Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 372(26): 2509-20, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26028255

ABSTRACT

BACKGROUND: Somatic mutations have the potential to encode "non-self" immunogenic antigens. We hypothesized that tumors with a large number of somatic mutations due to mismatch-repair defects may be susceptible to immune checkpoint blockade. METHODS: We conducted a phase 2 study to evaluate the clinical activity of pembrolizumab, an anti-programmed death 1 immune checkpoint inhibitor, in 41 patients with progressive metastatic carcinoma with or without mismatch-repair deficiency. Pembrolizumab was administered intravenously at a dose of 10 mg per kilogram of body weight every 14 days in patients with mismatch repair-deficient colorectal cancers, patients with mismatch repair-proficient colorectal cancers, and patients with mismatch repair-deficient cancers that were not colorectal. The coprimary end points were the immune-related objective response rate and the 20-week immune-related progression-free survival rate. RESULTS: The immune-related objective response rate and immune-related progression-free survival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mismatch repair-deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of 18 patients) for mismatch repair-proficient colorectal cancers. The median progression-free survival and overall survival were not reached in the cohort with mismatch repair-deficient colorectal cancer but were 2.2 and 5.0 months, respectively, in the cohort with mismatch repair-proficient colorectal cancer (hazard ratio for disease progression or death, 0.10 [P<0.001], and hazard ratio for death, 0.22 [P=0.05]). Patients with mismatch repair-deficient noncolorectal cancer had responses similar to those of patients with mismatch repair-deficient colorectal cancer (immune-related objective response rate, 71% [5 of 7 patients]; immune-related progression-free survival rate, 67% [4 of 6 patients]). Whole-exome sequencing revealed a mean of 1782 somatic mutations per tumor in mismatch repair-deficient tumors, as compared with 73 in mismatch repair-proficient tumors (P=0.007), and high somatic mutation loads were associated with prolonged progression-free survival (P=0.02). CONCLUSIONS: This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab. (Funded by Johns Hopkins University and others; ClinicalTrials.gov number, NCT01876511.).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , DNA Mismatch Repair , Neoplasm Metastasis/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis/genetics
2.
J Natl Compr Canc Netw ; 13(10): 1225-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26483062

ABSTRACT

BACKGROUND: The immune system plays an important role in cancer surveillance and therapy. Chemoradiation can cause severe treatment-related lymphopenia (TRL) (<500 cells/mm3) that is associated with reduced survival. MATERIALS AND METHODS: Data from 4 independent solid tumor studies on serial lymphocyte counts, prognostic factors, treatment, and survival were collected and analyzed. The data set included 297 patients with newly diagnosed malignant glioma (N=96), resected pancreatic cancer (N=53), unresectable pancreatic cancer (N=101), and non-small cell lung cancer (N=47). RESULTS: Pretreatment lymphocyte counts were normal in 83% of the patient population, and no patient had severe baseline lymphopenia. Two months after initiating chemoradiation, 43% developed severe and persistent lymphopenia (P=.001). An increased risk for death was attributable to TRL in each cancer cohort (gliomas: hazard rate [HR], 1.8; 95% CI, 1.13-2.87; resected pancreas: HR, 2.2; 95% CI, 1.17-4.12; unresected pancreas: HR, 2.9; 95% CI, 1.53-5.42; and lung: HR, 1.7; 95% CI, 0.8-3.61) and in the entire study population regardless of pathologic findings (HR, 2.1; 95% CI, 1.54-2.78; P<.0001). Severe TRL was observed in more than 40% of patients 2 months after initiating chemoradiation, regardless of histology or chemotherapy regimen, and was independently associated with shorter survival from tumor progression. CONCLUSIONS: Increased attention and research should be focused on the cause, prevention, and reversal of this unintended consequence of cancer treatment that seems to be related to survival in patients with solid tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/complications , Chemoradiotherapy/adverse effects , Glioma/complications , Lymphopenia/pathology , Pancreatic Neoplasms/complications , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Disease-Free Survival , Female , Glioma/drug therapy , Glioma/radiotherapy , Humans , Lymphocyte Count , Lymphopenia/chemically induced , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy
3.
Nat Rev Cancer ; 5(6): 459-67, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15905855

ABSTRACT

The identification of key signalling pathways involved in immune-system regulation, along with the development of early pancreatic tumours in mouse models have provided new opportunities for pancreatic cancer treatment and prevention. Immunotherapy for pancreatic cancer is one approach that is at a crucial crossroads, as therapeutics that are designed to target pancreatic-cancer-associated antigens and regulatory signalling molecules are entering clinical trials.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Immunotherapy/trends , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Clinical Trials as Topic , Humans , Immunotherapy/methods , Pancreatic Neoplasms/prevention & control , Signal Transduction
4.
Ann Surg ; 253(2): 328-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21217520

ABSTRACT

PURPOSE: Surgical resection provides the only possibility of cure for pancreas cancer. A standard adjuvant approach has not been established. We tested the safety and efficacy of a granulocyte-macrophage colony-stimulating factor (GM-CSF)-based immunotherapy administered in patients with resected pancreatic adenocarcinoma. PATIENTS AND METHODS: A single institution phase II study of 60 patients with resected pancreatic adenocarcinoma was performed. Each immunotherapy treatment consisted of a total of 5 × 108 GM-CSF-secreting cells distributed equally among 3 lymph node regions. The first immunotherapy treatment was administered 8 to 10 weeks after surgical resection. Subsequently, patients received 5-FU based chemoradiation. Patients who remained disease-free after completion of chemoradiotherapy received treatments 2 to 4, each 1 month apart. A fifth and final booster was administered 6 months after the fourth immunotherapy. The primary endpoint was disease free survival and secondary endpoints were overall survival and toxicity, and the induction of mesothelin specific T cell responses. RESULTS: The median disease-free survival is 17.3 months (95% CI, 14.6-22.8) with median survival of 24.8 months (95% CI, 21.2-31.6). The administration of immunotherapy was well tolerated. In addition, the post-immunotherapy induction of mesothelin-specific CD8+ T cells in HLA-A1+ and HLA-A2+patients correlates with disease-free survival. CONCLUSIONS: An immunotherapy approach integrated with chemoradiation is safe and demonstrates an overall survival that compares favorably with published data for resected pancreas cancer. These data suggest additional boost immunotherapies given at regular intervals beyond 1 year postsurgery should be tested in future studies, and provide the rationale for conducting a multicenter phase II study.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Pancreatic Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cancer Vaccines/radiation effects , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Combined Modality Therapy , Disease-Free Survival , Female , GPI-Linked Proteins/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Hypersensitivity, Delayed/immunology , Male , Mesothelin , Middle Aged , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Survival Rate , T-Lymphocytes/immunology , Transfection
5.
Clin Cancer Res ; 14(5): 1455-63, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316569

ABSTRACT

PURPOSE: The combination of chemotherapy and immunotherapy has not been examined in patients with advanced pancreatic cancer. We conducted a study of two granulocyte macrophage colony-stimulating factor-secreting pancreatic cancer cell lines (CG8020/CG2505) as immunotherapy administered alone or in sequence with cyclophosphamide in patients with advanced pancreatic cancer. EXPERIMENTAL DESIGN: This was an open-label study with two cohorts: cohort A, 30 patients administered a maximum of six doses of CG8020/CG2505 at 21-day intervals; and cohort B, 20 patients administered 250 mg/m(2) of cyclophosphamide i.v. 1 day before the same immunotherapy given as in cohort A. The primary objective was to evaluate safety and duration of immunity. Secondary objectives included time to disease progression and median overall survival. RESULTS: The administration of CG8020/CG2505 alone or in sequence with cyclophosphamide showed minimal treatment-related toxicity. Median survival values in cohort A and cohort B were 2.3 and 4.3 months, respectively. CD8(+) T-cell responses to HLA class I-restricted mesothelin epitopes were identified predominantly in patients treated with cyclophosphamide + CG8020/CG2505 immunotherapy. CONCLUSION: Granulocyte macrophage colony-stimulating factor-secreting pancreatic cancer cell lines CG8020/CG2505 alone or in sequence with cyclophosphamide showed minimal treatment-related toxicity in patients with advanced pancreatic cancer. Also, mesothelin-specific T-cell responses were detected/enhanced in some patients treated with CG8020/CG2505 immunotherapy. In addition, cyclophosphamide-modulated immunotherapy resulted in median survival in a gemcitabine-resistant population similar to chemotherapy alone. These findings support additional investigation of cyclophosphamide with CG8020/CG2505 immunotherapy in patients with advanced pancreatic cancer.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents, Alkylating/therapeutic use , Cyclophosphamide/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunotherapy , Pancreatic Neoplasms/therapy , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Cohort Studies , Combined Modality Therapy , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Pilot Projects , Survival Rate , Tissue Distribution
6.
Clin Cancer Res ; 14(17): 5602-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765554

ABSTRACT

PURPOSE: To determine the maximum tolerated dose (MTD), characterize the principal toxicities, and assess the pharmacokinetics of EKB-569, an oral selective irreversible inhibitor of the epidermal growth factor receptor tyrosine kinase, in combination with capecitabine in patients with advanced colorectal cancer. EXPERIMENTAL DESIGN: Patients were treated with EKB-569 daily for 21 days and capecitabine twice daily for 14 days of a 21-day cycle. The dose levels of EKB-569 (mg/day) and capecitabine (mg/m(2) twice daily) assessed were 25/750, 50/750, 50/1,000 and 75/1,000. An expanded cohort was enrolled at the MTD to better study toxicity and efficacy. Samples of plasma were collected to characterize the pharmacokinetics of the agents. Treatment efficacy was assessed every other cycle. RESULTS: A total of 37 patients, the majority of whom had prior chemotherapy, received a total of 163 cycles of treatment. Twenty patients were treated at the MTD, 50 mg EKB-569, daily and 1,000 mg/m(2) capecitabine twice daily. Dose-limiting toxicities were diarrhea and rash. No patients had complete or partial responses but 48% had stable disease. The conversion of capecitabine to 5-fluorouracil was higher for the combination of EKB-569 and capecitabine (321+/-151 ng*h/mL) than for capecitabine alone (176+/-62 ng*hours/mL; P=0.0037). CONCLUSION: In advanced colorectal cancer, 50 mg EKB-569 daily can be safely combined with 1,000 mg/m(2) capecitabine twice a day. A statistically significant increase in plasma levels of 5-fluorouracil for the combination of EKB-569 and capecitabine may be due to the single-dose versus multiple-dose exposure difference, variability in exposure or a potential drug interaction.


Subject(s)
Aminoquinolines/administration & dosage , Aniline Compounds/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Adult , Aged , Aged, 80 and over , Aminoquinolines/adverse effects , Aminoquinolines/pharmacokinetics , Aniline Compounds/adverse effects , Aniline Compounds/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Capecitabine , Colorectal Neoplasms/drug therapy , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Drug Administration Schedule , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Humans , Male , Middle Aged
7.
Clin Cancer Res ; 25(18): 5493-5502, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31126960

ABSTRACT

PURPOSE: Limited options exist for patients with advanced pancreatic cancer progressing after 1 or more lines of therapy. A phase II study in patients with previously treated metastatic pancreatic cancer showed that combining GVAX pancreas (granulocyte-macrophage colony-stimulating factor-secreting allogeneic pancreatic tumor cells) with cyclophosphamide (Cy) and CRS-207 (live, attenuated Listeria monocytogenes expressing mesothelin) resulted in median overall survival (OS) of 6.1 months, which compares favorably with historical OS achieved with chemotherapy. In the current study, we compared Cy/GVAX + CRS-207, CRS-207 alone, and standard chemotherapy in a three-arm, randomized, controlled phase IIb trial. PATIENTS AND METHODS: Patients with previously treated metastatic pancreatic adenocarcinoma were randomized 1:1:1 to receive Cy/GVAX + CRS-207 (arm A), CRS-207 (arm B), or physician's choice of single-agent chemotherapy (arm C). The primary cohort included patients who had failed ≥2 prior lines of therapy, including gemcitabine. The primary objective compared OS between arms A and C in the primary cohort. The second-line cohort included patients who had received 1 prior line of therapy. Additional objectives included OS between all treatment arms, safety, and tumor responses. RESULTS: The study did not meet its primary efficacy endpoint. At the final study analysis, median OS [95% confidence interval (CI)] in the primary cohort (N = 213) was 3.7 (2.9-5.3), 5.4 (4.2-6.4), and 4.6 (4.2-5.7) months in arms A, B, and C, respectively, showing no significant difference between arm A and arm C [P = not significant (NS), HR = 1.17; 95% CI, 0.84-1.64]. The most frequently reported adverse events in all treatment groups were chills, pyrexia, fatigue, and nausea. No treatment-related deaths occurred. CONCLUSIONS: The combination of Cy/GVAX + CRS-207 did not improve survival over chemotherapy. (ClinicalTrials.gov ID: NCT02004262)See related commentary by Salas-Benito et al., p. 5435.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Adult , Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Granulocyte-Macrophage Colony-Stimulating Factor , Humans
8.
J Chromatogr B Analyt Technol Biomed Life Sci ; 869(1-2): 142-5, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18534927

ABSTRACT

A liquid chromatography/tandem mass spectrometric (LC/MS/MS) assay was developed for the quantitative determination of salirasib (S-trans,trans-farnesylthiosalicylic acid, FTS) in human plasma. Sample pretreatment involved liquid-liquid extraction with methyl t-butyl ether of 0.5-mL aliquots of lithium heparin plasma spiked with the internal standard, S-trans,trans-5-fluoro-farnesylthiosalicylic acid (5-F-FTS). Separation was achieved on Waters X-Terra C(18) (50 mm x 2.1 mm i.d., 3.5 microm) at room temperature using isocratic elution with acetonitrile/10 mM ammonium acetate buffer mobile phase (80:20, v/v) containing 0.1% formic acid at a flow rate of 0.20 mL/min. Detection was performed using electrospray MS/MS by monitoring the ion transitions from m/z 357.2-->153.0 (salirasib) and m/z 375.1-->138.8 (5-F-FTS). Calibration curves were linear in the concentration range of 1-1000 ng/mL. A 5000 ng/mL sample that was diluted 1:10 (v/v) with plasma was accurately quantitated. The values for both within day and between day precision and accuracy were well within the generally accepted criteria for analytical method (<8.0%). This assay was subsequently used for the determination of salirasib concentrations in plasma of cancer patients after oral administration of salirasib at a dose of 400 mg.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Farnesol/analogs & derivatives , Salicylates/blood , Tandem Mass Spectrometry/methods , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/isolation & purification , Farnesol/administration & dosage , Farnesol/blood , Farnesol/isolation & purification , Humans , Salicylates/administration & dosage , Salicylates/isolation & purification
9.
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
10.
Mol Cancer Ther ; 4(11): 1645-52, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16275985

ABSTRACT

Recent advances in our understanding of the complex signaling pathways involved in immune system regulation, along with analyses of genetic differences between tumors and their normal cellular counterparts, have accelerated development of immune-based strategies for cancer treatment and prevention. More clinically relevant animal models have shown that successful immune-based strategies will require the integration of interventions that target specific tumor antigens with regulators of the antitumor immune response. Immunotherapy for cancer is at a critical crossroad, as therapeutics designed to target cancer-associated antigens and regulatory signaling molecules enter clinical trials. We outline here a paradigm for early-stage clinical development of immunotherapy combinations that use vaccines to drive tumor antigen-specific responses while simultaneously targeting immune regulatory pathways.


Subject(s)
Immunotherapy/methods , Neoplasms/genetics , Neoplasms/microbiology , Neoplasms/therapy , Antigens, Neoplasm/metabolism , Clinical Trials as Topic , Disease Progression , Humans , Immune System , Molecular Biology , Signal Transduction
11.
Methods Mol Med ; 103: 299-327, 2005.
Article in English | MEDLINE | ID: mdl-15542914

ABSTRACT

The management of patients with pancreatic cancer is a multidisciplinary approach that presents enormous challenges to the clinician. Overall 5-yr survival for all patients remains < 3%. Symptoms of early pancreas cancer are nonspecific. As such, only a fraction of patients are candidates for surgery. While surgical resection provides the only curative option, most patients will develop tumor recurrence and die of their disease. To date, the clinical benefits of chemotherapy and radiation therapy have been important but have led to modest improvements. Tumor vaccines have the potential to specifically target the needle of pancreas cancer cells amidst the haystack of normal tissue. The discovery of pancreas tumor-specific antigens and the subsequent ability to harness this technology has become an area of intense interest for tumor immunologists and clinicians alike. Without knowledge of specific antigen targets, the whole tumor cell represents the best source of immunizing antigens. This chapter will focus on the development of whole tumor cell vaccine strategies for pancreas cancer.


Subject(s)
Cancer Vaccines , Cytokines/immunology , Pancreatic Neoplasms/immunology , Antigens, Neoplasm/immunology , Humans , Immunotherapy/methods , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Survival Analysis
12.
J Gastrointest Cancer ; 46(1): 5-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25451139

ABSTRACT

PURPOSE: CNS metastasis of pancreatic cancer is extremely rare, although systemic metastasis is very common. We present eight such cases with various forms of nervous system involvement. METHODS: Data was gathered from chart review of 800 patients with pancreatic cancer treated between 2004 and 2012 of which eight patients are described with CNS metastases. RESULTS: The median age of patients was 61.5 years and the median time to develop CNS metastasis was 29 months. Interestingly, two patients had no other sites of metastasis. The treatment modalities tried included resection followed by radiation, resection alone, or whole brain radiation.


Subject(s)
Adenocarcinoma/pathology , Central Nervous System Neoplasms/secondary , Pancreatic Neoplasms/pathology , Aged , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms
13.
J Clin Oncol ; 33(16): 1745-53, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25918295

ABSTRACT

The balance between tumor-promoting and tumor-suppressing immune responses and the difference between them ultimately determine whether a cancer escapes immune recognition mechanisms. Defining the complex relationships between the tumor itself, the tumor environment, and the immune system has been critical in facilitating the development of successful immunotherapies. This review explores the role of oncogenes in inducing cancer-associated inflammation, the local and systemic factors that lead to immune suppression, and immunotherapy approaches to overcome immune privilege.


Subject(s)
Gastrointestinal Neoplasms/therapy , Immunotherapy , Animals , Antibodies/therapeutic use , Cancer Vaccines/therapeutic use , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/immunology , Humans , Immunotherapy/methods , Immunotherapy, Adoptive , Inflammation Mediators/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Oncogenes/genetics , Risk Factors , Treatment Outcome , Tumor Escape , Tumor Microenvironment
15.
Methods Mol Biol ; 980: 175-203, 2013.
Article in English | MEDLINE | ID: mdl-23359154

ABSTRACT

Management of patients with pancreatic cancer is a multidisciplinary approach that presents enormous challenges to the clinician. Overall 5-year survival for all patients remains <3%. Symptoms of early pancreas cancer are nonspecific. As such, only a fraction of patients are candidates for surgery. While surgical resection provides the only curative option, most patients will develop tumor recurrence and die of their disease. To date, the clinical benefits of chemotherapy and radiation therapy have been important but have led to modest improvements. Tumor vaccines have the potential to specifically target the needle of pancreas cancer cells amidst the haystack of normal tissue. The discovery of pancreas tumor-specific antigens and the subsequent ability to harness this technology has become an area of intense interest for tumor immunologists and clinicians alike. Without knowledge of specific antigen targets, the whole tumor cell represents the best source of immunizing antigens. This chapter will focus on the development of whole tumor cell vaccine strategies for pancreas cancer.


Subject(s)
Cancer Vaccines/immunology , Cytokines/immunology , Pancreatic Neoplasms/immunology , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cytokines/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Immunotherapy , Mice , NIH 3T3 Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Retroviridae/genetics , Transduction, Genetic
16.
Rare Tumors ; 3(1): e5, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21464878

ABSTRACT

Primary pancreatic small cell carcinoma (SCC) is rare, with just over 30 cases reported in the literature. Only 7 of these patients underwent surgical resection with a median survival of 6 months. Prognosis of SCC is therefore considered to be poor, and the role of adjuvant therapy is uncertain. Here we report two institutions' experience with resectable pancreatic SCC. Six patients with pancreatic SCC treated at the Johns Hopkins Hospital (4 patients) and the Mayo Clinic (2 patients) were identified from prospectively collected pancreatic cancer databases and re-reviewed by pathology. All six patients underwent a pancreaticoduodenectomy. Clinicopathologic data were analyzed, and the literature on pancreatic SCC was reviewed. Median age at diagnosis was 50 years (range 27-60). All six tumors arose in the head of the pancreas. Median tumor size was 3 cm, and all cases had positive lymph nodes except for one patient who only had five nodes sampled. There were no perioperative deaths and three patients had at least one postoperative complication. All six patients received adjuvant therapy, five of whom were given combined modality treatment with radiation, cisplatin, and etoposide. Median survival was 20 months with a range of 9-173 months. The patient who lived for 9 months received chemotherapy only, while the patient who lived for 173 months was given chemoradiation with cisplatin and etoposide and represents the longest reported survival time from pancreatic SCC to date. Pancreatic SCC is an extremely rare form of cancer with a poor prognosis. Patients in this surgical series showed favorable survival rates when compared to prior reports of both resected and unresectable SCC. Cisplatin and etoposide appears to be the preferred chemotherapy regimen, although its efficacy remains uncertain, as does the role of combined modality treatment with radiation.

17.
Radiat Oncol ; 6: 126, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21951377

ABSTRACT

BACKGROUND: The role of adjuvant chemoradiation therapy for ampullary carcinoma is unknown. Previous literature suggests that certain populations with high risk factors for recurrence may benefit from adjuvant chemoradiation. We combined the experience of two institutions to better delineate which patients may benefit from adjuvant chemoradiation. METHODS: Patients who underwent curative surgery for ampullary carcinoma at the Johns Hopkins Hospital (n=290; 1992-2007) and at the Mayo Clinic (n=130; 1977-2005) were reviewed. Patients with <60 days of follow-up, metastatic disease at surgery, or insufficient pathologic data were excluded. The final combined study consisted of 186 patients (n=104 Johns Hopkins, n=82 Mayo). Most patients received 5-FU based chemoradiation with conformal radiation. Cox proportional hazards models were used for survival analysis. RESULTS: Median overall-survival was 39.9 months with 2- and 5-year survival rates of 62.4% and 39.1%. On univariate analysis, adverse prognostic factors for overall survival included T3/T4 stage disease (RR=1.86, p=0.002), node positive status (RR=3.18, p<0.001), and poor histological grade (RR=1.69, p=0.011). Patients who received adjuvant chemoradiation (n=66) vs. surgery alone (n=120) showed a higher rate of T3/T4 stage disease (57.6% vs. 30.8%, P<0.001), lymph node involvement (72.7% vs. 30.0%, P<0.001), and close or positive margins (4.6% vs. 0.0%, P=0.019). Five year survival rates among node negative and node positive patients were 58.7% and 18.4% respectively. When compared with surgery alone, use of adjuvant chemoradiation improved survival among node positive patients (mOS 32.1 vs. 15.7 mos, 5 yr OS: 27.5% vs. 5.9%; RR=0.47, P=0.004). After adjusting for adverse prognostic factors on multivariate analysis, patients treated with adjuvant chemoradiation demonstrated a significant survival benefit (RR=0.40, P<0.001). Disease relapse occurred in 37.1% of all patients, most commonly metastatic disease in the liver or peritoneum. CONCLUSIONS: Node-positive patients with resected ampullary adenocarcinoma may benefit from 5-FU based adjuvant chemoradiation. Since a significant proportion of patients develop metastatic disease, there is a need for more effective systemic treatment.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Chemoradiotherapy/methods , Chemotherapy, Adjuvant/methods , Common Bile Duct Neoplasms/drug therapy , Common Bile Duct Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Ampulla of Vater/pathology , Antineoplastic Agents/therapeutic use , Female , Fluorouracil/therapeutic use , Humans , Male , Maryland , Middle Aged , Minnesota , Neoplasm Metastasis , Proportional Hazards Models , Radiotherapy, Conformal/methods , Retrospective Studies , Risk Factors , Treatment Outcome
19.
J Clin Oncol ; 27(16): 2697-704, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19380450

ABSTRACT

PURPOSE: Positron emission tomography (PET) with [(18)F]fluorodeoxyglucose (FDG-PET) has increasingly been used to evaluate the efficacy of anticancer agents. We investigated the role of FDG-PET as a predictive marker for response to mammalian target of rapamycin (mTOR) inhibition in advanced solid tumor patients and in murine xenograft models. PATIENTS AND METHODS: Thirty-four rapamycin-treated patients with assessable baseline and treatment FDG-PET and computed tomography scans were analyzed from two clinical trials. Clinical response was evaluated according to Response Evaluation Criteria in Solid Tumors, and FDG-PET response was evaluated by quantitative changes and European Organisation for Research and Treatment of Cancer (EORTC) criteria. Six murine xenograft tumor models were treated with temsirolimus. Small animal FDG-PET scans were performed at baseline and during treatment. The tumors were analyzed for the expression of pAkt and GLUT1. RESULTS: Fifty percent of patients with increased FDG-PET uptake and 46% with decreased uptake had progressive disease (PD). No objective response was observed. By EORTC criteria, the sensitivity of progressive metabolic disease on FDG-PET in predicting PD was 19%. Preclinical studies demonstrated similar findings, and FDG-PET response correlated with pAkt activation and plasma membrane GLUT1 expression. CONCLUSION: FDG-PET is not predictive of proliferative response to mTOR inhibitor therapy in both clinical and preclinical studies. Our findings suggest that mTOR inhibitors suppress the formation of mTORC2 complex, resulting in the inhibition of Akt and glycolysis independent of proliferation in a subset of tumors. Changes in FDG-PET may be a pharmacodynamic marker for Akt activation during mTOR inhibitor therapy. FDG-PET may be used to identify patients with persistent Akt activation following mTOR inhibitor therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Fluorodeoxyglucose F18 , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Positron-Emission Tomography , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Radiopharmaceuticals , Sirolimus/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Female , Glucose Transporter Type 1/metabolism , Humans , Mice , Mice, Nude , Neoplasms/enzymology , Neoplasms/pathology , Patient Selection , Phosphorylation , Predictive Value of Tests , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tomography, X-Ray Computed , Treatment Outcome , Xenograft Model Antitumor Assays
20.
Cancer Chemother Pharmacol ; 63(1): 181-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18379785

ABSTRACT

PURPOSE: To determine the maximum tolerated dose (MTD) of trabectedin plus gemcitabine administered on a weekly schedule in patients with advanced solid tumors. METHODS: Patients with ECOG performance status 0-1 and adequate organ function were enrolled. On days 1, 8, and 15 of a 28-day cycle, patients received gemcitabine (starting dose, 800 mg/m(2)) followed by trabectedin (starting dose, 0.3 mg/m(2)). Strict liver function test treatment criteria were employed to avoid hepatic toxicity seen in previous trabectedin studies. Plasma samples were collected during cycles 1 and 2 for pharmacokinetic analyses. RESULTS: Fifteen patients received >or=1 dose, with a median of two treatment cycles (range 1-10). The most common drug-related toxicity was hepatic. Dose reductions were required for trabectedin in four (27%) patients and gemcitabine in six (40%) patients. Cycle delays/dose holds were required in 11 (73%) patients and doses above trabectedin 0.4 mg/m(2) and gemcitabine 1,000 mg/m(2), which is the recommended phase II dose, were not feasible. Seven patients maintained stable disease after two cycles. Gemcitabine and trabectedin pharmacokinetics were not altered substantially with concomitant administration. CONCLUSIONS: Given the lack of pharmacokinetic interaction and potential efficacy of trabectedin and gemcitabine combination therapy, further study is warranted with alternate schedules.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Chemical and Drug Induced Liver Injury/etiology , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Dioxoles/administration & dosage , Dioxoles/adverse effects , Dioxoles/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematologic Diseases/chemically induced , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/adverse effects , Tetrahydroisoquinolines/pharmacokinetics , Trabectedin , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL