Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
ESMO Open ; 8(5): 101618, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37625194

ABSTRACT

Synovial sarcoma (SS) is a rare and aggressive disease that accounts for 5%-10% of all soft tissue sarcomas. Although it can occur at any age, it typically affects younger adults and children, with a peak incidence in the fourth decade of life. In >95% of cases, the oncogenic driver is a translocation between chromosomes X and 18 that leads to the formation of the SS18::SSX fusion oncogenes. Early and accurate diagnosis is often a challenge; optimal outcomes are achieved by referral to a specialist center for diagnosis and management by a multidisciplinary team as soon as SS is suspected. Surgery with or without radiotherapy and/or chemotherapy can be effective in localized disease, especially in children. However, the prognosis in the advanced stages is poor, with treatment strategies that have relied heavily on traditional cytotoxic chemotherapies. Therefore, there is an unmet need for novel effective management strategies for advanced disease. An improved understanding of disease pathology and its molecular basis has paved the way for novel targeted agents and immunotherapies that are being investigated in clinical trials. This review provides an overview of the epidemiology and characteristics of SS in children and adults, as well as the patient journey from diagnosis to treatment. Current and future management strategies, focusing particularly on the potential of immunotherapies to improve clinical outcomes, are also summarized.

2.
Ann Oncol ; 34(7): 615-625, 2023 07.
Article in English | MEDLINE | ID: mdl-37105265

ABSTRACT

BACKGROUND: The current treatment paradigm of imatinib-resistant metastatic gastrointestinal stromal tumor (GIST) does not incorporate KIT/PDGFRA genotypes in therapeutic drug sequencing, except for PDGFRA exon 18-mutant GIST that is indicated for avapritinib treatment. Here, circulating tumor DNA (ctDNA) sequencing was used to analyze plasma samples prospectively collected in the phase III VOYAGER trial to understand how the KIT/PDGFRA mutational landscape contributes to tyrosine kinase inhibitor (TKI) resistance and to determine its clinical validity and utility. PATIENTS AND METHODS: VOYAGER (N = 476) compared avapritinib with regorafenib in patients with KIT/PDGFRA-mutant GIST previously treated with imatinib and one or two additional TKIs (NCT03465722). KIT/PDGFRA ctDNA mutation profiling of plasma samples at baseline and end of treatment was assessed with 74-gene Guardant360® CDx. Molecular subgroups were determined and correlated with outcomes. RESULTS: A total of 386/476 patients with KIT/PDGFRA-mutant tumors underwent baseline (pre-trial treatment) ctDNA analysis; 196 received avapritinib and 190 received regorafenib. KIT and PDGFRA mutations were detected in 75.1% and 5.4%, respectively. KIT resistance mutations were found in the activation loop (A-loop; 80.4%) and ATP-binding pocket (ATP-BP; 40.8%); 23.4% had both. An average of 2.6 KIT mutations were detected per patient; 17.2% showed 4-14 different KIT resistance mutations. Of all pathogenic KIT variants, 28.0% were novel, including alterations in exons/codons previously unreported. PDGFRA mutations showed similar patterns. ctDNA-detected KIT ATP-BP mutations negatively prognosticated avapritinib activity, with a median progression-free survival (mPFS) of 1.9 versus 5.6 months for regorafenib. mPFS for regorafenib did not vary regardless of the presence or absence of ATP-BP/A-loop mutants and was greater than mPFS with avapritinib in this population. Secondary KIT ATP-BP pocket mutation variants, particularly V654A, were enriched upon disease progression with avapritinib. CONCLUSIONS: ctDNA sequencing efficiently detects KIT/PDGFRA mutations and prognosticates outcomes in patients with TKI-resistant GIST treated with avapritinib. ctDNA analysis can be used to monitor disease progression and provide more personalized treatment.


Subject(s)
Antineoplastic Agents , Circulating Tumor DNA , Gastrointestinal Stromal Tumors , Humans , Adenosine Triphosphate/therapeutic use , Antineoplastic Agents/therapeutic use , Circulating Tumor DNA/genetics , Disease Progression , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/diagnosis , Imatinib Mesylate , Mutation , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/therapeutic use
3.
Ann Oncol ; 31(11): 1506-1517, 2020 11.
Article in English | MEDLINE | ID: mdl-32891793

ABSTRACT

Sarcomas are a heterogeneous group of malignancies with mesenchymal lineage differentiation. The discovery of neurotrophic tyrosine receptor kinase (NTRK) gene fusions as tissue-agnostic oncogenic drivers has led to new personalized therapies for a subset of patients with sarcoma in the form of tropomyosin receptor kinase (TRK) inhibitors. NTRK gene rearrangements and fusion transcripts can be detected with different molecular pathology techniques, while TRK protein expression can be demonstrated with immunohistochemistry. The rarity and diagnostic complexity of NTRK gene fusions raise a number of questions and challenges for clinicians. To address these challenges, the World Sarcoma Network convened two meetings of expert adult oncologists and pathologists and subsequently developed this article to provide practical guidance on the management of patients with sarcoma harboring NTRK gene fusions. We propose a diagnostic strategy that considers disease stage and histologic and molecular subtypes to facilitate routine testing for TRK expression and subsequent testing for NTRK gene fusions.


Subject(s)
Sarcoma , Tropomyosin , Adult , Gene Fusion , Humans , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors , Receptor, trkA/genetics , Sarcoma/diagnosis , Sarcoma/drug therapy , Sarcoma/genetics
4.
Ann Oncol ; 30(10): 1647-1652, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31373348

ABSTRACT

BACKGROUND: A previous analysis of 113 National Comprehensive Cancer Network® (NCCN®) recommendations reported that NCCN frequently recommends beyond Food and Drug Administration (FDA)-approved indications (44 off-label recommendations) and claimed that the evidence for these recommendations was weak. METHODS: In order to determine the strength of the evidence, we carried out an in-depth re-analysis of the 44 off-label recommendations listed in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). RESULTS: Of the 44 off-label recommendations, 14 were later approved by the FDA and/or are supported by randomized controlled trial (RCT) data. In addition, 13 recommendations were either very minor extrapolations from the FDA label (n = 8) or were actually on-label (n = 5). Of the 17 remaining extrapolations, 8 were for mechanism-based agents applied in rare cancers or subsets with few available treatment options (median response rate = 43%), 7 were based on non-RCT data showing significant efficacy (>50% response rates), and 2 were later removed from the NCCN Guidelines because newer therapies with better activity and/or safety became available. CONCLUSION: Off-label drug use is a frequent component of care for patients with cancer in the United States. Our findings indicate that when the NCCN recommends beyond the FDA-approved indications, the strength of the evidence supporting such recommendations is robust, with a significant subset of these drugs later becoming FDA approved or supported by RCT. Recommendations without RCT data are often for mechanism-based drugs with high response rates in rare cancers or subsets without effective therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Approval , Evidence-Based Medicine , Neoplasms/drug therapy , Off-Label Use/standards , Patient Care Management/standards , Practice Guidelines as Topic/standards , Humans , Neoplasms/pathology , Off-Label Use/legislation & jurisprudence , Off-Label Use/statistics & numerical data , Prognosis , Randomized Controlled Trials as Topic , United States , United States Food and Drug Administration
5.
Ann Oncol ; 29(9): 1995-2002, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30084934

ABSTRACT

Background: Treatment options for soft tissue sarcoma (STS) patients aged ≥65 years (elderly) can be limited by concerns regarding the increased risk of toxicity associated with standard systemic therapies. Trabectedin has demonstrated improved disease control in a phase III trial (ET743-SAR-3007) of patients with advanced liposarcoma or leiomyosarcoma after failure of anthracycline-based chemotherapy. Since previous retrospective analyses have suggested that trabectedin has similar safety and efficacy outcomes regardless of patient age, we carried out a subgroup analysis of the safety and efficacy observed in elderly patients enrolled in this trial. Patients and methods: Patients were randomized 2 : 1 to trabectedin (n = 384) or dacarbazine (n = 193) administered intravenously every-3-weeks. The primary end point was overall survival (OS); secondary end points were progression-free survival (PFS), time-to-progression, objective response rate (ORR), duration of response, symptom severity, and safety. A post hoc analysis was conducted in the elderly patient subgroup. Results: Among 131 (trabectedin = 94; dacarbazine = 37) elderly patients, disease characteristics were well-balanced and consistent with those of the total study population. Treatment exposure was longer in patients treated with trabectedin versus dacarbazine (median four versus two cycles, respectively), with a significantly higher proportion receiving prolonged therapy (≥6 cycles) in the trabectedin arm (43% versus 23%, respectively; P = 0.04). Elderly patients treated with trabectedin showed significantly improved PFS [4.9 versus 1.5 months, respectively; hazard ratio (HR)=0.40; P = 0.0002] but no statistically significant improvement in OS (15.1 versus 8.0 months, respectively; HR = 0.72; P = 0.18) or ORR (9% versus 3%, respectively; P = 0.43). The safety profile for elderly trabectedin-treated patients was comparable to that of the overall trabectedin-treated study population. Conclusions: This subgroup analysis of the elderly population of ET743-SAR-3007 suggests that elderly patients with STS and good performance status can expect clinical benefit from trabectedin similar to that observed in younger patients. Trial registration: www.clinicaltrials.gov, NCT01343277.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/administration & dosage , Leiomyosarcoma/drug therapy , Liposarcoma/drug therapy , Trabectedin/administration & dosage , Administration, Intravenous , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/adverse effects , Drug Administration Schedule , Female , Humans , Kaplan-Meier Estimate , Leiomyosarcoma/mortality , Leiomyosarcoma/pathology , Liposarcoma/mortality , Liposarcoma/pathology , Male , Middle Aged , Progression-Free Survival , Time Factors , Trabectedin/adverse effects , Young Adult
6.
Ann Oncol ; 27(9): 1794-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27371698

ABSTRACT

BACKGROUND: This investigator-initiated trial provided the justification for the phase III GRID study resulting in worldwide regulatory approval of regorafenib as a third-line therapy for patients with metastatic gastrointestinal stromal tumors (GIST). We report the genotype analyses, long-term safety, and activity results from this initial trial of regorafenib in GIST. PATIENTS AND METHODS: The trial was conducted between February 2010 and January 2014, among adult patients with metastatic GIST, after failure of at least imatinib and sunitinib. Patients received regorafenib orally, 160 mg once daily, days 1-21 of a 28-day cycle. Clinical benefit rate (CBR), defined as complete or partial response (PR), or stable disease lasting ≥16 weeks per RECIST 1.1, progression-free survival (PFS), overall survival (OS), long-term safety data, and metabolic response by functional imaging were assessed. RESULTS: Thirty-three patients received at least one dose of regorafenib. The median follow-up was 41 months. CBR was documented in 25 of 33 patients [76%; 95% confidence interval (CI) 58% to 89%], including six PRs. The median PFS was 13.2 months (95% CI 9.2-18.3 months) including four patients who remained progression-free at study closure, each achieving clinical benefit for more than 3 years (range 36.8-43.5 months). The median OS was 25 months (95% CI 13.2-39.1 months). Patients whose tumors harbored a KIT exon 11 mutation demonstrated the longest median PFS (13.4 months), whereas patients with KIT/PDGFRA wild-type, non-SDH-deficient tumors experienced a median 1.6 months PFS (P < 0.0001). Long-term safety profile is consistent with previous reports; hand-foot skin reaction and hypertension were the most common reasons for dose reduction. Notably, regorafenib induced objective responses and durable benefit in SDH-deficient GIST. CONCLUSIONS: Long-term follow-up of patients with metastatic GIST treated with regorafenib suggests particular benefit among patients with primary KIT exon 11 mutations and those with SDH-deficient GIST. Dose modifications are frequently required to manage treatment-related toxicities. CLINICAL TRIAL NUMBER: NCT01068769.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Gastrointestinal Stromal Tumors/drug therapy , Phenylurea Compounds/administration & dosage , Proto-Oncogene Proteins c-kit/genetics , Pyridines/administration & dosage , Adult , Aged , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Genotype , Humans , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/adverse effects , Indoles/administration & dosage , Indoles/adverse effects , Male , Middle Aged , Mutation , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Sunitinib
7.
Ann Oncol ; 24(6): 1703-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23385197

ABSTRACT

BACKGROUND: This expanded access program (EAP) was designed to provide trabectedin access for patients with incurable soft tissue sarcoma (STS) following progression of disease with standard therapy. The outcomes of trial participants accrued over approximately 5 years are reported. PATIENTS AND METHODS: Adult patients with advanced STS of multiple histologies, including leiomyosarcoma and liposarcoma (L-sarcomas), following relapse or disease progression following standard-of-care chemotherapy, were enrolled. Trabectedin treatment cycles (1.5 mg/m(2), intravenously over 24 h) were repeated q21 days. Objective response, overall survival (OS), and safety were evaluated. RESULTS: Of 1895 patients enrolled, 807 (43%) had evaluable objective response data, with stable disease reported in 343 (43%) as best response. L-sarcoma patients exhibited longer, OS compared with other histologies [16.2 months (95% confidence interval (CI) 14.1-19.5) versus 8.4 months (95% CI 7.1-10.7)], and a slightly higher objective response rate [6.9% (95% CI 4.8-9.6) versus 4.0% (95% CI 2.1-6.8)]. The median treatment duration was 70 days representing a median of three treatment cycles; 30% of patients received ≥ 6 cycles. Safety and tolerability in this EAP were consistent with prior clinical trial data. CONCLUSION: Results of this EAP are consistent with previous reports of trabectedin, demonstrating disease control despite a low incidence of objective responses in advanced STS patients after failure of standard chemotherapy. CLINICALTRIALS.GOV: NCT00210665.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Compassionate Use Trials/trends , Dioxoles/administration & dosage , Global Health/trends , Sarcoma/drug therapy , Sarcoma/pathology , Tetrahydroisoquinolines/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Compassionate Use Trials/mortality , Dioxoles/adverse effects , Disease Progression , Female , Health Services Accessibility/trends , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Sarcoma/mortality , Tetrahydroisoquinolines/adverse effects , Trabectedin , Treatment Failure , Treatment Outcome , Young Adult
8.
Ann Oncol ; 24(1): 257-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22910841

ABSTRACT

BACKGROUND: To determine efficacy and safety of bevacizumab, a recombinant humanized antibody against vascular endothelial growth factor (VEGF), in the treatment of metastatic or locally advanced angiosarcoma and epithelioid hemangioendotheliomas. PATIENTS AND METHODS: In this single-arm phase II trial, 32 patients were enrolled and they received bevacizumab 15 mg/kg IV infusion in 21-day cycles. Patients had disease that was deemed not surgically resectable, Eastern Cooperative Oncology Group (ECOG) performance status of ≤1, adequate organ function and had not received any radiation treatment in the last 28 days. RESULTS: Of the 30 patients evaluated for efficacy and toxic effect, four (two angiosarcoma and two epithelioid hemangioendothelioma; 17%) had a partial response. Fifteen patients (11 angiosarcoma and 4 epithelioid hemangioendothelioma; 50%) showed stable disease with a mean time to progression of 26 weeks. Bevacizumab was well tolerated with only one grade 4 adverse event. Expected known toxic effects of the drug were manageable. CONCLUSION: Bevacizumab is an effective and well-tolerated treatment for metastatic or locally advanced angiosarcoma and epithelioid hemangioendotheliomas. Further phase III studies of bevacizumab in combination with other chemotherapeutic agents and/or radiation treatment are warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Hemangioendothelioma, Epithelioid/drug therapy , Hemangiosarcoma/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Bevacizumab , Female , Humans , Male , Middle Aged , Survival Analysis , Treatment Outcome , Young Adult
9.
Ann Oncol ; 24(2): 537-542, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23117071

ABSTRACT

BACKGROUND: The growth modulation index (GMI) is the ratio of time to progression with the nth line (TTP(n)) of therapy to the TTP(n)(-1) with the n-1th line. GMI >1.33 is considered as a sign of activity in phase II trials. PATIENTS AND METHODS: This retrospective analysis evaluated the concordance between the GMI and the efficacy outcomes in 279 patients with advanced soft tissue sarcoma (ASTS) treated with trabectedin 1.5 mg/m² (24-h infusion every 3 weeks) in four phase II trials. RESULTS: One hundred and forty-two (51%) patients received one prior line and 137 ≥ 2 lines. The median TTP(n) was 2.8 months (range 0.2-26.8), whereas the median TTP(n)(-1) was 4.0 months (0.3-79.5). The median GMI was 0.6 (0.0-14.4). Overall, 177 patients (63%) had a GMI <1; 21 (8%) a GMI equal to 1-1.33 and 81 (29%) a GMI >1.33, which correlated with the median overall survival in those patients (9.1, 13.9 and 23.8 months, respectively, P = 0.0005). A high concordance rate between the GMI and response rate (P < 0.0001) and progression-free survival (PFS, P < 0.0001) was observed. Good performance status (PS) was the only factor associated with GMI >1.33 (PS = 0; P < 0.04). CONCLUSIONS: A high GMI was associated with favorable efficacy outcomes in patients treated with trabectedin. Further research is needed to assess GMI as an indicator in this setting.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Cell Proliferation/drug effects , Dioxoles/therapeutic use , Sarcoma/drug therapy , Tetrahydroisoquinolines/therapeutic use , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/therapeutic use , Disease-Free Survival , Doxorubicin/therapeutic use , Female , Humans , Male , Middle Aged , Retrospective Studies , Salvage Therapy , Sarcoma/metabolism , Sarcoma/mortality , Sarcoma/pathology , Trabectedin , Treatment Outcome , Young Adult
11.
Ann Oncol ; 23(5): 1335-1340, 2012 May.
Article in English | MEDLINE | ID: mdl-21994214

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) and desmoid tumors (DTs) are two rare mesenchymal tumor. Anecdotal reports of individuals with both diseases led us to make the hypothesis that the association is a nonrandom event as the probability would be extremely low to observe such cases if they were independent events. PATIENTS AND METHODS: We evaluated the existence of patients with GIST and DT in a large multicenter cohort at 10 institutions in the United States, Australia and Europe. Data on gender, age at diagnosis, KIT, PDGFRA, CTNNB1 mutation status and follow-up time after diagnosis were collected. RESULTS: We identified 28 patients diagnosed with both tumors. DT was diagnosed after GIST in 75% of patients and concomitantly in 21%. In only one case (4%), GIST was diagnosed after DT. KIT or PDGFRA mutations were detected in 12 of 14 GIST, 9 in KIT exon 11, 2 in KIT exon 9 and 1 in PDGFRA. CONCLUSION: A statistical analysis of these 28 cases suggests a nonrandom association between GIST and DT. Further studies may be able to elucidate the underlying biology responsible for this association.


Subject(s)
Fibromatosis, Aggressive/complications , Fibromatosis, Aggressive/epidemiology , Gastrointestinal Stromal Tumors/complications , Gastrointestinal Stromal Tumors/epidemiology , Adult , Aged , Aged, 80 and over , Australia/epidemiology , Cohort Studies , Europe/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Retrospective Studies , United States/epidemiology
12.
Cancer Chemother Pharmacol ; 69(4): 977-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22119758

ABSTRACT

PURPOSE: Patients with advanced GIST following standard imatinib and sunitinib often have good performance status and need additional therapy. This study tested nilotinib, a second-generation tyrosine kinase inhibitor, in patients with advanced GIST refractory to standard therapies. METHODS: This single-center open-label phase II study has a primary objective to determine progression-free survival at 6 months. Using a novel statistical design, 17 patients were to be enrolled; if ≥ 10 were progression free (PF) at 2 months, 19 additional patients would be enrolled. The therapy was considered of benefit if ≥ 13 of 36 patients were PF at 6 months. All patients signed informed consent and entry criteria included normal cardiac function. Exploratory analyses correlating genotype with response were also performed. RESULTS: Thirteen patients were treated; 2 had received agents after imatinib and sunitinib. Treatment was well tolerated with one grade 4 anemia attributed to nilotinib. No measurable responses were observed; median time to progression was 2 months. One patient remained on study with stable disease for 12 months. Mutation testing is available from 10 primary tumors with 7 exon 11 mutations, 1 exon 9 mutation, and 2 without KIT/PDGFR mutations. Two samples from recurrent disease had 2 mutations, both primary exon 11 mutations with an additional exon 17 mutation, including the patient with prolonged stable disease. CONCLUSIONS: Nilotinib was well tolerated in these patients with advanced GIST. Accrual was halted due to insufficient clinical benefit. However, nilotinib may provide benefit to specific subsets of advanced GIST with exon 17 mutations.


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Aged , Benzamides , Disease-Free Survival , Drug Resistance, Neoplasm , Exons , Female , Gastrointestinal Stromal Tumors/enzymology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Indoles/pharmacology , Male , Middle Aged , Mutation , Piperazines/pharmacology , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sunitinib
13.
Ann Oncol ; 22(11): 2523-2529, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21464155

ABSTRACT

BACKGROUND: The objective of the study was to describe patterns of care of patients with gastrointestinal stromal tumors (GISTs) in the United States in the tyrosine kinase inhibitor (TKI) era. PATIENTS AND METHODS: From November 2004 through March 2009, data were collected regarding demographics, diagnostic history, treatment, relapse, and survival of 882 patients with GIST from 122 community and academic medical practices. RESULTS: The most common first-line treatment for the 719 patients presenting with localized GIST was surgery (87%). Use of adjuvant imatinib increased after June 2007; 47% of patients enrolled in the registry considered by the investigator to be at high risk for recurrence received adjuvant imatinib after June 2007 versus 18% before. Overall, 56% of patients received imatinib and 11% received sunitinib. The utilization of targeted therapy increased over time (45% and 0.4% of patients received imatinib and sunitinib, respectively, in 2006 versus 56% and 11%, respectively, in 2009). CONCLUSIONS: These are the first GIST registry data from the TKI era. The use of targeted therapy for GIST has increased in accordance with updated treatment guidelines. Diagnosis of GIST has evolved with increased use of KIT testing. The duration of targeted therapy in the adjuvant therapy setting is similar in community and academic practices.


Subject(s)
Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/therapy , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/therapy , Practice Patterns, Physicians' , Academic Medical Centers , Adolescent , Adult , Aged , Aged, 80 and over , Benzamides , Female , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/epidemiology , Hospitals, Community , Humans , Imatinib Mesylate , Indoles/therapeutic use , Male , Middle Aged , Molecular Targeted Therapy , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Registries , Sunitinib , United States/epidemiology , Young Adult
14.
Br J Cancer ; 99(9): 1415-25, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18841159

ABSTRACT

Inappropriate signalling through the EGFR and ErbB2/HER2 members of the epidermal growth factor family of receptor tyrosine kinases is well recognised as being causally linked to a variety of cancers. Consequently, monoclonal antibodies specific for these receptors have become increasingly important components of effective treatment strategies for cancer. Increasing evidence suggests that ErbB3 plays a critical role in cancer progression and resistance to therapy. We hypothesised that co-targeting the preferred ErbB2/ErbB3 heterodimer with a bispecific single-chain Fv (bs-scFv) antibody would promote increased targeting selectivity over antibodies specific for a single tumour-associated antigen (TAA). In addition, we hypothesised that targeting this important heterodimer could induce a therapeutic effect. Here, we describe the construction and evaluation of the A5-linker-ML3.9 bs-scFv (ALM), an anti-ErbB3/ErbB2 bs-scFv. The A5-linker-ML3.9 bs-scFv exhibits selective targeting of tumour cells in vitro and in vivo that co-express the two target antigens over tumour cells that express only one target antigen or normal cells that express low levels of both antigens. The A5-linker-ML3.9 bs-scFv also exhibits significantly greater in vivo targeting of ErbB2'+'/ErbB3'+' tumours than derivative molecules that contain only one functional arm targeting ErbB2 or ErbB3. Binding of ALM to ErbB2'+'/ErbB3'+' cells mediates inhibition of tumour cell growth in vitro by effectively targeting the therapeutic anti-ErbB3 A5 scFv. This suggests both that ALM could provide the basis for an effective therapeutic agent and that engineered antibodies selected to co-target critical functional pairs of TAAs can enhance the targeting specificity and efficacy of antibody-based cancer therapeutics.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antigens, Neoplasm/immunology , Immunoglobulin Fc Fragments/therapeutic use , Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Animals , Cell Line, Tumor , Dimerization , Humans , Male , Mice , Mice, Inbred ICR , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis
15.
Ann Oncol ; 19(10): 1802-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18497430

ABSTRACT

BACKGROUND: To determine the maximum tolerated dose (MTD), safety, potential pharmacokinetic (PK) interactions, and effect on liver histology of trabectedin in combination with pegylated liposomal doxorubicin (PLD) for advanced malignancies. PATIENTS AND METHODS: Entry criteria for the 36 patients included normal liver function, prior doxorubicin exposure <250 mg/m(2), and normal cardiac function. A 1-h PLD (30 mg/m(2)) infusion was followed immediately by one of six trabectedin doses (0.4, 0.6, 0.75, 0.9, 1.1, and 1.3 mg/m(2)) infused over 3 h, repeated every 21 days until evidence of complete response (CR), disease progression, or unacceptable toxicity. Plasma samples were obtained to assess PK profiles. RESULTS: The MTD of trabectedin was 1.1 mg/m(2). Drug-related grade 3 and 4 toxic effects were neutropenia (31%) and elevated transaminases (31%). Six patients responded (one CR, five partial responses), with an overall response rate of 16.7%, and 14 had stable disease (less than a 50% reduction and less than a 25% increase in the sum of the products of two perpendicular diameters of all measured lesions and the appearance of no new lesions) >4 months (39%). Neither drug had its PK affected significantly by concomitant administration compared with trabectedin and PLD each given as a single agent. CONCLUSION: Trabectedin combined with PLD is generally well tolerated at therapeutic doses of both drugs in pretreated patients with diverse tumor types and appears to provide clinical benefit. These results support the need for additional studies of this combination in appropriate cancer types.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dioxoles/administration & dosage , Dioxoles/adverse effects , Dioxoles/blood , Dioxoles/pharmacokinetics , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/analogs & derivatives , Female , Humans , Male , Middle Aged , Neoplasms/blood , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/adverse effects , Tetrahydroisoquinolines/blood , Tetrahydroisoquinolines/pharmacokinetics , Trabectedin
16.
Sarcoma ; 8(4): 107-11, 2004.
Article in English | MEDLINE | ID: mdl-18521404

ABSTRACT

PATIENTS: Soft tissue sarcomas are uncommon malignancies with few therapeutic options for recurrent or metastatic disease. Dolastatin-10 (Dol-10) is a pentapeptide anti-microtubule agent that binds to tubulin sites distinct from vinca alkaloids. Based on the novel mechanism of action, limited activity of other anti-microtubular agents, and anti-neoplastic activity in pre-clinical screening of Dol-10, this multi-institutional phase II study was conducted to determine the objective response rate of Dol-10 in recurrent or metastatic soft tissue sarcomas that had not been treated with chemotherapy outside of the adjuvant setting. METHODS: Dol-10 was given intravenously at a dose of 400 mug/m(2) and repeated every 21 days. Toxicities were assessed using the Common Toxicity Criteria (version 2.0). Radiographic studies and tumor measurements were repeated every two cycles to assess response [Miller AB, et al. Cancer 1981; 47(1): 207]. RESULTS: Dol-10 was associated with hematological toxicity and with some vascular toxicities. There was no significant gastrointestinal, hepatic or renal toxicity. There was one death on study due to respiratory failure. There were no objective responses in 12 patients treated with Dol-10. DISCUSSION: Based on this phase II trial, further study of Dol-10 on this schedule is not recommended in advanced or metastatic soft tissue sarcomas.

17.
Clin Cancer Res ; 7(5): 1181-91, 2001 May.
Article in English | MEDLINE | ID: mdl-11350882

ABSTRACT

Granulocyte macrophage colony-stimulating factor (GM-CSF) has been shown to be an effective vaccine adjuvant because it enhances antigen processing and presentation by dendritic cells. ALVAC-CEA B7.1 is a canarypox virus encoding the gene for the tumor-associated antigen carcinoembryonic antigen (CEA) and for a T-cell costimulatory molecule, B7.1. After an initial dose escalation phase, this study evaluated vaccination with 4.5 x 10(8) plaque-forming units ALVAC-CEA B7.1 alone (n = 30) or with GM-CSF (n = 30) in patients with advanced CEA-expressing tumors to determine whether the addition of the adjuvant GM-CSF enhances induction of CEA-specific T-cells. Patients were vaccinated with vaccine intradermally every other week for 8 weeks. GM-CSF was given s.c. for 5 days beginning 2 days before vaccination. Patients with stable or responding disease after four immunizations received monthly boost injections alone or with GM-CSF. Biopsies of vaccine sites were obtained 48 h after vaccination to evaluate leukocytic infiltration and CEA expression. Induction of peripheral blood CEA-specific T-cell precursors was assessed in HLA-A2 positive patients by an ELISPOT assay looking for the production of IFN-gamma. Therapy was well tolerated. All of the patients had evidence of leukocytic infiltration and CEA expression in vaccine biopsy sites. In the patients receiving GM-CSF, leukocytic infiltrates were greater in cell number but were less likely to have a predominant lymphocytic infiltrate compared with patients receiving vaccine in the absence of the cytokine adjuvant. After four vaccinations, CEA-specific T-cell precursors were statistically increased in HLA-A2 positive patients who received vaccine alone. However, the GM-CSF plus vaccine cohort of HLA-A2 positive did not demonstrate a statistically significant increase in their CEA-specific T-cell precursor frequencies compared with baseline results. The number of prior chemotherapy regimens was negatively correlated with the generation of a T-cell response, whereas there was a positive correlation between the number of months from the last chemotherapy regimen and the T-cell response. ALVAC-CEA B7.1 is safe in patients with advanced, recurrent adenocarcinomas that express CEA, is associated with the induction of a CEA-specific T-cell response in patients treated with vaccine alone but not with vaccine and GM-CSF, and can lead to disease stabilization for up to 13 months.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Vaccines, Synthetic/therapeutic use , Adult , Aged , Biopsy , Cancer Vaccines/adverse effects , Chemotherapy, Adjuvant , Cohort Studies , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Immunity/drug effects , Male , Middle Aged , Neoplasms/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome , Vaccines, Synthetic/adverse effects
18.
Clin Cancer Res ; 6(6): 2219-28, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873071

ABSTRACT

Coordinated presentation of antigen and costimulatory molecules has been shown to result in the induction of an antigen-specific T-cell response rather than the development of anergy. This study evaluated the vaccine ALVAC-CEA B7.1, a canary pox virus that has been engineered to encode the gene for the tumor-associated antigen carcinoembryonic antigen (CEA) and B7.1, a T-cell costimulatory molecule. Patients with CEA-expressing tumors were immunized with 2.5 x 10(7) (n = 3), 1.0 x 10(8) (n = 6), and 4.5 x 10(8) (n = 30) plaque-forming units intradermally every other week for 8 weeks. Patients with stable or responding disease received monthly boost injections. Biopsies of vaccine sites were obtained 48 h after vaccination to evaluate leukocytic infiltration and CEA expression. Induction of CEA-specific T-cell precursors was assessed by an ELISPOT assay looking for the production of IFN-gamma. Therapy was well tolerated, without significant toxicity attributable to vaccine. All patients had evidence of leukocytic infiltration and CEA expression in vaccine biopsy sites. Six patients with elevated serum CEA values at baseline had declines in their levels lasting 4-12 weeks. These patients all had stable disease after four vaccinations. After four vaccinations, patients who were HLA-A-2-positive demonstrated increases in their CEA-specific T-cell precursor frequencies to a CEA-A2-binding peptide from baseline. The number of prior chemotherapy regimens was inversely correlated with the ability to generate a T-cell response. ALVAC-CEA B7.1 is safe in patients with advanced, recurrent adenocarcinomas that express CEA, and it is associated with the induction of a CEA-specific T-cell response.


Subject(s)
Adenocarcinoma/therapy , Avipoxvirus/genetics , B7-1 Antigen/genetics , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/genetics , Vaccines, Synthetic/therapeutic use , Adenocarcinoma/immunology , Adult , Aged , B7-1 Antigen/toxicity , Biopsy , Carcinoembryonic Antigen/blood , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/biosynthesis , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Neoplasms/immunology , Neoplasms/therapy , Pilot Projects , Regression Analysis , T-Lymphocytes/immunology , Treatment Outcome , Vaccines, Synthetic/toxicity
19.
Semin Oncol Nurs ; 16(4 Suppl 1): 2-12, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11151455

ABSTRACT

OBJECTIVES: To review the immune response and tumor immunology, and to provide an update on the success and obstacles to targeted therapy using monoclonal antibodies and antibody conjugates. DATA SOURCES: Research articles and textbooks. CONCLUSIONS: Ongoing studies are exploiting the targeting properties of the immune system to improve anticancer therapy. Both monoclonal antibodies and immunoconjugates have shown promise in treatment of specific diseases. IMPLICATIONS FOR NURSING PRACTICE: The rapid growth of molecular techniques has allowed for the development of new anticancer therapies. Since nurses are intimately involved in the delivery of such therapy as well as in educating patients regarding risks and benefits, they must be knowledgeable.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy , Neoplasms/nursing , Neoplasms/therapy , Humans , Neoplasm Metastasis , Neoplasms/pathology , Oncology Nursing
20.
BioDrugs ; 12(3): 209-36, 1999 Sep.
Article in English | MEDLINE | ID: mdl-18031177

ABSTRACT

Research advances and promising clinical outcomes with immunotherapeutics has led to a resurgence of incorporating monoclonal antibodies in cancer treatment. Unconjugated, conjugated and multi-target constructs are emerging as a conventional form of therapy along with the classical trio of surgery, radiation and chemotherapy. The recent major accomplishments in monoclonals include: first, the development of human and chimeric structures negating the induction of humoral responses to murine counterparts which limited use; second, protein engineering has improved the affinity and specificity of the antibody to its target; third, technics have been designed to select monoclonal antibodies imparting a biological consequence (function) following binding; and, lastly, recombinant proteins are being created with multiple epitopic specificities and/or fusion with other biologically active proteins such as toxins and cytokines/growth factors. Clinical efficacy in the treatment of haematological malignancies has secured a role for monoclonals in routine treatment. Evidence of clinical responses in patients with metastatic solid tumours is leading to the next generation of trials in the adjuvant setting. This paper presents an overview of the clinical experience with monoclonal antibodies in cancer treatment over the past 5 years. Our aim is to highlight the successes and advances, as well as noting limitations of antibody therapeutics. The advances seen support a continued effort to optimise the creation, selection and use of immunotherapeutics in the battle against cancer.

SELECTION OF CITATIONS
SEARCH DETAIL