused the withdrawal of 2 patients. Another patient had elevated liver function test results and a stroke after his loading dose of cetuximab. Grade 3 or 4 toxicity developed in 3 of the remaining 5 patients treated with the level 1 dose. Therefore, no further dose escalations were planned. Grade 3 toxicities included nausea, vomiting, ileus, and pneumonitis. One patient had grade 4 diarrhea., Conclusions: The combination of cetuximab, gemcitabine, and radiation resulted in significant toxicity. A recommended phase II dose could not be determined.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.