Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Thorac Oncol ; 2(7): 638-44, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17607120

ABSTRACT

INTRODUCTION: We undertook this phase II study to measure postoperative drug delivery and toxicity of cisplatin plus docetaxel in patients with resected stage I-III non-small cell lung cancer. METHODS: The primary endpoint was amount of cisplatin delivered over a planned four cycles of adjuvant chemotherapy. Statistical design required a cohort to close if the regimen proved unlikely to improve cisplatin delivery compared with published phase III data. The first cohort was treated with docetaxel 35 mg/m2 intravenously (IV) on days 1, 8, and 15, and cisplatin 80 mg/m2 IV on day 15, every 4 weeks for four planned cycles. A second cohort was treated with docetaxel 75 mg/m2 IV plus cisplatin 80 mg/m2 IV on day 1 every 3 weeks for four planned cycles. RESULTS: Sixteen patients were treated with weekly docetaxel and cisplatin every 4 weeks, with five of 16 (31%) unable to complete three cycles. Subsequently, 11 patients were treated with docetaxel and cisplatin every 3 weeks, with six of 11 (55%) unable to complete three cycles. Among the 11 patients who failed to complete three cycles, the reasons for stopping included one or more of the following: fatigue (n = 8), nausea (n = 4), febrile neutropenia (n = 1), hypotension (n = 1), and nephrotoxicity (n = 1). CONCLUSIONS: The combination of cisplatin at 80 mg/m2 with docetaxel 35 mg/m2 weekly or 75 mg/m2 every 3 weeks is no better tolerated than older chemotherapy regimens. The most common reason to stop chemotherapy was intolerable fatigue. These results suggest that the most common dose-limiting toxicities are attributable to the cisplatin, given similar problems were encountered whether the docetaxel was delivered as a single dose every 3 weeks or as a lower weekly dose.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Lung Neoplasms/drug therapy , Taxoids/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/surgery , Chemotherapy, Adjuvant , Cisplatin/administration & dosage , Disease-Free Survival , Docetaxel , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Injections, Intravenous , Lung Neoplasms/mortality , Lung Neoplasms/surgery , Male , Maryland/epidemiology , Middle Aged , New York/epidemiology , Radiation-Sensitizing Agents , Retrospective Studies , Survival Rate , Taxoids/administration & dosage , Treatment Outcome
2.
Cancer ; 110(3): 599-605, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17577220

ABSTRACT

BACKGROUND: Preclinical studies have demonstrated that the inhibition of the PI3K/Akt/mTOR pathway restores gefitinib sensitivity in resistant cancer cell lines. A phase 1 study was conducted of the combination of everolimus, an mTOR inhibitor, and gefitinib to determine a daily dose of everolimus with gefitinib in patients with advanced nonsmall-cell lung cancer (NSCLC). METHODS: Oral everolimus and gefitinib were both administered daily to patients with progressive NSCLC. Patients were enrolled in 3-patient cohorts at everolimus dose levels of 5 and 10 mg daily. All patients received gefitinib 250 mg daily. RESULTS: Ten patients were enrolled. The maximum tolerated dose of everolimus was 5 mg when administered daily with gefitinib 250 mg. Two patients who were treated at the 10 mg dose level of everolimus experienced dose-limiting toxicity, including grade 5 hypotension and grade 3 stomatitis. Pharmacokinetic studies demonstrated no consistent, significant interaction on the t(max), C(max), and AUC(0-8h) of either agent. Two partial radiographic responses were identified among the 8 response-evaluable patients. CONCLUSIONS: For further study, everolimus at a dose of 5 mg daily in combination with daily gefitinib 250 mg is recommended. The 2 radiographic responses identified are encouraging. A phase 2 trial in patients with NSCLC is under way.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Dose-Response Relationship, Drug , Everolimus , Female , Gefitinib , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Quinazolines/administration & dosage , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Survival Rate , United States/epidemiology
3.
Clin Cancer Res ; 13(9): 2692-8, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473201

ABSTRACT

PURPOSE: Pralatrexate is a rationally designed antifolate with greater preclinical antitumor activity than methotrexate. Pralatrexate was synergistic with paclitaxel and with docetaxel in mouse xenograft experiments. This phase 1 study was designed to determine the maximum tolerated dose and toxicity of pralatrexate plus paclitaxel or docetaxel in patients with advanced cancer. EXPERIMENTAL DESIGN: Pralatrexate was administered i.v. every 2 weeks (days 1 and 15) in a 4-week cycle. Depending on the taxane used and dose being tested, the taxane was administered on days 1 and 15; days 2 and 16; or days 1, 8, and 15. In the latter part of the study, patients in the docetaxel arm were treated with vitamin B(12) and folic acid supplementation to mitigate toxicity and allow pralatrexate dose escalation. RESULTS: For the combination of pralatrexate plus paclitaxel without vitamin supplementation, dose-limiting stomatitis and peripheral neuropathy were encountered at the lowest dose levels tested. For pralatrexate plus docetaxel plus vitamin supplementation, pralatrexate 120 mg/m(2) plus docetaxel 35 mg/m(2) administered on the same day every other week was defined as the maximum tolerated dose and schedule, with dose-limiting toxicities at higher dose combinations including stomatitis and asthenia. Significant antitumor activity was observed for this combination in patients with non-small-cell lung cancer. CONCLUSIONS: Pralatrexate (120 mg/m(2)) plus docetaxel (35 mg/m(2)) plus vitamin supplementation is well tolerated with signs of efficacy against non-small-cell lung cancer that merit phase 2 testing.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Folic Acid Antagonists/adverse effects , Neoplasms/drug therapy , Paclitaxel/adverse effects , Taxoids/adverse effects , Adult , Aged , Aminopterin/administration & dosage , Aminopterin/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Dietary Supplements , Docetaxel , Female , Folic Acid Antagonists/administration & dosage , Homocysteine/blood , Humans , Male , Methylmalonic Acid/blood , Middle Aged , Paclitaxel/administration & dosage , Taxoids/administration & dosage , Vitamins/administration & dosage
4.
Cancer Chemother Pharmacol ; 59(4): 467-75, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16896930

ABSTRACT

PURPOSE: Based on our mouse xenograft model demonstrating that intermittent high-dose gefitinib sensitizes tumors to subsequent treatment with taxanes, we initiated this phase I trial to explore docetaxel in combination with escalating doses of intermittent gefitinib (Iressa) given prior to docetaxel. METHODS: This was a phase I study where patients with advanced cancer were treated with escalating doses of gefitinib (1,000, 1,500, 2,250, 3,000 mg) on days 1 and 2 followed by docetaxel (75 mg/m2) on day 3 of a 21 day cycle. Gefitinib pharmacokinetic data were obtained on days 1, 2, and 3 of cycles 1 and 2 at each dose level. RESULTS: 18 patients were enrolled in this study with the most frequent tumor types being non-small cell lung cancer and head and neck squamous cell cancer. The dose-limiting toxicity was neutropenia (n=1 at dose level 2, n=2 at dose level 4). Rash, diarrhea, and fatigue were the most common grade 1-2 toxicities. Pharmacokinetic data indicated no accumulation of gefitinib between cycles 1 and 2 and no clear correlation between gefitinib plasma levels and toxicity. Partial responses were observed in one patient with head and neck squamous cell carcinoma and one patient with anaplastic thyroid cancer. CONCLUSION: The recommended dose for phase II studies is gefitinib 2,250 mg on days 1 and 2, followed by docetaxel 75 mg/m2 on day 3.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Quinazolines/administration & dosage , Taxoids/administration & dosage , Adult , Aged , Docetaxel , Female , Gefitinib , Humans , Male , Middle Aged , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Taxoids/adverse effects , Taxoids/pharmacokinetics
5.
Cancer ; 104(10): 2149-55, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16208701

ABSTRACT

BACKGROUND: Trastuzumab is a monoclonal antibody directed against the human epidermal growth factor receptor-2 (HER-2). Nonsmall cell lung carcinoma (NSCLC) overexpresses HER-2 protein in approximately 20% of cases. In the current study, the authors combined trastuzumab with weekly taxanes in an attempt to improve outcomes over standard chemotherapy in patients with advanced NSCLC. METHODS: The primary objective was to determine whether docetaxel plus trastuzumab or paclitaxel plus trastuzumab was the superior regimen based on response and toxicity, and to determine whether either regimen was appropriate for further testing in a randomized Phase III trial. After stratification based on the results of HER-2 immunohistochemistry, chemotherapy-naive patients were randomized to receive trastuzumab plus docetaxel or trastuzumab plus paclitaxel. The study was designed so patients with or without HER-2 overexpression would be distributed equally between the study arms. RESULTS: Immunohistochemistry for HER-2 protein expression was attempted for 182 pathologic samples from 169 patients. Twenty-eight of the 179 evaluable samples (16%) revealed 2+ or 3+ staining. The objective response rate was 23% (7 of 30 patients) in the patients treated with docetaxel plus trastuzumab and 32% (11 of 34 patients) in the patients treated with paclitaxel plus trastuzumab (P=0.76, Wilcoxon test). No difference was noted in the median survival (16 mos vs. 14 mos) or 1-year survival (57% vs. 55%) (P=0.998). Toxicities were mild in both treatment arms. No difference with regard to response rates or survival was noted between HER-2-positive (2+ or 3+) and HER-2-negative (0-1+) patients. CONCLUSIONS: The expression of HER-2 protein in patients with advanced NSCLC in this study was found to be similar to that reported in previous series. The response rates and toxicities for patients treated with docetaxel and trasuzumab or paclitaxel and trasuzumab were not significantly different, though survival in both arms was better than expected. HER-2 expression status did not appear to affect outcomes for this uniform group of patients who were treated in a comparable fashion. Because of the infrequency of HER-2 overexpression, and the absence of improved outcomes in patients with NSCLC who were treated with trastuzumab plus chemotherapy in other studies, neither regimen tested will be advanced to a Phase III trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/mortality , Docetaxel , Female , Humans , Immunohistochemistry , Lung Neoplasms/mortality , Male , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Receptor, ErbB-2/biosynthesis , Survival Analysis , Taxoids/administration & dosage , Taxoids/adverse effects , Trastuzumab , Treatment Outcome
6.
Cancer ; 103(10): 2128-31, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15812822

ABSTRACT

BACKGROUND: Imatinib inhibits the c-kit tyrosine kinase, which, accounts for its activity in gastrointestinal stromal tumors. The presence of c-kit protein expression in small cell lung carcinoma (SCLC) tumor specimens, as well as in vitro data supporting the role of c-kit in autocrine and paracrine growth stimulation specifically in SCLC, provided a rationale for studying imatinib in this disease. The authors conducted a Phase II single-institution study of imatinib in patients with recurrent SCLC whose tumor specimens expressed c-kit protein. METHODS: Patients with progressive SCLC after one or two previous chemotherapy regimens consented to have their tumor specimens screened by immunoperoxidase stain (CD117, Dako Corporation, Carpinteria, CA) for c-kit protein expression. If present, individuals were then eligible for treatment with an imatinib dose of 400 mg orally twice daily (total, 800 mg per day). RESULTS: The presence of c-kit protein was assessable in 36 of 39 (92%) tumor samples. Twenty-eight (78%) tumor samples had immunohistochemical staining for c-kit protein. Twelve patients were enrolled in the treatment portion of the current study. No responses were observed, and all patients had disease progression by Week 4. Edema, fatigue, nausea, and electrolyte abnormalities were the primary toxicities. CONCLUSIONS: Imatinib did not have antitumor activity against SCLC, even with c-kit protein present in tumor specimens. The dismal prognosis for these patients with progressive SCLC emphasized the urgent need for continued studies of new therapies in this population.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Small Cell/metabolism , Lung Neoplasms/metabolism , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/analysis , Pyrimidines/therapeutic use , Antineoplastic Agents/adverse effects , Benzamides , Carcinoma, Small Cell/drug therapy , Disease Progression , Edema/chemically induced , Fatigue/chemically induced , Female , Gene Expression Regulation, Neoplastic , Humans , Imatinib Mesylate , Immunoenzyme Techniques , Lung Neoplasms/drug therapy , Male , Middle Aged , Nausea/chemically induced , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Piperazines/adverse effects , Prognosis , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/adverse effects , Treatment Failure
7.
J Clin Oncol ; 23(1): 165-74, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15557594

ABSTRACT

PURPOSE: The use of gefitinib, the first drug approved to inhibit the epidermal growth factor receptor tyrosine kinase, is indicated in patients with non-small-cell lung cancer with tumors progressive after chemotherapy. The unique mechanism of action of this agent leads to distinctive patterns of response and toxicity in persons with lung cancer. Many of the principles of management relevant to gefitinib are distinct from those with conventional cytotoxic drugs. To meet this need, we present practical guidelines on the use of gefitinib in patients with non-small-cell lung cancer. METHODS: This article reviews gefitinib's indications, dosing, response phenomena, and patterns of relapse in individuals with radiographic response. RESULTS: We present our recommendations for the management of rash and diarrhea caused by this agent. CONCLUSION: This information can guide practitioners and help them inform their patients about what to expect when they receive gefitinib.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Diarrhea/chemically induced , Diarrhea/drug therapy , Drug Interactions , Exanthema/chemically induced , Exanthema/drug therapy , Eye/drug effects , Female , Gefitinib , Humans , Lung Diseases, Interstitial/chemically induced , Lung Neoplasms/diagnostic imaging , Male , Patient Education as Topic , Quinazolines/administration & dosage , Quinazolines/pharmacology , Quinazolines/toxicity , Radiography
8.
Clin J Oncol Nurs ; 8(4): 385-92, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15354925

ABSTRACT

Treatment of solid tumors with chemotherapy regimens commonly is associated with debilitating or life-threatening side effects. Careful patient management, appropriate and prompt management of side effects, and interruption of therapy frequently are required for patients receiving chemotherapy. Furthermore, the systemic toxicity associated with chemotherapy may result in irreversible and incapacitating side effects, such as peripheral neuropathy, that lead to poor quality of life in patients. Gefitinib (Iressa, ZD1839, AstraZeneca Pharmaceuticals LP, Wilmington, DE) is a biologically based, molecular targeted therapy with a novel mechanism of action: selective inhibition of the epidermal growth factor receptortyrosine kinase (EGFR-TK) activity. Once-daily oral treatment with gefitinib is well tolerated. In clinical trials, treatment with gefitinib resulted in durable tumor responses and improvement in lung cancer-related symptoms in patients with advanced non-small cell lung cancer who had received prior chemotherapy. Trials are under way to explore the full potential of gefitinib and additional EGFR-TK inhibitors for other solid tumors and in other treatment settings, including prevention. Biologically based, molecular-targeted therapies such as gefitinib are providing new treatment options for patients and adding a new dimension to clinical practice for oncology nurses.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/nursing , Carcinoma, Non-Small-Cell Lung/physiopathology , Dose-Response Relationship, Drug , Epidermal Growth Factor/antagonists & inhibitors , Gefitinib , Humans , Lung Neoplasms/nursing , Lung Neoplasms/physiopathology , Patient Education as Topic , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/adverse effects , Quinazolines/pharmacology
9.
Clin Cancer Res ; 10(18 Pt 1): 6094-100, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15447995

ABSTRACT

PURPOSE: Immunotherapy directed toward cell surface antigens may provide a novel approach to the eradication of chemoresistant micrometastatic disease in patients with small-cell lung cancer (SCLC). Studies in SCLC cell lines and human tissues suggest that the ganglioside fucosyl GM1 is an abundant yet specific target. A prior clinical study demonstrated the potent immunogenicity of fucosyl GM-1 derived from bovine thyroid gland, conjugated to keyhole limpet hemocyanin (KLH) and administered with QS-21 adjuvant. EXPERIMENTAL DESIGN: We tested the immunogenicity of three different doses of a synthetic version of fucosyl-GM1 in patients with SCLC after a major response to initial therapy. The primary end point was to establish the lowest effective dose capable of inducing antibody production. RESULTS: Five of six patients at the 30-microg dose and three of five patients at the 10-microg dose mounted IgM responses of 1:80 or greater. These antibodies were confirmed by flow cytometry in seven of eight cases. None of the patients at the 3-microg dose had titers above 1:80. One patient at the 30-microg dose had an IgG response with a titer of 1:80. The sera from six of the eight responders induced potent complement-mediated cytotoxicity of tumor cells. CONCLUSIONS: Vaccination with the synthetic fucosyl GM1-KLH conjugate induces an IgM antibody response against fucosyl GM1 and tumor cells expressing fucosyl GM1, comparable with the response induced by the bovine derivative. We plan to combine synthetic fucosyl GM1 vaccine at a dose of 30 microg with vaccines against three other antigens-GM2, Globo H, and polysialic acid-to test in patients with SCLC after initial chemotherapy.


Subject(s)
Cancer Vaccines , Carcinoma, Small Cell/therapy , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/therapeutic use , Hemocyanins/therapeutic use , Immunotherapy/methods , Lung Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Neoplasm/chemistry , Carbohydrate Sequence , Cattle , Cell Separation , Chromatography, Thin Layer , Complement System Proteins , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immune Tolerance , Immunoglobulin G/chemistry , Male , Middle Aged , Models, Chemical , Molecular Sequence Data , Time Factors
10.
J Clin Oncol ; 22(6): 1103-9, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15020612

ABSTRACT

PURPOSE: Gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, induces radiographic regressions and symptomatic improvement in patients with non-small-cell lung cancer (NSCLC). Phase II trials suggested female sex and adenocarcinoma were associated with response. We undertook this analysis to identify additional clinical and pathologic features associated with sensitivity to gefitinib. PATIENTS AND METHODS: We reviewed medical records, pathologic material, and imaging studies of all 139 NSCLC patients treated on one of three consecutive studies of gefitinib monotherapy performed at our institution. We identified patients experiencing a major objective response and compared their clinical and pathologic features with the others. Univariate and multivariable analyses were performed on potential predictive features associated with sensitivity to gefitinib. RESULTS: Of 139 patients, 21 (15%; 95% CI, 9% to 21%), experienced a partial radiographic response. Variables identified as significant in univariate analysis included adenocarcinoma versus other NSCLC (19% v 0%; P=.004), adenocarcinoma with bronchioloalveolar features versus other adenocarcinomas (38% v 14%; P<.001), never smoker status versus former/current (36% v 8%; P<.001), and Karnofsky performance status > or =80% versus < or =70% (22% v 8%; P=.03). Multivariable analysis revealed the presence of adenocarcinoma with any bronchioloalveolar features (P=.004) and being a never smoker (P=.006) were independent predictors of response. CONCLUSION: Our data suggest that individuals in whom gefitinib is efficacious are more likely to have adenocarcinomas of the bronchioloalveolar subtype and to be never smokers. These observations may provide clues to mechanisms determining sensitivity to this agent and suggest that NSCLC has a different biology in patients who never smoked and those with bronchioloalveolar carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Bronchogenic/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , Smoking , Adult , Aged , Aged, 80 and over , Bronchoalveolar Lavage Fluid/cytology , Carcinoma, Bronchogenic/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Epidermal Growth Factor/antagonists & inhibitors , Female , Gefitinib , Humans , Logistic Models , Lung Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , Retrospective Studies
11.
Clin Cancer Res ; 10(3): 916-23, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871967

ABSTRACT

PURPOSE: Long chain polysialic acid (polySA) is a side chain on embryonal neural cell adhesion molecules that, in the adult, is largely restricted to small cell lung cancer (SCLC). Long chains of polySA are also expressed on group B meningococcus. In this clinical trial, we aimed to elicit an immune response against polysialic acid to target clinically inapparent residual disease in patients with SCLC who had successfully completed initial therapy. EXPERIMENTAL DESIGN: Patients were vaccinated with either 30 micro g unmodified polySA or N-propionylated-polySA (NP-polySA), conjugated to keyhole limpet hemocyanin (KLH) and mixed with 100 micro g of immunological adjuvant QS-21 at weeks 1, 2, 3, 4, 8, and 16. RESULTS: Of the 5 evaluable patients vaccinated with unmodified polySA, only 1 mounted an IgM antibody response to polySA. On the other hand, all 6 of the patients vaccinated with NP-polySA produced IgM antibodies to NP-polySA and these cross-reacted with unmodified polySA in all but 1 case. IgG antibodies to NP-polySA were observed in 5 of the patients, but these did not cross-react with polySA. The presence of IgM antibodies reactive with SCLC cell lines was confirmed in this group by flow cytometry. Complement-dependent lysis of tumor cells could not be demonstrated. However, postimmunization sera induced significant bactericidal activity against group B meningococcus when combined with rabbit complement. CONCLUSIONS: Vaccination with NP-polySA-KLH, but not polySA-KLH, resulted in a consistent high titer antibody response. We are now conducting a de-escalation dosing study with NP-polySA-KLH to better assess the immunogenicity, toxicities, and optimal dose of this vaccine. We plan to incorporate this vaccine as a component of a polyvalent vaccine with GM2, fucosylated GM1, and Globo H to target SCLC.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Small Cell/therapy , Hemocyanins/therapeutic use , Lung Neoplasms/therapy , Sialic Acids/therapeutic use , Vaccines, Conjugate/therapeutic use , Adjuvants, Immunologic/pharmacology , Aged , Antigens, Tumor-Associated, Carbohydrate/metabolism , Carbohydrate Sequence , Complement System Proteins , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/metabolism , Humans , Immunoglobulin G/chemistry , Immunoglobulin M/chemistry , Immunotherapy/methods , Male , Middle Aged , Models, Chemical , Molecular Sequence Data , Saponins/pharmacology , Time Factors
12.
Clin Cancer Res ; 9(6): 2072-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12796370

ABSTRACT

PURPOSE: 10-propargyl-10-deazaaminopterin (PDX) has superior antitumor efficacy in mouse xenograft models, likely attributable to increased uptake by the RFC-1 folate transporter and greater intracellular polyglutamylation. In a previous Phase I trial, stomatitis was the dose-limiting (and only clinically significant) toxicity of PDX. The recommended Phase II dose was 150 mg/m(2) i.v. every 2 weeks. Responses observed in patients with non-small cell lung cancer (NSCLC) in the Phase I trial prompted this Phase II trial. EXPERIMENTAL DESIGN: Patients had stage IIIB or IV NSCLC and either no previous chemotherapy or progression after initial response or stable disease to one previous chemotherapy regimen. Initially, PDX was administered at a dose of 150 mg/m(2) every 2 weeks. However, to decrease the frequency of stomatitis, the last 10 patients were treated at a dose of 135 mg/m(2). We planned to correlate PDX effects with folate and homocysteine levels and the expression of genes associated with folate transport and polyglutamylation. RESULTS: Thirty-nine patients were enrolled, 38 of whom were evaluable for response. Four patients had confirmed, major objective responses (10% based on intent to treat, 95% confidence interval 3-25) lasting 4, 9, 12, and 15 months. Twelve patients (31%) had stable disease. The median survival was 13.5 months. The predicted 1- and 2-year survival rates were 56 and 36%, respectively. Two patients (5%) suffered grade 4 stomatitis, and 6 (15%) had grade 3. No clinically significant myelosuppression occurred. No correlation between homocysteine or serum folate levels and severity of stomatitis was observed. Area under the curve (calculated using a limited sampling model) correlated with mucositis grade. A trend was noted between folate transporter expression and treatment effect. CONCLUSIONS: The broad applicability of this new antifolate with limited toxicity and proven efficacy in NSCLC encourage further development of this compound. Several trials are now underway combining PDX with other chemotherapeutic agents and testing its efficacy in other cancers.


Subject(s)
Aminopterin/analogs & derivatives , Aminopterin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Folic Acid Antagonists/therapeutic use , Lung Neoplasms/drug therapy , Membrane Transport Proteins , Adult , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carrier Proteins/genetics , Female , Homocysteine/blood , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Membrane Proteins/genetics , Middle Aged , RNA, Messenger/analysis
13.
J Clin Oncol ; 21(11): 2094-100, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12775734

ABSTRACT

PURPOSE: Gefitinib is an oral agent that inhibits the tyrosine kinase of the epidermal growth factor receptor. In phase I trials gefitinib was well tolerated and antitumor activity was seen in pretreated non-small-cell lung cancer (NSCLC) patients. Preclinical studies indicated enhanced effects when gefitnib was added to carboplatin or paclitaxel. This pilot trial combined gefitinib with carboplatin and paclitaxel to define the toxicities of the combination and assess drug-drug interactions in untreated advanced NSCLC patients. PATIENTS AND METHODS: Initially (part 1) patients were randomly assigned to receive intermittent gefitinib with cycle 1 or 2 of chemotherapy. Thereafter (part 2), the highest dose of gefitinib that was given without dose-limiting toxicity (DLT) from part 1 was administered continuously beginning with the first cycle of chemotherapy. Three sequentially enrolled cohorts received gefitinib 250 and 500 mg (intermittently) and 500 mg (continuously). RESULTS: We treated 24 patients; nine patients with 250 mg and 15 patients with 500 mg (nine patients continuous). Two occurrences of DLT were observed. One patient (500 mg, part 1) developed grade 3 rash and another patient (part 2) developed prolonged neutropenia. Steady-state gefitinib levels did not affect exposure to chemotherapy. In a limited sample, chemotherapy modestly increased the gefitinib area under concentration-time curve at steady-state and minimum steady-state trough concentration. Partial responses were observed in five of 24 patients. The median survival was 8 months. CONCLUSION: The gefitinib with carboplatin and paclitaxel regimen was generally well tolerated and no unanticipated toxicities or clinically relevant pharmacokinetic interactions were observed. Both doses of gefitinib were believed to be safe for further study with chemotherapy. This regimen was thus tested in a completed randomized phase III trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Area Under Curve , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Dose-Response Relationship, Drug , Epidermal Growth Factor/antagonists & inhibitors , Female , Gefitinib , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Paclitaxel/administration & dosage , Pilot Projects , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/administration & dosage , Quinazolines/adverse effects , Survival Rate , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...