Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cancer Lett ; 497: 221-228, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33127389

ABSTRACT

Recent research on genomic profiling of pancreatic ductal adenocarcinoma (PDAC) has identified many potentially actionable alterations. However, the feasibility of using genomic profiling to guide routine clinical decision making for PDAC patients remains unclear. We retrospectively reviewed PDAC patients between October 2013 and December 2017, who underwent treatment at the Johns Hopkins Hospital and had clinical tumor next-generation sequencing (NGS) through commercial resources. Ninety-two patients with 93 tumors tested were included. Forty-eight (52%) patients had potentially curative surgeries. The median time from the tissue available to the NGS testing ordered was 229 days (interquartile range 62-415). A total of three (3%) patients had matched targeted therapies based on genomic profiling results. Genomic profiling guided personalized treatment for PDAC patients is feasible, but the percentage of patients who receive targeted therapy is low. The main challenges are ordering NGS testing early in the clinical course of the disease and the limited evidence of using a targeted approach in these patients. A real-time department level genomic testing ordering system in combination with an evidence-based flagging system for potentially actionable alterations could help address these shortcomings.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/secondary , Molecular Targeted Therapy/standards , Mutation , Pancreatic Neoplasms/pathology , Precision Medicine , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , Combined Modality Therapy , Feasibility Studies , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Lymphatic Metastasis , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy , Prognosis , Retrospective Studies , Time Factors
2.
Oncotarget ; 11(21): 1929-1941, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32523648

ABSTRACT

PURPOSE: Metformin combined with the mTOR inhibitor rapamycin showed potential synergistic anti-tumor activity in preclinical studies in pancreatic ductal adenocarcinoma (PDA). This phase 1b study (NCT02048384) was conducted to evaluate the feasibility and activity of metformin +/- rapamycin in the maintenance setting for unselected patients with metastatic PDA (mPDA) treated with chemotherapy. MATERIALS AND METHODS: Eligible patients with stable or responding mPDA after ≥ 6 months on chemotherapy were randomized 1:1 to metformin alone (Arm A) or with rapamycin (Arm B), stratified by prior treatment with FOLFIRINOX. Fluorodeoxyglucose (FDG) PET scans and peripheral blood mononuclear cells were obtained for exploratory analyses. RESULTS: 22 subjects (11 per arm) received treatment per protocol. Median PFS/OS were 3.5 and 13.2 months respectively, with 2 year OS rate of 37%; there were no differences between arms. No responses were observed by RECIST; however, decreases in FDG avidity and/or CA19-9 were observed in several long-term survivors. Treatment related adverse events of Grade ≥ 3 occurred in 0% vs 27% of patients in Arm A vs B and were asymptomatic hematologic or electrolyte abnormalities that were not clinically significant. Improved survival was associated with low baseline neutrophil: lymphocyte ratio, baseline lack of assessable disease by PET, and greater expansion of dendritic cells following treatment. CONCLUSIONS: Metformin +/- rapamycin maintenance for mPDA was well-tolerated and several patients achieved stable disease associated with exceptionally long survival. Further prospective studies are needed to clarify the role of these agents in the maintenance setting and to enhance patient selection for such approaches.

3.
Clin Cancer Res ; 26(19): 5129-5139, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32591464

ABSTRACT

PURPOSE: This phase II study tested granulocyte-macrophage colony-stimulating factor (GM-CSF)-allogeneic pancreatic tumor cells (GVAX) and ipilimumab in metastatic pancreatic ductal adenocarcinoma (PDA) in the maintenance setting. PATIENTS AND METHODS: Patients with PDA who were treated with front-line chemotherapy consisting of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) in the metastatic setting and had ongoing response or stable disease after 8-12 doses were eligible. Patients were randomized 1:1 to treatment with GVAX and ipilimumab given every 3 weeks for four doses then every 8 weeks (Arm A) or to FOLFIRINOX continuation (Arm B). The primary objective was to compare overall survival (OS) between the two arms. RESULTS: Eighty-two patients were included in the final analysis (Arm A: 40; Arm B: 42). The study was stopped for futility after interim analysis. Median OS was 9.38 months [95% confidence interval (CI), 5.0-12.2] for Arm A and 14.7 months (95% CI, 11.6-20.0) for Arm B (HR, 1.75; P = 0.019). Using immune-related response criteria, two partial responses (5.7%) were observed in Arm A and four (13.8%) in Arm B. GVAX + ipilimumab promoted T-cell differentiation into effector memory phenotypes both in the periphery and in the tumor microenvironment and increased M1 macrophages in the tumor. CONCLUSIONS: GVAX and ipilimumab maintenance therapy did not improve OS over continuation of chemotherapy and resulted in a numerically inferior survival in metastatic PDA. However, clinical responses and biological effects on immune cells were observed. Further study of novel combinations in the maintenance treatment of metastatic PDA is feasible.


Subject(s)
Adenocarcinoma/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Combined Modality Therapy , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Irinotecan/administration & dosage , Irinotecan/adverse effects , Leucovorin/administration & dosage , Leucovorin/adverse effects , Macrophages/drug effects , Macrophages/immunology , Male , Middle Aged , Neoplasm Metastasis , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology
4.
Oncology (Williston Park) ; 32(10): 494-8, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30334238

ABSTRACT

This article provides guidance on how to incorporate immunotherapy into colorectal cancer care. We review the identification of appropriate patients, available immunotherapy agents, benefits and risks of therapy, and how to overcome potential barriers to access. With recent US Food and Drug Administration approvals of two programmed death 1 (PD-1) receptor inhibitors for use in colorectal cancer, and dual therapy with combined nivolumab (PD-1) and ipilimumab (cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] blocking antibody), it is vital that oncology providers be aware of how and when to implement these therapy options.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Colorectal Neoplasms/drug therapy , Ipilimumab/therapeutic use , Nivolumab/therapeutic use , Colorectal Neoplasms/genetics , Humans , Immunotherapy , Microsatellite Instability , Patient Selection
5.
Science ; 357(6349): 409-413, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28596308

ABSTRACT

The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Neoplastic Syndromes, Hereditary/immunology , Neoplastic Syndromes, Hereditary/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Mismatch Repair , Disease-Free Survival , Female , Humans , Male , Middle Aged , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/mortality , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...