Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nature ; 569(7754): 131-135, 2019 05.
Article in English | MEDLINE | ID: mdl-30996350

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis largely owing to inefficient diagnosis and tenacious drug resistance. Activation of pancreatic stellate cells (PSCs) and consequent development of dense stroma are prominent features accounting for this aggressive biology1,2. The reciprocal interplay between PSCs and pancreatic cancer cells (PCCs) not only enhances tumour progression and metastasis but also sustains their own activation, facilitating a vicious cycle to exacerbate tumorigenesis and drug resistance3-7. Furthermore, PSC activation occurs very early during PDAC tumorigenesis8-10, and activated PSCs comprise a substantial fraction of the tumour mass, providing a rich source of readily detectable factors. Therefore, we hypothesized that the communication between PSCs and PCCs could be an exploitable target to develop effective strategies for PDAC therapy and diagnosis. Here, starting with aĀ systematic proteomic investigation of secreted disease mediators and underlying molecular mechanisms, we reveal that leukaemia inhibitory factor (LIF) is a key paracrine factor from activated PSCs acting on cancer cells. Both pharmacologic LIF blockade and genetic Lifr deletion markedly slow tumour progression and augment theĀ efficacy of chemotherapy to prolong survival of PDAC mouse models, mainly by modulating cancer cell differentiation and epithelial-mesenchymal transition status. Moreover, in both mouse models and human PDAC, aberrant production of LIF in the pancreas is restricted to pathological conditions and correlates with PDAC pathogenesis, and changes in the levels ofĀ circulating LIF correlate well with tumour response to therapy. Collectively, these findings reveal a function of LIF in PDAC tumorigenesis, and suggest its translational potential as an attractive therapeutic target and circulating marker. Our studies underscore how a better understanding of cell-cell communication within the tumour microenvironment can suggest novel strategies for cancer therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Leukemia Inhibitory Factor/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Paracrine Communication , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/diagnosis , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Humans , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/blood , Male , Mass Spectrometry , Mice , Pancreatic Neoplasms/diagnosis , Paracrine Communication/drug effects , Receptors, OSM-LIF/deficiency , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism , Tumor Microenvironment
3.
Br J Cancer ; 122(11): 1630-1637, 2020 05.
Article in English | MEDLINE | ID: mdl-32238921

ABSTRACT

BACKGROUND: In this first-in-human, Phase 1 study of a microRNA-based cancer therapy, the recommended Phase 2 dose (RP2D) of MRX34, a liposomal mimic of microRNA-34a (miR-34a), was determined and evaluated in patients with advanced solid tumours. METHODS: Adults with various solid tumours refractory to standard treatments were enrolled in 3 + 3 dose-escalation cohorts and, following RP2D determination, expansion cohorts. MRX34, with oral dexamethasone premedication, was given intravenously daily for 5 days in 3-week cycles. RESULTS: Common all-cause adverse events observed in 85 patients enrolled included fever (% all grade/G3: 72/4), chills (53/14), fatigue (51/9), back/neck pain (36/5), nausea (36/1) and dyspnoea (25/4). The RP2D was 70 mg/m2 for hepatocellular carcinoma (HCC) and 93 mg/m2 for non-HCC cancers. Pharmacodynamic results showed delivery of miR-34a to tumours, and dose-dependent modulation of target gene expression in white blood cells. Three patients had PRs and 16 had SD lasting ≥4 cycles (median, 19 weeks, range, 11-55). CONCLUSION: MRX34 treatment with dexamethasone premedication demonstrated a manageable toxicity profile in most patients and some clinical activity. Although the trial was closed early due to serious immune-mediated AEs that resulted in four patient deaths, dose-dependent modulation of relevant target genes provides proof-of-concept for miRNA-based cancer therapy. CLINICAL TRIAL REGISTRATION: NCT01829971.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , MicroRNAs/administration & dosage , MicroRNAs/adverse effects , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacokinetics , Female , Humans , Liposomes/adverse effects , Liposomes/pharmacokinetics , Male , Maximum Tolerated Dose , MicroRNAs/pharmacokinetics , Middle Aged , Nanoparticles/administration & dosage , Nanoparticles/adverse effects
4.
Invest New Drugs ; 33(1): 148-58, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25260842

ABSTRACT

BACKGROUND: This phase Ib study used a parallel, multi-arm design to examine tasisulam-sodium (hereafter tasisulam), a drug with complex pharmacology, combined with standard chemotherapies in patients with advanced solid tumors, with the ultimate goal of accelerating drug development. METHODS: Patients received escalating doses of tasisulam (3 + 3 schema; target Cmax 300-400 Āµg/mL) every 28 days plus 1,000 mg/m(2) gemcitabine HCl (days 1 and 15), 60 mg/m(2) docetaxel, 200 mg/m(2)/day temozolomide, 75 mg/m(2) cisplatin, or 150 mg/day erlotinib. Following dose-escalation, patients were enrolled into specific tumor subtype arms, chosen based on the established activity of the standard agent. Because tasisulam is highly albumin-bound, patients in the tumor-specific confirmation arms were dosed targeting specific albumin-corrected exposure ranges (AUCalb) identified during dose-escalation (3,500 h*Āµg/mL [75th percentile] for docetaxel, temozolomide, and cisplatin; 4,000 h*Āµg/mL for gemcitabine and erlotinib). RESULTS: A total of 234 patients were enrolled. The safety profile of tasisulam with standard chemotherapies was sufficient to allow enrollment into the dose-confirmation phase in all arms. The primary dose-limiting toxicities were hematologic (thrombocytopenia and neutropenia). The most common grade ≥3 drug-related treatment-emergent adverse event was neutropenia, with the highest incidence in the docetaxel arm. CONCLUSIONS: The multi-arm design allowed the efficient determination of the maximum tolerated dose of tasisulam across multiple combinations, and a preliminary characterization of pharmacokinetics, safety, and potential efficacy. Although enrollment into all planned groups was not completed due to termination of compound development, these data support the feasibility of this approach for accelerated cancer drug development, even for drugs with complex pharmacology.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzamides/administration & dosage , Benzamides/adverse effects , Benzamides/blood , Benzamides/pharmacokinetics , Cisplatin/administration & dosage , Cisplatin/adverse effects , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Docetaxel , Erlotinib Hydrochloride , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Quinazolines/administration & dosage , Quinazolines/adverse effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Taxoids/administration & dosage , Taxoids/adverse effects , Temozolomide , Young Adult , Gemcitabine
5.
Cancer ; 119(16): 3043-51, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23775486

ABSTRACT

BACKGROUND: The efficacy of afatinib, an irreversible ErbB Family Blocker, was evaluated in patients who had 1 of 4 categories of solid tumors with epidermal growth factor receptor/human epidermal growth factor receptor 2 (EGFR/HER2) gene amplification or EGFR-activating mutations. METHODS: Patients with previously treated but ErbB inhibitor-naive esophagogastric, biliary tract, urothelial tract, or gynecologic cancers (lung cancers were excluded) harboring EGFR/HER2 gene amplification or high polysomy were identified by fluorescence in situ hybridization (FISH). Tumors were also screened for EGFR mutations. The primary endpoint was the objective response rate; secondary endpoints included the clinical benefit rate, pharmacokinetics, and safety. RESULTS: Of 385 prescreened patients, 38 had FISH-positive tumors (10 with EGFR amplification and 29 with HER2 amplification or high polysomy [1 tumor had EGFR/HER2 high polysomy]; none had EGFR-activating mutations), and 20 patients received treatment with afatinib 50 mg daily. The objective response rate was 5% (1 of 20 patients), and the best objective response included 1 complete response. Eight patients experienced stable disease. The most frequently reported adverse events were diarrhea, rash, and decreased appetite. The trial closed early because of slow recruitment. CONCLUSIONS: Single-agent afatinib activity was limited, yet encouraging, in selected tumors that were screened prospectively for target activation. The implementation of a biomarker-driven approach using a low-frequency biomarker for patient selection across multiple tumor types can be challenging.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Quinazolines/therapeutic use , Adult , Afatinib , Aged , Aged, 80 and over , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasms/enzymology , Neoplasms/genetics , Prospective Studies , Quinazolines/pharmacokinetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Treatment Outcome
6.
Cancer Chemother Pharmacol ; 88(3): 485-497, 2021 09.
Article in English | MEDLINE | ID: mdl-34097100

ABSTRACT

PURPOSE: Trifluridine/tipiracil (FTD/TPI) is approved for advanced colorectal and gastric/gastroesophageal cancer; however, data in patients with renal impairment (RI) are limited. This phase I study evaluated FTD/TPI in patients with advanced solid tumors and varying degrees of RI to develop dosing guidance. METHODS: Patients were enrolled into normal renal function (CrCl ≥ 90Ā mL/min), mild RI (CrCl 60-89Ā mL/min), or moderate RI (CrCl 30-59Ā mL/min) cohorts and administered the recommended FTD/TPI dose (35Ā mg/m2 twice daily, days 1-5 and 8-12; 28-day cycle). Based on interim pharmacokinetics/safety data, patients with severe RI (CrCl 15-29Ā mL/min) were enrolled and received FTD/TPI 20Ā mg/m2 twice daily. RESULTS: Forty-three patients (normal renal function [n = 12]; mild RI [n = 12]; moderate RI [n = 11]; severe RI [n = 8]) were enrolled and treated. At steady state, compared to values in patients with normal renal function, FTD area under the curve (AUC) was not significantly different in patients with RI, but TPI AUC was significantly higher and increased with RI severity. FTD/TPI safety profile was consistent with prior experience, but grade ≥ 3 adverse events (AEs) were more frequent in the RI cohorts (83.3% [mild], 90.9% [moderate], 75.0% [severe], and normal [50.0%]). Hematologic AEs (anemia and neutropenia) were more frequent with RI. Overall, seven patients discontinued because of unrelated, nonhematologic AEs. CONCLUSION: FTD/TPI is safe and tolerable at the recommended 35Ā mg/m2 dose in patients with mild/moderate RI and at the reduced 20Ā mg/m2 dose in patients with severe RI. TRIAL REGISTRATION: NCT02301117, registration date: November 21, 2014.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Kidney Diseases/physiopathology , Neoplasms/drug therapy , Pyrrolidines/administration & dosage , Thymine/administration & dosage , Trifluridine/administration & dosage , Adult , Aged , Aged, 80 and over , Anemia/chemically induced , Anemia/epidemiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Cohort Studies , Drug Combinations , Female , Humans , Male , Middle Aged , Neutropenia/chemically induced , Neutropenia/epidemiology , Pyrrolidines/adverse effects , Pyrrolidines/pharmacokinetics , Severity of Illness Index , Thymine/adverse effects , Thymine/pharmacokinetics , Trifluridine/adverse effects , Trifluridine/pharmacokinetics
7.
Proc (Bayl Univ Med Cent) ; 31(2): 210-212, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29706823

ABSTRACT

Carcinosarcoma is a rare subtype of pancreatic neoplasm including both carcinomatous and sarcomatous components. Fewer than 30 cases have been reported to the Surveillance, Epidemiology, and End Results Program database. Given such rarity, definitive treatment guidelines are not well defined. We report a case of pancreatic carcinosarcoma diagnosed in our institution, review tumor clinicopathological characteristics, and describe our medical and surgical management strategy.

8.
J Clin Pharmacol ; 57(6): 751-759, 2017 06.
Article in English | MEDLINE | ID: mdl-28070894

ABSTRACT

TAS-102 (trifluridine/tipiracil) is composed of an antineoplastic thymidine-based nucleoside analogue trifluridine (FTD), and a thymidine phosphorylase inhibitor, tipiracil (TPI), at a molar ratio of 1:0.5 (weight ratio, 1:0.471). A phase 1 study evaluated relative bioavailability of TAS-102 tablets compared with an oral solution containing equivalent amounts of FTD and TPI. In an open-label, 2-sequence, 3-period, crossover bioavailability study (part 1), patients 18 years or older with advanced solid tumors were randomized to receive TAS-102 tablets (60 mg; 3 Ɨ 20-mg tablets) on day 1 and TAS-102 oral solution (60 mg) on days 8 and 15, or the opposite sequence. In an extension (part 2), all patients received TAS-102 tablets. Of the 46 patients treated in the crossover study, 38 were evaluable in the crossover bioavailability pharmacokinetic population. For area under the concentration-time curve (AUC)0-∞ and AUC0-last for FTD and TPI, and maximum plasma concentration (Cmax ) for TPI, the 90% confidence intervals (CIs) of the geometric mean ratios were within the 0.80 to 1.25 boundary for demonstration of bioequivalence; for FTD Cmax , the lower limit of the 90%CI was 0.786. The most frequently reported treatment-related grade 3 or 4 adverse events were neutropenia (7 patients) and decreased neutrophil count (3 patients). Although the lower limit of the 90%CI for the geometric mean ratio of FTD Cmax was slightly lower than 0.80, the bioavailability of the TAS-102 tablet is considered clinically similar to that of a TAS-102 oral solution. TAS-102 was well tolerated in this population of patients with advanced solid tumors.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasms/metabolism , Trifluridine/pharmacokinetics , Uracil/analogs & derivatives , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Biological Availability , Cross-Over Studies , Drug Combinations , Female , Humans , Male , Middle Aged , Neoplasms/blood , Neoplasms/drug therapy , Pyrrolidines , Solutions , Tablets , Thymine , Treatment Outcome , Trifluridine/administration & dosage , Trifluridine/adverse effects , Trifluridine/therapeutic use , Uracil/administration & dosage , Uracil/adverse effects , Uracil/pharmacokinetics , Uracil/therapeutic use
9.
Clin Ther ; 38(4): 778-93, 2016 04.
Article in English | MEDLINE | ID: mdl-27041409

ABSTRACT

PURPOSE: Tenosynovial giant cell tumor (TGCT), a rare locally aggressive neoplasm of the synovium of joints and tendon sheaths, is associated with joint destruction, inflammation, pain, and swelling, in part due to colony-stimulating factor 1 receptor-bearing macrophages recruited to the tumor by genetic elevation of colony-stimulating factor 1 activity. The most common treatment is surgery, although promising pharmacologic treatments are in development. Patient-reported outcome (PRO) instruments are critical end points in demonstrating the clinical relevance of standard oncologic outcome measures and the overall impact of novel pharmacologic therapies in nonmalignant neoplastic conditions such as TGCT. The content validity of PROs relevant to patients with TGCT has not been formally investigated, and instruments to evaluate such outcomes do not exist for this condition. METHODS: PRO instruments of potential relevance were evaluated by using a literature review and by clinical and PRO experts.Ā Patients with TGCT were recruited through clinical sites and the InternetĀ for participationĀ in qualitative research interviews to identify predominant symptoms and to test the relevance and content validity of several PRO measures. Select PRO measures were included in a Phase I clinical trial, and preliminary results of the PRO end points are reported descriptively. FINDINGS: Of the 22 subjects who participated in qualitative interviews, 73% were female, and their mean age was 42.5 years (range, 27-56 years). The TGCTs (19 diffuse and 3 localized) were located in the knee (nĀ =Ā 15), hip (nĀ =Ā 3), ankle (nĀ =Ā 2), elbow (nĀ =Ā 1), and forearm (nĀ =Ā 1). The most common symptoms cited were pain (82%), swelling (86%), stiffness (73%), reduced range of motion (64%), and joint instability (64%), which were consistent with clinical expert input and with the content of instruments chosen by PRO experts. The worst pain numeric rating scale, Patient Reported Outcomes Measurement Information System physical functioning items, and the Western Ontario and McMaster Universities Osteoarthritis Index, as well as a worst stiffness numeric rating scale developed for TGCT, were confirmed as meaningful measures of TGCT patient symptoms and were well understood in qualitative interviews. Results from the Phase I trial showed trends of improvement in both pain and stiffness over time. IMPLICATIONS: This study is the first to gather information directly from patients with TGCT regarding their symptom experiences. Pain, stiffness, and physical functioning are important treatment outcomes in patients with TGCT. We have identified content-valid PRO measures of these concepts, which are included in an ongoing Phase III TGCT clinical trial with pexidartinib (PLX3397) (NCT02371369).


Subject(s)
Giant Cell Tumor of Tendon Sheath , Patient Reported Outcome Measures , Adult , Female , Humans , Male , Middle Aged , Treatment Outcome
10.
Clin Cancer Res ; 10(10): 3333-41, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15161687

ABSTRACT

In preclinical tumor models, inhibition of nuclear factor-kappaB (NF-kappaB) has been associated with increased sensitivity to chemotherapeutic agents such as irinotecan (CPT-11). This is based on the fact that a variety of chemotherapy agents such as CPT-11 activate NF-kappaB to result in the expression of genes such as c-IAP1 and c-IAP2 that might be responsible for the inhibition of chemotherapy-induced apoptosis. In this study, RNA interference [small interfering RNA (siRNA)] was used to down-regulate the NF-kappaB p65 subunit in the HCT116 colon cancer cell line, and its role, in the presence and absence of CPT-11, was assessed on cell growth and apoptosis. Reduction of endogenous p65 by siRNA treatment significantly impaired CPT-11-mediated NF-kappaB activation, enhanced apoptosis, and reduced colony formation in soft agar. Furthermore, the in vivo administration of p65 siRNA reduced HCT116 tumor formation in xenograft models in the presence but not the absence of CPT-11 administration. These data indicate that the administration of siRNA directed against the p65 subunit of NF-kappaB can effectively enhance in vitro and in vivo sensitivity to chemotherapeutic agents.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Down-Regulation , NF-kappa B/biosynthesis , RNA Interference , Agar/chemistry , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Baculoviral IAP Repeat-Containing 3 Protein , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Genes, Reporter , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Irinotecan , Mice , NF-kappa B/chemistry , NF-kappa B/genetics , Neoplasm Transplantation , Oligonucleotides/chemistry , Proteins/metabolism , RNA/chemistry , RNA, Small Interfering/metabolism , Time Factors , Transfection , Ubiquitin-Protein Ligases
11.
Cancer Chemother Pharmacol ; 75(1): 183-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25417902

ABSTRACT

PURPOSE: To identify the maximum tolerated dose (MTD) and recommended Phase II dose of MEK/AKT inhibitor combination of trametinib and afuresertib. PATIENTS AND METHODS: Eligibility criteria were advanced solid tumors, 18 years or older, Eastern Cooperative Oncology Group performance status 0 or 1, and adequate organ function. Exclusion criteria included Type 1 diabetes, active GI disease, leptomeningeal disease, or current evidence/risk of retinal venous occlusion/central serous retinopathy. Clinical safety parameters and response were evaluated and analyzed. RESULTS: Twenty patients were enrolled. Dose-limiting toxicities (Grade 2 esophagitis; Grade 3 aspartate aminotransferase increased, mucosal inflammation and hypokalemia) were reported at starting dose (1.5 mg trametinib/50 mg afuresertib once daily continuously), exceeding the MTD. Subsequent de-escalation cohorts (1.5 mg/25 mg or 1.0 mg/50 mg trametinib/afuresertib) were defined as MTDs for continuous dosing. Intermittent dosing schedule [1.5 mg trametinib (continuous)/50 mg afuresertib (Days 1-10 every 28 days)] was evaluated and considered tolerable. No patients were enrolled in Phase II. The most common adverse events reported (≥10 % of all patients) included: diarrhea (60 %), dermatitis acneiform (55 %), maculo-papular rash (45 %), fatigue (30 %), dry skin (25 %), nausea (25 %), dyspnea (20 %), and vomiting (20 %). One partial response (BRAF wild-type melanoma) was reported; four patients had stable disease as best response. CONCLUSION: Continuous daily dosing of trametinib/afuresertib combination was poorly tolerated. Evaluation of intermittent dose schedule showed greater tolerability. Given the interest in combination treatment regimens of MAPK and PI3K/AKT pathway inhibitors, further study of intermittent dose schedule or combination of trametinib with more selective inhibitors may be warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Multiple Myeloma/drug therapy , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Pyrazoles/adverse effects , Pyridones/adverse effects , Pyrimidinones/adverse effects , Thiophenes/adverse effects , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cohort Studies , Dose-Response Relationship, Drug , Early Termination of Clinical Trials , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/enzymology , Neoplasms/blood , Neoplasms/enzymology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Pyrimidinones/therapeutic use , Thiophenes/administration & dosage , Thiophenes/pharmacokinetics , Thiophenes/therapeutic use , Tumor Burden/drug effects
12.
Oncology (Williston Park) ; 18(14 Suppl 14): 46-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15685835

ABSTRACT

The combination of irinotecan (Camptosar), epirubicin, and capecitabine (Xeloda) has shown an acceptable toxicity profile. In this open-label phase I study, irinotecan was administered IV at a fixed dose of 250 mg/m2 on day 1 in combination with capecitabine at a fixed dose of 1500 mg/m2 for days 2 to 7 and epirubicin starting at a dose of 40 mg/m2 and escalating by 10 mg/m2 in cohorts of three patients for those with metastatic adenocarcinomas. With the addition of granulocyte colony-stimulating factor (G-CSF [Neupogen]) to the regimen, patients received epirubicin at clinically relevant doses after dose-escalation. Results of the topoisomerase activity will be reported with the final results of this phase I study. The dose-limiting toxicity has not yet been reached. This combination regimen in patients with upper gastrointestinal malignancies and breast cancer will be investigated as part of phase II studies, once the dose-limiting toxicity is determined. The appropriate sequencing of the regimen to maximize clinical efficacy will also be determined.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Deoxycytidine/analogs & derivatives , Adenocarcinoma/secondary , Antibiotics, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Capecitabine , Deoxycytidine/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Epirubicin/administration & dosage , Female , Fluorouracil/analogs & derivatives , Humans , Irinotecan , Male , Treatment Outcome
13.
Cancer Chemother Pharmacol ; 74(5): 917-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25149088

ABSTRACT

PURPOSE: The purpose of the study was to evaluate safety and determine the maximum tolerated dose (MTD) of MEDI-575, a fully human monoclonal antibody that selectively binds to platelet-derived growth factor receptor-α (PDGFRα), in patients with advanced solid tumors. METHODS: This phase I multicenter, open-label, single-arm study enrolled adults in a 3 + 3 dose escalation design to receive MEDI-575 (3, 6, 9, 12, or 15 mg/kg) once weekly (QW) until toxicity or disease progression occurred. One 0.5-mg/kg dose was given before the first dose in the 3-mg/kg cohort to determine pharmacokinetics (PK) and pharmacodynamics under unsaturated conditions. After completion of dose escalation in the QW cohorts, patients were enrolled in two additional cohorts and received MEDI-575 25 or 35 mg/kg every 3 weeks (Q3W). Secondary measures included assessments of PK, immunogenicity, and antitumor activity. RESULTS: A total of 35 patients received MEDI-575 QW (n = 23) or Q3W (n = 12). Most treatment-related adverse events were grade 1 or 2 in severity across all dose levels, with fatigue (n = 12) and nausea (n = 8) being reported most frequently. With no reports of dose-limiting toxicities (DLTs), the MTD was not reached. MEDI-575 exhibited a nonlinear PK profile and increased plasma platelet-derived growth factor-AA levels in a dose-dependent manner with limited immunogenicity. Stable disease was reported as the best tumor response in 9 of 29 evaluable patients; however, no objective responses were reported. CONCLUSION: Administration of MEDI-575 QW or Q3W resulted in a favorable safety profile, including a lack of DLTs, but without evidence of antitumor activity in patients with refractory solid tumors.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Neoplasms/drug therapy , Receptor, Platelet-Derived Growth Factor alpha/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Area Under Curve , Cohort Studies , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Fatigue/chemically induced , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Neoplasms/metabolism , Neoplasms/pathology , Survival Analysis , Treatment Outcome , Vomiting/chemically induced
14.
Cancer Chemother Pharmacol ; 73(4): 695-702, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24488322

ABSTRACT

PURPOSE: Figitumumab (CP-751,871) is a human IgG2 monoclonal antibody that binds and down-regulates insulin-like growth factor receptor-1 (IGF-1R) and inhibits activation of this receptor by IGF-1 and IGF-2. This nonrandomized, open-label, single-arm, phase II trial evaluated the antitumor activity and safety of figitumumab in patients with metastatic colorectal cancer that was refractory to ≥2 systemic therapies. METHODS: Cohorts A and B received intravenous figitumumab 20 and 30 mg/kg in 3-week cycles, respectively. Both received loading doses (20 or 30 mg/kg) on days 1 and 2 of cycle 1. The primary endpoint was 6-month survival (null hypothesis for each cohort, H0: p6 mo surv = 0.45). Secondary endpoints included progression-free survival (PFS), overall survival (OS), objective response, safety, and pharmacokinetics. RESULTS: A total of 168 patients (Cohort A, n = 85; Cohort B, n = 83) received figitumumab. Estimated 6-month survival was 49.4 % (95 % CI 38.8-60.0) in Cohort A and 44.1 % (95 % CI 33.4-54.9) in Cohort B. Median OS was 5.8 and 5.6 months, respectively; median PFS was 1.4 months in both cohorts. No objective partial or complete responses occurred. The respective rates of treatment discontinuation due to treatment-related adverse events (AEs) were 5 and 7 %. The most common grade 3/4 nonhematologic AEs in both cohorts were hyperglycemia and asthenia. No grade 4 hematologic laboratory abnormalities occurred. Most deaths were reported as due to progressive disease; none were due to figitumumab. CONCLUSION: Six-month survival data do not support further study of figitumumab 20 or 30 mg/kg in this patient population.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cohort Studies , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Survival Analysis
15.
J Thorac Oncol ; 8(11): 1425-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24084441

ABSTRACT

INTRODUCTION: On the basis of the promising activity of cetuximab and radiation therapy for head and neck cancers, we evaluated the efficacy of this regimen followed by surgery in patients with resectable esophageal cancer. This was a phase II, open-label, single-arm, multicenter study of patients with potentially resectable esophageal cancer. METHODS: Patients received two weekly doses of cetuximab followed by weekly cetuximab combined with radiation therapy for 6 weeks. After a 6- to 8-week rest, patients' primary tumor was resected. The main objective was to evaluate pathologic complete response (pCR) rate in the primary tumor after cetuximab and radiation therapy. RESULTS: Thirty-nine patients completed the study. Most patients were men (93%), median age was 64 years, performance status was 0 to 1 (95%), patients had a histology of adenocarcinoma (78%), and tumors were located in the esophagus (63%). Grade 3 toxicities in more than 5% of patients included dysphagia (17%), anorexia and dehydration (7%), and dyspnea, fatigue, hypernatremia (5%). Grade 5 aspiration occurred in 2% (1 patient). Four patients died, two from disease progression, one from aspiration pneumonia postsurgery, and one from septic shock. Thirty-one patients (76%) underwent esophagectomy. The pCR rate was 36.6% by intention-to-treat and 48% for patients who underwent esophagectomy. The pCR by histology was 6 of 9 (67%) for squamous cell carcinomas and 9 of 32 (28%) for adenocarcinoma. Earlier-stage disease was associated with increased pCR (IIA 70%, IIB 29%, III 28%). CONCLUSIONS: Cetuximab and radiation therapy results in a pCR rate that seems at least comparable with that of chemotherapy and radiation therapy. This regimen may be better tolerated than preoperative chemotherapy and radiation therapy in patients with resectable esophageal cancers.


Subject(s)
Adenocarcinoma/therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/therapy , Esophagogastric Junction/pathology , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Cetuximab , Combined Modality Therapy , Esophageal Neoplasms/pathology , Esophagectomy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Radiotherapy Dosage , Remission Induction
16.
Clin Cancer Res ; 16(6): 1812-23, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215551

ABSTRACT

PURPOSE: Myeloid-derived suppressor cells (MDSC) are one of the major factors responsible for immune suppression in cancer. Therefore, it would be important to identify effective therapeutic means to modulate these cells. EXPERIMENTAL DESIGN: We evaluated the effect of the synthetic triterpenoid C-28 methyl ester of 2-cyano-3,12-dioxooleana-1,9,-dien-28-oic acid (CDDO-Me; bardoxolone methyl) in MC38 colon carcinoma, Lewis lung carcinoma, and EL-4 thymoma mouse tumor models, as well as blood samples from patients with renal cell cancer and soft tissue sarcoma. Samples were also analyzed from patients with pancreatic cancer treated with CDDO-Me in combination with gemcitabine. RESULTS: CDDO-Me at concentrations of 25 to 100 nmol/L completely abrogated immune suppressive activity of MDSC in vitro. CDDO-Me reduced reactive oxygen species in MDSCs but did not affect their viability or the levels of nitric oxide and arginase. Treatment of tumor-bearing mice with CDDO-Me did not affect the proportion of MDSCs in the spleens but eliminated their suppressive activity. This effect was independent of antitumor activity. CDDO-Me treatment decreased tumor growth in mice. Experiments with severe combined immunodeficient-beige mice indicated that this effect was largely mediated by the immune system. CDDO-Me substantially enhanced the antitumor effect of a cancer vaccines. Treatment of pancreatic cancer patients with CDDO-Me did not affect the number of MDSCs in peripheral blood but significantly improved the immune response. CONCLUSIONS: CDDO-Me abrogated the immune suppressive effect of MDSCs and improved immune responses in tumor-bearing mice and cancer patients. It may represent an attractive therapeutic option by enhancing the effect of cancer immunotherapy.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Oleanolic Acid/analogs & derivatives , Aged , Aged, 80 and over , Animals , Arginase/metabolism , Clinical Trials, Phase I as Topic , Dendritic Cells/pathology , Dendritic Cells/transplantation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Middle Aged , Myeloid Cells/pathology , Myeloid Cells/transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nitric Oxide/metabolism , Oleanolic Acid/pharmacology , Prognosis , Reactive Oxygen Species/metabolism
17.
Am J Clin Oncol ; 31(3): 219-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18525298

ABSTRACT

OBJECTIVES: Preclinical studies using sequences of topoisomerase I and II inhibitors suggested synergism; preliminary clinical studies, resulting in enhanced antitumor responses, confirm this in selected malignancies. This study determined the maximum-tolerated dose (MTD), toxicity, and pharmacokinetics of irinotecan (CPT-11), capecitabine, and epirubicin in patients with metastatic adenocarcinoma of lung, breast, or gastrointestinal tract. Correlation of topoisomerase IIbeta was also done. METHODS: Eligibility criteria included the following: documented adenocarcinoma of the lung, breast, or gastrointestinal tract, <3 prior chemotherapy regimens, Eastern Cooperative Oncology Group (ECOG) performance status 0 to 2, age > or =18 years, adequate organ function, and signed informed consent. Irinotecan was administered at 250 mg/m2 intravenously day 1 every 21-day cycle, but was reduced to 180 mg/m2 in cohort 2 due to toxicity. Capecitabine was administered at 750 mg/m2 twice daily days 2 to 14 in cohort 1 but only on days 2 to 7 from cohort 2 due to early neutropenia and to allow for prophylactic granulocyte colony-stimulating factor (GCSF) support. Epirubicin was administered at 40 mg/m2 in cohort 1, but reduced to 30 mg/m2 in cohort 2, then reescalated until the MTD was reached. RESULTS: Toxicity was assessed in 21 patients; response was assessed in 17 patients. The most common grade 3 to 4 toxicities included neutropenia (57.1%) and anemia (28.6%). The MTD of epirubicin was 50 mg/m2. In evaluable patients, there were 2 partial responses (11.8%) and 13 stable disease (76.5%); these correlated well with topoisomerase IIbeta. CONCLUSIONS: The recommended doses for phase II studies: irinotecan 180 mg/m2 day 1, epirubicin 50 mg/m2 day 2, and capecitabine 750 mg/m2 twice daily days 2 to 7 of each 21-day cycle. This combination is reasonably active and warrants evaluation in the phase II setting.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Epirubicin/administration & dosage , Gastrointestinal Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Topoisomerase II Inhibitors , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Administration, Oral , Adult , Aged , Alopecia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Capecitabine , DNA Topoisomerases, Type I/blood , DNA Topoisomerases, Type II/blood , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Drug Resistance, Neoplasm , Drug Synergism , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Gastrointestinal Diseases/chemically induced , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematologic Diseases/chemically induced , Humans , Infusions, Intravenous , Irinotecan , Male , Maximum Tolerated Dose , Middle Aged , Treatment Outcome
18.
Support Cancer Ther ; 3(4): 247-50, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-18632501

ABSTRACT

Bevacizumab is the first successful example of targeting the vasculature for the treatment of solid tumors. Although generally well tolerated in combination with different chemotherapy regimens, bevacizumab has side effects that are unique to this class of agents. In this review, we discuss the side effects associated with bevacizumab and potential treatments to ameliorate these toxicities.

19.
Acta Pharmacol Sin ; 27(10): 1375-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17007746

ABSTRACT

AIM: Clinical treatment of solid tumors with docetaxel, flavopiridol, or 5-fluorouracil (5-FU) often encounters undesirable side effects and drug resistance. This study aims to evaluate the potential role of combination therapy with docetaxel, flavopiridol, or 5-FU in modulating chemosensitivity and better understand how they might be used clinically. METHODS: HCT116 colon cancer cells were treated with docetaxel, flavopiridol, and 5-FU in several different administrative schedules in vitro, either sequentially or simultaneously. Cell survival was measured by MTT assay. The activity of caspase-3 was determined by caspase-3 assays and the soft agar colony assay was used to test the colony formation of HCT116 cells in soft agar. We also established xenograft models to extend in vitro observations to an in vivo system. RESULTS: The maximum cytotoxicity was found when human colon cancer HCT116 cells were treated with docetaxel for 1 h followed by flavopiridol for 24 h and 5-FU for another 24 h. This sequential combination therapy not only inhibits tumor cell growth more strongly compared to other combination therapies but also significantly reduces colony formation in soft agar and augments apoptosis of HCT116 cells. Sequencing of docetaxel followed 1 h later by flavopiridol, followed 24 h later by 5-FU in xenograft models, also resulted in delayed tumor growth and higher survival rate. CONCLUSION: These results highlight the importance of an administrative schedule when combining docetaxel with flavopiridol and 5-FU, providing a rationale explanation for its development in clinical trials.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/pathology , Adenocarcinoma/drug therapy , Animals , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Docetaxel , Female , Flavonoids/administration & dosage , Fluorouracil/administration & dosage , HCT116 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Piperidines/administration & dosage , Taxoids/administration & dosage
20.
Acta Pharmacol Sin ; 26(7): 780-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15960883

ABSTRACT

NF-kappaB, a family of related transcription factors, has been a focus of intense scientific research during the past decade. Multiple stimuli, both extracellular and intracellular, lead to its activation. The NF-kappaB pathway regulates expression of a diverse array of genes involved in different biological processes. Various pathological states are characterized by the dysregulated NF-kappaB pathway. Recently, NF-kappaB activation has been connected with multiple aspects of oncogenesis and serves as an important mechanism to regulate cell survival in response to chemotherapy by activating different genes that inhibit apoptosis. Several methods of inhibiting NF-kappaB activation, such as antisense oligonucleotides, proteosome inhibitors and RNA interference (RNAi) are currently under investigation. RNAi represents a powerful tool to better define the role of specific genes in different signal transduction pathways and has recently been used to define the function of genes that regulate the NF-kappaB pathway. This review discusses the emerging role of RNAi to dissect the function of regulatory factors in the NF-kappaB pathway and its potential use as a targeted therapy.


Subject(s)
NF-kappa B , RNA Interference , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Animals , Enzyme Activation , Humans , Interleukin-12/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/physiology , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL