Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Breast Cancer Res Treat ; 54(3): 225-33, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10445421

ABSTRACT

Anthracyclines are among the most active agents in metastatic breast cancer. Mitoxantrone demonstrated a different toxicity profile when compared to doxorubicin. We performed a phase I/II study of single-agent high-dose mitoxantrone therapy for advanced breast cancer. Nineteen patients who had a diagnosis of metastatic breast cancer received treatment at the M.D. Anderson Cancer Center between June 1986 and December 1987. The patients received escalating doses of mitoxantrone until a maximum tolerated dose (MTD), defined as grade 3 or 4 nonhematologic toxicity or infection, was obtained. The starting dose of 25 mg/m2, given by short intravenous infusion, was escalated by 25% in each five-patient cohort if each patient in the previous cohort tolerated the initial course and 2 or fewer patients reached the MTD. The median cumulative dose of mitoxantrone was 93 mg/m2 (range, 25-205) and the maximum single dose was 39 mg/m2. Myelosuppression was the dose limiting toxicity. The median duration of granulocyte count < or = 250/microl was 5-7 days. Four patients (22%) had infections that required hospitalization, 3 patients (17%) had cardiac toxicity. One patient (6%) achieved a complete response, and 3 (17%) had a partial response, with an overall response rate of 22.3%. No apparent dose-response relationship was observed in our study. The mitoxantrone dosage recommended for phase II studies is 25 mg/m2 every 3-4 weeks. We conclude that high-dose mitoxantrone therapy for metastatic breast cancer was relatively well tolerated but was not associated with a higher response rate than that of standard dose mitoxantrone.


Subject(s)
Breast Neoplasms/drug therapy , Mitoxantrone/administration & dosage , Mitoxantrone/adverse effects , Adult , Algorithms , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Humans , Middle Aged , Neoplasm Metastasis/drug therapy
2.
Blood ; 90(6): 2188-95, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9310469

ABSTRACT

IDEC-C2B8 is a chimeric monoclonal antibody (MoAb) directed against the B-cell-specific antigen CD20 expressed on non-Hodgkin's lymphomas (NHL). The MoAb mediates complement and antibody-dependent cell-mediated cytotoxicity and has direct antiproliferative effects against malignant B-cell lines in vitro. Phase I trials of single doses up to 500 mg/m2 and 4 weekly doses of 375 mg/m2 showed clinical responses with no dose-limiting toxicity. We conducted a phase II, multicenter study evaluating four weekly infusions of 375 mg/m2 IDEC-C2B8 in patients with relapsed low-grade or follicular NHL (Working Formulation groups A-D). Patients were monitored for adverse events, antibody pharmacokinetics, and clinical response. Thirty-seven patients with a median age of 58 years (range, 29 to 81 years) were treated. All patients had relapsed after chemotherapy (median of 2 prior regimens) and 54% had failed aggressive chemotherapy. Infusional side effects (grade 1-2) consisting of mild fever, chills, respiratory symptoms, and occasionally hypotension were observed mostly with the initial antibody infusion and were rare with subsequent doses. Peripheral blood B-cell depletion occurred rapidly, with recovery beginning 6 months posttreatment. There were no significant changes in mean IgG levels and infections were not increased over what would be expected in this population. Clinical remissions were observed in 17 patients (3 complete remissions and 14 partial remissions), yielding an intent to treat response rate of 46%. The onset of these tumor responses was as soon as 1 month posttreatment and reached a maximum by 4 months posttreatment. In the 17 responders, the median time to progression was 10.2 months (5 patients exceeding 20 months). Likelihood of tumor response was associated with a follicular histology, with the ability to sustain a high serum level of antibody after the first infusion, and with a longer duration of remission to prior chemotherapy. One patient developed a detectable but not quantifiable immune response to the antibody that had no clinical significance. IDEC-C2B8 in a dose of 375 mg/m2 weekly for 4 weeks has antitumor activity in patients with relapsed low-grade or follicular NHL. Results with this brief, outpatient treatment compare favorably with results with standard chemotherapy, and IDEC-C2B8 has a better safety profile. Further studies evaluating IDEC-C2B8 in other types of lymphoma either alone or combined with chemotherapy are warranted.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Lymphoma, Non-Hodgkin/therapy , Adult , Aged , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Murine-Derived , B-Lymphocytes/cytology , Female , Humans , Immunotherapy , Lymphocyte Depletion , Male , Middle Aged , Recombinant Fusion Proteins , Rituximab , Survival Analysis
3.
Cancer Invest ; 15(5): 403-10, 1997.
Article in English | MEDLINE | ID: mdl-9316621

ABSTRACT

Recombinant human interleukin-1 beta (rhIL-1 beta) was evaluated in a phase 1 clinical trial in which patients with metastatic or unresectable solid tumors received carboplatin and etoposide in cycle 1 and carboplatin, etoposide, and rhIL-1 beta in cycle 2. Recombinant hIL-1 beta was given intravenously for 5 days in one of three schedules: (1) immediately postchemotherapy, (2) delayed for 5 days after chemotherapy, or (3) concurrently with chemotherapy. Four dose levels of rhIL-1 beta were evaluated: 20, 50, 100, and 200 ng/kg. The doses of carboplatin and etoposide were not changed between cycle 1 and cycle 2 so that the effect of rhIL-1 beta on chemotherapy-induced hemato-toxicity was evaluated; 54 patients were entered on study and 42 patients received at least two cycles of therapy and were thus evaluable for rhIL-1 beta toxicity and for the effect of rhIL-1 beta on hematotoxicity of carboplatin and etoposide. The major toxicities of rhIL-1 beta were chills, rigors, headache, fatigue, and hypotension. The maximum tolerated dose of rhIL-1 beta was not determined since the toxicities at all dose levels were similar. However, only 3/8 patients at the 200 ng/kg level received all 5 IL-1 beta infusions. We compared the effect of rhIL-1 beta on hematotoxicity of carboplatin/etoposide by comparing peripheral blood count parameters between cycles 1 and 2: rhIL-1 beta given postchemotherapy significantly increased absolute neutrophil count (AND) nadirs and improved neutrophil recovery times regardless of rhIL-1 beta dose level. Platelet count parameters were also improved when rhIL-1 beta was given postchemotherapy although these changes did not reach statistical significance. Thus, IL-1 beta exhibited extensive hematological effects but the usefulness of this agent in clinical practice will be limited by extensive toxicity at all tested dose levels.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Etoposide/administration & dosage , Female , Humans , Interleukin-1/administration & dosage , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/blood , Neutropenia/chemically induced , Recombinant Proteins/administration & dosage , Thrombocytopenia/chemically induced
4.
Kidney Int ; 50(3): 1044-50, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8872982

ABSTRACT

Rates of secretion of granulocyte-macrophage colony stimulating factor (GM-CSF) were measured in 50 primary cell cultures derived from cancerous and normal human kidneys. Mean rates of GM-CSF secretion measured by TF-1 cell proliferation assay (N = 21) and by ELISA (N = 31) were 2.5 and 7.8 ng/10(6) cells/24 hr, respectively. There was no significant difference between the mean rates of GM-CSF secretion by cancerous and normal renal cells. GM-CSF was also secreted by primary renal cell cultures grown in serum-free medium and by renal cell lines. GM-CSF mRNA was detected by RT-PCR in cultured renal cells, but not in undissociated kidney tissue. Rates of GM-CSF secretion were reduced up to 99% under conditions where the cellular density or substratum more closely resembled the in vivo environment. Some cultured human renal carcinoma cells (RCC) secreted GM-CSF at levels that occasionally overlapped the levels produced by the GM-CSF gene-modified human RCC vaccine now in phase I trial. The data indicate that GM-CSF is not expressed in vivo, and that stable GM-CSF secretion is induced by the dissociation and culture of human renal cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Kidney/cytology , Adenocarcinoma , Carcinoma, Renal Cell , Cell Count , Cell Line/cytology , Cell Line/metabolism , Cell Line/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-1/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/physiology
5.
Am J Hematol ; 51(4): 289-95, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8602629

ABSTRACT

This is a double-blind randomized placebo-controlled trial to evaluate the efficacy and safety of granulocyte-macrophage colony-stimulating-factor (GM-CSF) after dose-intensive cyclophosphamide, etoposide, and cisplatin (DICEP). Fifty-six patients with lymphoma or breast carcinoma were randomized to receive GM-CSF 250 microg/m2 or placebo subcutaneously (SC) every 12 hr after each course of DICEP until recovery of absolute neutrophil count (ANC) of 1.5 x 10(9)/L. Each patient was to receive three courses of DICEP. There were 28 patients in each group. The median duration of ANC below 0.5 x 10(9)/L was 10 versus 12 days for Course 1 (P = 0.010), 10 versus 12 days for Course 2 (P = 0.248), and 16.5 versus 15 days for Course 3 (P = 0.126); platelet counts below 20 x 10(9)/L was 4 versus 4 days for Course 1 (P = 0.586), 8.5 versus 7 days for Course 2 (P = 0.013), and 23.5 versus 10.5 days for Course 3 (P = 0.104); hospitalization for patients readmitted with cytopenic fever were 4 versus 8 days for Course 1 (P = 0.035); 7 versus 6 days for Course 2 (P = 0.692); and 8 versus 12 days for Course 3 (P = 0.884) in the GM-CSF and placebo group, respectively. GM-CSF significantly shortens the duration of neutropenia and readmission only during the first course of DICEP. There was a delay in platelet recovery and an increase in transfusion requirement during subsequent courses in the GM-CSF group. The result cautions the routine use of lineage specific hematopoietic growth factors in supporting repeated cycles of dose-intensive chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunologic Factors/therapeutic use , Neutropenia/prevention & control , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bacterial Infections/etiology , Bacterial Infections/prevention & control , Blood Transfusion , Breast Neoplasms/drug therapy , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Double-Blind Method , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Gastrointestinal Diseases/chemically induced , Humans , Length of Stay , Leukocyte Count , Lymphoma, Non-Hodgkin/drug therapy , Male , Middle Aged , Neutropenia/chemically induced , Neutropenia/therapy , Platelet Count , Thrombocytopenia/chemically induced , Treatment Outcome
6.
Cancer ; 73(8): 2087-98, 1994 Apr 15.
Article in English | MEDLINE | ID: mdl-8156514

ABSTRACT

BACKGROUND: This study continued the development and psychometric testing of the Lung Cancer Symptom Scale (LCSS), a disease- and site-specific instrument primarily measuring the physical and functional dimensions of quality of life for individuals with lung cancer. The instrument contains two scales, one for patients and a counterpart for health professionals as observers. METHODS: Feasibility, reliability, construct validity, and criterion-related validity were evaluated with 207 patients with non-small cell lung cancer (NSCLC) from six cancer centers. Within an interview with an observer, patients completed part of a battery of instruments by self-report and were interviewed for the remaining measures. Observers also completed measures after the interview. RESULTS: Feasibility, reliability, and validity were well supported for this lung cancer population. Feasibility was demonstrated by patient and staff compliance in completion at all six cancer centers. Internal consistency was good, with coefficient alphas of 0.82 for the patient scale and 0.75 for the observer scale. Construct validity was supported by 1. contrasted groups approach: regression lines (with 95% confidence bands) were obtained between the Karnofsky performance scale (KPS) and each of the two LCSS scales; 2. as a refinement, relationship testing: significant correlations between the LCSS and KPS for each item (except hemoptysis for the patient scale); and 3. multitrait-multimethod approach: good reliability (alphas ranging from 0.75 to 0.93), good convergent validity for the two LCSS scales (r = 0.77), and a good discriminant validity pattern from the Brief Symptom Inventory (BSI). Criterion-related validity with relevant gold standard measures (American Thoracic Society Questionnaire [ATS] and McGill Pain questionnaire, KPS, Profiles of Mood States [POMS], and Sickness Impact Profile [SIP]) was supported with significant correlations (0.40-0.67 for the LCSS patient scale; 0.54-0.65 for the LCSS observer scale). CONCLUSIONS: These psychometric properties demonstrate that the LCSS patient and observer scales are feasible, reliable, and valid quality of life measures that are ready for research and clinical use with lung cancer populations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/psychology , Clinical Trials as Topic , Lung Neoplasms/drug therapy , Lung Neoplasms/psychology , Multicenter Studies as Topic , Psychometrics/methods , Quality of Life , Adult , Aged , Aged, 80 and over , Feasibility Studies , Female , Humans , Male , Middle Aged , Reproducibility of Results
7.
Cancer Invest ; 12(1): 1-11, 1994.
Article in English | MEDLINE | ID: mdl-8281458

ABSTRACT

Patients whose lymphoma is resistant to standard treatment regimens continue to do poorly, with only an occasional patient achieving long-term remission even with bone marrow transplantation. Twenty-three patients with primarily refractory (11), or refractory relapsed (12) non-Hodgkin's lymphoma were treated with repeated cycles of dose-intensive cyclophosphamide, etoposide, and cisplatin (DICEP) without bone marrow transplantation. Each cycle of DICEP consisted of cyclophosphamide (2500 mg/m2/day, days 1-2), etoposide (500 mg/m2/day, days 1-3), and cisplatin (50 mg/m2/day, days 1-3). Twelve patients (52%) have achieved a complete response and 6 (26%)r a partial response. Three of the complete responders remain continuously free of disease for 19, 29, and 32 months, and 3 more are disease-free at 58, 59, and 65 months after receiving further therapy. Three-year survival for all patients is 45%. Patients with a good initial performance status (Zubrod 0 or 1) had a 58% complete response rate and a 2-year survival rate of 53%. One of 19 patients with an initial performance status of 0 or 1 had treatment-related mortality. Repeated cycles of DICEP can produce long-term responses in patients with refractory non-Hodgkin's lymphoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, Non-Hodgkin/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Drug Resistance , Etoposide/administration & dosage , Female , Follow-Up Studies , Humans , Lymphoma, Non-Hodgkin/mortality , Male , Middle Aged , Survival Rate , Treatment Outcome
8.
Cancer Chemother Pharmacol ; 33(4): 340-6, 1994.
Article in English | MEDLINE | ID: mdl-8281628

ABSTRACT

Dose intensification has the potential to increase the response frequency of chemosensitive tumors to chemotherapy. G-CSF and GM-CSF offer the possibility of dose-intensifying chemotherapy without prohibitive myelosuppression. A phase I study was undertaken to identify the maximum tolerated dose (MTD) of carboplatin that could be administered with a fixed dose of doxorubicin, 60 mg/m2, administered every 28 days. Further escalation of the carboplatin dose was then attempted, with the concomitant addition of GM-CSF 10 mg/kg per day on days 1-21. We had 21 patients, 13 with prior therapy, who were eligible. In all, 60 courses of therapy were delivered, all with doxorubicin and with carboplatin doses of 250 mg/m2, 325 mg/m2 and 400 mg/m2. At carboplatin 400 mg/m2 and doxorubicin 60 mg/m2, thrombocytopenia was dose limiting. The addition of GM-CSF did not allow further escalation. Of the 6 patients treated with carboplatin 400 mg/m2, doxorubicin 60 mg/m2, and GM-CSF, grade 4 granulocytopenia and thrombocytopenia were seen in 4 and 5 patients, respectively. The severity of thrombocytopenia was related to the calculated carboplatin AUC and also to baseline platelet count and prior therapy. In addition, the interaction of GM-CSF and chemotherapy, especially carboplatin-based, may be more complex than originally anticipated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Marrow Diseases/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Diseases/chemically induced , Carboplatin/administration & dosage , Doxorubicin/administration & dosage , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Thrombocytopenia/prevention & control
9.
Ann Oncol ; 5(1): 43-7, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8172792

ABSTRACT

BACKGROUND: In an attempt to improve dose intensity and therapeutic effectiveness in breast cancer, GM-CSF was incorporated into a multi-drug every other week chemotherapy regimen that had been previously reported to have promising activity, but whose dose-limiting toxicity was neutropenia. PATIENTS AND METHODS: A Phase I-II study in patients with locally advanced or metastatic breast cancer was initiated using GM-CSF and a 5-drug chemotherapy regimen employing oral cyclophosphamide daily for 7 days and doxorubicin, vincristine, methotrexate, 5-fluorouracil, and leucovorin IV every 2 weeks for 10 courses. In the first 8 patients, GM-CSF in escalating doses (1-20 micrograms/kg s.c. per day) was given on days 8-13 of each 2 week cycle. In the last 12 patients, GM-CSF was given on days 3-14 of each cycle in an attempt to improve its effectiveness by prolonging treatment duration. RESULTS: The regimen was poorly tolerated. Only 10 patients completed all 10 courses of treatment, and most of those required dose delays and/or reductions. GM-CSF failed to reduce neutropenia when given by either schedule. Furthermore, thrombocytopenia was severe and progressive, especially with the more prolonged GM-CSF schedule in which the mean lowest nadir platelet count was 15,000/microliters. Anemia, fatigue, mucositis, and neutropenic fevers were also common, and dose escalations were not possible in any patient. Central venous catheter complications were also common. Complete or partial remissions were observed in 15 of 20 patients, but response durations were brief. CONCLUSIONS: GM-CSF in two different schedules failed to ameliorate myelosuppression when used in combination with this multiple drug, every other week regimen. Neutropenia and, especially, thrombocytopenia remained dose limiting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Neutropenia/chemically induced , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Hematologic Tests , Humans , Middle Aged
10.
Cancer ; 72(11 Suppl): 3381-6, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8242571

ABSTRACT

The introduction of hematopoietic cytokines into the clinic has been rapid with three currently approved by the Food and Drug Administration and perhaps a dozen more in clinical trials. Combinations of cytokines have been relatively unexplored in the clinics. Knowledge about the use of cytokines with chemotherapy has grown considerably in the past few years. The literature relating to use of cytokines in support of standard chemotherapy, marrow failure syndromes associated with malignant disease, and dose intensification not requiring progenitor cell replacement has been reviewed. This review does not address the use of cytokines to support bone marrow transplantation and collection of progenitor cells. Hematopoietic cytokines shorten the duration of cytopenia and decrease infectious complications when used to support standard chemotherapy regimens. However, several randomized trials have failed to show a benefit of such cytokine-supported treatment programs in terms of antitumor response, palliation, or survival. One exception is granulocyte-macrophage colony stimulating factor used with aggressive but standard chemotherapy for high-risk non-Hodgkin lymphoma. Cytokines decrease the infectious complications of myelodysplastic syndromes and may decrease transfusion requirements. Marked escalations in chemotherapy doses are possible with specific chemotherapy regimens. These increase complete remission rates although no benefit in survival or palliation has yet been proven. Cytokines have not been effective in allowing escalation of doses with certain chemotherapy agents and regimens. Cytokines decrease hematopoietic toxicity with standard or escalated chemotherapy doses. Randomized trials are now beginning to define the true patient benefit of this capability.


Subject(s)
Hematopoietic Cell Growth Factors/therapeutic use , Neoplasms/therapy , Contraindications , Drug Administration Schedule , Hematopoietic Cell Growth Factors/administration & dosage , Humans , Neoplasms/drug therapy
11.
Invest New Drugs ; 11(1): 61-5, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8349438

ABSTRACT

Eighty-six patients with advanced colorectal, gastric or pancreatic carcinoma and no prior exposure to chemotherapy were treated with brequinar sodium. Brequinar was administered at a median weekly dose of 1200 mg/m2 intravenously. The toxicity was moderate, with thirty patients (35%) experiencing grade 3 or 4 toxicity. Objective responses were observed in 1/32 evaluable colorectal and 2/29 evaluable gastric carcinoma patients. There were no objective responses in 17 evaluable pancreatic cancer patients. We conclude that, at this dose and schedule, brequinar does not have sufficient activity in these gastrointestinal malignancies to warrant further evaluation.


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Colorectal Neoplasms/drug therapy , Drugs, Investigational/therapeutic use , Pancreatic Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Biphenyl Compounds/adverse effects , Drugs, Investigational/adverse effects , Female , Humans , Male , Middle Aged
12.
Biotechnol Ther ; 4(3-4): 183-95, 1993.
Article in English | MEDLINE | ID: mdl-8292969

ABSTRACT

Substantial increases in dose-intensity of chemotherapy yield a severalfold increase in complete remission rates and durable responses in several types of malignant disease. Hematopoietic colony-stimulating factors decrease the duration of the resultant severe neutropenia but optimal dosing regimens of these cytokines have not yet been determined. This study was designed to explore the use of both yeast-derived and E. coli-derived GM-CSF given pre- and postchemotherapy with an intensive combination chemotherapy regimen. The chemotherapeutic regimen consisted of cyclophosphamide 5000 mg/m2, etoposide 1500 mg/m2, and cisplatin 150 mg/m2 (DICEP). Patients receiving either yeast-derived GM-CSF (6.5 days) or E. coli-derived GM-CSF (6.0 days) had a shorter duration of severe granulocytopenia with an absolute granulocyte count below 300/microL than patients receiving no GM-CSF (11.0 days, p = 0.0001). Administration of GM-CSF for 6 days immediately preceding DICEP did not further shorten the duration of cytopenia. E. coli-derived GM-CSF given at doses above 5 micrograms/kg was poorly tolerated and offered no hematologic advantage. Lower doses (3 micrograms/kg) of the E. coli product were better tolerated but still produced more toxicities than yeast-derived GM-CSF. The yeast-derived product produced no local skin reactions and decreased the incidence of nonhematologic and all grade 3 or 4 toxicities compared to the control group.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Neutropenia/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Escherichia coli , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Male , Middle Aged , Neoplasms/complications , Neutropenia/chemically induced , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Yeasts
13.
Semin Hematol ; 29(4 Suppl 3): 27-32, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1492231

ABSTRACT

GM-CSF decreases the hematopoietic toxicity of both nonablative and marrow ablative intensive chemotherapy regimens. Data are limited at this time, particularly in regard to optimal schedules and GM-CSF dosing regimens with particular chemotherapy programs. The importance of GM-CSF scheduling in relationship to chemotherapy is becoming more clear. The dose-intensive regimens supported by GM-CSF produce a high complete remission rate in several types of malignancy. Evidence that these remissions will be durable is still lacking. Future directions will include filling some of these voids in our knowledge, as well as exploring various cytokine combinations for improved hematopoietic recovery and other chemotherapeutic regimens for improved antitumor effect. The near complete cytoreduction produced by these dose-intensive regimens may also create a better setting for attempts at therapeutic immunomodulation. Yet another, but more difficult, task will be to identify those situations where progenitor cell replacement is beneficial and appropriate.


Subject(s)
Agranulocytosis/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Adult , Agranulocytosis/chemically induced , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cell Count/drug effects , Child , Clinical Trials as Topic , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Male , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombocytopenia/chemically induced , Thrombocytopenia/therapy
15.
J Clin Oncol ; 10(9): 1460-9, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1517789

ABSTRACT

PURPOSE: This trial evaluated the optimum dosing regimen for recombinant human granulocyte-macrophage colony-stimulating factor (rhu GM-CSF) to support a dose-intensive chemotherapy regimen given without progenitor cell replacement. PATIENTS AND METHODS: Fifty-one patients with refractory malignancy received cyclophosphamide 2,500 mg/m2 on days 1 and 2, etoposide 500 mg/m2 on days 1, 2, and 3, and cisplatin 50 mg/m2 on days 1, 2, and 3. Patients were hospitalized from cycle days 1 to 4 for chemotherapy and readmitted for cytopenic temperatures above 38.5 degrees C. Cycles were repeated every 35 days in patients who responded to a total of three cycles. GM-CSF was given at doses of 250 to 1,000 micrograms/m2 by continuous intravenous infusion (CIV) or subcutaneously starting on cycle days 3 to 6. Two nonrandomized control groups are used. RESULTS: The optimum regimen of GM-CSF for shortening the duration of leukopenia (WBC count less than 300/microL) was 500 micrograms/m2 given CIV. Duration of leukopenia was 5.9 days compared with 13.2 and 10.2 days in the controls (P less than .05). The optimum regimens for shortening duration of hospitalization, however, were 500 and 750 micrograms/m2/d given as divided (twice daily) subcutaneous injections. Durations of hospitalization were 9.6 and 9.8 days compared with 15.7 and 22.2 days in the controls (P less than .08). At the higher GM-CSF dose, only 36% of patients required readmission for cytopenic fever. Toxicities of GM-CSF at clinically useful doses were minimal. Twelve patients had complete response (24%) and 22 partial response (43%). CONCLUSIONS: This dose-intensive regimen can be given safely without progenitor replacement. rhu GM-CSF decreases the duration of severe leukopenia and decreases the need for hospitalization and antibiotic therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Leukopenia/drug therapy , Neoplasms/drug therapy , Adult , Aged , Blood Cell Count , Drug Administration Schedule , Drug Evaluation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Injections, Subcutaneous , Leukopenia/chemically induced , Male , Middle Aged , Pilot Projects , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Research Design , Treatment Outcome
16.
Cancer ; 70(4 Suppl): 913-20, 1992 Aug 15.
Article in English | MEDLINE | ID: mdl-1379115

ABSTRACT

BACKGROUND: Three hematopoietic colony-stimulating factors (CSF) are currently approved for clinical use and several more are being used in clinical trials. These factors have impressive quantitative and/or functional effects on their respective target cell lines, but their best clinical uses have not been defined. METHODS: The literature regarding the use of CSF in standard chemotherapy is reviewed data and from early Phase I-II studies the use of granulocyte-macrophage CSF (GM-CSF) in support of dose intensification without progenitor cell replacement are presented. Dose intensive cyclophosphamide (5000 mg/m2), etoposide (1500 mg/m2), and cisplatin (150 mg/m2) (DICEP) were administered during 3 days, followed by CSF until hematologic recovery was achieved. Patients had advanced types of cancer unlikely to respond to standard chemotherapy. RESULTS: CSF can shorten the duration of leukocyte nadirs when given with standard chemotherapy, but a benefit to patients in terms of improved survival or quality of life has not been shown. Dose intensification has increased partial and complete response rates in several tumor types. The higher complete remission rate may translate into improved survival; no convincing data have been published to date supporting that possibility. The duration of severe leukopenia (absolute neutrophil count, less than 100/microliters) after DICEP can be shortened from 8.5 days to 5-6 days with either granulocyte CSF (G-CSF) or GM-CSF. GM-CSF has been shown to decrease the duration of hospital stay from 18.7 to 9.6 days and to decrease the need for readmittance to the hospital for cytopenic fever. A high complete remission rate (35%) was seen in patients with breast cancer and durable complete remissions were achieved in patients with refractory non-Hodgkin lymphoma. CONCLUSIONS: CSF shortens the duration of postchemotherapy leukocyte nadirs and allows maintenance of dose when this is essential for a beneficial outcome. Substantial dose escalations are also possible with CSF support and can produce a high complete response rate that offers hope for improved survival in selected patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Bone Marrow Transplantation , Cytokines/therapeutic use , Dose-Response Relationship, Drug , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans
17.
J Immunother (1991) ; 11(3): 176-83, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1515422

ABSTRACT

Thirty-seven patients with advanced malignancies were treated sequentially with recombinant interferon-gamma (rIFN-gamma) and recombinant interleukin-2 (rIL-2) in an outpatient dose escalation clinical trial. rIFN-gamma (0.1 or 0.25 mg/m2/day) was administered by intramuscular injection, days 1-7 and rIL-2 (12, 18, or 24 x 10(6) IU/m2/day) was administered by a 15-min intravenous bolus, days 8-12. Common toxicities encountered included fever, chills, fatigue, neutropenia, and elevations of SGOT, bilirubin, or creatinine. Hypotension and cardiac and pulmonary toxicities were rare. With repeated cycles of therapy, nausea/vomiting and diarrhea associated with the administration of rIL-2 were seen in greater frequency. There were no treatment-related deaths, and no patient required intensive care unit admission for toxicity management. A complete response was observed in one of 11 patients with renal cancer and a partial response was observed in one of seven patients with malignant melanoma. Due to problems with drug supply, further dose escalation could not be continued, and maximum tolerated doses (MTD) were not determined by strict criteria. However, the combination of rIFN-gamma, 0.25 mg/m2/day, and rIL-2, 24 x 10(6) IU/m2/day, appeared to be beyond the MTD, as three of six patients at this dose level could not complete one cycle of therapy due to toxicity. It is unlikely that higher doses of either agent would be tolerated, and for further study using this schedule, we recommend the doses: rIFN-gamma, 0.1 mg/m2/day, and rIL-2, 24 x 10(6) IU/m2/day.


Subject(s)
Immunologic Factors/therapeutic use , Interferon-gamma/therapeutic use , Interleukin-2/therapeutic use , Neoplasms/therapy , Adult , Aged , Combined Modality Therapy , Drug Evaluation , Female , Humans , Immunologic Factors/adverse effects , Interferon-gamma/adverse effects , Interleukin-2/adverse effects , Male , Middle Aged , Recombinant Proteins
18.
J Clin Oncol ; 10(1): 134-42, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1727915

ABSTRACT

PURPOSE: This multicenter, double-blind, randomized trial was performed to determine the efficacy and safety of pamidronate disodium (APD) in comparison to etidronate disodium (EHDP) in the treatment of cancer-related hypercalcemia. PATIENTS AND METHODS: Sixty-five male and female adult patients with cancer and corrected calcium levels of greater than or equal to 12.0 mg/dL after 24 hours of hydration were randomized to receive either 60 mg APD given as a single 24-hour infusion or 7.5 mg/kg EHDP given as a 2-hour infusion daily for 3 days. RESULTS: APD normalized corrected calcium levels in 70% (21 of 30) of patients, whereas EHDP did so in 41% (14 of 34) of patients (P = .026). The mean corrected serum calcium level decreased from 14.6 to 10.5 mg/dL in the APD-treated group and from 13.8 to 11.6 mg/dL in the EHDP-treated group within the first week of treatment. There was no difference in response to APD in patients without versus those with bone metastases (78% v 67%). Both drugs were well tolerated. CONCLUSION: This study demonstrated that a single 60-mg infusion of APD is safe and more effective than EHDP given at the dose of 7.5 mg/kg for 3 days in the treatment of cancer-related hypercalcemia.


Subject(s)
Diphosphonates/therapeutic use , Etidronic Acid/therapeutic use , Hypercalcemia/drug therapy , Neoplasms/complications , Adult , Aged , Analysis of Variance , Calcium/blood , Diphosphonates/administration & dosage , Diphosphonates/adverse effects , Double-Blind Method , Etidronic Acid/administration & dosage , Etidronic Acid/adverse effects , Female , Humans , Hypercalcemia/etiology , Infusions, Intravenous , Male , Middle Aged , Neoplasms/blood , Pamidronate
19.
Gynecol Oncol ; 44(1): 66-70, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1730428

ABSTRACT

The use of intravenous melphalan at higher doses is limited by severe myelosuppression. It was postulated that GM-CSF would permit the use of higher dose melphalan with only moderate myelosuppression easily manageable in an outpatient setting. Therefore, a phase I study of intravenous melphalan utilizing GM-CSF (recombinant granulocyte-macrophage colony-stimulating factor) support was initiated. Intravenous melphalan at doses of 15-45 mg/m2 was administered every 28 days. GM-CSF was utilized at doses of 10-20 micrograms/kg/day subcutaneously Days 2-21 on a 28-day cycle. Twenty-five patients received 53 courses of therapy. The dose-limiting toxicities were severe or life-threatening granulocytopenia and thrombocytopenia. Utilizing 20 micrograms/kg/day GM-CSF, the maximum tolerated dose (MTD) of melphalan is 30 mg/m2 and, with 10 mg/kg/day GM-CSF, the maximum tolerated melphalan dose is only 20 mg/m2. One patient with ovarian cancer achieved a partial response. Because the reported MTD of intravenous melphalan without GM-CSF is 30 mg/m2, GM-CSF has not allowed sufficient escalation of the intravenous melphalan dose for routine outpatient use.


Subject(s)
Genital Neoplasms, Female/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/toxicity , Melphalan/toxicity , Adenocarcinoma/drug therapy , Adult , Agranulocytosis/chemically induced , Agranulocytosis/prevention & control , Anemia/chemically induced , Drug Evaluation , Endometrial Neoplasms/drug therapy , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Leiomyosarcoma/drug therapy , Melphalan/pharmacokinetics , Melphalan/therapeutic use , Ovarian Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Thrombocytopenia/chemically induced , Thrombocytopenia/prevention & control
20.
Breast Cancer Res Treat ; 20 Suppl: S15-23, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1687203

ABSTRACT

Substantial intensification of chemotherapy doses is a promising approach to the treatment of refractory malignancy currently receiving increasing attention. For the past 4 years we have used 3 repeated cycles of a combination of cyclophosphamide (5 g/m2), etoposide (1500 mg/m2), and cisplatin (150 mg/m2) without replacement of progenitor cells and with and without colony-stimulating factor support. The duration of threatening levels of granulocytopenia with this regimen averages 10.2 days, although an occasional patient has prolonged recovery (range, 5-20 days) and most patients require antibiotic therapy for cytopenic fever. We have not yet identified the optimal dose of GM-CSF, but 500 micrograms/m2 significantly shortens the duration of cytopenia (ANC less than 300/mm3) to 5.9 days with a resultant decrease in incidence and duration of cytopenic fever (from 10.8 to 1.7 days), use of antibiotics (from 10.8 to 7.6 days), and duration of hospitalization (from 22.2 to 16.3 days). Seventeen patients with metastatic breast cancer have received this regimen to date with a 35% complete response (CR) rate and a 53% partial response (PR) rate. Most of these patients were refractory to standard therapy. Four of six (67%) not refractory to standard therapy have achieved complete responses that are ongoing at 3.5 to 10.4 months. We conclude that dose-intensive therapy is an option that needs more careful exploration early in the treatment of advanced breast cancer and that GM-CSF decreases morbidity and risk of dose-intensive regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Evaluation , Female , Humans , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL